Biomech of Human Motion1
Biomech of Human Motion1
Orthopaedic
Biomechanics
Cheng-Kung Cheng
Savio L-Y. Woo
Editors
123
Frontiers in Orthopaedic Biomechanics
Cheng-Kung Cheng • Savio L-Y. Woo
Editors
Frontiers in Orthopaedic
Biomechanics
Editors
Cheng-Kung Cheng Savio L-Y. Woo
School of Biomedical Engineering Musculoskeletal Research Center,
Shanghai Jiao Tong University Department of Bioengineering
Shanghai University of Pittsburgh
China Pittsburgh, PA
USA
This Springer imprint is published by the registered company Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
Foreword
Kerong Dai
Ninth People’s Hospital
Shanghai, China
Shanghai Jiao Tong University School of Medicine
Shanghai, China
v
Contents
Biomechanics of Bone and Cartilage������������������������������������������������������ 1
Yi-Xian Qin, Minyi Hu, and Xiaofei Li
Biomechanics of Skeletal Muscle and Tendon�������������������������������������� 37
Yuan-Hung Chao and Jui-Sheng Sun
Biomechanics of Ligaments�������������������������������������������������������������������� 75
Jie Yao, Zizhan Lian, Bin Yang, and Yubo Fan
Hand and Wrist Biomechanics �������������������������������������������������������������� 89
Steven Regal, Steven Maschke, and Zong-Ming Li
Biomechanics of the Elbow �������������������������������������������������������������������� 105
Su-Ya Lee and Fong-Chin Su
Biomechanics of the Shoulder���������������������������������������������������������������� 131
Min Zhang and Chih-Hwa Chen
Biomechanics of Spine ���������������������������������������������������������������������������� 147
Lizhen Wang, Zhongjun Mo, Yuanjun Zhu, Enze Zhou,
and Yubo Fan
Biomechanics of the Hip�������������������������������������������������������������������������� 169
Bolun Liu, Jia Hua, and Cheng-Kung Cheng
Biomechanics of the Knee ���������������������������������������������������������������������� 189
Huizhi Wang, Bolun Liu, Xinzheng Qi, Savio L-Y. Woo,
and Cheng-Kung Cheng
Biomechanics of Foot and Ankle������������������������������������������������������������ 219
Duo Wai-Chi Wong, Ming Ni, Yan Wang, and Ming Zhang
vii
viii Contents
Biomechanics of Human Motion������������������������������������������������������������ 265
Rongshan Cheng, Zhongzheng Wang, Cong Wang, Fuping Li,
Yifei Yao, Yan Yu, and Tsung-Yuan Tsai
Biomechanics of the Fracture Fixation�������������������������������������������������� 301
Yingze Zhang, Hongde Wang, Tianrui Wang, Wei Chen,
and Yanbin Zhu
Biomechanical Principles in Designing Custom-Made
Hip Prosthesis������������������������������������������������������������������������������������������ 339
Jia Hua
Biomechanics of Orthopedic Rehabilitation������������������������������������������ 357
Ayman A. Mohamed, Yih-Kuen Jan, Ian M. Rice, Fang Pu,
and Cheng-Kung Cheng
Biomechanics of Osteo-Synthetics���������������������������������������������������������� 397
Chia-Ying James Lin, Heesuk Kang, and Scott J. Hollister
Introduction
ix
x Introduction
explain how the musculoskeletal system works, what causes musculoskeletal inju-
ries, and how various treatment methods can improve the outcome.
There are 15 chapters in this book. Following Introduction, Chaps. 1–10 detail
important biomechanical concepts relevant to surgical and clinical practice, includ-
ing cartilage, muscles and tendons, ligaments, and joints. Chapter 11 introduces the
biomechanics of human motion, and the following three chapters discuss various
orthopaedic injuries and treatment methods. The last chapter focuses on orthopae-
dic bio-synthetics. Each chapter is contributed by experts in the respective research
fields and presents advanced approaches to orthopaedic biomechanics. Key features
of each chapter are highlighted as follows. The introductory section outlines the
target and contents of the book. Chapter 1 discusses how mechanical biology and
biomechanical signal transduction attenuate musculoskeletal degeneration, as
knowledge of specific cellular responses is critical to understanding the underlying
mechanism which has the potential to regulate bone and cartilage adaptation.
Chapter 2 introduces the anatomy of muscles and tendons and demonstrates how
these tissues act to produce mechanical force. Chapter 3 presents mechanical meth-
ods for measuring forces in ligaments, in addition to detailing the mechanism of
ligament injury and treatment methods available. Chapter 4 provides a review of the
anatomy and biomechanical function of the hand and wrist. This chapter also dis-
cusses the pathomechanics of the hand and wrist after injury, i.e. arthritis of the
thumb, distal radius fracture, and malunion carpal tunnel syndrome. The functional
and musculoskeletal anatomy of the elbow is illustrated with pictures in Chap. 5.
This chapter also details the pathomechanics of the elbow, including elbow fracture
and arthritis. Chapter 6 contains a systematical review of the musculoskeletal and
functional anatomy of the shoulder, as well as a description of shoulder arthroplasty.
Chapter 7 emphasizes on the repair of the lamina and related ligaments, using exam-
ples to illustrate the design of spinal arthroplasty to restore the function of the spine
and improve the stability and longevity of implants. In Chap. 8, significant param-
eters relating to the function and kinematics of the hip are introduced, as well as an
analysis of reaction forces in the hip joint. Chapter 9 focuses on knee disorders,
namely knee osteoarthritis and osteotomy, and discusses the design of knee arthro-
plasties with the aim of restoring knee function. Chapter 10 includes a biomechani-
cal assessment of the foot and ankle based on computational models, in addition to
detailing methods of applying finite element analysis to the study of injuries, pros-
thetic design, surgical approaches, and orthotics. Chapter 11 discusses the structural
anatomy that contributes to stability and mobility of joints. In this chapter, common
pathologies and hip arthroplasties are introduced. Chapter 12 focuses on the intro-
duction of fracture fixation methods. Biomechanical principles of screw and plate
fixation for fracture healing are described comprehensively in this section. Chapter
13 introduces the concept of customized hip stems for individual patients based on
their unique bone geometry, deformities, and pathological conditions. This chapter
may be of interest to surgeons and researchers who regularly interact with such
custom implants but need additional information on the development methods.
Chapter 14 details methods for improving the effectiveness of orthopaedic rehabili-
tation. This section investigates the biomechanical principles of exercises as well as
Introduction xi
appropriate assistive devices (splints, walkers, crutches, and canes) for orthopaedic
rehabilitation. The final chapter, Chap. 15, focuses on the introduction of osteo-
synthetics to the field of biomedical engineering. Design parameters such as implant
microstructural and materials for osteo-synthetics are discussed, as well as a new
design strategy termed Topology Optimization. This final chapter presents novel
research that could readily be applied in clinical settings, such as materials that pos-
sess the solid-fluid bi-phasic properties of native bone.
We are truly grateful for the contributions made by all the authors of this book.
Each is a distinguished leader in their respective areas of orthopaedic biomechanics.
Their wealth of knowledge and experience makes this book a true representation of
“the Frontiers in Orthopaedic Biomechanics”. We thank each author for presenting
the materials in each chapter in a comprehensive manner. We are also indebted to
Springer Nature and the editorial team for their kind assistance with making this
publication possible.
1 Background
Bone mineral density (BMD) and muscle strength are highly biomechanically
related to each other [1, 2]. High physical activity level has been associated with
high bone mass and low fracture risks, and is therefore recommended to reduce
fractures at old age [3–7]. As a direct consequence of exposure to microgravity,
astronauts experience several physiological changes, which can have serious medi-
cal complications. Most immediate and significant are the musculoskeletal implica-
tions in bone and muscles [8–11]. Results of the joint Russian/US studies of the
effect of microgravity on bone tissue from 4.5 to 14.5 month-long missions have
demonstrated that bone mineral density (BMD, g/cm2) and mineral content (BMC,
g) are decayed in the whole body of the astronauts [12]. The greatest BMD losses
have been observed in the skeleton of the lower body, i.e., in pelvic bones
(−11.99 ± 1.22%) and in the femoral neck (−8.17 ± 1.24%), while there is no evi-
table decay found in the skull region. Overall changes in bone mass of the whole
skeleton of male cosmonauts during the period of about 6 months on a mission
made up −1.41 ± 0.41% and suggest the mean balance of calcium over flight equals
to −227 ± 62.8 mg/day. On average, the magnitude and rate of the loss are stagger-
ing; astronauts lose bone mineral in the lower appendicular skeleton at a rate
approaching 2% per month with muscle atrophy [5, 6, 13–15]. In simulated or
actual microgravity, human postural muscles undergo substantial atrophy: after
about 270 days, the muscle mass attains a constant value of about 70% of the initial
one. Most animal studies have reported preferential atrophy of slow-twitch fibers
whose mechanical properties change towards the fast type. After microgravity, the
maximal force of several muscle groups has showed a substantial decrease (6–25%
of pre-flight values) [8, 10, 16–19]. The mechanism that explains both muscle and
bone decays in the function disuse environment is still unclear. In recent years,
considerable attention has focused on identifying particular parameters and exer-
cise paradigms to ameliorate the deficits of muscle atrophy and bone density.
Perhaps microcirculation and interstitial fluid flow that link with exercise and mus-
cle contraction can identify the interrelationship between muscle and bone flow in
response to loading and disuse environment. The headward shift of body fluids and
the removal of gravitational loading from bone and muscles have led to progressive
changes in the musculoskeletal systems. The underlying factor producing these
changes may be primarily due to the fluid flow and circulations in both muscular
and bone tissues.
The ability of musculoskeletal tissues to respond to changes in its functional
milieu is one of the most intriguing aspects of such living tissue, and certainly con-
tributes to its success as a structure. The ability of bone and muscle to rapidly
accommodate changes in its functional environment ensures that sufficient skeletal
mass is appropriately placed to withstand the rigors of functional activity, an attri-
bute described as Wolff’s law [20, 21]. This adaptive capability of musculoskeletal
tissues suggests that biophysical stimuli may be able to provide a site-specific,
exogenous treatment for controlling both bone mass and morphology. The premise
1 Biomechanics of Bone and Cartilage 3
Similar to most of the biological tissues, bone has its unique structure and serves as
a functional unit [45–47] (Fig. 1.1). In general, the basic structure of mature long
bone includes both cortical (or compact) bone and trabecular (or cancellous) bone.
Within the mature cortical bone, the skeleton shows the lamellar structure. Such a
plate structure is centered with the Haversian canal, mostly aligned longitudinally
and connected with the Volkmann’s canals that is running horizontally in the cor-
tex. These two conduits are rich in vascular tissue; two-thirds of the vascular supply
is provided through the medullary cavity, and the other one-third is provided
through the periosteum. Bone cells that present in the bone lacunae are intercon-
nected by the canaliculi, tubules structure, and communicate with each other
through the gap junction. Each concentric structure is consisted of a capillary tube,
a concentric plate structure, lacuna, and canaliculi. Such a unique bone unit is
called osteon.
The Haversian canals, canaliculi, and lacunae occupy 13.3% of the volume of the
cortical bone [48, 49]. The rest solid portion is the matrix, which is occupied by
mineral material containing the hydroxyapatite crystal and collagen fiber. The solid
bone matrix of the bone contains pores of the order of 0.01–0.1 μm in diameter
[48–52]. The primary constituents of an osteon are collagen (organic), hydroxyapa-
tite (inorganic), and fluid. Fluid flows through the various microstructural spaces to
transfer metabolites to the osteocytes, ensuring that bone tissue remains viable. This
continuous perfusion allows the remodeling processes to continue in perpetuity. The
requisite nutrition of the cells and essential disposal of their waste products are car-
ried by this dynamic fluid stream. Because of the intricate microstructure of cortical
bone, transportation between cells would be very poor if it relied solely on fluid
diffusion. Indeed, a complex network of very thin (~1 μm) and quite long (up to
100 μm) canaliculi must employ a more active policy of nutrient allocation and
signal dispersion [48–52].
4 Y.-X. Qin et al.
Bone (femur)
Lamellae
Periosteum
Marrow Cavity
Endosteum
Fig. 1.1 Cortical bone and osteon structure. The general structure of cortical bone includes mar-
row cavity, periosteum, and endosteum. The microstructure includes Haversian Canals, Volkmann’s
canal, osteocyte, and lacuna and canaliculi system
The mechanical strength of biological tissue or a sample can be defined and quanti-
fied by its mechanical parameters, such as stress, strain, and modulus. These
mechanical parameters are defined as follows.
3.1 Stress
3.2 Strain
External loading on a structure can induce the change of dimension of the structure,
which is called deformation. The strain is defined as the structural deformation
against the original shape of the structure caused by an applied load. There are two
6 Y.-X. Qin et al.
types of strains, one of which is the normal strain, which causes a change in the
length of the specimen, and another is the shear strain, which causes a change in the
angular relationship within the structure. Strain has no unit quantity, but is usually
expressed in %, ε, and με.
3.3 Elastic Modulus
It is the relation between strain and stress, such as the ratio of stress to strain, as the
basic material constants that reflect the mechanical properties of a material. The
relationship between stress and strain can be defined as stress/strain = elastic modu-
lus, the value of which is the ratio of stress to strain. The unit of elastic modulus is
also Pa, MPa, or GPa. The larger the value of elastic modulus, the higher the strength
of the material (Fig. 1.2).
Bone intensity is referred to the ability of bone tissue to resist structural damage
after being loaded. It is a comprehensive index that integrates bone structure, bone
mass, and material properties.
The mechanical properties of the tissue can be quantified by various mechanical
loading and evaluations. The mechanical strength testing instruments can be used to
obtain the test results of the tissue strength, stiffness, hardness, and toughness. The
mechanical properties of bone are usually assessed by mechanical testing of the
130
Ultimate stress
120
110
100
90
80
Stress (MPa)
Yield point
70
60
50 E, slope of linear portion
40
30
20
10
0
0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5 6.0
Strain (%)
Fig. 1.3 Long bone under loading of 3-point bending, 4-point bending, and torsion
bone using a 3- or 4-point bending device (Fig. 1.3). The shear mechanical test of
bone can be achieved by torsional testing (Fig. 1.3). The choice of the test type is
determined by various technical and physiological factors. For example, in the study
of healing of long bone fractures, bending and torsion tests are a logical choice
because they can test the bending and torsional strength of the tissue. The torsional
test subjects each cross-section of the bone to the same torque, while the 4-point
bending test creates a uniform bending moment [53] for the entire epiphysis.
For the torsional test, an additional parameter, twisting to failure (fracture), can
be used as a measure of the callus ductility. Although it can only be measured once
for a given callus, it is possible to obtain multiple measurements on hardness and
stiffness before reaching failure. Multilevel testing methods have been reported to
test the loading model by performing a noninvasive load on bone. Within these
methods, bending strength or torsional and compressive stiffness can be quantified
in multiple planes.
High physical activity level has been associated with high bone mass and low frac-
ture risks, and is therefore recommended to reduce fractures [3, 4, 6]. The ability of
musculoskeletal tissues to respond to changes in its functional milieu is one of the
most intriguing aspects of such living tissue, and certainly contributes to its success
as a structure. Bone and muscle rapidly accommodate changes in its functional
environment to ensure that sufficient skeletal mass is appropriately placed to with-
stand the regions of functional activity, an attribute described as Wolff’s law [20,
21]. This adaptive capability of musculoskeletal tissues suggests that biophysical
stimuli may be able to provide a site-specific, exogenous treatment to control both
bone mass and morphology. The premise of mechanical influence on bone morphol-
ogy has become a basic tenet of bone physiology [24–26]. Based on the Muscle
Pump Theory, vascular arteries and veins within skeletal muscles are compressed
upon muscle contraction, and therefore increasing the arteriovenous pressure gradi-
ent and promoting capillary filtration [54–56].
8 Y.-X. Qin et al.
10000
Net bone ±s.e.
formation
m=3.5
Anabolic 1000
m=5
Microstrain
m=2.5
Low exercise
m=4.5
Med exercise
100 High freq loading m=1
Maintain Electrical field
Bone therapy
resorption
LIPUS
10
0.1 1 10 100 1000 10000 100000 1000000
Number of Daily Loading Cycles
Resorb
Fig. 1.4 Nonlinear interrelationship of cycle number and daily strain magnitude following a gov-
erning equation (Eq. 1.1). It appears that bone mass can be retained through a number of distinct
strategies: (1) bone is preserved with extremely low daily loading cycles and extremely high fre-
quency, i.e., 4 cycles per day of 2000 με, (2) bone mass is maintained with 100 cycles per day of
1000 με, or 10,000s of cycles of signals well below 100 με (each represented as a star), (3) bone is
responded to extremely high frequency and extremely low strain magnitude, such as electrical field
induced muscle contraction and low-intensity pulsed ultrasound (LIPUS). It is proposed that fall-
ing below this “daily strain energy” would stimulate bone loss, or any combination exceeding this
relationship would stimulate net bone gain [53]
1 Biomechanics of Bone and Cartilage 9
loading cycles and the peak bone strain magnitude generated in the body can be
generated using curve fitting analysis [53].
where S represents the peak bone strain, and C represents the total daily loading
cycles. Such higher loading cycles can be achieved using higher frequency daily
loading regimen.
It is well accepted that overloading will damage the bone and lead to failure, just
as too much light, noise, or pressure will overwhelm sight, hearing, and touch,
respectively. Although the skeleton’s primary responsibility is structural support in
nature, its overall responsibilities are broader than first presumed, and even include,
a critical role of the acoustic sensory organ in elephants [3]. Emphasizing this point,
bone’s adaptation to mechanical signals is nonlinear, such that it can be influenced
by a very few high-magnitude strain events, or by many thousands of low-magni-
tude strain events.
To adapt to the changing demands of mechanics, bone mass and bone morphol-
ogy can be regulated via bone remodeling at specific sites. This crucial process of
structural remodeling of the bone involves bone resorption and the subsequent bone
formation. However, difficulties to determine specific mechanical components will
hamper our understanding of bone remodeling related diseases, as well as limiting
our judgments on bone fractures and healing capacity. Therefore, continuous stud-
ies of the bone remodeling process, for example, to determine the mechanical model
of this remodeling process, can ultimately benefit the intervention on prevention
and treatment of musculoskeletal disorders.
becular BMD in both hip and femur regions was greater than 2% per month, while
there was only minimally decrease in cortical bone [72]. Similar results were
observed in animal studies. Burr et al. [73] showed an increase in the bone turnover
rate in cast-immobilized animals that received muscle stimulation for 17 days.
Lower extremity muscle volume was also altered by disuse. Exposure to a
6-month space mission resulted in a decrease in muscle volume of 10% in the quad-
riceps and 19% in the gastrocnemius and soleus [6, 74]. Computed tomography
measurements of the muscle cross-sectional area (CSA) indicated a decrease of
10% in the gastrocnemius and 10–15% in the quadriceps after short-term missions
[15, 75]. Similar results were concluded after SCI, where patients suffered signifi-
cant 21%, 28%, and 39% reductions in CSA at the quadriceps femoris, soleus, and
gastrocnemius muscles, respectively [76, 77]. In addition to the effects on whole
muscle volume, muscle fiber characteristics were also modified due to inactivity
[78–80]. There are two primary muscle fibers: slow (type I) fibers play an important
role in maintaining body posture while fast (type II) fibers are responsive during
physical activity. Under disuse conditions, all fiber types were decreased in size,
16% for type I and 23–36% for type II [79–81]. The atrophied soleus muscles also
underwent a shift from type I (−8% in fiber numbers) to type II fibers [79, 81, 82].
Clinical muscle stimulation has been examined extensively in SCI patients to
strengthen skeletal muscle and alleviate muscle atrophy with promising outcomes
[83–86]. A few physical training studies further investigated this electrical stimulation
technique to determine its effect on osteopenia. These studies showed mixed results
with respect to bone density data [83, 87–90]. Using dual-energy X-ray absorptiom-
etry (DXA), BeDell et al. found no change in BMD of the lumbar spine and femoral
neck regions after functional electrical stimulation-induced cycling exercise, while
Mohr et al. showed a 10% increase in BMD in the proximal tibia following 12 months
of similar training [91–93]. In a 24-week study of SCI patients in whom 25 Hz electri-
cal stimulation was applied to the quadriceps muscles daily, Belanger and colleagues
reported a 28% recovery of BMD in the distal femur and proximal tibia, along with
increased muscle strength [92]. A number of reported animal studies also indicated
that muscle stimulation can not only enhance muscle mass, but bone mineral density
as well [94, 95]. Both animal and human studies seem to strongly support that func-
tional disuse can result in significant bone loss and muscle atrophy.
A recent study has revealed that induced marrow fluid pressure and bone strain by
muscle stimulation were dependent on dynamic loading parameters and optimized
at certain loading frequencies [96]. Adult Sprague Dawley retired breeder rats with
a mean body weight of 387 ± 41 g (Taconic, NY) were used to measure the relation-
ships between ImP, bone strain, and induced muscle contraction. Rats were anesthe-
tized using standard isoflurane inhalation. A micro-cardiovascular pressure
transducer (Millar Instruments, SPR-524, Houston, TX) was inserted into the femo-
1 Biomechanics of Bone and Cartilage 11
ral marrow cavity, guided via a 16-gauge catheter. A single element strain gauge
(120 Ω, factor 2.06, Kenkyojo Co., Tokyo) was firmly attached onto the lateral
surface of the same femur at the mid-diaphyseal region, with minimal disruption to
the quadriceps, for measuring bone strains during the loading. The MS was applied
at various frequencies (1, 2.5, 5, 10, 15, 20, 30, 40, 50, 60, and 100 Hz) to induce
muscle stimulation.
Normal heartbeat generated approximately 5 mmHg of ImP in the femur at a
frequency of 5.37 ± 0.35 Hz. The ImP value (peak-peak) was increased significantly
by dynamic MS at 5, 10, 15, 20, 30, and 40 Hz (p < 0.05 for 5, 10, 30, and 40 Hz,
p < 0.01 for 15 and 20 Hz). The response trend of the ImP against frequency was
nonlinear; the ImP reached a maximum value of 45 ± 9.3 mmHg (peak-peak) at
20 Hz (Fig. 1.5), although there was no significant difference between 10, 20, and
a 60 b b
a
50 a
PRESSURE (mmHg)
a
40
a
30
20
10
0
0 10 20 30 40 50 60 70 80 90 100
FREQUENCY (Hz)
b
160 a
140
STRAIN (microstrain)
a
120
100 b
80
60
40 c
c c
20 c c
0
0 10 20 30 40 50 60 70 80 90 100
FREQUENCY (Hz)
Fig. 1.5 (a) ImP in rat femur increased significantly with electrical frequency at 5, 10, 15, 20, 30,
and 40 Hz. In the loading spectrum from 1 to 100 Hz, stimulation at 1 Hz generated an ImP of
18 mmHg. A maximum ImP of 45 mmHg was measured at 20 Hz, which was 2.5-folds higher than
1 Hz. ap < 0.05 vs. baseline ImP; bp < 0.01 vs. baseline ImP. (b) Bone surface strain measurement.
Dynamic muscle stimulation applied at various frequencies significantly increased bone strain. In
the loading spectrum from 1 to 100 Hz, stimulation at 1 Hz produced a strain of 62 με. Peak strain
of 128 με was recorded at 10 Hz stimulation. The strain magnitude was reduced by >75% of the
peak strain for stimulation frequencies greater than 30 Hz. ap < 0.01 vs. 1, 2.5, and 5 Hz; bp < 0.01
vs. 10 Hz; cp < 0.001 vs. stimulation 20 Hz and below
12 Y.-X. Qin et al.
30 Hz. In the range from 5 to 40 Hz, the ImP was shown approximately 60% of the
maximum ImP. The MS generated ImP in the marrow cavity with values of
17.4 ± 6.2, 24 ± 5.4, 37.5 ± 11.0, 26.3 ± 11.1, and 3.7 ± 1.5 mmHg at frequencies of
1, 5, 10, 40, and 100 Hz, respectively.
The response of matrix strain to the MS frequency also was nonlinear (Fig. 1.5).
The MS generated femoral matrix strains of 61.8 ± 6.2, 87.5 ± 5.1, 128.4 ± 19.2,
78.3 ± 6.8, 18.7 ± 1.3, and 10.1 ± 1.8 με at frequencies of 1, 5, 10, 20, 40, and 100 Hz,
respectively. While the ImP trend indicated that the peak ImP value was observed at
20 Hz, the maximum matrix strain was measured at 10 Hz. Bone strain induced by
MS at 10 Hz was significantly different (p < 0.01) comparing to all other stimulations
with the exception of 15 Hz. In addition, the strains generated by MS above 30 Hz
were significantly lower than those values loaded at and below 20 Hz (p < 0.005), in
which matrix strains, when loaded above 30 Hz, decreased by more than 75% of the
peak strain measured at 10 Hz. For frequencies from 40 to 100 Hz, MS induced
matrix strain were less than 20 με. These results suggest that MS with a relatively
high rate and small magnitude can trigger significant fluid pressure in the skeleton.
These findings were verified in an in vivo experiment under functional disuse condi-
tions [97]. Fifty-six 6-month-old female Sprague-Dawley retired breeder rats
(Taconic, NY) were used to investigate the effects of frequency-dependent dynamic
muscle stimulation (MS) on skeletal adaptation under disuse environment. Animals
were randomly assigned to seven groups with n = 8 per group: (1) baseline control,
(2) age-matched control, (3) HLS, (4) HLS + 1 Hz MS, (5) HLS + 20 Hz MS, (6)
HLS + 50 Hz MS, and (7) HLS + 100 Hz MS. Functional disuse was induced by an
HLS, setup modified from Morey-Holton and Globus [98, 99]. An approximately
30° head-down tilt was set to prevent contact of the animal’s hind limbs with the
cage bottom. The body weight of each animal was weighed 3 times per week
throughout the study.
Throughout the entire experimental period, the body weights were not signifi-
cantly different between groups at the beginning of the study, with an average of
320 ± 47 g. Age-matched control animals were able to maintain a steady body
weight throughout the study, with only a −0.15% difference between the start and
end date. Animals subjected to 4-week functional disuse lost a significant amount of
body mass. These weight reductions were similar in HLS and HLS + MS groups,
with −10% for HLS (p < 0.05), −8% for 1 Hz (p = 0.07), −9% for 20 Hz (p < 0.05),
−11% for 50 Hz (p < 0.01), and −8% for 100 Hz (p = 0.09).
Trabecular bone structure changes by MS stimulation seem to be sensitive to the
fluid pressure magnitude experienced by the tissue, where a larger response occurred
at the region near the marrow cavity, and attenuated at the region near the growth
1 Biomechanics of Bone and Cartilage 13
plate. For example, M1 is the distal metaphyseal region 1.5 mm above the growth
plate. The lack of weight-bearing activity for 4 weeks significantly reduced trabecu-
lar bone quantity and quality, demonstrated by a 70% decreases in BV/TV, an 86%
decrease in Conn.D, a 28% decrease in Tb.N, a 57% increases in SMI, and a 43%
increase in Tb.Sp compared with baseline (p < 0.001). Similar results were observed
when compared with age-matched control (p < 0.001); decreases in BV/TV (66%),
Conn.D (86%), and Tb.N (26%), as well as increases in SMI (39%) and Tb.Sp
(39%) were observed. Trabecular BV/TV in electrically stimulated animals, with
the exception of 1 Hz, was significantly greater than that of disused bone. Animals
with MS at 20 Hz and 50 Hz showed an increase in BV/TV by 143% (p < 0.05) and
147% (p < 0.01), respectively. Stimulation at 100 Hz showed an 86% increase in
BV/TV, but this change was not statistically different from the HLS group. The
other outcome measures of Conn.D, Tb.N, and Tb.Sp were also significantly affected
by MS at 20, 50, and 100 Hz frequencies. There were up to 600% and 38% increases
for Conn.D and Tb.N, and up to a 36% decrease for trabecular separation (20 Hz
p < 0.01, 50 Hz p < 0.001, and 100 Hz p < 0.05). SMI and Tb.Th were not affected
by the stimulus, regardless of its frequency. The animals subjected to 4 weeks of
1 Hz MS showed the same level of bone loss and structural deterioration as did the
HLS animals without MS, and showed significant differences compared to
stimulation at higher frequencies. M3, the distal metaphyseal portion directly above
the growth plate, is a region with the most abundant trabecular network with
0.3 ± 0.05 BV/TV and 4.72 ± 0.64 Tb.N (Fig. 1.6). Disuse induced a 38% bone loss,
75% decrease in Conn.D, 30% reduction in Tb.N, and 43% more spacing within this
region. Similar to the results reported for the M2 portion, 50 Hz MS resulted in the
greatest preventive effects against disuse osteopenia, with increased BV/TV (40%;
p < 0.05), Conn.D (305%; p < 0.001), and Tb.N (41%; p < 0.001), and reduced
Tb.Sp (31%; p < 0.001). While BV/TV was not significantly altered by MS at 20 Hz
(+26%) and 100 Hz (+20%), trabecular qualities, Conn.D, Tb.N, and Tb.Sp, were
improved (up to 226%, 28%, and 24%, respectively, p < 0.001). Like the other
metaphyseal regions, SMI and Tb.Th were not affected by the stimulation. With the
exception of 1 Hz, stimulation frequencies at 20, 50, and 100 Hz had greater effects
on the trabecular bone 2.25 cm away from the growth plate, closer to the diaphysis.
Also, BV/TV inhibition at M1 was significantly higher (p < 0.05) than that of M3
with 20 Hz MS. Although following a trend similar to that of the above indices, at
50 and 20 Hz MS, the percent changes of the μCT measurements were not statisti-
cally significant between the three metaphyseal regions.
While trabecular bone responded to MS showing structural property changes, the
epiphyseal trabecular bone was not significantly affected by the 4-week HLS. The
percentage changes were minor versus the metaphyseal regions, with −5% BV/TV,
−44% Conn.D, −7% Tb.N, and +4% Tb.Sp. In this region, MS did not induce any
measurable effect on the bone volume and trabecular integrity at any stimulation
frequency. All stimulated values were comparable to age-matched and HLS ani-
mals, with up to 10% greater in BV/TV, 8% greater in Tb.N, and a 9% reduction in
Tb.Sp. These changes were not statistically significant.
14 Y.-X. Qin et al.
0.25 80
70
***
0.2 * ** **
60
Conn.D (1/mm^3)
**
50
0.15
BV/TV
40
#+ #+
0.1 30 #+ #+
20
0.05 10
0
0 Baseline Age- HLS 1Hz 20Hz 50Hz 100Hz
Baseline Age- HLS 1Hz 20Hz 50Hz 100Hz matched **
matched
0.5 #+ #+
4.5
0.45
4 ** *** ** *** *
0.4
#+ #+
Tb.Sp (mm)
3.5 0.35 ***
3 0.3
Tb.N
2.5 0.25
2 0.2
1.5 0.15
1 0.1
0.5 0.05
0 0
Baseline Age- HLS 1Hz 20Hz 50Hz 100Hz Baseline Age- HLS 1Hz 20Hz 50Hz 100Hz
matched matched
Fig. 1.6 MS induced ImP maintained bone mass at higher loading frequency, indicated by μCT
images of trabecular bone in distal femur. Graphs show mean ± SD values for bone volume frac-
tion (BV/TV, %), connectivity density (Conn.D, 1/mm3), trabecular number (Tb.N, 1/mm), and
separation (Tb.Sp, mm). MS at 50 Hz produced a significant change in all indices, compared to
HLS. #p < 0.001 vs. baseline; +p < 0.001 vs. age-matched; ∗p < 0.05 vs. HLS and 1 Hz MS;
∗∗
p < 0.01 vs. HLS and 1 Hz MS; ∗∗∗p < 0.001 vs. HLS and 1 Hz MS
The μCT data were correlated with the histomorphometry analyses. In the
metaphyseal trabecular bone, BV/TV measured by the 2-D histomorphometric
method was 43% lower in the HLS group than in age-matched controls (p < 0.001).
Animals subjected to MS also experienced 22–29% of bone loss (p < 0.01). The
result was correlated with the BV/TV values from the μCT analysis, giving an R2
value of 0.84 (p < 0.05). In other bone formation indices, HLS animals also showed
significant decline in MS/BS (76%, p < 0.001), MAR (80%, p < 0.001), and BFR/
BS (92%, p < 0.001). Disuse had an insignificant effect on the trabecular BV/TV
(−10%) at the epiphyseal region, similar to the results of the μCT analysis. Bone
formation indices were reduced due to HLS (52% for MS/BS, 147% for MAR, and
59% for BFR/BS), and daily MS failed to prevent such reduction of bone formation
activity.
These data imply that MS, applied at a high frequency with low magnitude and
for a short duration, is able to mitigate bone loss induced by the functional disuse.
There was, however, no evidence to suggest that such loading would enhance over-
all new bone formation, e.g., the total bone mass was less than age-match animals.
However, further studies related to cellular activities, e.g., osteoclast and osteo-
blast, and linked either bone resorption and bone formation, may be necessary to
further explore the balance of resorption and formation in such functional dis-
use model.
1 Biomechanics of Bone and Cartilage 15
Bone remodeling involves all related cell types, i.e., osteoblast, osteoclast, osteo-
cyte, T-cells, B-cells, megakaryocyte, and lining cells. Thus, all these cells are
potentially mechano-sensitive and even interrelated. These cells respond to mechan-
ical loading and can express specific molecular pathways. This section will discuss
several potential pathways involved in mechanical stimulation induced adaptation.
To explore the interrelation among overall bone cells, a cluster of bone forming and
bone resorption cells among dynamic and temporal adaptation structures are pro-
posed, known as “basic multicellular units” (BMUs) [100]. Bone adaptation occurs
constantly and each cycle may take over several weeks. Such processes are performed
with a combination of resorption and formation. Each phase can involve targeted
molecular and gene activations. An active BMU consists of a leading front of bone-
resorbing osteoclasts. Reversal cells, of unclear phenotype, follow the osteoclasts,
covering the newly exposed bone surface, and prepare it for deposition of replace-
ment bone, following deposition of an unmineralized bone matrix known as osteoid.
Related molecular factors are represented in this temporal sequence (Fig. 1.7).
In response to mechanical loading, the first stage of remodeling reflects the
detection of initiating triggering signals such as fluid flow and any other of physical
stimulation, e.g., pressure, electrical, and acoustic waves. Prior to activation, the
resting bone surface is covered with bone-lining cells, including preosteoblasts. B
cells are present in the bone marrow and secrete osteoprotegerin (OPG), which sup-
presses osteoclastogenesis.
During the Activation phase, the endocrine bone-remodeling signal parathyroid
hormone (PTH) binds to the PTH receptor on preosteoblasts. Damage to the miner-
alized bone matrix results in localized osteocyte apoptosis, reducing the local
transforming growth factor β (TGF-β) concentration and its inhibition of
osteoclastogenesis.
In the resorption phase, in response to PTH signaling, preosteoclasts are recruited
to the bone surface. Additionally, osteoblast expression of OPG is decreased, and
the production of CSF-1 and RANKL is increased to promote the proliferation of
osteoclast precursors and differentiation of mature osteoclasts. Mature osteoclasts
anchor to RGD-binding sites, creating a localized microenvironment (sealed zone)
that facilitates degradation of the mineralized bone matrix.
In the Reversal phase, reversal cells engulf and remove demineralized undigested
collagen from the bone surface. Transition signals are generated that halt bone
resorption and stimulate the bone formation process.
During the Formation phase, formation signals and molecules arise from the
degraded bone matrix, mature osteoclasts, and potentially reversal cells. PTH and
16 Y.-X. Qin et al.
Hematopoietic
stem cells
Mesenchymal
stem cells
RANKL Monocyte
Csf-1
Preosteoclast
OPG
PTH Preosteoblast
Osteoclast
Macrophages Osteoblasts
Bone-lining cells
sclerostin Osteoid
New bone
Old bone
Osteocytes
Osteocytes
Fig. 1.7 Bone remodeling cycle and its associated molecular pathways. Three primary cell types
are involved in bone modeling and remodeling, including osteoblast, osteoclast, and osteocyte. The
remodeling cycle of bone is composed of sequential phases of resorption and formation, including
the activation of precursor cells, osteoclasts activation generating bone resorption, bone formation
by osteoblasts after mineral removal and reversal, and mineralization. The osteoblasts that are
buried within the newly formed mineral matrix yield to osteocytes. Other osteoblasts that rest on
the bone surface become bone lining cells [47]
The data from disuse osteopenia and clinical osteoporosis have shown significant
reduction of bone density and structural integrity, culminating in an elevated risk of
skeletal fracture. Concurrently, a marked reduction in the available bone-marrow-
1 Biomechanics of Bone and Cartilage 17
derived population of mesenchymal stem cells (MSCs) [101] jeopardizes the regenera-
tive potential that is critical to recovery from bone loss, musculoskeletal injury, and
diseases. A potential way to combat the deterioration involves harnessing the sensitivity
of bone to mechanical signals, which is crucial in defining, maintaining, and recovering
bone mass. As discussed above, bone cells, i.e., osteoblast, osteoclast, and osteocyte,
may sense external mechanical loading directly and perform the balance of formation
and resorption in the remodeling process; specific mechanotransductive signals may
also bias MSC differentiation towards osteoblastogenesis and away from adipogenesis.
Mechanical targeting of the bone marrow stem-cell pool might, therefore, represent a
novel, drug-free means of slowing the age-related decline of the musculoskeletal system.
Considering the importance of exercise in stemming both osteoporosis and obe-
sity, combined with the fact that MSCs are progenitors of both osteoblasts and adi-
pocytes (fat cells), as well as the anabolic response of the skeletal system to
mechanical loadings, it was hypothesized that mechanical signals anabolic to bone
would invariably cause a parallel decrease in fat production. In an in vivo setting,
7-week-old C57BL/6J mice on a normal chow diet were randomized to undergo low
magnitude high-frequency loading (90 Hz at 0.2 g for 15 min/day) or placebo treat-
ment [102]. At 15 weeks, with no differences in food consumption between groups,
in vivo CT scans showed that the abdominal fat volume of mice subjected to loading
was 27% lower than that of controls (p < 0.01) [103, 104]. Wet weights of visceral
and subcutaneous fat deposits in loading mice were correspondingly lower.
Confirmed by fluorescent labeling and flow cytometry studies [103, 104], these data
indicated that mechanical signals influence not only the resident bone cell (osteo-
blast/osteocyte) population, but also their progenitors, biasing MSC differentiation
towards bone (osteoblastogenesis) and away from fat (adipogenesis). In a follow-up
test of this hypothesis, mice fed on a high-fat diet were subjected to low magnitude
loading or placebo treatment [103, 104]. Suppression of adiposity by the mechani-
cal signals was accompanied by a “mechanistic response” at the molecular level
showing that loading significantly influenced MSC commitment to either an osteo-
genic (Runx2, a transcription factor central to osteoblastogenesis) or adipogenic
(peroxisome proliferator-activated receptor [PPAR]γ, a transcription factor central
to adipogenesis) fate. Runx2 expression was greater and PPARγ expression was
decreased in mice that underwent LMMs compared with controls. The PPARγ tran-
scription factor, when absent or present as a single copy, facilitates osteogenesis at
least partly through enhanced canonical Wnt signaling [105, 106], a pathway criti-
cally important to MSC entry into the osteogenic lineage and expansion of the
osteoprogenitor pool. Notably, low magnitude mechanical loading treatment also
resulted in a 46% increase in the size of the MSC pool (p < 0.05) [103, 104]. These
experiments, although not obviating a role for the osteoblast/osteocyte syncytium,
provide evidence that bone marrow stem cells are capable of sensing exogenous
mechanical signals and responding with an alteration in cell fate that ultimately
influences both the bone and fat phenotype. Importantly, the inverse correlation of
bone and fat phenotype has increasing support in the clinical literature. Although
controversial, and despite the presumption that conditions such as obesity will
inherently protect the skeleton owing to increased loading events, data in humans
evaluating bone–fat interactions indicate that an ever-increasing adipose burden
comes at the cost of bone structure and increased risk of fracture [107].
18 Y.-X. Qin et al.
Osteocytes, cells embedded within the mineralized matrix of bone, are becoming the
target of intensive investigation [100, 108–110]. Osteoblasts are defined as cells that
make bone matrix, and are thought to translate mechanical loading into biochemical
signals that affect bone modeling and remodeling. The interrelationship between
osteoblasts and osteocytes would be expected to have the same lineage, yet these cells
also have distinct differences, particularly in their responses to mechanical loading
and utilization of the various biochemical pathways to accomplish their respective
functions. Among many factors, Wnt/β-catenin signaling pathway may be recognized
as an important regulator of bone mass and bone cell functions [100, 110]. While
osteocytes are embedded within the mineral matrix, Wnt/β-catenin signaling pathway
may serve as a transmitter to transfer mechanical signals sensed by osteocytes to the
surface of bone. Further, new data suggest that the Wnt/β-catenin pathway in osteo-
cytes may be triggered by crosstalk with the prostaglandin pathway in response to
loading which then leads to a decrease in expression of negative regulators of the
pathway such as Sclerostin (Sost) and Dickkopf-related protein 1 (Dkk-1) [66, 110].
Figure 1.8 indicates the potential pathway in response to mechanical loading.
It has been shown that the Wnt pathway is closely involved in bone cell differen-
tiation, proliferation, and apoptosis [110, 111]. Regulation of the Wnt/β-catenin sig-
naling pathway is vested largely in proteins that either act as competitive binders of
Wnts, notably the secreted frizzled-related proteins (sFRP) family, or act at the level
of low-density lipoprotein receptor-related protein 5 (LRP5), including the
osteocyte-specific protein, sclerostin (the Sost gene product), and the Dkk proteins,
particularly Dkk-1 and Dkk-2 [110–114]. Sclerostin has been shown to be made by
mature osteocytes and inhibits Wnt/β-catenin signaling by binding to LRP5 and
preventing the binding of Wnt. Dkk-1 is highly expressed in osteocytes [112–114].
Clinical trial studies using antibodies to sclerostin have also shown increased bone
mass, suggesting that targeting of these negative regulators of Wnt/β-catenin signal-
ing pathway might be anabolic treatments for diseases such as osteoporosis [112].
Finally, mechanical loading has been shown to reduce sclerostin levels in bone
[112], suggesting that one of the targets of the pathways, activated by the early
events after mechanical loading, is the genes encoding these negative modulators of
the Wnt/β-catenin signaling pathway.
There is still much to be learned regarding how the bone cells, i.e., osteocyte,
sense and transmit signals in response to or absence of loading and further elevate
the activity of other cells. Although fluid shear stress is proposed as a triggering
force, the identity of these particular mechanical signals is still a challenging area to
study. The osteocyte is joining the osteoblast and osteoclast as targets for therapeu-
tics to treat or prevent bone disease. Clearly targeting the Wnt/β-catenin pathway in
osteocytes because of its central role in bone mass regulation and bone formation in
response to mechanical loading may prove useful for designing new paradigms and
pharmaceuticals to treat bone disease in the future.
1 Biomechanics of Bone and Cartilage 19
10
Lrp5
3
EP2/4
4 Wnt
11 Fz
Inte Akt
grin
s 5
ILK
GSK-3β
β-Cat
Cs43 HC
2 ERα β-Cat
6
9
7
PGE2
Sost↓
β-Cat
Dkk↓
1 Tcf
8
Wnt↑
ε
Fig. 1.8 Mechanical loading, e.g., fluid shear stress, induced Wnt signaling activation [110]. The
mechanical load applied to bone (ε) is perceived by the osteocyte through an unknown mechanism,
although fluid flow induced through the lacunar-canalicular system may be a critical component of
this perception, “step 1.” Perception of load (strain) triggers a number of intracellular responses
including the release of PGE2, “2” through a poorly understood mechanism into the lacunar-
canalicular fluid where it can act in an autocrine and/or paracrine fashion. Connexin-43 hemichan-
nels (CX43 HC) in this PGE2 and integrin proteins appear to be involved. Binding of PGE2 to its
EP2 and/or EP4 receptor, “3,” leads to a downstream inhibition of GSK-3β, “5” (likely mediated
by Akt, “4”) and the intracellular accumulation of free β-catenin, “6.” (Integrin activation can also
lead to Akt activation and GSK-3β inhibition.) New evidence suggests that ER may participate in
the nuclear translocation of β-catenin, “7” which leads to changes in the expression of a number of
key target genes “8.” One of the apparent consequences is the reduction in sclerostin and Dkk1, “9”
with increased expression of Wnt, “10” (which one or ones is unknown). The net result of these
changes is to create a permissive environment for the binding of Wnt to Lrp5-Fz and amplification
of the load signal, “11.” (With permission of Bone)
LRP5 has been shown to have important functions in the mammalian skeleton.
Experimental evidence has pointed LRP5 as a critical factor in translating mechan-
ical signals into the proper skeletal response. For example, loss-of-function muta-
tions in LRP5 have been reported to cause the autosomal recessive human disease
Osteoporosis-Pseudoglioma syndrome (OPPG), which leads to a significant reduc-
tion of BMD, and are more susceptible to skeletal fracture and deformity [115–
20 Y.-X. Qin et al.
The newly discovered MicroRNAs (miRNAs) are short noncoding RNAs, which
can be complementary to messenger RNA (mRNA) sequences to silence gene
expression by either degradation or inhibitory translation of target transcripts [123,
124]. Regulation of Runx2, bone morphogenic protein (BMP), and Wnt signaling
pathways is by far the most well-studied miRNA-related osteoblast function.
Positive and negative regulations of miRNAs on Runx2 expression have been shown
to affect skeletal morphogenesis and osteoblastogenesis [125]. Inhibition of osteo-
blastogenesis can result from miRNA-135 and miRNA-26a regulated BMP-2/Smad
signaling pathway [126]. Activation of Wnt signaling through miRNA-29a-targeted
Wnt inhibitors is upregulated during osteoblast differentiation [127]. In addition,
studies have been done to investigate the miRNA function on self-renewal and lin-
eage determination for tissue regeneration via human stromal stem cells [128, 129].
Moreover, extensive studies have also been done to access the effects of miRNAs on
osteogenic functions in committed cell lines including osteoprogenitors, osteo-
blasts, and osteocytic cell lines. In general, actions of miRNA may affect bone cell
differentiation in either positive or negative ways [123, 129].
Recent research has gained interests in studying the transcription and microRNA
regulation to better understand gene expression regulation in a mechanical loading
model. Transcription factors can bind to motifs in the promoter of genes and directly
affect their expression; therefore, mechanotransduction in bone may result in tran-
scription factors alteration for regulation. Using a predictive bioinformatics algo-
rithm, a recent study investigated the time-dependent regulatory mechanisms that
governed mechanical loading-induced gene expression in bone. Axial loading was
performed on the right forelimb in rodents. A linear model of gene expression was
created and 44 transcription factor binding motifs and 29 microRNA binding sites
were identified to predict the regulated gene expression across the time course. It
1 Biomechanics of Bone and Cartilage 21
may be important in controlling the loading-induced bone formation process via the
time-dependent regulatory mechanisms.
As a kind of connective tissue, articular cartilage is a white, smooth, and thin layer
that covers the surfaces of bones in diarthrodial joints [94]. Its primary function is
to bear physical stress, distribute the mechanical load, absorb the shock, and lubri-
cate the friction between bones in a joint during daily activities [141–143].
Articular cartilage has a highly organized layered structure. And it can be divided
into four zones based on the cell shape, size, and collagen direction (as shown in
Fig. 1.9) [142, 143]: (1) superficial zone (10–20%), elongated chondrocytes and
thin collagen fibrils are parallel to the surface of joints in this zone; (2) middle zone
(40–60%), spherical chondrocytes and larger collagen fibrils are randomly ori-
ented; (3) deep zone (30–40%), spherical chondrocytes and larger collagen fibrils
are perpendicular to the joint surface; (4) calcified zone (5%), the main components
of the calcified zone are calcium salts deposition without chondrocytes or colla-
gens [143].
22 Y.-X. Qin et al.
Tidemark
Calcified Zone (~5%)
Subchondral bone
Fig. 1.9 The stratified structure of articular cartilage and the arrangement of chondrocytes and
collagen fibrils in each zone. There are three primary layers in normal articular cartilage. The cell
shapes are shown specific shapes in various zones, e.g., flat in the superficial zone, round in the
middle zone, and column-like in the deep zone
Compared with other tissues such as bone and muscle, articular cartilage in diar-
throdial joints does not contain blood vessels, lymphatic vessels, and nerves, and
therefore the nutrients are transported to the chondrocytes from the synovial fluid in
joint movements. The capacity of articular cartilage self-repair is very limited, and
could not be able to heal on its own. During daily activities, articular cartilage expe-
riences the mechanical environment changes caused by joint movements. And it can
respond to a variety of mechanical stress, strain, and pressure generated by normal
load-bearing activities in the walking, running, and other daily exercises [145]. The
1 Biomechanics of Bone and Cartilage 23
Collagen Fabric
Negatively
charged sGAG
Proteoglycan
Chondrocyte
Extracellular Matrix
Fig. 1.10 The proteoglycan, collagen, and chondrocyte in the extracellular matrix of the articular
cartilage
chondrocytes and ECM play a unique role in the anabolism and catabolism of artic-
ular cartilage, and are responsible for the maintenance of the structural integrity and
mechanical function of articular cartilage.
Chondrocytes are sparsely embedded in the ECM [141–143, 145]. They have a
low level of metabolic activity and replication due to the limited nutrition transporta-
tion from the synovial fluid [145]. For each chondrocyte, it can establish its micro-
environment and trap itself in its matrix, which could prevent their migration to the
adjacent area of cartilage. And the communication between cells and direct cell-to-
cell signal transduction is limited under this microenvironment trap. The survival of
chondrocytes in ECM mainly depends on the mechanical and chemical environment
provided by articular cartilage, and they can regulate ECM in their vicinity area by
sensing the mechanical force in their surrounding microenvironment and responding
to mechanical stimuli caused by the physical load of joints, such as compressive,
tensile, and shear stress.
Moreover, the negatively charged sGAG in ECM can attract ions, and provide
articular cartilage with the osmotic imbalance in the ionic interstitial fluid, which
leads to absorb water and induces a swelling pressure to resist compression loads
[143, 145]. The collagen network in ECM enables articular cartilage to resist ten-
sion and shear forces. The mechanical balance between sGAG and collagen in ECM
is responsible for the stable micromechanical environment for chondrocytes [143,
145]. Taken together, the chondrocytes could sense mechanical signals to synthe-
size and maintain the turnover of ECM. In turn, the ECM could protect chondro-
cytes from potentially damaging biomechanical forces and enables articular
cartilage to support the physical load in our daily life. The interactions between
chondrocytes and ECM are crucial for the homeostasis of articular cartilage.
24 Y.-X. Qin et al.
Articular cartilage could maintain its structural integrity and mechanical func-
tion under joint normal loading conditions during routine daily activity.
However, the homeostasis of articular cartilage is interrupted when it is exposed
to insufficient or excessive mechanical loading conditions, and could cause car-
tilage degeneration with a consequence of joint diseases, such as osteoarthritis.
Previous studies have found that the synthesis of proteoglycan decreased and
cartilage degeneration was observed under joint overuse conditions in marathon
runners or animal with running exercises (as shown in Table 1.1) [147–153].
Besides, prolonged joint disuse or joint immobilization can cause joint atrophy
and AC degeneration [153–157]. Progressive thinning of AC and the degrada-
tion of the extracellular matrix (ECM) has been reported in both immobilized
patients and unload animal models [153–157]. And the animal studies also dem-
onstrated that the thickness of cartilage became thinner and the underlying sub-
chondral bone increased in osteoarthritis group induced by anterior cruciate
ligament transection; moreover, in hind limb suspension group, the irregular
and lose distributions of chondrocytes in cartilage were found due to mechani-
cal functional disuse. However, low-intensity ultrasound stimulation could gen-
erate acoustic pressure waves with mechanical energy, and it could mitigate the
cartilage degeneration [5, 143, 145] (as shown in Fig. 1.11).
Table 1.1 Safranin O intensity of cartilage in the lateral middle section of tibia (ratio = experimental/
control) [147]
Immobilization/remobilization Running
Strenuous Endurance
Moderate (20 km/ (40 km/
Cartilage Immobilization Remobilization Remobilization (4 km/day, day, 15 day, 15
zones for 11 weeks for 15 weeks for 50 weeks 15 weeks) weeks) weeks)
Superficial 0.58∗ 0.82 0.81 0.81 0.95 0.62∗∗
zone
Middle 0.80 1.00 0.81 0.90 1.02 1.00
zone
Deep zone 0.82 0.95 0.93 0.90 1.00 1.00
Calcified 1.00 1.00 0.80 1.20 1.01 0.98
zone
Changes of Safranin O intensity in different cartilage zones of the canine knee joints in the inter-
mediate section of the latéral condyle of the tibia under immobilization/remobilization conditions
and running conditions. The two-tailed Mann-Whitney U-test or Wilcoxon’s matched-pairs signed-
ranks tests was used for statistical analysis
∗
p < 0.05
∗∗
p < 0.01
1 Biomechanics of Bone and Cartilage 25
100 µm
Age-matched OA OA+US
100 µm
Fig. 1.11 The toluidine blue and fast green staining of cartilage in right tibial plateau in different
groups (OA osteoarthritis, US ultrasound, HLS hind limb suspension) [158]
12 Discussion
These data have suggested that dynamic stimulation generates fluid pressure in bone
with simultaneously low-level bone strain. MS adjacent to the rat femur induces a
peak ImP at 20–30 Hz. The increase in bone fluid pressure suggests that hyperten-
sion in the skeletal nutrient vessels may increase ImP and regulate fluid flow in bone
[56]. Similarly, the bone strain was highest at approximately 10 Hz. Stimulus-
induced bone fluid and matrix deformation are dependent on stimulation frequency.
It appears that the oscillatory MS stimulates relatively high fluid pressure at the
frequency range between 20 Hz and 50 Hz in the tested frequencies up to 100 Hz.
In such optimized loading rate (e.g., 20–50 Hz), relatively high ImP and relatively
low bone strain were observed in response to the MS loading, which may be critical
to regulating fluid flow, and adaptation in bone as a functional loading frequency-
dependent manner. It is also noted that both ImP and strain at 10 Hz and 20 Hz are
26 Y.-X. Qin et al.
higher than the value at a lower frequency, i.e., 1 Hz. There is a significant strain
difference between 10 Hz and 20 Hz, while no significant ImP difference was
observed between 10 Hz and 20 Hz. At an optimal frequency, MS can produce high
fluid pressure gradients within the femoral marrow cavity and a high strain value in
bone. It is possible that loading generated matrix strain and fluid pressure in bone
may have combined effects in adaptation if loaded at relatively high frequencies.
Dynamic muscle stimulation was able to inhibit bone loss and trabecular archi-
tectural deterioration caused by a lack of daily weight-bearing activities. The impor-
tance of selecting an effective loading regimen was investigated in this study, in
which a wide range of stimulation frequencies was tested to determine their effects
on skeletal adaptive responses. Throughout this study, we have referred 1 Hz as low
frequency, 20 Hz and 50 Hz as mid frequency, and 100 Hz as high frequency. From
our results, we concluded that the effectiveness of dynamic MS was greatly depen-
dent on the stimulation frequency. The optimized frequency, which resulted in a
strong adaptive response in the disuse osteopenia model, was in the range of 50 Hz.
While the strain level generated at 50 Hz by MS was relatively low, e.g., approxi-
mately 10 με, the adaptive response may be mainly contributed by induced fluid
flow, and uncoupled to strain [159]. The degree of effectiveness of MS in attenuating
bone loss varied in different regions of the distal femur. Such spatial response may
also depend on the fluid pressure generated in the local region as a dose-dependent
manner. While low-frequency MS was unsuccessful in preventing osteopenia, mid-
frequency MS applied to the quadriceps was able to maintain trabecular bone mass.
These results were consistent with previous in vivo results in which mechanical
loading at frequencies between 20 Hz and 50 Hz was shown to be anti-catabolic to
bone [53, 160–162]. This sensitivity was even more apparent in trabecular bone,
perhaps due to the increased surface area in the trabecular network, which exposes
it to rapid changes in fluid pressure [163]. For example, trabecular osteoblast sur-
face in the tibia was increased by 26% when an MS protocol at 10 Hz was applied
for 3 weeks [162]. Likewise, whole body vibration at 45 Hz increased the rate of
formation of the growing skeleton by 30% [164].
Both ImP and matrix strain have indicated a nonlinear response in the MS spec-
trum between 1 Hz and 100 Hz, though peaked differently at 20 Hz (ImP) and 10 Hz
(strain). While no obvious muscle fatigue was observed, perhaps due to the rest
period during the stimulation, the mechanism behind such a nonlinear response is
not clear. From the characteristics of tissue material point of view, e.g., the visco-
elastic property of the tissues, both muscle and bone could quickly dampen the
response at high frequencies through the MS loading. But, due to the difference in
densities and viscosities between muscle and bone, MS induced ImP, and matrix
strain could result in different frequency responses or optimized/resonant patterns
for different tissues against the loading. In addition, mechanotransduction of MS
through different connective tissues may attenuate the high-frequency response in
bone, e.g., via the connective pathway from muscle, tendon to bone, thus resulting
in peak strain and peak ImP at varied frequencies. Future research on such complex
interrelations between muscle kinematics, bone fluid flow, and matrix strain is nec-
essary to elucidate the mechanism further.
1 Biomechanics of Bone and Cartilage 27
Even in the absence of bone matrix strain, previous data have shown that ImP
alone can induce bone adaptation [161]. Using a turkey ulna osteotomy model, dis-
use alone resulted in a 5.7% loss of cortical bone. Direct fluid loading at 20 Hz for
4 weeks increased cortical bone mass by 18% by enhancing the formation of bone
at both periosteal and endosteal surfaces [161]. Transcortical fluid pressure gradient
and total bone formation were strongly and positively correlated. Bone fluid flow
plays an important role in triggering bone remodeling [165–167]. Strong evidence
suggests that interstitial fluid flow in bone interacts strongly with external muscular
activities via various mechanisms [168, 169]. According to a muscle pump hypoth-
esis, an arteriovenous pressure gradient enhances muscle perfusion [54, 170]. This
process may, in turn, increase the hydraulic pressure in skeletal nutrient vessels and
amplify the capillary filtration in bone tissue [54–56].
As a clinical application, MS on spinal cord-injured patients can cause a partial
reversal of disuse osteopenia and recovery of muscular strength [92]. Other in vivo
studies have also reported positive effects of using muscle stimulation to inhibit
muscle atrophy. Immobilization studies using MS at 50–100 Hz have shown to min-
imize the reduction of the cross-sectional area of muscle fiber and to restore
mechanical properties [171, 172]. Previous data showed that stimulation of distal
nerve stumps had similar action potential response between normal and innervated
muscle [173]. Although the response of ImP and bone mass by MS under such
periphery nerve block conditions still remains unknown, MS could serve as a miti-
gating agent to retain bone mass under chronic nerve damage conditions, e.g., spinal
cord injury. Taken together with the results from our current study, dynamic MS
may be applied as both a skeletal therapy and a muscular therapy to prevent osteo-
penia and sarcopenia.
Loading-induced fluid flow in the musculoskeletal tissues may ultimately
enhance interstitial flow and mechanotransduction in bone. Furthermore, dynamic
loading, if applied at an optimal frequency, has been shown to have preventive
potential in osteopenia in a functional disuse environment as a biomechanically
based intervention for preventing and treating osteoporosis and muscle atrophy.
Acknowledgments This research was supported by the National Institutes of Health (R01
AR52379, AR61821, and AR49286), and the National Space Biomedical Research Institute
through a NASA Cooperative Agreement NCC 9-58. The authors are grateful to Ms. Alyssa Tuthill
for her technical assistance and all the members in the Orthopaedic Bioengineering Research Lab,
particularly for their tireless assistance in the study.
References
1. Ozdurak RH, Duz S, Arsal G, Akinci Y, Kablan N, Isikli S, Korkusuz F. Quantitative forearm
muscle strength influences radial bone mineral density in osteoporotic and healthy males.
Technol Health Care. 2003;11:253–61.
2. Srinivasan S, Weimer DA, Agans SC, Bain SD, Gross TS. Low-magnitude mechanical load-
ing becomes osteogenic when rest is inserted between each load cycle. J Bone Miner Res.
2002;17:1613–20.
28 Y.-X. Qin et al.
3. Gerdhem P, Ringsberg KA, Akesson K, Obrant KJ. Influence of muscle strength, physical
activity and weight on bone mass in a population-based sample of 1004 elderly women.
Osteoporos Int. 2003;14:768–72.
4. Gerdhem P, Ringsberg KA, Magnusson H, Obrant KJ, Akesson K. Bone mass cannot be pre-
dicted by estimations of frailty in elderly ambulatory women. Gerontology. 2003;49:168–72.
5. LeBlanc A. Summary of research issues in human studies. Bone. 1998;22:117S–8S.
6. LeBlanc A, Lin C, Shackelford L, Sinitsyn V, Evans H, Belichenko O, Schenkman B,
Kozlovskaya I, Oganov V, Bakulin A, Hedrick T, Feeback D. Muscle volume, MRI relaxation
times (T2), and body composition after spaceflight. J Appl Physiol. 2000;89:2158–64.
7. LeBlanc AD, Evans HJ, Johnson PC, Jhingran S. Changes in total body calcium balance with
exercise in the rat. J Appl Physiol. 1983;55:201–4.
8. Convertino VA, Sandler H. Exercise countermeasures for spaceflight. Acta Astronaut.
1995;35:253–70.
9. Fluckey JD, Dupont-Versteegden EE, Montague DC, Knox M, Tesch P, Peterson CA, Gaddy-
Kurten D. A rat resistance exercise regimen attenuates losses of musculoskeletal mass during
hindlimb suspension. Acta Physiol Scand. 2002;176:293–300.
10. Keller TS, Strauss AM, Szpalski M. Prevention of bone loss and muscle atrophy during
manned space flight. Microgravity Q. 1992;2:89–102.
11. McPhee JC, White RJ. Physiology, medicine, long-duration space flight and the NSBRI. Acta
Astronaut. 2003;53:239–48.
12. Grigoriev A, Morukov B, Stupakov G, Bobrovnik E. Influence of bisphosphonates on cal-
cium metabolism and bone tissue during simulation of the physiological effects of micro-
gravity. J Gravit Physiol. 1998;5:69–70.
13. LeBlanc A, Marsh C, Evans H, Johnson P, Schneider V, Jhingran S. Bone and muscle atrophy
with suspension of the rat. J Appl Physiol. 1985;58:1669–75.
14. LeBlanc A, Schneider V. Countermeasures against space flight related bone loss. Acta
Astronaut. 1992;27:89–92.
15. LeBlanc A, Rowe R, Schneider V, Evans H, Hedrick T. Regional muscle loss after short dura-
tion spaceflight. Aviat Space Environ Med. 1995;66:1151–4.
16. Convertino VA. Mechanisms of microgravity induced orthostatic intolerance: implications
for effective countermeasures. J Gravit Physiol. 2002;9:1–13.
17. Serova LV. Microgravity and aging of animals. J Gravit Physiol. 2001;8:137–8.
18. Mayet-Sornay MH, Hoppeler H, Shenkman BS, Desplanches D. Structural changes in arm
muscles after microgravity. J Gravit Physiol. 2000;7:S43–4.
19. Larina IM, Tcheglova IA, Shenkman BS, Nemirovskaya TL. Muscle atrophy and hormonal
regulation in women in 120 day bed rest. J Gravit Physiol. 1997;4:121–2.
20. Wolff J. Das Gesetz Der Transformation Der Knochen. Berlin, Hirschwald; 1892.
21. Wolff J. The law of bone remodeling. Berlin: Springer; 1986.
22. Evans FG. The mechanical properties of bone. Artif Limbs. 1969;13:37–48.
23. Evans FG. Factors affecting the mechanical properties of bone. Bull N Y Acad Med.
1973;49:751–64.
24. Evans FG, Vincentelli R. Relations of the compressive properties of human cortical bone to
histological structure and calcification. J Biomech. 1974;7:1–10.
25. Martin RB, Burr DB. Structure, function and adaptation of compact bone. New York:
Raven; 1989.
26. Stokes IA. Analysis of symmetry of vertebral body loading consequent to lateral spinal cur-
vature. Spine. 1997;22:2495–503.
27. Donaldson CL, Hulley SB, Vogel JM, Hattner RS, Bayers JH, McMillan DE. Effect of pro-
longed bed rest on bone mineral. Metabolism. 1970;19:1071–84.
28. Gross TS, Edwards JL, McLeod KJ, Rubin CT. Strain gradients correlate with sites of perios-
teal bone formation. J Bone Miner Res. 1997;12:982–8.
29. Qin YX, Otter MW, Rubin CT, McLeod KJ. The influence of intramedullary hydrostatic pres-
sure on transcortical fluid flow patterns in bone. Trans Ortho Res Soc. 1997;22:885.
1 Biomechanics of Bone and Cartilage 29
30. Rubin CT, Lanyon LE. Regulation of bone formation by applied dynamic loads. J Bone Joint
Surg Am. 1984;66:397–402.
31. Turner CH. Site-specific skeletal effects of exercise: importance of interstitial fluid pressure.
Bone. 1999;24:161–2.
32. Jones HH, Priest JD, Hayes WC, Tichenor CC, Nagel DA. Humeral hypertrophy in response
to exercise. J Bone Joint Surg Am. 1977;59:204–8.
33. Judex S, Zernicke RF. High-impact exercise and growing bone: relation between high strain
rates and enhanced bone formation. J Appl Physiol. 2000;88:2183–91.
34. Krolner B, Toft B, Pors NS, Tondevold E. Physical exercise as prophylaxis against involu-
tional vertebral bone loss: a controlled trial. Clin Sci (Lond). 1983;64:541–6.
35. Nilsson BE, Westlin NE. Bone density in athletes. Clin Orthop. 1971;77:179–82.
36. Stokes IA, Aronsson DD, Spence H, Iatridis JC. Mechanical modulation of intervertebral disc
thickness in growing rat tails. J Spinal Disord. 1998;11:261–5.
37. Joyner MJ, Lennon RL, Wedel DJ, Rose SH, Shepherd JT. Blood flow to contracting human
muscles: influence of increased sympathetic activity. J Appl Physiol. 1990;68:1453–7.
38. Joyner MJ. Does the pressor response to ischemic exercise improve blood flow to contracting
muscles in humans? J Appl Physiol. 1991;71:1496–501.
39. Joyner MJ, Nauss LA, Warner MA, Warner DO. Sympathetic modulation of blood
flow and O2 uptake in rhythmically contracting human forearm muscles. Am J Physiol.
1992;263:H1078–83.
40. Joyner MJ, Wieling W. Increased muscle perfusion reduces muscle sympathetic nerve activ-
ity during handgripping. J Appl Physiol. 1993;75:2450–5.
41. Joyner MJ, Dietz NM. Nitric oxide and vasodilation in human limbs. J Appl Physiol.
1997;83:1785–96.
42. Joyner MJ, Proctor DN. Muscle blood flow during exercise: the limits of reductionism. Med
Sci Sports Exerc. 1999;31:1036–40.
43. Joyner MJ. Blood pressure and exercise: failing the acid test. J Physiol. 2001;537:331.
44. Hicks A, McGill S, Hughson RL. Tissue oxygenation by near-infrared spectroscopy
and muscle blood flow during isometric contractions of the forearm. Can J Appl Physiol.
1999;24:216–30.
45. Mak AF, Huang L, Wang Q. A biphasic poroelastic analysis of the flow dependent subcuta-
neous tissue pressure and compaction due to epidermal loadings: issues in pressure sore. J
Biomech Eng. 1994;116:421–9.
46. Qin YX, Lin W, Rubin C. The pathway of bone fluid flow as defined by in vivo intramed-
ullary pressure and streaming potential measurements. Ann Biomed Eng. 2002;30(5):
693–2.
47. Qin YX, Hu M. Mechanotransduction in musculoskeletal tissue regeneration: effects of fluid
flow, loading, and cellular-molecular pathways. Biomed Res Int. 2014;2014:863421.
48. Frost HM. Intermediary organization of the skeleton. Boca Raton, FL: CRC Press; 1986.
49. Frost HM. Bone’s mechanostat: a 2003 update. Anat Rec. 2003;275A:1081–101.
50. Cowin SC. Bone mechanics. Boca Raton, FL: CRC Press; 1989.
51. Cowin SC. Bone poroelasticity. J Biomech. 1999;32:217–38.
52. Piekarski K, Demetriades D, Mackenzie A. Osteogenetic stimulation by externally applied dc
current. Acta Orthop Scand. 1978;49:113–20.
53. Qin Y-X, Rubin CT, McLeod KJ. Non-linear dependence of loading intensity and cycle num-
ber in the maintenance of bone mass and morphology. J Orthop Res. 1998;16:482–9.
54. Laughlin MH. The muscle pump: what question do we want to answer? J Appl Physiol.
2005;99:774.
55. Otter MW, Qin YX, Rubin CT, McLeod KJ. Does bone perfusion/reperfusion initiate bone
remodeling and the stress fracture syndrome? Med Hypotheses. 1999;53:363–8.
56. Winet H. A bone fluid flow hypothesis for muscle pump-driven capillary filtration:
II. Proposed role for exercise in erodible scaffold implant incorporation. Eur Cell Mater.
2003;6:1–10.
30 Y.-X. Qin et al.
57. Lanyon LE, Rubin CT. Static vs dynamic loads as an influence on bone remodelling. J
Biomech. 1984;17:897–905.
58. Rubin CT, Lanyon LE. Kappa Delta Award paper. Osteoregulatory nature of mechani-
cal stimuli: function as a determinant for adaptive remodeling in bone. J Orthop Res.
1987;5:300–10.
59. Biering-Sorensen F, Bohr HH, Schaadt OP. Longitudinal study of bone mineral content in the
lumbar spine, the forearm and the lower extremities after spinal cord injury. Eur J Clin Invest.
1990;20:330–5.
60. Dauty M, Perrouin VB, Maugars Y, Dubois C, Mathe JF. Supralesional and sublesional bone
mineral density in spinal cord-injured patients. Bone. 2000;27:305–9.
61. Garland DE, Adkins RH, Stewart CA, Ashford R, Vigil D. Regional osteoporosis in women
who have a complete spinal cord injury. J Bone Joint Surg Am. 2001;83-A:1195–200.
62. Lazo MG, Shirazi P, Sam M, Giobbie-Hurder A, Blacconiere MJ, Muppidi M. Osteoporosis
and risk of fracture in men with spinal cord injury. Spinal Cord. 2001;39:208–14.
63. Ryg J, Rejnmark L, Overgaard S, Brixen K, Vestergaard P. Hip fracture patients at risk of
second hip fracture: a nationwide population-based cohort study of 169,145 cases during
1977-2001. J Bone Miner Res. 2009;24:1299–307.
64. Vestergaard P, Krogh K, Rejnmark L, Mosekilde L. Fracture rates and risk factors for frac-
tures in patients with spinal cord injury. Spinal Cord. 1998;36:790–6.
65. Vestergaard P, Rejnmark L, Mosekilde L. Osteoarthritis and risk of fractures. Calcif Tissue
Int. 2009;84:249–56.
66. Armstrong VJ, Muzylak M, Sunters A, Zaman G, Saxon LK, Price JS, Lanyon LE. Wnt/beta-
catenin signaling is a component of osteoblastic bone cell early responses to load-bearing and
requires estrogen receptor alpha. J Biol Chem. 2007;282:20715–27.
67. Heaney RP. Pathophysiology of osteoporosis. Endocrinol Metab Clin North Am.
1998;27:255–65.
68. Ingram RR, Suman RK, Freeman PA. Lower limb fractures in the chronic spinal cord injured
patient. Paraplegia. 1989;27:133–9.
69. Szollar SM, Martin EM, Sartoris DJ, Parthemore JG, Deftos LJ. Bone mineral density and
indexes of bone metabolism in spinal cord injury. Am J Phys Med Rehabil. 1998;77:28–35.
70. Macdonald JH, Evans SF, Davie MW, Sharp CA. Muscle mass deficits are associated with bone
mineral density in men with idiopathic vertebral fracture. Osteoporos Int. 2007;18:1371–8.
71. McCarthy I, Goodship A, Herzog R, Oganov V, Stussi E, Vahlensieck M. Investigation of
bone changes in microgravity during long and short duration space flight: comparison of
techniques. Eur J Clin Invest. 2000;30:1044–54.
72. Lang T, LeBlanc A, Evans H, Lu Y, Genant H, Yu A. Cortical and trabecular bone mineral loss
from the spine and hip in long-duration spaceflight. J Bone Miner Res. 2004;19:1006–12.
73. Burr DB, Frederickson RG, Pavlinch C, Sickles M, Burkart S. Intracast muscle stimula-
tion prevents bone and cartilage deterioration in cast-immobilized rabbits. Clin Orthop.
1984;189:264–78.
74. LeBlanc A, Rowe R, Evans H, West S, Shackelford L, Schneider V. Muscle atrophy during
long duration bed rest. Int J Sports Med. 1997;18(Suppl 4):S283–5.
75. Narici M, Kayser B, Barattini P, Cerretelli P. Effects of 17-day spaceflight on electrically
evoked torque and cross-sectional area of the human triceps surae. Eur J Appl Physiol.
2003;90:275–82.
76. Gorgey AS, Dudley GA. Skeletal muscle atrophy and increased intramuscular fat after
incomplete spinal cord injury. Spinal Cord. 2007;45:304–9.
77. Shah PK, Stevens JE, Gregory CM, Pathare NC, Jayaraman A, Bickel SC, Bowden M,
Behrman AL, Walter GA, Dudley GA, Vandenborne K. Lower-extremity muscle cross-
sectional area after incomplete spinal cord injury. Arch Phys Med Rehabil. 2006;87:772–8.
78. Riley DA, Slocum GR, Bain JL, Sedlak FR, Sowa TE, Mellender JW. Rat hindlimb
unloading: soleus histochemistry, ultrastructure, and electromyography. J Appl Physiol.
1990;69:58–66.
1 Biomechanics of Bone and Cartilage 31
79. Roy RR, Pierotti DJ, Garfinkel A, Zhong H, Baldwin KM, Edgerton VR. Persistence of motor
unit and muscle fiber types in the presence of inactivity. J Exp Biol. 2008;211:1041–9.
80. Zhong H, Roy RR, Siengthai B, Edgerton VR. Effects of inactivity on fiber size and myo-
nuclear number in rat soleus muscle. J Appl Physiol. 2005;99:1494–9.
81. Stewart BG, Tarnopolsky MA, Hicks AL, McCartney N, Mahoney DJ, Staron RS, Phillips
SM. Treadmill training-induced adaptations in muscle phenotype in persons with incomplete
spinal cord injury. Muscle Nerve. 2004;30:61–8.
82. Zhou MY, Klitgaard H, Saltin B, Roy RR, Edgerton VR, Gollnick PD. Myosin heavy
chain isoforms of human muscle after short-term spaceflight. J Appl Physiol. 1995;78:
1740–4.
83. Griffin L, Decker MJ, Hwang JY, Wang B, Kitchen K, Ding Z, Ivy JL. Functional electrical
stimulation cycling improves body composition, metabolic and neural factors in persons with
spinal cord injury. J Electromyogr Kinesiol. 2009;19:614–22.
84. Lim PA, Tow AM. Recovery and regeneration after spinal cord injury: a review and summary
of recent literature. Ann Acad Med Singapore. 2007;36:49–57.
85. Rodgers MM, Glaser RM, Figoni SF, Hooker SP, Ezenwa BN, Collins SR, Mathews T,
Suryaprasad AG, Gupta SC. Musculoskeletal responses of spinal cord injured individuals
to functional neuromuscular stimulation-induced knee extension exercise training. J Rehabil
Res Dev. 1991;28:19–26.
86. Shields RK, Dudley-Javoroski S. Musculoskeletal adaptations in chronic spinal cord injury:
effects of long-term soleus electrical stimulation training. Neurorehabil Neural Repair.
2007;21:169–79.
87. Dudley-Javoroski S, Littmann AE, Iguchi M, Shields RK. Doublet stimulation protocol to
minimize musculoskeletal stress during paralyzed quadriceps muscle testing. J Appl Physiol.
2008;104:1574–82.
88. Dudley-Javoroski S, Shields RK. Muscle and bone plasticity after spinal cord injury:
review of adaptations to disuse and to electrical muscle stimulation. J Rehabil Res Dev.
2008;45:283–96.
89. Dudley-Javoroski S, Shields RK. Dose estimation and surveillance of mechanical loading
interventions for bone loss after spinal cord injury. Phys Ther. 2008;88:387–96.
90. Yang YS, Koontz AM, Triolo RJ, Cooper RA, Boninger ML. Biomechanical analysis of func-
tional electrical stimulation on trunk musculature during wheelchair propulsion. Neurorehabil
Neural Repair. 2009;23:717–25.
91. BeDell KK, Scremin AM, Perell KL, Kunkel CF. Effects of functional electrical stimulation-
induced lower extremity cycling on bone density of spinal cord-injured patients. Am J Phys
Med Rehabil. 1996;75:29–34.
92. Belanger M, Stein RB, Wheeler GD, Gordon T, Leduc B. Electrical stimulation: can it
increase muscle strength and reverse osteopenia in spinal cord injured individuals? Arch Phys
Med Rehabil. 2000;81:1090–8.
93. Mohr T. [Electric stimulation in muscle training of the lower extremities in persons with
spinal cord injuries]. Ugeskr Laeger. 2000;162:2190–4.
94. Allen MR, Hogan HA, Bloomfield SA. Differential bone and muscle recovery follow-
ing hindlimb unloading in skeletally mature male rats. J Musculoskelet Neuronal Interact.
2006;6:217–25.
95. Swift JM, Nilsson MI, Hogan HA, Sumner LR, Bloomfield SA. Simulated resistance training
during hindlimb unloading abolishes disuse bone loss and maintains muscle strength. J Bone
Miner Res. 2010;25:564–74.
96. Qin YX, Lam H. Intramedullary pressure and matrix strain induced by oscillatory skeletal
muscle stimulation and its potential in adaptation. J Biomech. 2009;42:140–5.
97. Lam H, Qin YX. The effects of frequency-dependent dynamic muscle stimulation on inhibi-
tion of trabecular bone loss in a disuse model. Bone. 2008;43:1093–100.
98. Morey-Holton ER, Globus RK. Hindlimb unloading of growing rats: a model for predicting
skeletal changes during space flight. Bone. 1998;22:83S–8S.
32 Y.-X. Qin et al.
99. Morey-Holton ER, Globus RK. Hindlimb unloading rodent model: technical aspects. J Appl
Physiol. 2002;92:1367–77.
100. Raggatt LJ, Partridge NC. Cellular and molecular mechanisms of bone remodeling. J Biol
Chem. 2010;285:25103–8.
101. Ozcivici E, Luu YK, Adler B, Qin YX, Rubin J, Judex S, Rubin CT. Mechanical signals as
anabolic agents in bone. Nat Rev Rheumatol. 2010;6:50–9.
102. Rubin CT, Capilla E, Luu YK, Busa B, Crawford H, Nolan DJ, Mittal V, Rosen CJ, Pessin
JE, Judex S. Adipogenesis is inhibited by brief, daily exposure to high-frequency, extremely
low-magnitude mechanical signals. Proc Natl Acad Sci U S A. 2007;104:17879–84.
103. Luu YK, Pessin JE, Judex S, Rubin J, Rubin CT. Mechanical signals as a non-invasive means
to influence mesenchymal stem cell fate, promoting bone and suppressing the fat phenotype.
Bonekey Osteovision. 2009;6:132–49.
104. Luu YK, Capilla E, Rosen CJ, Gilsanz V, Pessin JE, Judex S, Rubin CT. Mechanical stimula-
tion of mesenchymal stem cell proliferation and differentiation promotes osteogenesis while
preventing dietary-induced obesity. J Bone Miner Res. 2009;24:50–61.
105. Kawaguchi H, Akune T, Yamaguchi M, Ohba S, Ogata N, Chung UI, Kubota N, Terauchi Y,
Kadowaki T, Nakamura K. Distinct effects of PPARgamma insufficiency on bone marrow
cells, osteoblasts, and osteoclastic cells. J Bone Miner Metab. 2005;23:275–9.
106. Liu J, Wang Y, Pan Q, Su Y, Zhang Z, Han J, Zhu X, Tang C, Hu D. Wnt/beta-catenin pathway
forms a negative feedback loop during TGF-beta1 induced human normal skin fibroblast-to-
myofibroblast transition. J Dermatol Sci. 2012;65:38–49.
107. Taes YE, Lapauw B, Vanbillemont G, Bogaert V, De Bacquer D, Zmierczak H, Goemaere S,
Kaufman JM. Fat mass is negatively associated with cortical bone size in young healthy male
siblings. J Clin Endocrinol Metab. 2009;94:2325–31.
108. Bonewald LF. Osteocytes: a proposed multifunctional bone cell. J Musculoskelet Neuronal
Interact. 2002;2:239–41.
109. Bonewald LF. Osteocytes as dynamic multifunctional cells. Ann N Y Acad Sci.
2007;1116:281–90.
110. Bonewald LF, Johnson ML. Osteocytes, mechanosensing and Wnt signaling. Bone.
2008;42:606–15.
111. Robinson JA, Chatterjee-Kishore M, Yaworsky PJ, Cullen DM, Zhao W, Li C, Kharode Y,
Sauter L, Babij P, Brown EL, Hill AA, Akhter MP, Johnson ML, Recker RR, Komm BS, Bex
FJ. Wnt/beta-catenin signaling is a normal physiological response to mechanical loading in
bone. J Biol Chem. 2006;281:31720–8.
112. Ke HZ, Richards WG, Li X, Ominsky MS. Sclerostin and Dickkopf-1 as therapeutic targets
in bone diseases. Endocr Rev. 2012;33:747–83.
113. Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, Harris SE, Wu D. Sclerostin binds to LRP5/6
and antagonizes canonical Wnt signaling. J Biol Chem. 2005;280:19883–7.
114. Li X, Liu P, Liu W, Maye P, Zhang J, Zhang Y, Hurley M, Guo C, Boskey A, Sun L, Harris SE,
Rowe DW, Ke HZ, Wu D. Dkk2 has a role in terminal osteoblast differentiation and mineral-
ized matrix formation. Nat Genet. 2005;37:945–52.
115. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, Wang H, Cundy T,
Glorieux FH, Lev D, Zacharin M, Oexle K, Marcelino J, Suwairi W, Heeger S, Sabatakos G,
Apte S, Adkins WN, Allgrove J, Arslan-Kirchner M, Batch JA, Beighton P, Black GC, Boles
RG, Boon LM, Borrone C, Brunner HG, Carle GF, Dallapiccola B, De Paepe A, Floege B,
Halfhide ML, Hall B, Hennekam RC, Hirose T, Jans A, Juppner H, Kim CA, Keppler-Noreuil
K, Kohlschuetter A, LaCombe D, Lambert M, Lemyre E, Letteboer T, Peltonen L, Ramesar
RS, Romanengo M, Somer H, Steichen-Gersdorf E, Steinmann B, Sullivan B, Superti-Furga
A, Swoboda W, van den Boogaard MJ, Van Hul W, Vikkula M, Votruba M, Zabel B, Garcia
T, Baron R, Olsen BR, Warman ML. LDL receptor-related protein 5 (LRP5) affects bone
accrual and eye development. Cell. 2001;107:513–23.
116. Robling AG, Turner CH. Mechanotransduction in bone: genetic effects on mechanosensitiv-
ity in mice. Bone. 2002;31:562–9.
1 Biomechanics of Bone and Cartilage 33
117. Robling AG, Turner CH. Mechanical signaling for bone modeling and remodeling. Crit Rev
Eukaryot Gene Expr. 2009;19:319–38.
118. Sawakami K, Robling AG, Ai M, Pitner ND, Liu D, Warden SJ, Li J, Maye P, Rowe DW,
Duncan RL, Warman ML, Turner CH. The Wnt co-receptor LRP5 is essential for skeletal
mechanotransduction but not for the anabolic bone response to parathyroid hormone treat-
ment. J Biol Chem. 2006;281:23698–711.
119. Kiel DP, Hannan MT, Barton BA, Bouxsein ML, Lang TF, Brown KM, Shane E, Magaziner
J, Zimmerman S, Rubin CT. Insights from the conduct of a device trial in older persons: low
magnitude mechanical stimulation for musculoskeletal health. Clin Trials. 2010;7:354–67.
120. Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne M, Folz C, Manning SP, Swain PM,
Zhao SC, Eustace B, Lappe MM, Spitzer L, Zweier S, Braunschweiger K, Benchekroun Y,
Hu X, Adair R, Chee L, FitzGerald MG, Tulig C, Caruso A, Tzellas N, Bawa A, Franklin B,
McGuire S, Nogues X, Gong G, Allen KM, Anisowicz A, Morales AJ, Lomedico PT, Recker
SM, Van Eerdewegh P, Recker RR, Johnson ML. A mutation in the LDL receptor-related protein
5 gene results in the autosomal dominant high-bone-mass trait. Am J Hum Genet. 2002;70:11–9.
121. Niziolek PJ, Warman ML, Robling AG. Mechanotransduction in bone tissue: The A214V and
G171V mutations in Lrp5 enhance load-induced osteogenesis in a surface-selective manner.
Bone. 2012;51:459–65.
122. Zhong Z, Zeng XL, Ni JH, Huang XF. Comparison of the biological response of osteoblasts
after tension and compression. Eur J Orthod. 2013;35:59–65.
123. Lisse TS, Chun RF, Rieger S, Adams JS, Hewison M. Vitamin D activation of functionally dis-
tinct regulatory miRNAs in primary human osteoblasts. J Bone Miner Res. 2013;28:1478–88.
124. Sengul A, Santisuk R, Xing W, Kesavan C. Systemic administration of an antagomir designed
to inhibit miR-92, a regulator of angiogenesis, failed to modulate skeletal anabolic response
to mechanical loading. Physiol Res. 2013;62:221–6.
125. Lian JB, Stein GS, van Wijnen AJ, Stein JL, Hassan MQ, Gaur T, Zhang Y. MicroRNA con-
trol of bone formation and homeostasis. Nat Rev Endocrinol. 2012;8:212–27.
126. Luzi E, Marini F, Sala SC, Tognarini I, Galli G, Brandi ML. Osteogenic differentiation
of human adipose tissue-derived stem cells is modulated by the miR-26a targeting of the
SMAD1 transcription factor. J Bone Miner Res. 2008;23:287–95.
127. Kapinas K, Kessler CB, Delany AM. miR-29 suppression of osteonectin in osteo-
blasts: regulation during differentiation and by canonical Wnt signaling. J Cell Biochem.
2009;108:216–24.
128. Mizuno Y, Yagi K, Tokuzawa Y, Kanesaki-Yatsuka Y, Suda T, Katagiri T, Fukuda T,
Maruyama M, Okuda A, Amemiya T, Kondoh Y, Tashiro H, Okazaki Y. miR-125b inhibits
osteoblastic differentiation by down-regulation of cell proliferation. Biochem Biophys Res
Commun. 2008;368:267–72.
129. Zhang JF, Fu WM, He ML, Xie WD, Lv Q, Wan G, Li G, Wang H, Lu G, Hu X, Jiang S, Li JN,
Lin MC, Zhang YO, Kung HF. MiRNA-20a promotes osteogenic differentiation of human
mesenchymal stem cells by co-regulating BMP signaling. RNA Biol. 2011;8:829–38.
130. Klein-Nulend J, Bacabac RG, Mullender MG. Mechanobiology of bone tissue. Pathol Biol
(Paris). 2005;53:576–80.
131. Sikavitsas VI, Temenoff JS, Mikos AG. Biomaterials and bone mechanotransduction.
Biomaterials. 2001;22:2581–93.
132. Ferreri SL, Talish R, Trandafir T, Qin YX. Mitigation of bone loss with ultrasound
induced dynamic mechanical signals in an OVX induced rat model of osteopenia. Bone.
2011;48:1095–102.
133. Hu M, Cheng J, Qin YX. Dynamic hydraulic flow stimulation on mitigation of trabecular
bone loss in a rat functional disuse model. Bone. 2012;51:819–25.
134. Uddin SMZ, Cheng J, Lin W, Qin YX. Low-intensity amplitude modulated ultrasound
increases osteoblastic mineralization. Cell Mol Bioeng. 2011;4:81–90.
135. Hu M, Serra-Hsu F, Bethel N, Lin L, Ferreri S, Cheng J, Qin YX. Dynamic hydraulic fluid
stimulation regulated intramedullary pressure. Bone. 2013;57(1):137–41.
34 Y.-X. Qin et al.
136. Qin YX, Lam H, Ferreri S, Rubin C. Dynamic skeletal muscle stimulation and its potential in
bone adaptation. J Musculoskelet Neuronal Interact. 2010;10:12–24.
137. Datta N, Pham QP, Sharma U, Sikavitsas VI, Jansen JA, Mikos AG. In vitro generated extra-
cellular matrix and fluid shear stress synergistically enhance 3D osteoblastic differentiation.
Proc Natl Acad Sci U S A. 2006;103:2488–93.
138. Mauney JR, Sjostorm S, Blumberg J, Horan R, O’Leary JP, Vunjak-Novakovic G, Volloch
V, Kaplan DL. Mechanical stimulation promotes osteogenic differentiation of human bone
marrow stromal cells on 3-D partially demineralized bone scaffolds in vitro. Calcif Tissue Int.
2004;74:458–68.
139. Milan JL, Planell JA, Lacroix D. Computational modelling of the mechanical environment
of osteogenesis within a polylactic acid-calcium phosphate glass scaffold. Biomaterials.
2009;30:4219–26.
140. Pioletti DP. Biomechanics and tissue engineering. Osteoporos Int. 2011;22:2027–31.
141. Corvelli M, Che B, Saeui C, Singh A, Elisseeff J. Biodynamic performance of hyaluronic
acid versus synovial fluid of the knee in osteoarthritis. Methods. 2015;84:90–8.
142. Sophia Fox AJ, Bedi A, Rodeo SA. The basic science of articular cartilage: structure, compo-
sition, and function. Sports Health. 2009;1:461–8.
143. Steward AJ, Liu Y, Wagner DR. Engineering cell attachments to scaffolds in cartilage tissue
engineering. JOM. 2011;63:77–82.
144. Kurz B, Lemke AK, Fay J, Pufe T, Grodzinsky AJ, Schunke M. Pathomechanisms of cartilage
destruction by mechanical injury. Ann Anat. 2005;187:473–85.
145. Pearle AD, Warren RF, Rodeo SA. Basic science of articular cartilage and osteoarthritis. Clin
Sports Med. 2005;24:1–12.
146. Bansal PN, Joshi NS, Entezari V, Grinstaff MW, Snyder BD. Contrast enhanced computed
tomography can predict the glycosaminoglycan content and biomechanical properties of
articular cartilage. Osteoarthritis Cartilage. 2010;18:184–91.
147. Arokoski JP, Jurvelin JS, Vaatainen U, Helminen HJ. Normal and pathological adaptations of
articular cartilage to joint loading. Scand J Med Sci Sports. 2000;10:186–98.
148. Guilak F, Nims RJ, Dicks A, Wu CL, Meulenbelt I. Osteoarthritis as a disease of the cartilage
pericellular matrix. Matrix Biol. 2018;71–72:40–50.
149. Hunter DJ, Arden N, Conaghan PG, Eckstein F, Gold G, Grainger A, Guermazi A, Harvey W,
Jones G, Hellio Le Graverand MP, Laredo JD, Lo G, Losina E, Mosher TJ, Roemer F, Zhang
W. Definition of osteoarthritis on MRI: results of a Delphi exercise. Osteoarthritis Cartilage.
2011;19:963–9.
150. Hunter DJ, Beavers DP, Eckstein F, Guermazi A, Loeser RF, Nicklas BJ, Mihalko SL, Miller
GD, Lyles M, Devita P, Legault C, Carr JJ, Williamson JD, Messier SP. The Intensive Diet and
Exercise for Arthritis (IDEA) trial: 18-month radiographic and MRI outcomes. Osteoarthritis
Cartilage. 2015;23:1090–8.
151. Jorgensen AEM, Kjaer M, Heinemeier KM. The effect of aging and mechanical loading on
the metabolism of articular cartilage. J Rheumatol. 2017;44:410–7.
152. Mazor M, Best TM, Cesaro A, Lespessailles E, Toumi H. Osteoarthritis biomarker responses
and cartilage adaptation to exercise: a review of animal and human models. Scand J Med Sci
Sports. 2019;29:1072–82.
153. Guilak F. Biomechanical factors in osteoarthritis. Best Pract Res Clin Rheumatol.
2011;25:815–23.
154. Guilak F, Butler DL, Goldstein SA, Baaijens FP. Biomechanics and mechanobiology in func-
tional tissue engineering. J Biomech. 2014;47:1933–40.
155. Sun HB. Mechanical loading, cartilage degradation, and arthritis. Ann N Y Acad Sci.
2010;1211:37–50.
156. Trudel G, Uhthoff H, Laneuville O. Knee joint immobility induces Mcl-1 gene expression in
articular chondrocytes. Biochem Biophys Res Commun. 2005;333:247–52.
157. Vanwanseele B, Lucchinetti E, Stussi E. The effects of immobilization on the characteris-
tics of articular cartilage: current concepts and future directions. Osteoarthritis Cartilage.
2002;10:408–19.
1 Biomechanics of Bone and Cartilage 35
158. Li X, Sun Y, Zhou Z, Zhang D, Jiao J, Hu M, Hassan CR, Qin YX. Mitigation of articu-
lar cartilage degeneration and subchondral bone sclerosis in osteoarthritis progression using
low-intensity ultrasound stimulation. Ultrasound Med Biol. 2019;45:148–59.
159. Caulkins C, Ebramzadeh E, Winet H. Skeletal muscle contractions uncoupled from gravi-
tational loading directly increase cortical bone blood flow rates in vivo. J Orthop Res.
2009;27:651–6.
160. Garman R, Gaudette G, Donahue LR, Rubin C, Judex S. Low-level accelerations applied
in the absence of weight bearing can enhance trabecular bone formation. J Orthop Res.
2007;25:732–40.
161. Qin YX, Kaplan T, Saldanha A, Rubin C. Fluid pressure gradients, arising from oscillations
in intramedullary pressure, are correlated with the formation of bone and inhibition of intra-
cortical porosity. J Biomech. 2003;36:1427–37.
162. Zerath E, Canon F, Guezennec CY, Holy X, Renault S, Andre C. Electrical stimulation of
leg muscles increases tibial trabecular bone formation in unloaded rats. J Appl Physiol.
1995;79:1889–94.
163. Qin YX, Lin W, Rubin CT. Load-induced bone fluid flow pathway as defined by in-vivo
intramedullary pressure and streaming potentials measurements. Ann Biomed Eng.
2002;30:693–702.
164. Xie L, Jacobson JM, Choi ES, Busa B, Donahue LR, Miller LM, Rubin CT, Judex S. Low-
level mechanical vibrations can influence bone resorption and bone formation in the growing
skeleton. Bone. 2006;39:1059–66.
165. Wang L, Fritton SP, Cowin SC, Weinbaum S. Fluid pressure relaxation depends upon osteonal
microstructure: modeling an oscillatory bending experiment. J Biomech. 1999;32:663–72.
166. Wang L, Fritton SP, Weinbaum S, Cowin SC. On bone adaptation due to venous stasis. J
Biomech. 2003;36:1439–51.
167. Wang L, Ciani C, Doty SB, Fritton SP. Delineating bone’s interstitial fluid pathway in vivo.
Bone. 2004;34:499–509.
168. Stevens HY, Meays DR, Frangos JA. Pressure gradients and transport in the murine femur
upon hindlimb suspension. Bone. 2006;39:565–72.
169. Valic Z, Buckwalter JB, Clifford PS. Muscle blood flow response to contraction: influence of
venous pressure. J Appl Physiol. 2005;98:72–6.
170. Laughlin MH, Joyner M. Closer to the edge? Contractions, pressures, waterfalls and blood
flow to contracting skeletal muscle. J Appl Physiol. 2003;94:3–5.
171. Kim SJ, Roy RR, Zhong H, Suzuki H, Ambartsumyan L, Haddad F, Baldwin KM, Edgerton
VR. Electromechanical stimulation ameliorates inactivity-induced adaptations in the medial
gastrocnemius of adult rats. J Appl Physiol. 2007;103:195–205.
172. Qin L, Appell HJ, Chan KM, Maffulli N. Electrical stimulation prevents immobilization atro-
phy in skeletal muscle of rabbits. Arch Phys Med Rehabil. 1997;78:512–7.
173. O’Gara T, Urban W, Polishchuk D, Pierre-Louis A, Stewart M. Continuous stimulation of
transected distal nerves fails to prolong action potential propagation. Clin Orthop Relat Res.
2006;447:209–13.
Chapter 2
Biomechanics of Skeletal Muscle
and Tendon
Abstract Skeletal muscle is the biological tissue able to transform chemical energy
to mechanical energy. Skeletal muscle has three basic performance parameters that
describe its function: structure and composition, force production, and movement
production. From a mechanical perspective, the musculotendinous unit behaves as
an elastic-contractile component (muscle fibers) in series with another elastic com-
ponent (tendons) to move human body. Due to their unique hierarchical structure
and composition, tendons possess characteristic biomechanical properties, includ-
ing high mechanical strength and viscoelasticity, which enable them to carry and
transmit mechanical loads (muscular forces) effectively. Tendons are also mechano-
responsive by adaptively changing their structure and function in response to altered
mechanical loading conditions. The production of movement and force is the
mechanical outcome of skeletal muscle contraction. The focus of this chapter is on
the biomechanical behavior of skeletal muscle and tendon as they contribute to
function of the musculoskeletal system.
Y.-H. Chao
School and Graduate Institute of Physical Therapy, College of Medicine, National Taiwan
University, Taipei City, Taiwan
e-mail: yuanhungchao@ntu.edu.tw
J.-S. Sun (*)
Department of Orthopedic Surgery, College of Medicine, National Taiwan University,
Taipei City, Taiwan
Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei City, Taiwan
1 Skeletal Muscle
1.1.1 Muscle Fiber
The fiber of the skeletal muscle is a long, cylindrical, multinucleated cell that is
filled with smaller units of filaments. The largest of the filaments is the myofibril,
which is composed of the sarcomeres that are arranged from end to end through the
length of the myofibril. Each sarcomere also contains filaments, which is known as
myofilaments. There are two types of myofilaments within each sarcomere, namely,
thicker myofilaments and thinner myofilaments. The thicker one is composed of
myosin protein molecules. The thinner one is composed of molecules of the protein
actin. Sliding of the actin myofilament on the myosin chain is the basic mechanism
of muscle contraction.
Connective tissue within the muscle, called intramuscular connective structure
(IMCT), consists of predominantly collagen protein and proteoglycans [1]. It covers
skeletal muscle fiber in layers, providing mechanical support for blood vessels and
nerves deep in every muscle fiber. As muscle fiber generates mechanical tension, the
IMCT transmits muscular passive force [2–4]. Extending from tendon, epimysium
embraces every individual muscle. Fascicles within the muscle are layered by the
perimysium, and individual muscle fibers are covered by the endomysium. IMCT
comprises various proportion of collagen fiber, proteoglycans, and other substances.
According to the location and ingredients, IMCT plays various functional roles.
Endomysium with better elasticity can adapt with more deformation. Perimysium
with better stiffness can transmit force produced by muscle fibers more efficiently.
Endomysium is directly in contact with the sarcolemma and muscle fiber,
accounting for only 0.47–1.2% of the muscle dry weight. The main collagen fiber
type of endomysium is type III, IV, V, and lesser percentage of type I is also found.
Due to lesser proportion of type I collagen, endomysium is more elastic and less
stiff for transmitting force. When force is produced by muscle fibers, the adjacent
endomysium adapts itself with various angle between collagen fibers of endomy-
sium and muscle fibers. Because of this adaptation, the endomysium can resist elon-
gation deformation up to 150% change of its original physiological length. This
adaptation and continuity of endomysium let muscle fibers not recruited during a
movement render tendon transmitting force without changing length.
Different with endomysium, perimysium is multilayered, consisting of collagen
fibers type I, III, IV, V, VI, and XII and proteoglycans, elastin fiber [1, 4]. Due to
collagen fiber type I, perimysium is able to resist more traction force and has an
important role in transmitting force. According to studies on bovine muscle, peri-
mysium accounts for 0.4–4.8% muscle dry weight with different functions. The
ratio of weight of perimysium in different muscles is more various than endomy-
sium. This variance may indicate that perimysium plays a main role in defining
functional differences in multiple muscle groups [1].
2 Biomechanics of Skeletal Muscle and Tendon 39
The epimysium, formed by collagen fibers type I and III, with larger diameter is
thicker than endo- and perimysium [3]. It covers all muscle belly and consists of two
layers of wavy collagen fibrils in contact with tendon surface [1, 4]. When tension
takes place along the direction of the fiber, epimysium provides resistance. When
the force is orthogonal, the epimysium will show a discrete yielding. When epimy-
sium of two different muscles connects, it provides a membrane for vessels and
nerves to transport into muscles and a surface for sliding in all directions when
muscle contracts. Epimysium receives forces directly from perimysium and muscle
fibers sometimes, and transmits the force to the tendons or the aponeurotic expan-
sions. In conjunction with different muscles, it provides a fluent phase for sliding.
1.1.2 Mononuclear Cells
Fibroblast is the primary mononuclear cell in normal muscle ECM which is respon-
sible for producing collagen, fibronectin, MMPs, and PGs [5]. Being sensitive to
mechanical loading, fibroblasts are responsible for transforming mechanical signals
into altered gene expression. Once experienced an altered mechanical environment,
fibroblasts proximal to the myotendinous junction signal for increased collagen pro-
duction to help in the development and maintenance of tendon. Laminins and talins
are also produced to link the ends of muscle fibers to the basement membrane at the
myotendinous junction [6]. A recent study by Archile-Contreras et al. showed that
fibroblasts isolated from different muscle types possess distinct in vitro growth
capability and express different levels of MMP-2 [5]. In this animal study, fibro-
blasts cultured from semitendinosus and sternomandibularis muscles showed higher
growth capacity compared to that of longissimus dorsi. Active MMP-2 expression
was highest in semitendinosus fibroblast cultures and lowest in sternomandibularis
fibroblast cultures. The mechanical properties and collagen content were not ana-
lyzed in these muscles; therefore, these observed differences could not be correlated
with muscle function or ECM turnover.
Satellite cells (SCs) are shown by Guerin et al. to be capable of synthesizing and
secreting MMP-2 and MMP-9. This study suggested the skeletal muscle cells could
participate in remodeling the ECM during myogenesis and the regeneration of skel-
etal muscle through assisting MMPs’ migration through the ECM to muscle injury
site [7]. A recent study by Hoedt et al. even showed that myogenic differentiation
factor (myoD) could be induced by erythropoietin (Epo) treatment to prevent apop-
tosis of satellite cells in murine and in vitro models. In addition, satellite cell content
in type II myofibers as well as the content of MyoD+ SCs increased through endur-
ance training. Epo receptor mRNA expression in human SCs also suggested that
Epo treatment could regulate human SCs in vivo [8]. Crameri et al. showed that a
single bout of high intensity exercise could induce SCs re-entry into the cell growth
cycle though a single bout was not sufficient for the SCs to undergo terminal dif-
ferentiation [9].
40 Y.-H. Chao and J.-S. Sun
Skeletal muscle collagens, which accounts for 1–10% of muscle dry weight mass,
is the major structural protein in skeletal muscle ECM. In the various types of col-
lagens, type I and III predominate in adult endo-, peri-, and epimysium with type I
being the predominant collagen in perimysium and type III evenly distributed
between endomysium [10]. Type V collagen forms a core for type I collagen fibrils
in peri- and endomysium [11]. Type XII and XIV collagen are fibril-associated col-
lagens with interrupted triple helices (FACITS) localized primarily to perimysium
and appeared to link fibrillary collagen to other ECM components without precisely
known function [12]. Muscle basement membrane consists mainly of type I colla-
gen network though type VI, XV, and XVIII are also present [13]. Types XV and
XVIII are multiplexins that can bind growth factors and assist in linking the base-
ment membranes of glycoproteins and endomysium [14]. The basement membrane
and endomysium are closely connected, involving in the transmission of force from
the myofiber to the tendon [15].
Many of the proteoglycans (PGs) in muscle ECM belong to the family of small
leucine-rich proteoglycans (SLRPs), including decorin, biglycan, fibromodulin, and
lumican with decorin and biglycan being the most abundant. Other PGs comprised
approximately 30% in muscle ECM includes the heparin sulfate PGs collagen
XVIII, perlecan, and agrin which bind to growth factors. Growth factors can be
stored and released by negatively charged glycosaminoglycans (GAGs) in the base-
ment membrane and endomysium surrounding muscle fibers. Enzymes in the ECM
cleave the GAG chains with their associated growth factors allowing interaction in
cell signaling and mechanotransduction [16, 17].
Comprising the major structure of ECM, collagen and proteoglycans interacts
uniquely to maintain the structure and organization of the matrix. Decorin, the
major PG in the perimysium, has been proposed to be a regulator of type I collagen
fibrillogenesis due to its binding interaction with type I collagen. It inhibits the lat-
eral growth of collagen fibrils through its core protein presumably. Biglycan also
binds to type I collagen in a similar way with decorin. The binding and the location
of collagen crosslinks might be key factors determining the structural organization
of the ECM since collagen fibril diameter in tendon becomes irregular in the absence
of these two materials. In mice absent of biglycan, mild muscular dystrophic pheno-
type is found implicating that these PGs are vital in maintaining normal tissue func-
tion and mechanical properties [18].
Various glycoproteins in the ECM functions in linking type IV collagen (base-
ment membrane) and sarcolemma (muscle fiber membrane). Laminins are bound by
intergrins and α-dystroglycan where integrins are bound to fibronectin [13].
Laminins are also bound to type IV collagen directly and indirectly linked to fibro-
nectin or nidogen. These glycoproteins interact to provide lateral force transmis-
sions from the myofiber, and with the network formed with type IV collagen, form
the basic structure of basement membrane [19].
Matricellular proteins in skeletal muscle include osteopontin, secreted protein
acidic and rich in cysteine (SPARC), thrombospondin, and tenascin-C. Osteopontin
2 Biomechanics of Skeletal Muscle and Tendon 41
possesses cytokine-like functions that appears during muscle regeneration but sel-
dom observed in normal skeletal muscle. It is typically secreted by inflammatory
cells and may also be secreted by myoblasts [20]. SPARC has been observed to bind
collagen and accompany its interaction in extracellular matrix [21]. The role of
thrombospondin is assumed to prevent excessive capillary formation in skeletal
muscle ECM as evidenced in the mice model [22]. Tenascin-C is indicated to play
a role in maintaining neuromuscular junction architecture by binding to perlecan
and agrin [23]. The major function of these proteins is not structural, but they are
vital in ECM signaling and maintaining ECM organization.
Skeletal muscle ECM composition and its mechanical properties are regulated
by matrix remodeling enzymes. Matrix metalloproteinase (MMP) levels are gener-
ally low in uninjured muscle. In this, MMP-2 and MMP-9 are gelatinases expressed
in muscle that degrade type IV collagen, fibronectin, proteoglycans, and laminin.
Collagenases MMP-1 and MMP-13 degrade types I and III collagens. Membrane
type I MMP not only activates MMP-2 but also functions proteolytically near mus-
cle cell surface. Tissue inhibitors of matrix metalloproteinases (TIMPs) 1–3 bind to
active MMPs or stabilize inactive MMPs to inhibit their enzymatic activities.
Through these enzymes, ECM of skeletal muscle is strictly regulated [13].
To define ECM mechanical properties, Meyer et al. first showed in their study
that isolated muscle fiber stress–strain behavior was linear, and that of muscle bun-
dle was nonlinear. To further differentiate and distinguish the source of nonlinearity
(ECM itself vs. numerous individual muscle fibers with different linear material
properties), numerous single fibers were isolated from ECM and grouped into a
bundle to compare among groups of multiple muscle fibers with and without
ECM. The results showed that the modulus of a fiber group was nearly the same as
that of single fibers while the modulus of bundle was about five times higher. These
results demonstrate that ECM is inherently stiffer than muscle fibers and its material
properties are highly nonlinear [24].
1.2 Passive Muscles
1.2.1 Elastic Behavior
The property of elasticity has a central role in a diverse range of physiological and
pathophysiological processes in skeletal muscle. Skeletal muscle is anisotropic due
to its microstructural arrangement. Its structure is divided into many levels—epimy-
sium, perimysium, and endomysium, as stated. Each muscle fiber contains actin,
myosin, and titin filaments. According to the sliding filament theory, muscle con-
traction occurs when actin and myosin interacts with a cross-bridge, and consume
one ATP (adenosine triphosphate) per cross-bridge cycle.
Elasticity describes a material property in which the material returns to its origi-
nal shape after stress applied and then removed. Muscle elastic behavior is nonlin-
ear. There are three main features to its viscoelasticity: First, when the muscle is
42 Y.-H. Chao and J.-S. Sun
stretched and relaxed, the tension decreases over time until the muscle reaches a
new steady state (stress relaxation). Second, when the muscle is stretched with a
constant tension, the length of muscle increases over time until it reaches a new
steady state (creep). Third, muscle exhibits different length–tension relationship
curve if repeatedly stretched. This is called “effect of cyclic loading” [25].
A relaxed skeletal muscle, referring to the passive muscle, will act as a deform-
able body when stretched beyond the resting length. However, the muscle deforma-
tion in response to the stretching does not follow Hooke’s law, which means it is not
linear. The passive muscle elasticity is mainly attributed to the connective tissues
content within the muscle [26]. The more proportion of the connective tissue within
the muscle, the stiffer the muscle will become [27]. Thus, lower extremity muscles,
containing more intramuscular connective tissue, are usually stiffer, compared to
the muscles from upper extremity. In addition, the muscles derived in vitro is usu-
ally shorter than the natural muscle length in vivo because of the lack of muscle tone
from neural supply and the difference in intrinsic mechanical properties.
When muscle is stretched to more than its resting length, it will exhibit passive
resistance to the stretching force without any metabolic energy requirement. The
resistance to deform is mainly from the following three structural components: con-
nective tissue within and around the muscle, intramuscular cross-links, and the non-
contractile proteins [26, 28]. The connective tissues provide general support of the
muscle structure. The binding sites of the cross-bridges between the actin and myo-
sin filaments provide the resistance to stretching during attached state. The giant
protein molecules such as titin help to maintain the integrity of sarcomere. By keep-
ing the myosin in the center between the Z discs, the proteins form the elastic con-
nections of the myosin and the Z-line. The part close to the fiber ends of the
sarcomere is less deformable than that at other sites. The length of the sarcomere
can be extended to a maximum of 3.5–4.0 μm [29].
However, from a macro perspective in joint motion, the changes in muscle–ten-
don complex length are much more than in the muscle length itself because the
tendons will generate little resistance to stretch in the toe region of the stress–strain
curve. On average, tendon changes account for almost more than half of the mus-
cle–tendon complex length changes [30]. Moreover, the passive resistance in joints
can be the interaction results of two components, including the joint angle and the
displacement. The passive resistance produced regarding the joint angle is negligi-
ble in the middle range of the motion, but significantly increase around the end
range of the motion [26]. The relationship between the passive resistance and the
joint angle is presented as the following equation:
( )
k e l (α −α0 ) − 1 , if α ≥ α
T =
0
0, if α < α 0
where T refers to the passive joint torque, k and l are constants, α is the joint angle,
and α0 is the joint angle threshold; joint angles below this value are slack and do not
produce much passive resistance. On the other hand, the antagonist muscle, such as
2 Biomechanics of Skeletal Muscle and Tendon 43
the triceps brachialis when performing elbow flexion, will not contribute much
resistance to the deformation during joint motion. Although the antagonist muscle
does not activate, a little tension exerted by the muscle tone still contains energy for
elastic deformation, which stores some space for structural extensibility without
generating too much resistance to deformation.
1.2.2 Viscoelastic Behavior
Passive muscles are viscoelastic material that exhibit both viscous and elastic char-
acteristics when subjected to tensile and compressive deformation [31].
Viscoelasticity is the stress and strain experienced in the passive muscle dependent
on the rate of loading; hence, the timing of the force application affects the strain
response of the passive muscle. The viscoelasticity displayed during tensile loading
is contributed by the passive properties of the protein titin [32, 33]. Lengthening
velocity affects the passive properties of muscle fibers significantly [34]. As length-
ening rate increases, the peak transient stress within the muscle fiber also increases,
even at very slow velocities. In passive muscles, a high rate of stretch results in a
higher stiffness than a slow stretch. This is one of the reasons that slow stretching
exercises are capable of minimizing the increase in force in the muscle–tendon unit
for a given amount of stretch.
There are other important properties of viscoelastic materials, including creep,
stress relaxation, and hysteresis. Hysteresis is the phenomenon that a stretched pas-
sive muscle returns to its pre-stretched length. It is found when length–tension char-
acteristics of passive muscle differ during loading (imposition of an external force)
and unloading response [35]. The hysteresis area between the loading and unload-
ing represents the amount of mechanical energy loss (dissipated to heat) in the
recovery from that stretch.
The more mechanical energy dissipated, the more obvious the hysteresis phe-
nomenon. In general, the relation between hysteresis and muscle stretch speed (or
rhythmic deformation frequency) can be presented as:
E = kω n
where E is the dissipated mechanical energy (the hysteresis area), ω is the oscilla-
tion frequency, and k and n are empirical constants. If n = 1, the dissipated energy is
proportional to velocity, and when n = 0, the dissipated energy is velocity indepen-
dent. In the latter case, the resistance acts like friction and is the same at any veloc-
ity. In human joints, the exponent n is typically close to zero. Therefore, muscle
hysteresis is relatively independent of oscillation frequency compared to “classic”
viscoelastic bodies, such as rubber. This indicates that the amount of mechanical
energy converted to heat is relatively independent of the rate of muscle length
change. Recent study has examined the effects of aging and gender on muscular
viscoelastic characteristics in athletes [36]. Increased tone and muscular stiffness
are found associated with aging, which can potentially lead to increased risk of falls,
44 Y.-H. Chao and J.-S. Sun
µ = ρν 2 λ 2
where μ is the shear modulus, ρ is the density of the tissue, v is frequency of the
vibration, λ is the local wavelength.
Elastography is a newly developed image-based technique for measuring passive
mechanical properties of skeletal muscles. Using MRI or ultrasound, stiffness and
elastic properties are determined based on one of the following two principles: (1)
For a given stress (force), stiffer tissue strains (deforms) less. (2) Mechanical waves
(specifically shear waves) travel (wave propagation) faster through stiffer tissue. In
vivo methods for measuring muscle mechanical properties are developing. There
are inverse finite analysis (FEA) for determining muscle mechanical properties in
human [42] and 3-in-1 Whole Animal Muscle System (Aurora Scientific) for mea-
suring in vivo, in situ, and in vitro muscle function. However, there are still some
uncertainties and limitations to these methods such that further studies are needed.
Understanding the normal mechanical properties of muscle is beneficial to differen-
tiate the abnormal pattern and figure out the problems. This must be especially
2 Biomechanics of Skeletal Muscle and Tendon 45
1.3 Active Muscles
The relationship between muscle force production and movement speed has been
described by Hill [43]. As pushing and lifting speed varies with the mass of the
object under fixed amount of force. Equation established by Hill simulates the ther-
mic generation during muscle contraction. This formation is: (v + b)
(F + a) = b(F0 + a), where F means the tension in the muscle, v means the velocity
of the contraction of the muscle, a is the Hill’s coefficient which correlated with the
heat generated during muscle shortening, and F0 means the tension of the muscle
when the contraction velocity is zero (isometric contraction).
In this model, where a and b are constant, the tension of the muscle and the
velocity of the contraction are inversely proportional. In fast contraction, the tension
generated by the muscle would be less than that in slow contraction under the same
environmental setting. While in vivo, the environment varies and influenced by
many factors. So, Hill-type model has been widely studied and modified into differ-
ent models implicated in different situations for more accurate simulation through-
out the years; however, the accuracy of the model in vivo remains unknown [44].
Biewener and his colleagues demonstrated a two-element Hill-type model in goats’
gastrocnemius muscles with EMG and found this model more accurate at predicting
muscle forces [44]. Further research to investigate more accurate models to simulate
the muscle force–velocity relationship within different situations is required since it
is multifactorial.
Features of muscle force–velocity in different types of muscle contractions are
distinguished. In concentric contraction, the maximal force production decreases as
the muscle contraction velocity increases. In isometric contraction, the length of the
muscle stays unchanged. Therefore, the velocity of muscle contraction is zero. The
eccentric contraction means the length change of the muscle is positive during mus-
cle contraction. The amount of force production increases while the contraction
velocity increases, which is converse to the situation in concentric contraction. The
maximum force generated by the eccentric contraction is about 1.5 times the value
of isometric contraction, and the plateau speed is also higher on isolated frog mus-
cles [45]. However, the experiment showed that the phenomenon did not have the
same effect on voluntary eccentric contraction on human body. In addition, EMG
activation did not change much while the eccentric velocity changes; it is even
lower than concentric contraction. Westling and his colleagues thought that might
be due to a protective neural inhibitory mechanism from too much tension on the
muscle [46]. Voukelatos et al. used electrical stimulation to train quadriceps mus-
cles for 3 weeks and found the overall torque output in eccentric contraction
46 Y.-H. Chao and J.-S. Sun
increases [47]. The results might indicate that the protective mechanism is change-
able. However, the enhancement of the peak value of voluntary eccentric contrac-
tion did not correlate with the increasing of angular velocity, which, to some extent,
might be due to the protective mechanism, though the mechanism remained unclear.
Typically, Hill-type model contains three elements: a contractile element with
force-length and force–velocity relations, a serial and a parallel elastic element. For
simulating the proper biomechanics in eccentric contraction, based on Hill-type
model, Haeufle et al. established a four-element model composed of a contractile
element, a parallel elastic element, a series elastic element, and a serial damping
element. This model considered serial damping and eccentric force–velocity rela-
tion. Thus, it is thought to be more suitable for simulating eccentric contraction in
human movements [48].
The decreasing of isometric force, maximum velocity of muscle shortening, and
an increase curvature of the force–velocity relationship are three main factors con-
tributing to the power loss in mammalian muscles after fatigue. The underlying
mechanism of muscle physiologic change needs further exploration. Jones [49]
reviewed recent articles and found there were two main discussion aspects: First,
the relationship between excitation-contraction coupling and force, and the other is
the slowing of contractile properties [50]. The contraction velocity was found slow-
ing down when muscle was fatigued comparing to the same force output generated
by non-fatigue muscle. On the other hand, under the same amount of velocity, the
force output was also decreased [51].
Muscle fascicle length is the other way of measuring muscle force. Arnold et al.
stated that the length and velocity of muscle fibers can have a large effect on muscle
force generation [55]. As muscle fibers increase in length, the number of sarcomeres
added to the ends of the myofiber increases. With longer muscle fascicle length,
more sarcomeres with cross-bridges produce more force.
Besides muscle growth with age, the influence of exercise on muscle plays an
important role between muscle size and muscle strength. In general, immobilization
can reduce muscle size and mass within 1–2 weeks. Kawakami et al. found that
subjects who performed isometric and bilateral leg extension exercise every day
after bed ridden for 20 days suffered less decrease in muscle strength and fiber
cross-sectional area [56]. The fiber cross-sectional area decreased in control group
by −7.8% while in exercise group it showed a lower tendency of −3.8% decrement.
This implies that exercise can reduce muscle atrophy and influence muscle size.
For more accurate evaluation of the change in muscle circumference (muscle
size), fiber cross-sectional area is usually employed. It can be measured using
anthropometric measurements or magnetic resonance imaging (MRI), with the lat-
ter being more accurate [53]. Therefore, MRI becomes the major tool for measuring
the fiber cross-sectional area.
Body movements depend on the contraction of muscles and the consequent motion
of the bone moving through the joint. When muscle contracts, the force produced
acts on muscle attachment (usually attached to bone). After receiving the force, the
bone starts moving around the joint. The relation between moment arm and force
can be simplified in the equation: T = F × R (Fig. 2.1), where T means the torque, F
means the force (N), R means movement arm, the perpendicular distance from the
pivot (rotation center) to the line of force F.
T = F×R
of force vector (F) and the position vector (r), which is from pivot to any point along
the line of F.
Mt = F × r
Force vector (F) could be divided into magnitude force (f) and unit force
vector (u).
F = fu
Mt = fu × r = f ( u × r )
Moment arm vector (d) comes from the cross of unit force vector (u) and posi-
tion vector (r). In this equation, moment arm vector (d), also called normalized
moment vector, means moment of force vector per unit scalar of force acts on the
bone. The direction of moment arm vector (d) can be defined by right-hand rule.
When circling the right hand second to fourth finger from the direction of position
vector (r) to the direction of unit force vector (u), the direction of the right-hand
thumb is the same as the direction of vector d.
In the muscle movement model, the magnitude of moment arm vector (d) was
equal to the perpendicular distance from the pivot (joint center) to the line of force.
However, when the force line was not perpendicular to the position vector (r), the
magnitude of moment arm vector (d) is equal to r sin θ, θ means the angle between
vectors r and u (when the force line is perpendicular to r, θ is 90° and sinθ = 1). In
the below picture, vector r and u are on the same plate o. d is moment arm vector.
As the cross production of r × u, moment arm vector d is perpendicular to the r and
u simultaneously. The direction of d is counterclockwise which can be defined by
right-hand rule (Fig. 2.2).
Mt = fd
d = (r × u )
d = r sin θ
When it comes to human movement, XYZ 3D system can be used to analyze the
muscle and joint action. Below is an elbow flexion example. Take elbow joint as a
center of 3D model, the movement arc produced by brachialis on humerus and ulna
bone is then defined. rA and rB are the position vectors of the brachialis origin and
insertion. F is the contraction force line and direction of the brachialis. u is the unit
force vector of F. d is the moment arm vector of the brachialis on elbow joint. d is
the magnitude of moment arm vector and it is equal to the length of the shortest
distance from center (elbow joint) to the brachialis (Fig. 2.3).
u can be calculated by brachialis position vector (rA − rB) and the length of
brachialis (|rA − rB|).
--- ---
- rA − rB
u = --- ---
rA − rB
the muscle with longer movement arm at the same shortened length of muscle con-
traction (Fig. 2.4).
In the figure, θ1 is the angle produced by the contraction of muscle 1. When the
muscle is relaxed, the bone is lying on the X-axis (X = 0). The blue line is the bone
situation when muscle 1 shortens (half of initial length). The yellow line is the bone
situation when the muscle 2 shortens (half of initial length), which has a longer
movement arm. Under the same shortening ratio (half of initial length in figure), θ1
is larger than θ2. Thus, if muscle 1 and muscle 2 have similar shortening speed,
muscle 1 can induce more angular excursions than muscle 2.
However, the interaction between the muscle force and the movement arm length
should be considered when discussing muscle function on joint movement in real
life. The force produced by muscle relies on the type and the number of muscle
fiber. In general, the muscle with more muscle fiber can produce larger force with
contraction. Muscle types can be morphologically divided into parallel and pennate
types. Parallel type muscle has liner muscle fiber. Parallel type has two subgroups,
fusiform and strap type. Because the parallel type muscle is composed of long
fibers, they can shorten more than the pennate muscle. More shortened length is
able to induce larger angular excursion. However, the pennate type muscle is easy
to induce large force in contraction. The movement arm could be confirmed with
anatomical analyzation or the calculation of movement arm vector.
The method of measuring movement arm can be simply divided into geometric
models and functional model. In early studies, geometric model was employed
using cadaver and anatomical approach that may not apply to the living models and
their complex movements. With technological advancement, high performance
computer, and precise imaging system (X-ray, MRI), the measurement of move-
ment arm improved. Thus, 3D geometric models and imaging geometric models
2 Biomechanics of Skeletal Muscle and Tendon 51
occurred. Researchers can observe muscle movement in the living situation, and
calculation through the computer software can be done. In functional model, the
actual movement arm is not important. The effective moment arm could be defined
through calculation of the equation. The effective moment arm could be defined
through calculation from the equation, Moment of Force = Force × Moment Arm.
Functional model applies when the joint type, muscle structure, muscle length, joint
angle are too complex to measure by geometric model.
The muscle moment arm vector is important in studying biomechanics and kine-
siology. Combining the muscle force, contraction speed joint angle, and muscle
moment arm human body movement can be analyzed precisely and in detail.
Advance theory of biomechanics and kinesiology could be thus derived.
Motor unit is considered the functional unit of the neuromuscular system which
work together to coordinate the contractions of a single muscle. A motor unit con-
sists of a motor neuron and the skeletal muscle fibers innervated by that motor
neuron’s axonal terminals [57]. A group of motor units within a muscle is called
motor pool. In human, there are no direct measures of the number of motor neurons
innervating any muscle [58]. It can be counted by anatomical [59] or electrophysi-
ological [60] method, but with some error. The number of muscle fibers within a
single motor unit varies both within a particular muscle and from muscle to muscle
as well. Muscles that act on the larger body masses usually contain more muscle
fibers and hence more motor units [57]. For example, triceps brachii muscle has
more muscle fibers in each motor unit than brachioradialis muscle.
Muscle produces force by regulating recruitment of motor units and by modulat-
ing the firing rates of recruited motor units [61]. Force production is controlled by
three neuromuscular mechanisms: the number and type of recruited motor unit and
their firing rate [62]. Increasing speed, force, or duration of movement involves
progressive recruitment of motor units with larger size and higher activation thresh-
olds. In 1965, Henneman et al. proposed a size principle of motor units recruitment
[63]. Neurons within the central nervous system vary widely in size. The larger cell
has larger surface area, and it should have some correlation with function. Finally,
they found the motor units were recruited from the smallest to the largest based on
the size of the load. As a result, the order is from slow twitch, low-force, fatigue-
resistant muscle fibers to fast twitch, high-force, and less fatigue-resistant muscle
fibers. The size principle is discussed in relation to the organization of the motor
neuron pools. “Fractionation,” “discharge zone,” “subliminal fringe,” “facilitation,”
and “occlusion” are redefined abbreviated terms of the sizes and excitabilities of the
cells in a pool. Cell size is assumed to be the determinant of the overall frequency
of discharge or usage of a motor neuron [63].
The relation between the recruitment and firing rate of motor units is organized
in an inverse hierarchical structure—the onion skin scheme [61]. Hence, the firing
rates of earlier-recruited motor units have a greater value than those of the
52 Y.-H. Chao and J.-S. Sun
later-recruited motor units at any time and force during an isometric muscle con-
traction [64–66]. Also, the same pool of all the motor units of a muscle receives a
common excited signal, known as common drive [64], and modulates their firing
rate in unison. The later-recruited, higher-threshold motor units have shorter-dura-
tion, higher-amplitude force twitches compared with the earlier-recruited, lower-
threshold motor units. The higher threshold motor units were driven by lower firing
rate because of the nature of fatigue. It seems that the neuromuscular system is
designed to optimize some combination of force and duration over which the force
is sustained. The firing rate has a large range from very low frequencies to produce
a series of single twitch contractions to high frequencies to produce a fused tetanic
contraction. In general, there will be a 2 to 4-fold change in force. The firing rate of
each individual motor unit increases with increased muscular effort until a maxi-
mum rate is reached [67].
As each individual level of analysis of the neuromuscular system structure has a
unique frequency, there are also robust relations in the frequency domain between
different levels [68]. Elble and Randall found a strong coherence between force
output and EMG at 8–12 Hz, and they also found coherence at a similar frequency
between single motor units and EMG [69]. Farmer et al. have found coherence
between motor units in 1–12 and 16–32 Hz bandwidths during volitional force
output [70].
Skeletal muscles are categorized into: (1) slow oxidative fibers, (2) fast oxidative–
glycolytic fibers, and (3) fast glycolytic fibers, in terms of energy utilization. They
can also be classified as type I fibers, type IIA fibers, and type IIB fibers, respec-
tively. Most skeletal muscles contain a mixture of all three fiber types. Their propor-
tions vary with the typical action of the muscle. Each type of muscle fiber harbors a
unique feature. Slow oxidative fibers are the smallest in diameter and the least pow-
erful type of muscle fibers. Fast oxidative–glycolytic fibers are intermediate in
diameter between the other two types of fibers. Fast glycolytic fibers are the largest
in diameter and produce most power. Other characteristics include myoglobin con-
tent, color, contraction velocity, method for generating ATP, fatigue resistance,
order of recruitment, and primary functions [71].
The above classification classifies muscle fiber types as being fast or slow based
on the speed of shortening and utilization of aerobic/anaerobic cellular respiration.
Muscle fibers can also be typed using two other different methods: (1) histochemi-
cal staining for myosin ATPase and (2) myosin heavy chain isoform identification
[72]. Classification for myosin ATPase staining, which based on staining intensities
and differences in pH sensitivity, leads to 7 muscle fiber types. According to the
speed of muscle contraction (from the slowest to the fastest): types I, IC, IIC, IIAC,
IIA, IIAB, and IIB. Difference of myosin heavy chain isoform in muscle is also used
to distinguish different muscle type. Although the human genome contains at least
10 genes for myosin heavy chains, only three are expressed in adult human limb
2 Biomechanics of Skeletal Muscle and Tendon 53
muscles [73]. MHC I, MHC IIA, and MHC IIB correspond to the isoforms identi-
fied by the speed of muscle shortening as types I, IIA, and IIB, respectively.
Skeletal muscle can be typed into different categories in different ways. However,
skeletal muscle not only changes in size in response to demands, but also converts
from one type to another (muscle fiber plasticity). Fiber conversions between type
IIB and type IIA are the most common, but type I to type II conversions are possible
in pathologic conditions. Aging can also influence the conversion between the pro-
portion of muscle fiber type. Older muscle was found to have a greater percentage
of fibers that co-express MHC I and MHC IIA (28.5%) compared with younger
muscle (5–10%) [74]. Exercise, the common prescription in physical therapy, is the
other way to change the proportion of muscle fiber type.
2 Tendon
Tendon cells, also named tenocytes, are the foremost fibroblast-like cells reside
between either the intrafascicles or the interfascicles. With a highly developed endo-
plasmic reticulum, tenocytes are responsible to sense and respond to the changes of
the microenvironment, including synthesis and degradation of the extracellular
matrix (ECM) [77]. Although tendon cells seem to have a similar morphology, the
54 Y.-H. Chao and J.-S. Sun
diversity of tenocytes is classified into several types according to their location and
cell morphology. The first type is the “chondrocytic” type. The chondrocytic type
presents in the entheses, where tendon attaches to bone. This type of tenocyte is
characterized with round nuclei with the ability to produce chondrocyte-like matrix,
such as type II collagen and aggrecan [78, 79]. The second type is the “external”
type. The external-type tenocytes are more similar to the synovial cells; they reside
in epitenon and interfascicles of endotenon and paratenon. Thus, they can provide
adequate lubrication to facilitate the gliding of collagen bundles [80]. Another type
is the “internal” type. The internal-type tendon cells can be found between the col-
lagen fibers; they have fusiform morphology and a spindle-shaped nucleus that are
responsible for producing the collagenous and non-collagenous ECM [80, 81]. Till
now, the role of different cell populations on tendon healing is still in debate. After
laceration of the flexor profundus tendons from adult monkeys, tenocytes from the
epitenon first migrated and proliferated to the wound site at day 4 while the response
of cells from the endotenon was postponed to day 9. Both cells demonstrated the
ability to synthesize type I procollagen [82]. Gelberman et al. revealed the epitenon
fibroblasts migrated to the injured spot and removed the cellular debris and collagen
fragments through phagocytosis while the collagen anabolism occurred within the
endotenon cell layer [83]. Recently, Cadby et al. indicated that both cells from peri-
tenon and endotenon had multi-differentiated potency; the peritenon-derived cells
migrated and proliferated more quickly with increased expression of alpha-smooth
muscle actin (α-SMA), a specific gene marker of myofibroblast [84]. From the
above information, tendon is consisted of multiple fibroblasts with different mor-
phologies and biochemical characteristics, and it has the ability of intrinsic healing.
In this decade, tendon stem/progenitor cells (TSPCs) were first named and iso-
lated from human and mouse tendons. These cells account for only 3–4% of total
cells and possess divergent characteristics compared with tendon cells. TSPCs har-
bor several criteria of stem cells, including clonogenicity, multipotency, and self-
renewal. Besides differentiating into tenocytes, TSPCs have the differentiating
potential toward osteogenesis, adipogenesis, and chondrogenesis. Consequently,
TSPCs are also named tendon-derived mesenchymal stem cells [85]. Similar to ten-
don cells, TSPCs can be classified into several types depending on the designated
niches. In the study of Mienaltowski, tendon stem cells were isolated from the
Achilles tendon and the surrounding peritenon (paratenon and epitenon) of C57Bl/6
mice. Interestingly, stem cells from tendon portion had higher colony forming units
and a higher expression of tendon-related gene markers, such as tenomodulin
(Tnmd) and scleraxis (Scx). Stem cells from peritenon displayed a higher level of
endomucin (Emcn), a vascular endothelial marker, as well as Cd133 and Musashi-1
(Msi1), pericyte stem cell markers. These findings indicated that tendon tissue con-
tains separate progenitor pools with distinct features. Thus, different kinds of TSPCs
may play distinct role in tendon healing. For example, intrinsic healing is related to
the expression of tendon-related gene markers while extrinsic repairing is achieved
through recruiting the perivascular cells from peritenon [86]. Although the applica-
tion of TSPCs into the research of tendon regeneration has drawn much interest, the
use of TSPCSs still has several dilemmas. Due to their scarcity and the tendency to
2 Biomechanics of Skeletal Muscle and Tendon 55
lose their phenotype and function with passaging, several factors have been found
to maintain their stemness [87]. For instance, matrix composition, substrate confor-
mation, growth factor and cytokine supplement, mechanical loading as well as oxy-
gen tension may affect their differentiated trend [88]. Thus, more studies are needed
for further investigation of TSPCs in tendon pathology and tissue engineering.
2.1.2 Extracellular Matrix
In human body, collagen is the most plentiful protein with 25 subtypes. Among
them, fibrillar types I, III, V, and XI collagen are essential for providing the connec-
tive tissue with its original stiffness and strength [76, 89]. In tendon, collagen com-
poses 60–85% of tendon dry weight and type I collagen accounts for 95% of total
collagen [90]. Besides type I collagen, there is little and varied amount of other
collagens, including type III, V, VI, XI, XII, and XIV [76]. Type I collagen is a triple
helix structure which comprises two α1 chains and one α2 chain. Thus, it is encoded
by two genes: Col1a1 and Col1a2. During collagen synthesis, procollagen is trans-
lated and folded into triple helix in the rough endoplasmic reticulum and then trans-
ported to the Golgi apparatus. Next, procollagen is modified into bundles and
packed into secretory vacuoles and then secreted to the extracellular matrix [91].
Type III collagen accounts for the remaining 5% of total collagen. Pertaining to
the fibrillar type collagen, the structure of type III collagen is similar to type I col-
lagen, wherein the difference is that type III collagen is composed by homotrimeric
molecules; type I collagen consists of heterotrimeric units. Coinciding with type I
collagen, Type III collagen is well known for its regulatory role in type I collagen
fibrogenesis. The interaction between type I and III collagen modulates the diameter
of collagen fibrils. Through tendon maturation, type III collagen assembles and
gathers into the endotenon or tendon sheath [91]. Increased type III collagen was
observed during tendon regeneration [92, 93].
Proteoglycans are a category of glycoprotein consisting of a core protein to
which at least one glycosaminoglycan (GAG) chain is attached. PGs can be divided
into two classes. One is the small leucine-rich proteoglycans (SLRPs), such as deco-
rin (Dcn), biglycan (Bgn), fibromodulin (Fmod), and lumican. The other type is the
large PGs, including aggrecan and versican [94]. Decorin is the most plentiful PGs
in tendon, accounting for 80% of total PGs content. It is a horse-hoof like protein
constituted by three domains. An N-terminal region is attached by a single dermatan
or chondroitin sulfate side chain. A central region is composed of ten leucine-rich
repeats which harbor a binding site of other proteins, including type I, II, III, VI
collagen, and TGF-β, etc. A C-terminal region contains several cysteine residues
[94, 95]. The central portion binds to the collagen fibril with the GAG side chain
lying parallel or perpendicular to the axis of the collagen fibril. By doing so, the
GAG side chain can interact with the other side chain of another Dcn molecule
which binds to a collagen fibril nearby, connecting to adjacent collagen fibril with
an inter-fibrillar bridge [96]. This is the way that Dcn maintains the spatial arrange-
ment of collagen fibrils and regulates the diameter of collagen fibrils. Dcn−/− mice
56 Y.-H. Chao and J.-S. Sun
showed disability in the regulation of collagen fibril diameter with increasing pres-
ence of enormous fibrils. In the studies of Halper and Reed et al., the packing of
collagen fibrils was sabotaged, thus resulting in a decrease of tendon mechanical
properties. These findings suggested that Dcn regulated the diameter of collagen
fibrils through inhibiting abnormal lateral fusion of collagen fibrils [94, 95]. With
two chondroitin or dermatan sulfate side chains, biglycan shares a similar collagen
binding site with Dcn. Knockout of Bgn or Dcn leads to morphological defects of
collagen fibrils. However, the lack of Bgn losses its ability to suppress bone mor-
phogenetic protein-2 (BMP-2), resulting in ectopic tendon ossification [18, 94].
With keratin side chains, lumican and fibromodulin share the same binding site on
fibrillar collagen, different from the binding site of Dcn and Bgn [94, 96].
Glycoproteins are a variety of proteins which are covalently attached by carbo-
hydrate groups. The glycoproteins mainly found in tendon include tenomodulin
(Tnmd), tenascin-C (Tnc), collagen oligomeric matrix component (COMP), and
lubricin. As a potential candidate for tendon markers, tenomodulin is a type II trans-
membrane glycoprotein which highly presents in late developing and mature ten-
dons. Tnmd−/− mice demonstrated a decrease in tendon cell density. Also, a reduce
in cell proliferation was found without the presence of cell apoptosis. As a regulator
of collagen architecture and fibrogenesis, Tnmd−/− mice showed uneven fibrillar
surfaces and abnormally increased diameter of collagen fibrils [97]. Knockout of
Tnmd in TSPCs inhibited self-renew capacity, and tendon stem cells tended to cease
cell cycle inducing early cellular senescence [98].
As a hexameric extracellular matrix glycoprotein, tenascin-C (Tnc) is highly pre-
sented in the circumstances below. High concentration of Tnc was found during the
embryogenesis of the musculoskeletal system, containing tendon, skin, ligament,
muscle, articular cartilage, and bone [99, 100]. However, the Tnc−/− mice did not
express a particular phenotype and revealed an ordinary wound healing process in
skin and nerve tissues [101, 102]. Interestingly, upregulation of Tnc was discovered
in mechanosensitive tissues that transmit forces from muscle to bone, such as myo-
tendinous junction and osteotendinous junction [100, 103]. Therefore, the expres-
sion of Tnc is modulated by mechanical stimulus and has been postulated with the
increase of elasticity in the ECM [104].
2.2 Basic Mechanics
2.2.1 Elastic Behavior
Elasticity is the tendency of tissue to resist deformation with an applied force [105].
Tendon is made up of collagen fibrils with highly consistent alignment. The colla-
gen fibrils are the basic load-bearing units. Traditional material testing can be used
to test the elasticity of the tendon. By fixing an isolated tendon specimen to a mate-
rial testing machine, recording the loading of tensile stretch applied to the tendon
and the amount of tendon elongation under each load, the elasticity of the tendon
2 Biomechanics of Skeletal Muscle and Tendon 57
µ = c2 ρ
where μ is the shear modulus, c is the shear wave propagation velocity, and ρ is the
density of the tissue. The density of all soft tissue can be assumed to be 1000 kg/m3.
Ultrasound and magnetic resonance imaging (MRI) are imaging modalities that
can also be used to detect shear waves [108]. When force is imposed on a stiffer
tendon, it will shorten rapidly and bear less load. A stiffer tendon will transmit more
of the imposed force to the muscle, while a more compliant tendon will be more
prone to involuntary oscillations. Tendon stiffness is influential by diet. In an animal
experiment, the rats fed on diets containing advanced glycation end products
(AGEs) for 88 days had stiffer tendons than those in the control group. Glycation
has effect on collagen cross-linking, and this may result in change in integrity and
elastic property of tendon tissue [110].
The slack of the muscle–tendon unit may influence the elastic behavior of tendon.
Fukutani et al. applied supramaximal electrical stimulation at the tibial nerves of eight
young healthy male volunteers [111]. The joint torque (plantar-flexion) was signifi-
cantly larger when the ankle joint was positioned in 20° of dorsiflexion than posi-
tioned in 20° of plantar-flexion. However, the magnitude of Achilles tendon elongation
measured by ultrasound was significantly smaller in the 20° dorsiflexed position.
58 Y.-H. Chao and J.-S. Sun
Assuming a 20° dorsiflexed position, the influence of the slack is zero and the tendon
elongation was resulted from muscle force only; at 20° plantar-flexed position, 20%
of the tendon elongation was resulted from muscle force, and 80% of tendon elonga-
tion could be caused by elimination of slack due to different positioning [111].
Konow et al. investigated the lateral gastrocnemius muscle–tendon interaction
during landing, and found that the muscle–tendon unit lengthened significantly dur-
ing the last one-third of landing with increased range of motion of the joints, while
the muscle fascicles behaved isometrically during the first two-thirds of landing
without any observable muscle–tendon unit lengthening [112]. This result indicates
that during the first two-thirds of landing, the tendon lengthened rapidly, storing the
energy through elongation, and it recoiled, releasing the energy and lengthened the
muscle fascicles during the last one-third of the landing. In such situations, tendon
plays a role in power attenuation, which may be a protective mechanism that reduces
the risk of muscle fascicle damage during rapid lengthening [112].
Farris et al. recruited young male participants to observe the muscle–tendon
interaction during walking with constant speed and walking with acceleration. The
amount of triceps surae muscle fascicles shortening was larger in early stance phase
during the accelerative walking than during the constant walking, and the amount of
fascicle shortening was not consistent to the muscle–tendon unit length change.
This indicates that the energy produced by the fascicle shortening was stored in the
tendon through elongation. In other words, the contractile work was stored in series
elastic elements in the form of elastic energy [113]. The tendon then released the
energy for the movement of the desired tasks. In the task of two-legged hopping,
both adults and children presented maximal tendon excursion and minimal muscle
excursion at their preferred self-selected hopping speed, which is in consistent with
the optimal energy-saving strategy in theory [114]. This experimental result implies
that elastic behavior of tendon plays an important role in movement efficiency.
2.2.2 Viscoelastic Behavior
2.3 Mechano-responses of Tendon
2.3.1 Physiological Mechano-responses
p remature roosters [132]. These findings indicated the turnover of collagen enhance-
ment but a decrease in quality after strenuous endurance exercise of collagen fiber.
A 12-week uphill running exercise increased the expression of type III collagen and
insulin-like growth factor I (IGF-I). Type III collagen forms thinner and disorga-
nized fiber. Increased type III collagen can be found in tendon lesion site [133, 134].
IGF-I is an effective stimulator of collagen anabolism and cell proliferation [135,
136]. These results indicate that exercise promotes metabolism of the ECM while
the following mechanical consequences still need to be defined.
Several models were employed to mimic the mechanical stimulus during the
locomotion of the body segment. In addition, tendons were loaded and unloaded,
causing changes in the influx and extrusion of the interstitial fluid, and the intersti-
tial fluid flow toward the organized extracellular matrix with low permeability
[137]. Thus, fluid shear stress was used to mimic the shear force the tendon with-
stood. Shear stress at 1666 μm/s, approximately 0.02–0.1 Pa, increased the intracel-
lular calcium concentration. Combining shear stress and uniaxial tension strain at
4% enhanced the percentage of tenocytes exhibiting transient calcium response
[137]. Shear stress at 0.41 Pa reduced the expression of Col I, Col III and several
profibrotic factors in rat palmar flexor tendon cells. Shear stress also downregulated
TGF-β2, TGF-β3 and TGF-β receptors, except for TGF-β1. Increase in the expres-
sion of vascular endothelial growth factor (VEGF) as well as MMPs, and decrease
in the expression of TIMPs, MMP inhibitors, were found under shear stress
condition. These findings suggested shear stress had an antifibrotic effect and pro-
moted tendon catabolism [138].
Besides shear stress, tensile loading has been exerted to evaluate how tenocytes
regulated cell behaviors and homeostasis of the ECM in response to tensile strain.
Yang et al. investigated the effect of cyclic mechanical stretching on the prolifera-
tion and collagen mRNA expression and protein production of human patellar ten-
don fibroblasts under serum-free condition [139]. The role of TGF-β1 in collagen
production by cyclically stretched tendon fibroblasts was also investigated. The
results suggest that increase in the cellular production of collagen type I is at least
in part mediated by TGF-β1. Uniaxial cyclic strain at the parameter of 4%, 0.5 Hz
enhanced the expression of tendon-related genes in TSCs, such as Col1 and Tnmd,
while 8%, 0.5 Hz strain upregulated the expression of LPL, Sox-9, and Runx2,
which, respectively, represented a specific gene marker of adipocytes, chondro-
cytes, and osteocytes. The findings were in accordance with the in vivo treadmill
running model. Moderated treadmill running elevated the expression of tendon-
related genes, including Col1 and Tnmd. The expression of IGF-1 was enhanced
and TSCs proliferation increased with moderate exercise as well. Additionally,
intensive exercise induced tendon anabolism through the expression of Col1 and
Tnmd with the concomitant increase of non-tendon-related genes [140, 141]. This
suggested that proper mechanical loading promoted the maintenance of tendon phe-
notype and facilitated TSCs differentiation into tendon lineage.
On the contrary, the following studies show different types of tendons and different
tendon tissue response under unloading or detraining. Kubo et al. found that the ten-
don structure and collagen content of Achilles tendon showed gradual improvement
62 Y.-H. Chao and J.-S. Sun
2.3.2 Pathological Mechano-responses
It is well established that appropriate mechanical loads are beneficial to enhance the
mechanical properties of the tendon. However, excessive mechanical loads on ten-
dons may result in tendon injuries. Tendon injuries are classified into acute and
chronic injury. Lacerations and ruptures are two common acute injuries. As for the
chronic tendon injury, tendinopathy can be the representative [144].
Tendinopathy, which is usually termed tendinitis, tendinosis, and paratenonitis,
has been found to be a result of a failed healing response according to recent histo-
pathological studies [145]. It may cause degenerative changes with increased levels
of prostaglandin E2 (PGE2), deoxyribonucleic acid, and protein synthesis in vitro
within tendon [146, 147]. Every single microtrauma a tendon experienced may
accumulate over time and cause tendinopathy. Previous study has found that loading
on the tendon fibroblasts will increase PGE2 and leukotriene B4 expression. The
correlation of tendon breakdown and the enhanced activities of vascular endothelial
growth factor (VEGF) and matrix metalloproteinase (MMP) was evidenced in the
study of Schwartz [148]. When comparing the histological characteristics of tendi-
nopathy to those of the normal tendons, it shows an increased number of tenocytes
and concentration of glycosaminoglycans in the ground substance, and disorganiza-
2 Biomechanics of Skeletal Muscle and Tendon 63
tion and fragmentation of the collagen, and neovascularization [145]. Moreover, the
increased expression of type III collagen, fibronectin, tenascin C, aggrecan, and
biglycan are the molecular changes found in the patients with chronic Achilles ten-
dinopathy. These changes might be an adaptive response to changes in mechanical
loading [149].
Overloading might be the cause of tendinopathy. Human patella tendon fibro-
blasts underwent cyclic uniaxial stretching of strain conditions: 4%, 8%, 12% ran-
domly for 4 h and rested 4 h to observe the inflammatory response of mediators
PGE2 and LTB4. Results showed that, under higher stress loads (12%) PGE2 and
LTB4 levels were higher with significant difference. This may imply the possible
mechanism of overuse inflammatory tendinitis [150, 151]. In tendon cells, repeti-
tive, small-magnitude stretching, e.g., 4% strain at a frequency of 0.5 Hz, increased
Col I, TGF-β production and reduced inflammatory mediators through downregula-
tion of the expression of COX-2 and PGE2. Whereas large-magnitude stretching,
e.g., 8% strain at a frequency of 0.5 Hz, also increased Col I, TGF-β and MMP-1
production but had a pro-inflammatory effect [139, 152, 153]. The above studies
indicated change in tendon anabolism with excessive mechanical loading may have
a pathological inference on tendinopathy.
Insufficient mechanical loads, such as disuse or immobilization can also have some
effects on tendons [146]. Immobilization has been associated with decreased levels of
extracellular matrix protein expression, alterations in tenocyte m
orphology, and loss of
normal extracellular matrix architecture, resulting in impaired function and healing
capacity [154–158]. Deprived of stress, tendon tissues underwent dramatic changes in
tendon cell shape, cell number, and collagen fiber alignment, and eventually caused
tendon degeneration [155]. Stress deprivation-initiated metabolic alterations, such as
decrease in anabolic activities but increase in catabolic activities of the tendon matrix
are likely responsible for certain degenerative changes in tendons [159].
Non-physiological loading may induce tendinopathy as well, by altering TSPC
function and fate at both the proliferation and differentiation levels. Tendon stem/
progenitor cells (TSPCs) is responsible for preserving adequate tenocyte numbers
in the tissue throughout life and replenishing them after injury. Studies supported
that mechanical loading critically affects the fate of TSPCs. An in vitro study
showed that mechanical loading at physiological levels promoted TSPC prolifera-
tion and differentiation into tenocytes, while excessive levels of loading led TSPCs
to differentiate into non-tenocytes, such as adipocytes, chondrocytes, and osteo-
cytes, in addition to tenocytes [159]. An in vivo study using treadmill running fur-
ther found that tendons subjected to repetitive strenuous mechanical loading
produced high levels of PGE2, which was associated with decreased TSPC prolif-
eration and induced TSPCs to differentiate into adipocytes and osteocytes [160].
These studies suggest that non-physiological loading may induce tendinopathy by
altering TSPC function and fate at both the proliferation and differentiation levels.
Better understanding of these mechanisms may provide a new strategy for the pre-
vention and treatment of tendinopathy.
Aging accounts for tendinopathy as well which is termed “age-related tendi-
nopathy.” Morphologically, increased lipid deposition and ossification and PGE2
64 Y.-H. Chao and J.-S. Sun
2.3.3 Mechanotransduction
suggested that tendon cell responded to tensile mechanical stimuli through increas-
ing the intracellular calcium level via the SACs. Taken together, these studies sug-
gest that calcium is an important mediator in cellular mechanotransduction.
MAPKs constitute key steps in many intracellular signaling pathways. They usu-
ally function as final effectors of signal transduction pathways directly to activate
transcription factors in the cytoplasm and nucleus. MAPKs are serine/threonine-
specific protein kinases that respond to extracellular stimuli (mitogens) and regulate
various cellular activities such as gene expression, mitosis, differentiation, and cell
survival/apoptosis. MAPK is crucial for the conversion of mechanical load to tissue
adaptation inducing signaling from the cytosol to the nucleus. It is well described
that several cell types and subsets of MAPKs such as extracellular signal-regulated
kinase 1 and 2 (ERK1/2), MAPK-p38, and stress-activated protein kinases/c-jun
NH2-terminal kinase (SAPK/JNK) can be activated by mechanical stress, as well as
by lowered pH, growth factors, hormones, and reactive oxygen species [1]. ERK-
mediated pathways are mostly involved in proliferation and differentiation and gen-
erally considered anti-apoptotic. JNK and p38-signaling pathways are activated by
stress stimuli, many of which induce apoptosis, but in some cellular systems they
have been implicated in proliferation and differentiation as well. Each is proved to
have major roles in the regulation of intracellular metabolism and gene expression
and integral actions in many areas including growth and development, disease,
apoptosis, and cellular responses to external stresses [176]. For example, cyclic
strain resulted in an immediate activation of JNK in patellar tendon fibroblasts
which was regulated by a magnitude-dependent response and appeared to be medi-
ated through a calcium-dependent mechanotransduction pathway [177]. Our recent
study demonstrated that mechanical stretch with 8% strain at 1 Hz rate improves
tenocyte characteristics via reversing the fibroblast-to-adipocyte phenotypic transi-
tion induced by high glucose through inhibition of Akt and activation of ERK [178].
References
1. Kjaer M. Role of extracellular matrix in adaptation of tendon and skeletal muscle to mechani-
cal loading. Physiol Rev. 2004;84(2):649–98.
2. McCormick RJ. The flexibility of the collagen compartment of muscle. Meat Sci.
1994;36(1–2):79–91.
3. Purslow PP. The structure and functional significance of variations in the connective tissue
within muscle. Comp Biochem Physiol A Mol Integr Physiol. 2002;133(4):947–66.
4. Turrina A, Martinez-Gonzalez MA, Stecco C. The muscular force transmission system: role
of the intramuscular connective tissue. J Bodyw Mov Ther. 2013;17(1):95–102.
5. Archile-Contreras AC, Mandell IB, Purslow PP. Phenotypic differences in matrix
metalloproteinase 2 activity between fibroblasts from 3 bovine muscles. J Anim Sci.
2010;88(12):4006–15.
6. Chiquet M, Gelman L, Lutz R, Maier S. From mechanotransduction to extracellular matrix
gene expression in fibroblasts. Biochim Biophys Acta. 2009;1793(5):911–20.
7. Guerin CW, Holland PC. Synthesis and secretion of matrix-degrading metalloproteases by
human skeletal muscle satellite cells. Dev Dyn. 1995;202(1):91–9.
66 Y.-H. Chao and J.-S. Sun
30. Herbert RD, Moseley AM, Butler JE, Gandevia SC. Change in length of relaxed muscle
fascicles and tendons with knee and ankle movement in humans. J Physiol. 2002;539(Pt
2):637–45.
31. Zatsiorsky VM, Prilutsky BI. Biomechanics of skeletal muscles. Champaign, IL: Human
Kinetics; 2012.
32. Wang K, McCarter R, Wright J, Beverly J, Ramirez-Mitchell R. Viscoelasticity of the sarco-
mere matrix of skeletal muscles. The titin-myosin composite filament is a dual-stage molecu-
lar spring. Biophys J. 1993;64(4):1161–77.
33. Toursel T, Stevens L, Granzier H, Mounier Y. Passive tension of rat skeletal soleus
muscle fibers: effects of unloading conditions. J Appl Physiol (Bethesda, MD: 1985).
2002;92(4):1465–72.
34. Rehorn MR, Schroer AK, Blemker SS. The passive properties of muscle fibers are velocity
dependent. J Biomech. 2014;47(3):687–93.
35. Ramos J, Lynch S, Jones D, Degens H. Hysteresis in muscle. Int J Bifurc Chaos.
2017;27(1):1730003.
36. Gervasi M, Sisti D, Amatori S, Andreazza M, Benelli P, Sestili P, et al. Muscular visco-
elastic characteristics of athletes participating in the European Master Indoor Athletics
Championship. Eur J Appl Physiol. 2017;117(8):1739–46.
37. Ryan ED, Herda TJ, Costa PB, Walter AA, Hoge KM, Stout JR, et al. Viscoelastic creep in
the human skeletal muscle–tendon unit. Eur J Appl Physiol. 2010;108(1):207–11.
38. Knudson D. Fundamentals of biomechanics. J Sports Sci Med. 2007;6(3):384.
39. Proske U, Morgan DL, Gregory JE. Thixotropy in skeletal muscle and in muscle spindles: a
review. Prog Neurobiol. 1993;41(6):705–21.
40. Proske U, Tsay A, Allen T. Muscle thixotropy as a tool in the study of proprioception. Exp
Brain Res. 2014;232(11):3397–412.
41. Sakanaka TE, Lakie M, Reynolds RF. Sway-dependent changes in standing ankle stiffness
caused by muscle thixotropy. J Physiol. 2016;594(3):781–93.
42. Silva MET, Brandao S, Parente MPL, Mascarenhas T, Natal Jorge RM. Biomechanical prop-
erties of the pelvic floor muscles of continent and incontinent women using an inverse finite
element analysis. Comput Methods Biomech Biomed Eng. 2017;20(8):842–52.
43. Hill AV. The heat of shortening and the dynamic constants of muscle. Proc R Soc Lond B Biol
Sci. 1938;126(843):136–95.
44. Biewener AA, Wakeling JM, Lee SS, Arnold AS. Validation of Hill-type muscle models in
relation to neuromuscular recruitment and force-velocity properties: predicting patterns of
in vivo muscle force. Integr Comp Biol. 2014;54(6):1072–83.
45. Harry JD, Ward AW, Heglund NC, Morgan DL, McMahon TA. Cross-bridge cycling theories
cannot explain high-speed lengthening behavior in frog muscle. Biophys J. 1990;57(2):201–8.
46. Westing SH, Seger JY, Thorstensson A. Effects of electrical stimulation on eccentric and
concentric torque-velocity relationships during knee extension in man. Acta Physiol Scand.
1990;140(1):17–22.
47. Voukelatos D, Kirkland M, Pain MTG. Training induced changes in quadriceps activation
during maximal eccentric contractions. J Biomech. 2018;73:66–72.
48. Haeufle DF, Gunther M, Bayer A, Schmitt S. Hill-type muscle model with serial damping and
eccentric force-velocity relation. J Biomech. 2014;47(6):1531–6.
49. Jones DA. Changes in the force-velocity relationship of fatigued muscle: implications for
power production and possible causes. J Physiol. 2010;588(Pt 16):2977–86.
50. Allen DG, Lamb GD, Westerblad H. Skeletal muscle fatigue: cellular mechanisms. Physiol
Rev. 2008;88(1):287–332.
51. Ruiter CJ, Didden WJ, Jones DA, Haan AD. The force-velocity relationship of human adduc-
tor pollicis muscle during stretch and the effects of fatigue. J Physiol. 2000;526(Pt 3):671–81.
52. Pearson AM. Muscle growth and exercise. Crit Rev Food Sci Nutr. 1990;29(3):167–96.
53. Tonson A, Ratel S, Le Fur Y, Cozzone P, Bendahan D. Effect of maturation on the relationship
between muscle size and force production. Med Sci Sports Exerc. 2008;40(5):918–25.
68 Y.-H. Chao and J.-S. Sun
54. Krivickas LS, Dorer DJ, Ochala J, Frontera WR. Relationship between force and size in
human single muscle fibres. Exp Physiol. 2011;96(5):539–47.
55. Arnold EM, Hamner SR, Seth A, Millard M, Delp SL. How muscle fiber lengths and veloci-
ties affect muscle force generation as humans walk and run at different speeds. J Exp Biol.
2013;216(Pt 11):2150–60.
56. Kawakami Y, Akima H, Kubo K, Muraoka Y, Hasegawa H, Kouzaki M, et al. Changes in
muscle size, architecture, and neural activation after 20 days of bed rest with and without
resistance exercise. Eur J Appl Physiol. 2001;84(1–2):7–12.
57. Buchthal F, Schmalbruch H. Motor unit of mammalian muscle. Physiol Rev.
1980;60(1):90–142.
58. Fuglevand AJ. Mechanical properties and neural control of human hand motor units. J
Physiol. 2011;589(Pt 23):5595–602.
59. Feinstein B, Lindegard B, Nyman E, Wohlfart G. Morphologic studies of motor units in nor-
mal human muscles. Acta Anat (Basel). 1955;23(2):127–42.
60. McComas AJ, Fawcett PR, Campbell MJ, Sica RE. Electrophysiological estimation
of the number of motor units within a human muscle. J Neurol Neurosurg Psychiatry.
1971;34(2):121–31.
61. De Luca CJ, Erim Z. Common drive of motor units in regulation of muscle force. Trends
Neurosci. 1994;17(7):299–305.
62. Akataki K, Mita K, Watakabe M, Ito K. Age-related change in motor unit activation strategy
in force production: a mechanomyographic investigation. Muscle Nerve. 2002;25(4):505–12.
63. Henneman E, Somjen G, Carpenter DO. Functional significance of cell size in spinal moto-
neurons. J Neurophysiol. 1965;28:560–80.
64. De Luca CJ, LeFever RS, McCue MP, Xenakis AP. Control scheme governing concurrently
active human motor units during voluntary contractions. J Physiol. 1982;329:129–42.
65. De Luca CJ, Contessa P. Hierarchical control of motor units in voluntary contractions. J
Neurophysiol. 2012;107(1):178–95.
66. De Luca CJ, Hostage EC. Relationship between firing rate and recruitment threshold of
motoneurons in voluntary isometric contractions. J Neurophysiol. 2010;104(2):1034–46.
67. De Luca CJ. Control properties of motor units. J Exp Biol. 1985;115:125–36.
68. Sosnoff JJ, Vaillancourt DE, Larsson L, Newell KM. Coherence of EMG activity and sin-
gle motor unit discharge patterns in human rhythmical force production. Behav Brain Res.
2005;158(2):301–10.
69. Elble RJ, Randall JE. Motor-unit activity responsible for 8- to 12-Hz component of human
physiological finger tremor. J Neurophysiol. 1976;39(2):370–83.
70. Farmer SF, Bremner FD, Halliday DM, Rosenberg JR, Stephens JA. The frequency content
of common synaptic inputs to motoneurones studied during voluntary isometric contraction
in man. J Physiol. 1993;470:127–55.
71. Scott W, Stevens J, Binder-Macleod SA. Human skeletal muscle fiber type classifications.
Phys Ther. 2001;81(11):1810–6.
72. Roy RR, Talmadge RJ, Hodgson JA, Oishi Y, Baldwin KM, Edgerton VR. Differential
response of fast hindlimb extensor and flexor muscles to exercise in adult spinalized cats.
Muscle Nerve. 1999;22(2):230–41.
73. Andersen JL, Terzis G, Kryger A. Increase in the degree of coexpression of myosin heavy
chain isoforms in skeletal muscle fibers of the very old. Muscle Nerve. 1999;22(4):449–54.
74. Staron RS. Human skeletal muscle fiber types: delineation, development, and distribution.
Can J Appl Physiol. 1997;22(4):307–27.
75. Nickisch F. Anatomy of the achilles tendon. In: Nunley JA, editor. The achilles tendon: treat-
ment and rehabilitation. New York: Springer New York; 2009. p. 2–16.
76. Thorpe CT, Birch HL, Clegg PD, Screen HRC. Chapter 1—Tendon physiology and mechani-
cal behavior: structure–function relationships. Tendon regeneration. Boston, MA: Academic
Press; 2015. p. 3–39.
2 Biomechanics of Skeletal Muscle and Tendon 69
77. Liu W, Wang B, Cao Y. Chapter 14—Engineered tendon repair and regeneration. Tendon
regeneration. Boston, MA: Academic Press; 2015. p. 381–412.
78. Benjamin M, Ralphs JR. Fibrocartilage in tendons and ligaments—an adaptation to compres-
sive load. J Anat. 1998;193(Pt 4):481–94.
79. Smith RKW, Goodship AE. Chapter 2.3—Tendon and ligament physiology: responses to
exercise and training A2—Hinchcliff, Kenneth W. In: Geor RJ, Kaneps AJ, editors. Equine
exercise physiology. Edinburgh: W.B. Saunders; 2008. p. 106–31.
80. Banes AJ, Donlon K, Link GW, Gillespie Y, Bevin AG, Peterson HD, et al. Cell populations
of tendon: a simplified method for isolation of synovial cells and internal fibroblasts: confir-
mation of origin and biologic properties. J Orthop Res. 1988;6(1):83–94.
81. Goodship AE, Birch HL, Wilson AM. The pathobiology and repair of tendon and ligament
injury. Vet Clin North Am Equine Pract. 1994;10(2):323–49.
82. Russell JE, Manske PR. Collagen synthesis during primate flexor tendon repair in vitro. J
Orthop Res. 1990;8(1):13–20.
83. Gelberman RH, Manske PR, Vande Berg JS, Lesker PA, Akeson WH. Flexor tendon repair
in vitro: a comparative histologic study of the rabbit, chicken, dog, and monkey. J Orthop
Res. 1984;2(1):39–48.
84. Cadby JA, Buehler E, Godbout C, van Weeren PR, Snedeker JG. Differences between the cell
populations from the peritenon and the tendon core with regard to their potential implication
in tendon repair. PLoS One. 2014;9(3):e92474.
85. Bi Y, Ehirchiou D, Kilts TM, Inkson CA, Embree MC, Sonoyama W, et al. Identification of
tendon stem/progenitor cells and the role of the extracellular matrix in their niche. Nat Med.
2007;13(10):1219–27.
86. Mienaltowski MJ, Adams SM, Birk DE. Regional differences in stem cell/progenitor cell
populations from the mouse achilles tendon. Tissue Eng A. 2013;19(1–2):199–210.
87. Tan Q, Lui PP, Rui YF. Effect of in vitro passaging on the stem cell-related properties of tendon-
derived stem cells-implications in tissue engineering. Stem Cells Dev. 2012;21(5):790–800.
88. Gaspar D, Spanoudes K, Holladay C, Pandit A, Zeugolis D. Progress in cell-based therapies
for tendon repair. Adv Drug Deliv Rev. 2015;84:240–56.
89. Humphrey JD, Dufresne ER, Schwartz MA. Mechanotransduction and extracellular matrix
homeostasis. Nat Rev Mol Cell Biol. 2014;15(12):802–12.
90. Kastelic J, Galeski A, Baer E. The multicomposite structure of tendon. Connect Tissue Res.
1978;6(1):11–23.
91. Banos CC, Thomas AH, Kuo CK. Collagen fibrillogenesis in tendon development: cur-
rent models and regulation of fibril assembly. Birth Defects Res C Embryo Today Rev.
2008;84(3):228–44.
92. Ahmed AS, Schizas N, Li J, Ahmed M, Ostenson CG, Salo P, et al. Type 2 diabetes
impairs tendon repair after injury in a rat model. J Appl Physiol (Bethesda, MD: 1985).
2012;113(11):1784–91.
93. Eriksen HA, Pajala A, Leppilahti J, Risteli J. Increased content of type III collagen at the
rupture site of human Achilles tendon. J Orthop Res. 2002;20(6):1352–7.
94. Halper J. Proteoglycans and diseases of soft tissues. Adv Exp Med Biol. 2014;802:49–58.
95. Reed CC, Iozzo RV. The role of decorin in collagen fibrillogenesis and skin homeostasis.
Glycoconj J. 2002;19(4–5):249–55.
96. Thorpe CT, Birch HL, Clegg PD, Screen HR. The role of the non-collagenous matrix in ten-
don function. Int J Exp Pathol. 2013;94(4):248–59.
97. Docheva D, Hunziker EB, Fassler R, Brandau O. Tenomodulin is necessary for tenocyte
proliferation and tendon maturation. Mol Cell Biol. 2005;25(2):699–705.
98. Alberton P, Dex S, Popov C, Shukunami C, Schieker M, Docheva D. Loss of tenomodulin
results in reduced self-renewal and augmented senescence of tendon stem/progenitor cells.
Stem Cells Dev. 2015;24(5):597–609.
99. Kardon G. Muscle and tendon morphogenesis in the avian hind limb. Development
(Cambridge, England). 1998;125(20):4019–32.
70 Y.-H. Chao and J.-S. Sun
100. Jarvinen TA, Kannus P, Jarvinen TL, Jozsa L, Kalimo H, Jarvinen M. Tenascin-C in the
pathobiology and healing process of musculoskeletal tissue injury. Scand J Med Sci Sports.
2000;10(6):376–82.
101. Forsberg E, Hirsch E, Frohlich L, Meyer M, Ekblom P, Aszodi A, et al. Skin wounds
and severed nerves heal normally in mice lacking tenascin-C. Proc Natl Acad Sci U S
A. 1996;93(13):6594–9.
102. Erickson HP. A tenascin knockout with a phenotype. Nat Genet. 1997;17(1):5–7.
103. Kannus P, Jozsa L, Jarvinen TA, Jarvinen TL, Kvist M, Natri A, et al. Location and dis-
tribution of non-collagenous matrix proteins in musculoskeletal tissues of rat. Histochem
J. 1998;30(11):799–810.
104. Riley GP, Harrall RL, Cawston TE, Hazleman BL, Mackie EJ. Tenascin-C and human tendon
degeneration. Am J Pathol. 1996;149(3):933–43.
105. Sigrist RMS, Liau J, Kaffas AE, Chammas MC, Willmann JK. Ultrasound elastography:
review of techniques and clinical applications. Theranostics. 2017;7(5):1303–29.
106. Proske U, Morgan DL. Tendon stiffness: methods of measurement and significance for the
control of movement. A review. J Biomech. 1987;20(1):75–82.
107. Kubo K, Kanehisa H, Fukunaga T. Effects of different duration isometric contractions on
tendon elasticity in human quadriceps muscles. J Physiol. 2001;536(Pt 2):649–55.
108. Sarvazyan AP, Rudenko OV, Swanson SD, Fowlkes JB, Emelianov SY. Shear wave elas-
ticity imaging: a new ultrasonic technology of medical diagnostics. Ultrasound Med Biol.
1998;24(9):1419–35.
109. Eby SF, Song P, Chen S, Chen Q, Greenleaf JF, An KN. Validation of shear wave elastogra-
phy in skeletal muscle. J Biomech. 2013;46(14):2381–7.
110. Grasa J, Calvo B, Delgado-Andrade C, Navarro MP. Variations in tendon stiffness due to
diets with different glycotoxins affect mechanical properties in the muscle-tendon unit. Ann
Biomed Eng. 2013;41(3):488–96.
111. Fukutani A, Misaki J, Isaka T. Relationship between joint torque and muscle fascicle shorten-
ing at various joint angles and intensities in the plantar flexors. Sci Rep. 2017;7(1):290.
112. Konow N, Azizi E, Roberts TJ. Muscle power attenuation by tendon during energy dissipa-
tion. Proc Biol Sci. 2012;279(1731):1108–13.
113. Farris DJ, Raiteri BJ. Elastic ankle muscle-tendon interactions are adjusted to produce accel-
eration during walking in humans. J Exp Biol. 2017;220(Pt 22):4252–60.
114. Waugh CM, Korff T, Blazevich AJ. Developmental differences in dynamic muscle-tendon
behaviour: implications for movement efficiency. J Exp Biol. 2017;220(Pt 7):1287–94.
115. Kubo K, Kanehisa H, Kawakami Y, Fukunaga T. Influence of static stretching on viscoelastic
properties of human tendon structures in vivo. J Appl Physiol (1985). 2001;90(2):520–7.
116. Seynnes OR, Bojsen-Moller J, Albracht K, Arndt A, Cronin NJ, Finni T, et al. Ultrasound-
based testing of tendon mechanical properties: a critical evaluation. J Appl Physiol (1985).
2015;118(2):133–41.
117. Cortes DH, Suydam SM, Silbernagel KG, Buchanan TS, Elliott DM. Continuous shear wave
elastography: a new method to measure viscoelastic properties of tendons in vivo. Ultrasound
Med Biol. 2015;41(6):1518–29.
118. Graf BK, Vanderby R Jr, Ulm MJ, Rogalski RP, Thielke RJ. Effect of preconditioning on the
viscoelastic response of primate patellar tendon. Arthroscopy. 1994;10(1):90–6.
119. Shepherd JH, Legerlotz K, Demirci T, Klemt C, Riley GP, Screen HR. Functionally distinct
tendon fascicles exhibit different creep and stress relaxation behaviour. Proc Inst Mech Eng
H. 2014;228(1):49–59.
120. Wren TAL, Lindsey DP, Beaupré GS, Carter DR. Effects of creep and cyclic loading on
the mechanical properties and failure of human achilles tendons. Ann Biomed Eng.
2003;31(6):710–7.
121. Suydam SM, Soulas EM, Elliott DM, Silbernagel KG, Buchanan TS, Cortes DH. Viscoelastic
properties of healthy achilles tendon are independent of isometric plantar flexion strength and
cross-sectional area. J Orthop Res. 2015;33(6):926–31.
2 Biomechanics of Skeletal Muscle and Tendon 71
166. Chiquet M. Regulation of extracellular matrix gene expression by mechanical stress. Matrix
Biol. 1999;18(5):417–26.
167. Fluck M, Carson JA, Gordon SE, Ziemiecki A, Booth FW. Focal adhesion proteins FAK and
paxillin increase in hypertrophied skeletal muscle. Am J Physiol. 1999;277(1 Pt 1):C152–62.
168. Fluck M, Tunc-Civelek V, Chiquet M. Rapid and reciprocal regulation of tenascin-C and
tenascin-Y expression by loading of skeletal muscle. J Cell Sci. 2000;113(Pt 20):3583–91.
169. Goodyear LJ, Chang PY, Sherwood DJ, Dufresne SD, Moller DE. Effects of exercise and
insulin on mitogen-activated protein kinase signaling pathways in rat skeletal muscle. Am J
Physiol. 1996;271(2 Pt 1):E403–8.
170. Milanini J, Vinals F, Pouyssegur J, Pages G. p42/p44 MAP kinase module plays a key role in
the transcriptional regulation of the vascular endothelial growth factor gene in fibroblasts. J
Biol Chem. 1998;273(29):18165–72.
171. Sackin H. Mechanosensitive channels. Annu Rev Physiol. 1995;57:333–53.
172. Jones BF, Wall ME, Carroll RL, Washburn S, Banes AJ. Ligament cells stretch-adapted on
a microgrooved substrate increase intercellular communication in response to a mechanical
stimulus. J Biomech. 2005;38(8):1653–64.
173. Wall ME, Banes AJ. Early responses to mechanical load in tendon: role for calcium sig-
naling, gap junctions and intercellular communication. J Musculoskel Neuronal Interact.
2005;5(1):70–84.
174. Wall ME, Dyment NA, Bodle J, Volmer J, Loboa E, Cederlund A, et al. Cell signaling in teno-
cytes: response to load and ligands in health and disease. Adv Exp Med Biol. 2016;920:79–95.
175. Nam HY, Balaji Raghavendran HR, Pingguan-Murphy B, Abbas AA, Merican AM, Kamarul
T. Fate of tenogenic differentiation potential of human bone marrow stromal cells by uni-
axial stretching affected by stretch-activated calcium channel agonist gadolinium. PLoS One.
2017;12(6):e0178117.
176. Cowan KJ, Storey KB. Mitogen-activated protein kinases: new signaling pathways function-
ing in cellular responses to environmental stress. J Exp Biol. 2003;206(Pt 7):1107–15.
177. Arnoczky SP, Tian T, Lavagnino M, Gardner K, Schuler P, Morse P. Activation of stress-
activated protein kinases (SAPK) in tendon cells following cyclic strain: the effects of strain
frequency, strain magnitude, and cytosolic calcium. J Orthop Res. 2002;20(5):947–52.
178. Wu YF, Huang YT, Wang HK, Yao CJ, Sun JS, Chao YH. Hyperglycemia augments the
adipogenic transdifferentiation potential of tenocytes and is alleviated by cyclic mechanical
stretch. Int J Mol Sci. 2017;19(1):E90.
Chapter 3
Biomechanics of Ligaments
Abstract The ligaments are important tissues that connect the bones in the
human body. The main function is to transmit the tensile load by playing an
important role in maintaining the stability and restraint excessive joint motion
in musculoskeletal system. Their unique composition and structure let liga-
ments to guide joints to articulate smoothly and to protect other soft tissues in
and around the joints. These ligaments have biomechanical properties designed
for this important function. As such, understanding the biomechanical behavior
of ligaments is important. Also, these fundamental knowledges are helpful in
preventing ligament injury and improving the treatment method. In this chapter,
the ligament composition, structure, and function are introduced. Then the bio-
mechanical properties of the ligaments and the method to obtain them are
described by using the anterior cruciate ligament (ACL) of the knee as an exam-
ple. And finally, injury, as well as current surgical treatment and postoperative
rehabilitation, is reviewed.
J. Yao · Z. Lian
Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School
of Biological Science and Medical Engineering, Beihang University, Beijing, China
Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
B. Yang
Orthopedics Department, Peking University International Hospital, Beijing, China
Y. Fan (*)
Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School
of Biological Science and Medical Engineering, Beihang University, Beijing, China
Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
National Research Center for Rehabilitation Technical Aids, Beijing, China
e-mail: yubofan@buaa.edu.cn
1.1 Composition
The ligament is a fibrous connective tissue that is mainly composed of fibroblasts. Its
extracellular matrix is rich in collagen, proteoglycans, and water, which determines the
biomechanical characteristics of the ligament. Water accounts for about 60–70% of the
mass of the cytoplasmic matrix. Collagen accounts for about 70–80% of its dry weight,
and is a most important component of bearing loading. In addition, other components
such as elastin and proteoglycan are also included in the cytoplasmic matrix [1, 2].
Collagen is the main component of the ligament extracellular matrix and is
mainly composed of type I collagen. In addition, type II, type III, type V, and type
XI collagen also account for a certain proportion. For example, Type II collagen
usually exists at the junction of the ligament and bone; the proportion of type III
collagen in the ligament gradually decreases as the embryo develops. A small
amount of type V and type XI collagen is often accompanied by type I, II, and III
collagen in fibrous cell surfaceome [3, 4].
The content of elastin varies among different types of ligaments, generally less
than 1% of dry weight. It can impart ligament elasticity and tensile strength together
with collagen. There is no continuous repetitive cycle throughout the peptide chain
in its amino acid sequence structure. But there are alternating hydrophobic and
hydrophilic segments [5]. Proteoglycans consist of small amounts of protein bound
to negatively charged polysaccharide chains referred to as GAGs. Proteoglycans
account for a small proportion of ligaments, about 0.5%. It can bind to water and
improve the stability and strength between ligament fibers. In addition, there are a
variety of other chemicals, including intracellular DNA, proteases, lipoproteins,
etc., which together maintain the survival of cells in the ligament.
The distribution of the biochemical components that make up the ligament is
nonuniform. For example, the collagen proportions of the four ligaments (anterior
cruciate ligament (ACL), posterior cruciate ligament (PCL), medial collateral liga-
ment (MCL), and lateral collateral ligament (LCL)) in knee joint are different, the
hydroxyproline densities are: 15.1 ± 0.4, 18.3 ± 0.3, 17.6 ± 0.4, 17.6 ± 0.4 mg/μm3.
On the same ligament, the collagen distribution is nonuniform. The collagen pro-
portion in the anterior sites of the above four ligaments is higher than the posterior
sites. Along the longitudinal axis of ACL and PCL, the collagen proportions at the
middle regions are greater than those at both ends near the bone [6]. Furthermore,
there are also capillaries in the ligament, which provide blood for the growth of the
ligament, as well as the repair after injury. Blood vessels growing in some ligaments
can also nourish surrounding bones and other soft tissues.
3 Biomechanics of Ligaments 77
1.2 Structure
Microscopically, the collagen of the ligament is a layered structure. The triple helix-
shaped collagen molecules aggregate to form collagen fibers. Macroscopically, the
ligaments can be divided into visceral ligament and joint ligament. They are distrib-
uted in different locations, which makes their structure varying. The visceral liga-
ment is formed by a single layer or double layers of peritoneal folds. It is connected
to the visceral layer of the liver, kidneys, and other organs. The ends of the joint
ligament are attached to the surface of the bone or fused to the joint capsule. At the
ligament’s insertion site on the bone, the collagen fibers bind to the bone cells and
form the skeletal junction of the ligament. The middle part of the ligament is a dense
connective tissue composed of a large number of fiber bundles. The length and
thickness of the fiber bundles are nonuniformly and irregularly distributed. The fiber
bundles interact with each other, and each bundle of fibers is at an angle of 0–30° to
the longitudinal direction of the ligament.
Take the PCL as an example. 95% of the fiber bundles are aligned and distributed
consistent with the longitudinal axis of the ligament. The other 5% is arranged
obliquely from the proximal posterior end to distal medial, at an angle of 15–20° to
the longitudinal axis of the ligament. The fiber bundles in the middle part of the liga-
ment are tightly connected to each other, and looks like a twisted rope. The fiber
bundles near the femoral and tibial attachments are relatively loose and can be sepa-
rated. These fiber bundles are composed of elastin and collagen, which are inter-
twined to form the overall structure of PCL.
1.3 Function
“toe” region of the loading-elongation curve. When the load is increased, the col-
lagen fibers are elongated, and the sliding between each other is reduced, the stiff-
ness of the ligament is increased. Ligament maintains approximately linear elastic
until the collagen fibers reach the yield threshold. In addition, the mutual sliding
action between the fiber bundles can contribute to the viscoelastic effect of the liga-
ment. All these characteristics provide the basis for the physiological function of the
ligament: at low loads, guide the path of joint motion; at high loads, restrict exces-
sive motion (displacement) between bones to protect other soft tissue.
2.1 Tensile Properties
The main function of the ligament is to withstand the tensile loads. Therefore, quan-
tification of the mechanical properties of the ligament in response to different load-
ing condition is important for understanding its function as well as the mechanism
of ligament injury healing. The uniaxial tensile test is usually used to measure the
tensile properties of the ligament [1]. Taking the anterior cruciate ligament of New
Zealand rabbits as an example, the general procedure is as follows:
After sacrifice of the subject, the knee joint is taken, including the femur and
tibia. Because subject’s ACL is short and difficult to fix, the “femur-ACL-tibia”
specimen is prepared. The skin, muscle, fat, meniscus, and other ligamentous tis-
sues are carefully removed. In order to facilitate the fixation of the sample, the
proximal femur and the distal tibia are fixed in a standard mold with resin material
(Fig. 3.1). The stiffness of the mold, resin, and bone has been tested to be much
greater than ACL, and thus their deformations could be neglected compared with
the ACL. Since the mechanical properties of ACL are significantly correlated with
water proportion, saline is used to keep ACL moist in both the preparation process
and the subsequent experimental procedures.
The Femur-ACL-Tibia specimen is loaded and measured using a dynamic
mechanical testing machine (Instron E10000, Illinois Tool Works Inc., USA) with a
sensor range of 250 N. ACL is kept coinciding with the load axis. ACL length under
1.0 N loading is measured as the initial length. First, a cycle loading is performed.
A loading magnitude of 1% in the nominal strain and a loading frequency of 1 Hz
are applied for 20 cycles. Second, a tensile test with a strain rate of 0.1%/s is applied.
To calculate the nominal stress, the entire ACL is embedded in paraffin. The average
cross-sectional area is calculated from five positions along the longitudinal axis of
the ligament. The nominal stress is calculated as the ratio of loading to the cross-
sectional area. The nominal strain is calculated as the change in length per unit of
the original length of the ligament.
The force-displacement curve of ACL is shown in Fig. 3.2. ACL exhibits typical
hyperelasticity under the uniaxial tension. The force-displacement curve can be
divided into toe region, linear region, and yield region. When the load is small, the
3 Biomechanics of Ligaments 79
Femur
ACL
Tibia
Fixation
Load-Displacement
200
180
160
Toe Region Linear Region
140
120
Load(N)
100
80
F Stif f ness = F/ L
60
40
L
20
0
0 1 2 3 4 5 6 7
Displacement(mm)
fiber bundles in the ligament are not fully stretched, and the ligament is relatively
flexible, which is in the toe region in the figure. The modulus of elasticity in the toe
region increases continuously, but it is at a smaller level than that in linear region.
As the stress increases, all the fiber bundles turn into the tension state, which is in
the linear region in the figure. Linear region is the main functional region of liga-
ment. Compared with the toe region, the modulus of linear region is greater and
nearly constant. As the load continues to increase, some fiber bundles break, and the
80 J. Yao et al.
ligament is damaged. Plastic deformation occurred in the region. With the increase
of the fiber bundles broken, the ligament modulus decreases.
The critical point between the toe region and the linear region is called the toe
region point. The larger the stress in the toe region, the larger the degree of curl-
ing of the fiber bundle in the relaxed state. This is instructive for the setting of
preloading in ligament reconstruction. The normal functional region of the liga-
ment is linear region. Therefore, the elastic modulus of the linear region is an
important parameter of the biomechanical property of the ligament, which is used
to represent the stiffness of different ligaments. In the above experiment, the rab-
bit ACL elastic modulus was about 61.3 MPa. The point between the linear region
and the yield region is called the elastic limit. When the ligament exceeds the
elastic limit, the ligament begins to injury. In the stress-strain curve, the point at
which the stress reaches the maximum value is called the tensile strength. When
the ligament is stretched exceeding its strength, the ligament is completely torn.
Therefore, strength refers to the ability of the ligament to resist damage. The
acute injury to the ligament is mostly due to the load exceeding the strength of the
ligament.
The mechanical properties of the ligament are related to age. Taking the human
ACL as an example, the linear region stiffness and ultimate load will decrease with
age [7]. The cadaveric study indicated that the linear region stiffness of young group
(20–35 years old) (242 ± 28 N/mm) was 10% higher than that of the middle-aged
group (40–50 years old) (220 ± 24 N/mm), and was 34% higher than that of elderly
group (60–97 years old) (180 ± 25 N/mm). The ultimate load of young group
(2160 ± 157 N) was 44% higher than that of the middle-aged group (1503 ± 83 N)
and 228% higher than that of the elderly group (658 ± 129 N). In addition, the
energy absorbed by the ligament injury has the similar tendency as the linear stiff-
ness. Ligaments of young group absorb more energy when destroyed than those of
older groups. In the elderly group, the incidence of collagen ligament destruction in
the middle of the ligament (12/18, 67%) was higher than that in the young group
(5/18, 28%) and the middle-age group (8/18, 44%). In the young group, the skeletal
avulsion at the ligament was the highest (9/18, 50%). Possible causes include: Older
donors’ knee joints have fewer types and amounts of daily activity than younger
donors, resulting in changes in joint geometry and ligament material properties.
In complex in vivo mechanical environments, the loading direction of the joint is
often not parallel to the longitudinal axis of the ligament, and thus also affects the
mechanical response of the ligament. Take the ACL as an example. The stiffness of
the femoral-ACL-tibia specimen parallel to the longitudinal axis of the ACL was
242 ± 28 N/mm, and the stiffness of the specimen parallel to the tibia shaft was
reduced to 218 ± 27 N/mm. The ultimate load of the specimen in the direction of the
longitudinal axis of the ACL is 35% higher than the direction of the tibia shaft. The
experimental results imply that the influence of the tensile direction of the specimen
plays a role in the mechanical properties of the ligament. In fact, the various parts of
the ligament have different lengths. Therefore, the entire ligament is not uniformly
elongated during the stretching. There are also differences in the way the ligaments
yield in different directions of stretching. When the ligament is loaded in the
3 Biomechanics of Ligaments 81
d irection of the longitudinal axis, the deformations of elastic fibers are relatively
uniform under the tensile load. The ultimate load in this direction can more accu-
rately reflect the strength of the ligament. Therefore, when testing the mechanical
properties of the ligament, it is necessary to ensure that the longitudinal axis of the
ligament coincides with the loading axis.
2.2 Viscoelastic Properties
The complex interaction of collagen, elastin, proteoglycan, and water in the liga-
ment results in a viscoelastic property of the ligament. This is manifested in the fact
that the mechanical response is related to the rate of loading and exhibits hysteresis,
creep, and stress relaxation. The viscoelastic properties of the ligaments are gener-
ally measured by uniaxial tensile tests at different loading rates. Taking the mea-
surement of ACL of New Zealand rabbits as an example, the general procedure is as
follows:
Femur-ACL-tibia specimens are prepared and fixed in the dynamic mechani-
cal testing machine (Instron E10000, Illinois Tool Works Inc., USA). The liga-
ment is kept in line with the loading direction. In daily joint motion, the ligaments
generally perform a periodic stretching motion at approximate 0.1–1 Hz.
Therefore, a sinusoidal test of 1% strain amplitude is performed at a frequency of
0.1 Hz and 1 Hz, and each frequency was performed for 20 cycles. Each set of
specimens is recovered for 60 min at 0% strain between trials, eliminating the
effects of historical deformation.
In the test, the soft tissue strain was large; therefore the true strain and true stress
were calculated:
ε = ln ( L / L0 ) (3.1)
F L
δ= (3.2)
A0 A0
where L is the measured length, L0 is the initial length, F is the measured tensile
force, and A0 is the cross-sectional area. The data is then fitted to the stress and strain
data of the sinusoidal test:
z = A sin ( 2π ft + ϕ ) + z0 (3.3)
There are several methods of quantifying the viscoelastic properties of the liga-
ment. The parameter “supplementary stress” can also describe this property [8].
Define the “supplementary stress” variable as:
σ y − σ 0.1
Supplementary stress =
σy
where y indicates a different strain rate value. σy indicates the stress at y%/s strain
rate, and σ0.1 indicates the stress at 0.1%/s strain rate. The supplementary stress
represents the percentage of stress at a high strain rate at a strain rate greater than
0.1%/s strain rate at the same strain level. The shape of the stress-strain curve does
not change with increasing strain rate, except that the toe region appears in the lower
strain range. As the strain rate increases, the “supplemental stress” increases signifi-
cantly. When the strain rate is 40%/s, the “supplemental stress” value can reach
70%. This phenomenon of bovine ACL is similar to the conclusion obtained from
the rabbit ACL test.
The viscoelastic effect of the ligament is closely related to its physiological func-
tion. When the joint movement speed is low, the ligament has a low strain rate and
is relatively flexible, then the muscle plays a greater role in stability and restraint of
joint. As the speed of joint motion increases, the stiffness of the ligament increases,
which can compensate the delay of the muscle response.
In addition, the toe region modulus is also one of the methods to reflect visco-
elastic properties [9]. In the uniaxial tensile tests with different loading rates, the
ratio of stress to strain at the critical point between the toe region and the linear
region is defined as the toe region modulus. The viscosity increases with the differ-
ence of the toe modulus at different strain rates. By this method, the change in vis-
cosity of the xenograft porcine superflexor tendon (pSFT) used in ACL reconstruction
before and after decellularization was measured. The natural pSFT has a significant
difference in the toe modulus (29.7, 104.3, 139.8 MPa) under the three different
strain rates (1%/s, 10%/s, 100%/s). However, there is no such difference in the toe
modulus (48.8, 63.1, and 68.1 MPa) after decellularization. This indicates that pSFT
after decellularization results in a decrease in viscosity.
3.1 Injury
There are many factors contributing to the injury of the ligament. Most ligament
injuries are caused by violent, traumatic, non-physiological activities, especially
joint ligaments. For example, medial collateral ligament may tear under violent
knee valgus, and ACL may tear under knee internal rotation. When external factors
cause the ligament to reach the elastic limit and exceed its tolerance, the ligament is
3 Biomechanics of Ligaments 83
injured. The injury may occur in the middle or the attachments of the ligament.
Damage to the joint ligaments can contribute to dislocation, maltracking, and insta-
bility of the joints, and lead to subsequent diseases of other soft tissue and bone,
such as osteoarthritis. In addition, genetic, endocrine, and microbial infections can
lead to ligamentous lesions that gradually damage the ligaments.
The risk of ligament injury is also related to the physiological structure of the
musculoskeletal system and the external environment. Taking ACL injury as an
example, the probability of ACL injury in the subject with narrow intercondylar
notch is greater. Since ACL is located in the joint cavity of the tibiofemoral joint,
and is sandwiched by the medial and lateral femoral condyles, when the knee joint
is subjected to internal and external loads, the two femoral condyles will press the
ACL, causing additional lateral force on the ACL and increasing the chance of ACL
tear [10–12]. In addition, ACL footprints are at the anteromedial aspect of the inter-
condylar area on the tibial plateau and the posteromedial aspect of the lateral femo-
ral condyle. For individuals with an anterior medial aspect of the tibial plateau that
is higher than the posterior lateral aspect, when the knee joint is subjected to a
vertical load, the tibia will be additionally subjected to the forward and medial
loading, thereby causing additional in situ force in ACL, and increasing the prob-
ability of ACL injury. A number of statistical studies have reported the correlation
between this knee anatomical feature and ACL injury [13–15].
Under different sports types and ground conditions, the injury rate of ACL is also
different. Renstrom et al. reported the rate of ACL injury in basketball, ice hockey,
football, and gymnastics. The ACL injury rate of women’s basketball and women’s
gymnastics was the highest, reaching nearly 5%, while the ACL injury rate for
men’s basketball and women’s ice hockey was only 0.7% [16]. At the same time, a
number of statistical studies have analyzed the correlation between ground environ-
ment and ACL injury rate; selecting the appropriate site according to the type of
exercise can reduce the probability of ACL injury [17–20]. In addition, Lambson
et al. found that different sole materials and textures provide different grips (such as
lateral, longitudinal, and torsional resistance) and have an impact on ACL injury [21].
3.2 Treatment
The treatment of ligament injury mainly includes conservative treatment and surgi-
cal treatment. The choice of the treatment methods mainly depends on the type and
severity of the ligament injury. For example, nearly 90% of knee joint ligament
injuries are ACL and MCL injuries [22]. MCL injuries usually heal spontaneous,
although scar tissue will appear between MCL and bones, resulting in nonideal
changes in MCL mechanical properties. However, the clinical treatment is usually
conservative, and the MCL will be repaired by resting. In contrast, ACL tears often
require surgery because ACL has fewer blood vessels and poor healing ability. If
nonsurgical conservative treatment is used, it may lead to subsequent meniscus and
cartilage lesions, resulting in severe instability of the knee joint.
84 J. Yao et al.
a b
Fig. 3.3 Rabbit ACL reconstruction. (a) Rabbit knee joint before ACL reconstruction. (b) Rabbit
knee joint after ACL reconstruction
86 J. Yao et al.
needle. The sutures at both ends of the graft are respectively passed through the
tunnel, and the graft is pulled into the tunnel. The joint stability is assessed accord-
ing to the tibiofemoral anterior-posterior translation. Finally, the sutures at both
ends of the graft were tied to the proximal femur and the distal end of the tibia. The
knee joint capsule and the epidermal incision were sutured and placed in the cage
after being injected with antibiotics.
3.4 Rehabilitation
4 Summary
The biomechanical properties of the ligaments are closely related to their physio-
logical functions. Therefore, the ligaments’ composition, structure, as well as their
biochemical and mechanical interactions with the surrounding tissue are varying in
different locations of the human body. Illustrating the relationship between the bio-
chemical and mechanical characters of ligaments is critical for understanding the
mechanism of ligament injury and improving the treatment strategies. This will
become an important direction for future research and will bring profound signifi-
cance to clinical application.
Statement on the Animal Benefit All animal treatments were approved by the
Animal Care Committee of Beihang University in accordance with the Regulation
of Administration of Affairs Concerning Experimental Animals of State Science
and Technology Commission of China.
Acknowledgement The authors would like to convey their appreciation to Prof. Savio L-Y. Woo
for his precious suggestions.
3 Biomechanics of Ligaments 87
References
1. Woo SL, Abramowitch SD, Kilger R, Liang R. Biomechanics of knee ligaments: injury, heal-
ing, and repair. J Biomech. 2006;39(1):1–20.
2. Mienaltowski MJ, Birk DE. Structure, physiology, and biochemistry of collagens. In: Progress
heritable soft connective tissue diseases, vol. 802. Dordrecht: Springer; 2014. p. 5–29.
3. Milz S, Jakob J, Buttner A, Tischer T, Putz R, Benjamin M. The structure of the coracoacro-
mial ligament: fibrocartilage differentiation does not necessarily mean pathology. Scand J Med
Sci Sports. 2008;18(1):16–22.
4. Smith SM, Thomas CE, Birk DE. Pericellular proteins of the developing mouse tendon: a
proteomic analysis. Connect Tissue Res. 2012;53(1):2–13.
5. Quapp KM. Material characterization of human medical collateral. Ligament. 1997;
6. Mommersteeg TJ, Blankevoort L, Kooloos JG, Hendriks JC, Kauer JM, Huiskes R. Nonuniform
distribution of collagen density in human knee ligaments. J Orthop Res. 1994;12(2):238–45.
7. Woo SL-Y, Hollis JM, Adams DJ, Lyon RM, Takai S. Tensile properties of the human femur-
anterior cruciate ligament-tibia complex. The effects of specimen age and orientation. Am J
Sports Med. 1991;19(3):217–25.
8. Pioletti DP, Rakotomanana LR, Leyvraz PF. Strain rate effect on the mechanical behavior of
the anterior cruciate ligament-bone complex. Med Eng Phys. 1999;21(2):95–100.
9. Edwards JH, Ingham E, Herbert A. Decellularisation affects the strain rate dependent and
dynamic mechanical properties of a xenogeneic tendon intended for anterior cruciate ligament
replacement. J Mech Behav Biomed Mater. 2019;91:18–23.
10. Souryal TO, Freeman TR. Intercondylar notch size and anterior cruciate ligament injuries in
athletes. A prospective study. Am J Sports Med. 1993;21(4):535–9.
11. Lund-Hanssen H, Gannon J, Engebretsen L, Holen KJ, Anda S, Vatten L. Intercondylar notch
width and the risk for anterior cruciate ligament rupture. A case-control study in 46 female
handball players. Acta Orthop Scand. 1994;65(5):529–32.
12. Souryal TO, Moore HA, Evans JP. Bilaterality in anterior cruciate ligament injuries: associated
intercondylar notch stenosis. Am J Sports Med. 1988;16(5):449–54.
13. Hashemi J, Chandrashekar N, Mansouri H, Gill B, Slauterbeck JR, Schutt RC, et al. Shallow
medial tibial plateau and steep medial and lateral tibial slopes new risk factors for anterior
cruciate ligament injuries. Am J Sport Med. 2010;38(1):54–62.
14. Todd MS, Lalliss S, Garcia E, DeBerardino TM, Cameron KL. The relationship between pos-
terior tibial slope and anterior cruciate ligament injuries. Am J Sport Med. 2010;38(1):63–7.
15. Khan MS, Seon JK, Song EK. Risk factors for anterior cruciate ligament injury: assessment
of tibial plateau anatomic variables on conventional MRI using a new combined method. Int
Orthop. 2011;35(8):1251–6.
16. Renstrom P, Ljungqvist A, Arendt E, Beynnon B, Fukubayashi T, Garrett W, et al. Non-contact
ACL injuries in female athletes: an International Olympic Committee current concepts state-
ment. Br J Sport Med. 2008;42(6):394–412.
17. Orchard J, Seward H, McGivern J, Hood S. Rainfall, evaporation and the risk of non-
contact anterior cruciate ligament injury in the Australian Football League. Med J Aust.
1999;170(7):304–6.
18. Olsen OE, Myklebust G, Engebretsen L, Holme I, Bahr R. Relationship between floor type and
risk of ACL injury in team handball. Scand J Med Sci Sports. 2003;13(5):299–304.
19. Orchard JW, Powell JW. Risk of knee and ankle: sprains under various weather conditions in
american football. Med Sci Sports Exerc. 2003;35(7):1118–23.
20. Orchard JW, Chivers I, Aldous D, Bennell K, Seward H. Rye grass is associated with
fewer non-contact anterior cruciate ligament injuries than Bermuda grass. Br J Sport Med.
2005;39(10):704–9.
21. Lambson RB, Barnhill BS, Higgins RW. Football cleat design and its effect on anterior cruci-
ate ligament injuries. A three-year prospective study. Am J Sports Med. 1996;24(2):155–9.
22. Woo SL-Y, Debski RE, Zeminski J, Abramowitch SD, Saw SS, Fenwick JA. Injury and repair
of ligaments and tendons. Annu Rev Biomed Eng. 2000;2:83–118.
Chapter 4
Hand and Wrist Biomechanics
Abstract The biomechanical function of the human hand and wrist is closely
related to the intricacy of its anatomy. The arrangement of the bones, ligament, and
muscles in the hand allows a complex array of tasks to be performed. This chapter
provides a brief review of the anatomy of the hand and wrist, normal biomechanical
function, and examples of pathomechanics.
The human hand and wrist are a set of complex anatomic tools that forms a tactile
connection to the world. The intricacies of hand and wrist anatomy permit a multi-
tude of different functions including the ability to grip an object, let go, flex and
extend and to allow the precise movements required for writing, playing sport, and
innumerable other actions. This chapter will briefly review some skeletal and liga-
mentous anatomy of the hand and wrist and then describe the complex biomechan-
ics of the muscles, ligaments, and joints. Lastly, a few examples of pathomechanics
are provided, including arthritic condition of the first carpometacarpal joint, distal
radius fracture, and carpal tunnel syndrome.
S. Regal · S. Maschke
Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, OH, USA
e-mail: regals@ccf.org; maschks@ccf.org
Z.-M. Li (*)
Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, OH, USA
Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, USA
Department of Physical Medicine and Rehabilitation, Cleveland Clinic, Cleveland, OH, USA
e-mail: liz4@ccf.org
The human hand comprises 27 bones (14 phalanges, 5 metacarpals, and 8 carpal bones)
that form the distal portion of the wrist joint. The “wrist joint,” or more accurately “wrist
joint complex,” contains numerous articulations linking the distal forearm to the hand
and consists of the distal radius and ulna articulating with the multi-articular carpal
bones which then articulate with the metacarpals. The carpal bones have been classi-
cally divided into proximal and distal rows based upon their kinematic behavior during
wrist motion. The distal row contains the trapezium, trapezoid, capitate, and hamate
which are tightly bound to each other by stout intercarpal ligaments that allow minimal
motion. The scaphoid, lunate, triquetrum, and pisiform form the proximal carpal row
and is described as an intercalated segment because no tendons insert on them and their
motion is dependent on mechanical forces from their adjacent articulations [1, 2].
Taleisnik [3] divided the carpal ligaments into intrinsic and extrinsic groups deter-
mined by the anatomical origination and insertion of the ligament. The extrinsic liga-
ments of the wrist bridge the carpal bones to the radius, ulna, or metacarpals. The
dorsal extrinsic ligaments consist of the dorsal radiocarpal (radiotriquetral) ligament
and the dorsal intercarpal ligament that form a sideways V. Both ligaments have
attachments to the lunate and are important secondary stabilizers of the scapholunate
joint [1]. The palmar extrinsic ligaments of the wrist have been shown to provide the
most restraint in dorsal/palmar translation of the carpus and include the radioscaph-
oid-capitate, long radiolunate, short radiolunate, ulnotriquetral, ulnolunate, and
ulnocapitate ligaments [4]. The space of Poirier is an area of capsular weakness that
is clinically important in dislocations about the lunate and is located between the
radioscaphocapitate and long radiolunate ligaments (Fig. 4.1). The radioscapholu-
nate ligament is not a true ligament but is a vascular conduit to the lunate [5, 6].
a b
There are nine intrinsic ligaments of the wrist that originate and insert on adja-
cent carpal bones. The two most important intrinsic ligaments are the scapholunate
and lunotriquetral ligaments which can be divided into dorsal, proximal, and palmar
parts. The thickest and strongest part of the scapholunate ligament is the dorsal por-
tion whereas the thickest and strongest part of the lunotriquetral ligament is located
palmarly.
2 Biomechanics
The hand and wrist contain four different types of synovial joints—plane, pivot,
ellipsoidal, and saddle. Plane joints allow simple gliding motion between flat articu-
lar surfaces such as in the intercarpal, intermetacarpal, and carpometacarpal joints
of the second to fourth digits. The interphalangeal joints of the digits are hinge
joints and only allow motion to occur in one plane 90° to the bones to permit flexion
and extension. An ellipsoidal joint formed by oval-shaped condyle and ellipsoidal
cavity allows motion in two planes, flexion/extension and abduction/adduction sec-
ondary to their reciprocal concave/convex articular surfaces like those found in the
radiocarpal and metacarpophalangeal joints. A saddle joint is formed by bones with
concave and convex articular surfaces and only occurs in the carpometacarpal joint
of the thumb, allowing motion in flexion/extension, abduction/adduction, and cir-
cumduction [7].
2.1 Wrist Motion
Normal wrist range of motion in the sagittal plan is about 65° of flexion and 55°
of extension while in the coronal plane there is 15° of radial deviation and 35° of
ulnar deviation. Research on the specifics of carpal motion and wrist kinematics
date back to the nineteenth century and several theories have been proposed. The
eight carpal bones of the wrist are commonly described as containing a proximal
and distal row; however, when discussing carpal kinematics, it is much more com-
plex. Navarro described the columnar theory in 1921 dividing the wrist into three
columns. The lateral column contained the scaphoid, trapezium, and trapezoid; the
central column contained the lunate, capitate, and hamate; and the medial column
contained the triquetrum and pisiform [8]. Taleisnik later modified this theory in
1976 and stated only the scaphoid was in the lateral column while the central col-
umn now consisted of the trapezium, trapezoid, lunate, capitate, and hamate. In
both theories, the central column moved concurrently during flexion/extension
and the medial and lateral columns rotated around the central column in radial/
ulnar deviation [3]. Years later, Craigen and Stanley proposed that carpal motion
is complex and variable and can be accounted for by both the row and columnar
theories [9].
92 S. Regal et al.
The proximal carpal row has no direct tendon attachment and moves passively
when muscle contraction initiates movement at the distal carpal row. In normal
wrists, there is very little motion between the bones in the distal carpal row which
essentially moves as one functional unit. During wrist flexion, the distal carpal row
synchronously flexes accompanied by a small amount of ulnar deviation. Conversely,
during wrist extension the distal row rotates into extension and radial deviation.
During radioulnar deviation, the proximal carpal row moves from a flexed position
in radial deviation to an extended position in ulnar deviation. More recently, studies
of carpal motion have been performed in vivo with computed tomography and mag-
netic resonance imaging. Moojen et al. [10] quantified in vivo carpal kinematics and
compared their results to in vitro studies. Their in vivo results were consistent with
prior studies in that that the majority of wrist flexion and extension occur at the
radiocarpal joint. Their results showed that in 60° of wrist flexion and extension, the
scaphoid contributes to 62% and 87% of the in-plane motion, respectively, while the
lunate contributes 31% and 66%. Kaufmann et al. [11] showed that the midcarpal
joint is responsible for 86% of radial deviation and 66% of ulnar deviation. In radio-
ulnar deviation, the scaphoid flexes and extends, respectively, while in wrist flexion/
extension the scaphoid moves in the same plane of motion. A combination of these
motions, radiodorsal/ulnopalmar, has been described as the dart thrower’s motion
and has been portrayed as the most frequently used motion during activities of daily
living [12–14]. A three-dimensional motion analysis of the carpal bones during the
dart thrower’s motion found that the majority of the motion occurs at the midcarpal
joint and that there is minimal motion between the scaphoid and lunate [13, 15, 16].
2.2 Muscular Biomechanics
Hand and wrist motion is multifaceted and relies not only on the bony articulations
but also on a dynamic balance of muscular forces. The hand is merely a functional
puppet that follows the brain’s desire to perform a specific task; the brain sends
signals to the hand via nerves and activates precise muscles to contract or relax with
instantaneous sensory feedback from visual cues and sensory receptors. This com-
plex, coordinated interaction happens constantly at a subconscious level.
Anatomically and functionally, the wrist is powered by 3 extensors and 3 flexor
muscles while the hand is powered by 9 extrinsic muscles and 17 intrinsic muscles
(Table 4.1). The amount of force a muscle can produce is somewhat proportional to
its cross-sectional area and the mechanical effect on a joint is determined by its
insertion and origin; the flexors insert on the volar aspect of the bone while the
extensors insert dorsally. The muscles of the hand and wrist are in constant harmo-
nious balance of synergism and antagonism. When the wrist is passively extended,
the finger flexor muscles are lengthened and involuntarily finger flexion occurs. In
contrast, passive wrist flexion causes reciprocal finger extension. The combination
of these motions has been termed “the tenodesis effect” and is a clinically useful
4 Hand and Wrist Biomechanics 93
exam technique when evaluating flexor tendon injuries [17]. Furthermore, position
of the wrist affects the amount of peak force that can be generated by the flexor
tendons [18, 19]. Using electromyography, Volz et al. [20] found that the greatest
grip strength was with the wrist in 20° of extension and weakest in 40° of flexion.
Hazelton et al. [18] found the highest force production at the interphalangeal joints
were greatest with the wrist in ulnar deviation, followed by extension and weakest
with wrist in flexion. Muscular action and joint forces will be further discussed dur-
ing the description of specific joints of the hand and wrist.
94 S. Regal et al.
2.3 Carpometacarpal Joints
There are five carpometacarpal (CMC) joints in the hand that consist of articulations
between the distal carpal row and the bases of the metacarpals. While there are simi-
larities between the joints of the thumb and the joints of the fingers, the thumb is
discussed separately for its function and motion differ significantly. The second
through fourth CMC joints are synovial joints with motion in one plane, flexion and
extension, while the fifth CMC joint is a saddle-like joint with 2 degrees of freedom;
flexion-extension and abduction-adduction. The second and third CMC joint are
immobile while the fifth CMC joint can move as much as 20° due to the bony con-
figuration of the base of the fifth metacarpal and hamate. The immobility of the
second and third CMC joint is a functional adaptation that facilitates the function of
the wrist extensors and radial wrist flexor (extensor carpi radialis longus and brevis,
flexor carpi radialis). These muscles insert on the bases of the second and third
metacarpals and with their rigid attachment to the carpals, the lever arm of the
muscle is increased.
A critical function of the CMC joints of the hand is its contribution to the palmar
arches. The hand consists of a longitudinal and two transverse arches that are essential
in power grip and pinch as well as the ability to cup and flatten the hand (Fig. 4.2). The
intermetacarpal and the deep transverse metacarpal ligaments of the proximal and
distal portions of the metacarpal, respectively, are necessary to maintain these arches
[21, 22]. The longitudinal arch is fixed due to the immobility of the second and third
CMC joints while the transverse arches facilitate conforming the hand to the shape of
an object being held. The proximal transverse arch is maintained by the shape of the
carpal bones as well as their encompassing ligaments, the flexor retinaculum (or
transverse carpal ligament) and the transverse intercarpal ligaments. The bony carpal
arch and the flexor retinaculum comprise what is known as the carpal tunnel. The
carpal tunnel not only allows the palm to increase its contact area with objects, it also
protects the finger flexors and median nerve. When the flexor retinaculum is incised
during a carpal tunnel release, there is widening of the arch but overall transverse
stability is maintained due to the intact transverse intercarpal ligaments [23].
The first CMC joint is an articulation between the trapezium and the first metacarpal
and has been described as a saddle joint that is concave in the sagittal plane and
convex in the coronal plane (Fig. 4.3). This unique joint allows flexion-extension,
abduction-adduction, and pronation-supination with the net effect of these motions
allowing circumduction or opposition-reposition. Though there is motion in three
anatomic planes, the first CMC joint is commonly considered to have 2 degrees of
freedom. The flexion-extension axis is through the trapezium and the abduction-
adduction axis run through the metacarpal base; both are constant through a range
of motion. Because these axes are not perpendicular to each other, for any position
of flexion-extension/abduction-adduction, a set degree of pronation-supination
occurs [24, 25]. The bony configuration of the thumb allows for increased range of
motion required for opposition. Opposition is the sequential movements of abduc-
tion, flexion, and adduction of the first metacarpal with concurrent rotation. This
compound movement brings the thumb out of the palm and rotates the metacarpal
to put the thumb into a position to grip an object together with a finger.
Stability of the first CMC joint is provided by seven ligaments, the dorsoradial
ligament, the posterior oblique ligament, the superficial and deep palmar oblique
ligament, the ulnar collateral ligament, the volar and dorsal first metacarpal ulnar
base–second metacarpal radial base intermetacarpal ligaments. The palmar oblique
ligament, also referred to as the beak ligament, originates on the palmar tubercle
of the trapezium and inserts on the ulnar side of the metacarpal base. As a result of
its intracapsular location, it plays a major role in CMC stabilization and resists
2.5 Metacarpophalangeal Joints
The interphalangeal joints of the hand are ginglymus, or hinge joints, comprised of
two phalanges with congruous articular surfaces that allow up to 10° of hyperexten-
sion and 100° of flexion [28]. The interphalangeal joints achieve stability from the
bony architecture as well as from the volar plate and collateral ligaments. On the
proximal interphalangeal joint, the proper collateral ligament (PCL) originates from
the proximal and dorsal sides of the proximal phalanx and inserts via Sharpey’s
fibers on the middle phalanx. The fibers at the origin are parallel to the phalanx and
the insertional fibers are more oblique, giving the ligament a fan-shaped appearance.
4 Hand and Wrist Biomechanics 97
a b
b
98 S. Regal et al.
The accessory collateral ligament (ACL) originates on the volar portion of the PCL
and inserts distally on dorsolateral side of the volar plate on the middle phalanx [29].
A recent three-dimensional computed tomography study of the proximal interpha-
langeal joint found that the dorsal portion of the PCL increased in length as the joint
went from 0° to 90° of flexion and was the most stabilizing structure while the joint
was in flexion. The volar portion of the PCL and distal portion of the ACL provided
joint stability while in extension [30].
Motion of the fingers is a dynamic balance between the extrinsic and intrinsic
muscles of the hand. The extrinsic muscles, those that originate proximal to the
hand, include the flexor digitorum profundus, flexor digitorum superficialis, exten-
sor digitorum communis, extensor indices proprius, and extensor digitorum minimi.
The lumbricals and interossei comprise the intrinsic muscles. The flexor tendons are
held closely to the volar surface of the phalanges by five annular pulleys and three
cruciate pulleys. Not only are these pulleys biomechanically critical to prevent bow-
stringing, they transmit force during finger flexion. The flexor digitorum profundus
inserts on the proximal portion of the distal phalanx; when the muscle belly con-
tracts, force is emitted along the tendon and flexion at the distal interphalangeal joint
follows. Flexion at the joint creates an angled pathway for the profundus tendon as
it exits the A4 pulley which transmits force to the middle phalanx and resultant flex-
ion of the proximal interphalangeal joint. The profundus tendon creates a similar
force as it exits the A2 pulley and causes a flexion moment at the metacarpophalan-
geal joint [7] (Fig. 4.6).
3 Pathological Biomechanics
instability. Pathologic motion ensues which often leads to disability and pain. With
knowledge of normal biomechanics, treatment can be made to manipulate the
mechanical environment or recreate it.
The thumb CMC joint is the second most common site for osteoarthritis (OA) in the
hand and is the most common joint in the upper extremity to undergo surgical
reconstruction for OA. Arthritis of the thumb CMC joint, also known as basal joint
arthritis or trapeziometacarpal arthritis, can be a debilitating disorder that results in
pain, decreased motion, and reduced strength. Basal joint arthritis affects 1 in 4
women and 1 in 12 men; such a gender factor is thought to be related to ligament
hypermobility secondary to hormonal differences or possibly to subtle differences
in trapezium morphology [26]. The etiology of basal joint arthritis is multifactorial
but it is widely believed to involve the loss of the integrity of the palmar oblique
ligament for multiple cadaveric studies have shown a direct correlation between the
stages of OA and integrity of the ligament [31]. As attenuation or hyperlaxity of the
ligament ensues, increased shear forces are placed on the joint leading to inflamma-
tion (synovitis) and degeneration. With progressive disease, the thumb becomes
adducted and subsequently often be dorsoradially subluxated due to the distal pull
of the adductor pollicus muscle. With the thumb adducted, the hand is at a mechani-
cal disadvantage to grip objects and will often pathologically compensate with fixed
metacarpophalangeal hyperextension, the so-called “Z-deformity” (Fig. 4.7) [32].
Patients with basal joint arthritis often experience pain, at times severe, and diffi-
culty performing daily activities such as hand writing or opening jars. Commonly,
patients complain of functional limitations secondarily to decreased strength and
motion. A recent three-dimensional motion analysis found that patients with OA at
the thumb CMC joint had a 50.6% decrease in the circumduction envelope com-
pared to those without arthritis [33].
Nonsurgical treatment for OA of the thumb CMC joint include activity modifi-
cation, nonsteroidal anti-inflammatories, splinting, and corticosteroid injections.
Surgical options include trapeziectomy with/without volar ligament reconstruc-
tion and with/without tendon interposition, metacarpal extension osteotomy,
arthroscopy, resection arthroplasty, implant arthroplasty, and arthrodesis (bone
fusion). The surgical procedure is chosen by extent of disease, age and activity
level of patient, and surgeon preference. For early disease, ligament reconstruction
and metacarpal extension osteotomy are common procedures. Ligament recon-
struction is frequently performed with a strip of the flexor carpi radialis passed
through a drill hole in the first metacarpal and sutured to itself, recreating the volar
oblique and dorsoradial ligaments. Koff et al. [34] performed a cadaveric biome-
chanical study that found ligament reconstruction to reduce laxity in the dorsovo-
lar, radioulnar, and pronosupination directions. Limited evidence is available for
metacarpal extension osteotomy but the biomechanical rationale of its clinical suc-
cess is shifting the joint contact forces to non-arthritic cartilage and reducing joint
laxity [35].
Trapeziectomy with/without volar ligament reconstruction and with/without ten-
don interposition, arthroplasty, and arthrodesis are common surgical options for
more severe disease. All options provide pain relief and improve function though
their biomechanical implications are varied. The native 2 degrees of freedom (DoF)
the trapeziometacarapal joint has is altered with surgical reconstruction. When
arthrodesis is performed, the joint has 0 DoF in contrast to arthroplasty procedures
such as ligament reconstruction tendon interposition (LRTI) and ball and socket
implant have 3 DoF. The change in DoF impacts the joint reaction forces at this
CMC joint. Comparing the above procedures in a biomechanical study, the arthro-
plasty group (3 DoF) had 12 times the joint reaction force than the native joint while
the fusion group (0 DoF) showed the lowest amount of joint forces [24]. Another
biomechanical study found that LRTI had kinematics similar to a native joint though
the pivot point and center of rotation were different. This same study found that the
ball and socket arthroplasty has a fixed axis of rotation which could possibly lead to
a higher rate of wear and loosening [36]. Though the treatment of basal joint arthri-
tis is varied, surgical reconstruction improves pain, increases function, and reliably
produces good to excellent outcomes in 90–95% of cases [32].
Distal radius fractures account for one-sixth of all emergency visits and up to 46%
of all skeletal fractures seen in a primary care setting. One of the most common
complications after a distal radius fracture is malunion, as high as 24% in those
treated in a cast [37]. A malunion is when a fractured bone heals in an abnormal
position. Numerous studies have evaluated the biomechanical effects and load
mechanics of radial shortening, loss of radial inclination and abnormal volar or
dorsal angulation. In the non-fractured distal radius with neutral ulnar variance,
4 Hand and Wrist Biomechanics 101
79% of the mechanical load of the wrist joint is through the radius and 21% is trans-
mitted across the ulna [38]. Wrists with 2 mm or more of ulnar plus variance showed
69% of the transmitted load through the radius and 31% of the load through the ulna
[37]. Pogue et al. [39] showed that greater than 2 mm of radial shortening resulted
in a significant increase in lunate contact area, and at 6–8 mm of radial shortening,
the ulna was noted to impinge on the triquetrum and/or the ulnar aspect of the
lunate. Palmer and Werner [38] found a 25% increase in load to the ulna with a posi-
tive 1 mm change to the ulnar variance.
Malalignment of distal radius fracture in the sagittal plane cause abnormal wrist
biomechanics and decreased functional results in patients [39, 40]. Fernandez [40]
reported that distal radius fracture malunions with greater than 25° dorsal angula-
tion were more likely to be symptomatic and the scaphoid/lunate fossa had increased
loads and more dorsal contact areas compared to the normal wrist. As the distal
radius articular surface moves from its native 10° volar tilt to 45° of dorsal angula-
tion, load through the ulna increases from 21% to 67% of the total load. Additionally,
it has been reported that malunited distal radius fractures may cause dynamic mid-
carpal instability, increased strain on the triangular fibrocartilage, decreased grip
strength, and median nerve neuropathy [41].
Treatment of distal radius malunions should be reserved for only those who are
symptomatic rather than just treating radiographic findings. Surgical options include
corrective osteotomy, wrist denervation, and wrist fusion. In a study with 195 cor-
rective osteotomies for dorsal malunion of the distal radius, the flexion-extension
arc improved by 49° and there was an increase in prono-supination and increase in
grip strength from 29 to 40 kg [41].
Carpal tunnel syndrome (CTS) is the most common compressive neuropathy of the
upper extremity with a prevalence of 2.6–5.8% in the general adult population.
There are more than 500,000 surgical procedures performed each year in the United
States with an economic impact exceeding 2 billion dollars. The carpal tunnel is
formed by the carpal bones dorsally and the transverse carpal ligament (TCL)
volarly, with nine flexor tendons and the median nerve passing through the tunnel.
The median nerve is vulnerable to compression due to the unyielding borders of the
tunnel; when the median nerve experiences compression, its blood flow is reduced
and symptoms of pain and paresthesias ensue in the median nerve distribution. Most
cases of CTS are idiopathic or unknown; however some cases have been attributed
to trauma, fluid overload states (pregnancy, renal disease, congestive heart failure),
and space occupying lesions in the carpal tunnel (ganglion cyst, anomalous tendon,
lipoma), all with the common pathway of compression on the median nerve [42].
Surgical release of the TCL is the standard treatment of choice for CTS when
nonoperative measures fail. When the TCL is released open or endoscopically, there
are many known and even more unknown biomechanical consequences. In an endo-
102 S. Regal et al.
scopic carpal tunnel release, the incision is performed from within the carpal tunnel
and only the TCL is incised, whereas in an open release, the skin, fat, and superficial
fascial layer are incised. The intact TCL helps maintain the concavity of the carpal
arch and acts as a pulley for the flexor tendons of the hand. After surgical release of
the TCL, the volume of the carpal tunnel is increased, thus decreasing the compres-
sion on the median nerve. Magnetic resonance imaging (MRI) studies have shown
the contents of the carpal tunnel to be displaced volarly after carpal tunnel release
(CTR). With anterior displacement of the flexor tendons, their moment arm is effec-
tively increased which theoretically may result in decreased grip strength. Several
in vitro biomechanical studies found that flexor tendon excursion was significantly
increased, as much as 26%, after CTR [43, 44]. Some surgeons lengthen and recon-
struct the TCL after a CTR; small clinical studies have shown less volar displace-
ment of carpal tunnel contents as well as earlier improved grip and pinch strength
compared to CTR without reconstruction. These results are perceived to be caused
by the mechanical advantage of the restoring the flexor pulley [45, 46].
Carpal arch flattening and widening of the carpal bones is another known conse-
quence of CTR, though not all authors agree on the extent of widening or its clinical
effects. Kwon et al. [47] compared carpal arch widths with radiographs before and
6 months after open CTR and found the carpal arch widened on average 1.8 mm
after surgery while previous MRI studies found as much as 2.7 mm of carpal arch
widening after release of the TCL. In a finite element study, Guo et al. [48] reported
after TCL release and an axial load was placed, all carpal bones became more radi-
ally deviated and the contact forces at the midcarpal joint were altered, and yet
long-term biomechanics and its clinical effects of dividing the TCL remain unclear.
Recently, Li and colleagues proposed a nonsurgical treatment of carpal tunnel
syndrome by biomechanically manipulating the carpal arch width as a means to
expand carpal tunnel area and decompress the median nerve [49–51].
References
1. Kijima Y, Viegas SF. Wrist anatomy and biomechanics. J Hand Surg Am. 2009;34(8):1555–63.
2. Kuo CE, Wolfe SW. Scapholunate instability: current concepts in diagnosis and management.
J Hand Surg Am. 2008;33(6):998–1013.
3. Taleisnik J. The ligaments of the wrist. J Hand Surg Am. 1976;1(2):110–8.
4. Katz DA, Green JK, Werner FW, Loftus JB. Capsuloligamentous restraints to dorsal and pal-
mar carpal translation. J Hand Surg Am. 2003;28(4):610–3.
5. Berger RA. The ligaments of the wrist. A current overview of anatomy with considerations of
their potential functions. Hand Clin. 1997;13(1):63–82.
6. Lamas C, Carrera A, Proubasta I, Llusa M, Majo J, Mir X. The anatomy and vascularity of the
lunate: considerations applied to Kienbock’s disease. Chir Main. 2007;26(1):13–20.
7. Gonzalez MH, Mohan V, Elhassan B, Amirouche F. Biomechanics of the digit. J Am Soc Surg
Hand. 2005;5(1):48–60.
8. Navarro A. Luxaciones del carpo. Anales de la Facultad de Medicina. 1921;6:113–41.
9. Craigen MA, Stanley JK. Wrist kinematics. Row, column or both? J Hand Surg Br.
1995;20(2):165–70.
4 Hand and Wrist Biomechanics 103
10. Moojen TM, Snel JG, Ritt MJ, Venema HW, Kauer JM, Bos KE. In vivo analysis of carpal
kinematics and comparative review of the literature. J Hand Surg Am. 2003;28(1):81–7.
11. Kaufmann R, Pfaeffle J, Blankenhorn B, Stabile K, Robertson D, Goitz R. Kinematics of the
midcarpal and radiocarpal joints in radioulnar deviation: an in vitro study. J Hand Surg Am.
2005;30(5):937–42.
12. Li ZM, Kuxhaus L, Fisk JA, Christophel TH. Coupling between wrist flexion-extension and
radial-ulnar deviation. Clin Biomech. 2005;20(2):177–83.
13. Moritomo H, Apergis EP, Garcia-Elias M, Werner FW, Wolfe SW. International Federation of
Societies for Surgery of the Hand 2013 Committee’s report on wrist dart-throwing motion. J
Hand Surg Am. 2014;39(7):1433–9.
14. Wolfe SW, Crisco JJ, Orr CM, Marzke MW. The dart-throwing motion of the wrist: is it unique
to humans? J Hand Surg Am. 2006;31(9):1429–37.
15. Crisco JJ, Coburn JC, Moore DC, Akelman E, Weiss AP, Wolfe SW. In vivo radiocarpal kine-
matics and the dart thrower’s motion. J Bone Joint Surg Am. 2005;87(12):2729–40.
16. Werner FW, Green JK, Short WH, Masaoka S. Scaphoid and lunate motion during a wrist dart
throw motion. J Hand Surg Am. 2004;29(3):418–22.
17. Shah KN, Hodax JD, Katarincic JA. Evaluating a flexor tendon laceration with the tenodesis
effect. J Pediatr. 2016;176:214–214.e1.
18. Hazelton FT, Smidt GL, Flatt AE, Stephens RI. The influence of wrist position on the force
produced by the finger flexors. J Biomech. 1975;8(5):301–6.
19. Li ZM. The influence of wrist position on individual finger forces during forceful grip. J Hand
Surg Am. 2002;27(5):886–96.
20. Volz RG, Lieb M, Benjamin J. Biomechanics of the wrist. Clin Orthop Relat Res.
1980;149:112–7.
21. Bade H, Schubert M, Koebke J. Functional morphology of the deep transverse metacarpal liga-
ment. Ann Anat. 1994;176(5):443–50.
22. Dzwierzynski WW, Matloub HS, Yan JG, Deng S, Sanger JR, Yousif NJ. Anatomy of the
intermetacarpal ligaments of the carpometacarpal joints of the fingers. J Hand Surg Am.
1997;22(5):931–4.
23. Garcia-Elias M, An KN, Cooney WP 3rd, Linscheid RL, Chao EY. Stability of the transverse
carpal arch: an experimental study. J Hand Surg Am. 1989;14(2 Pt 1):277–82.
24. Domalain MF, Seitz WH, Evans PJ, Li ZM. Biomechanical effect of increasing or decreas-
ing degrees of freedom for surgery of trapeziometacarpal joint arthritis: a simulation study. J
Orthop Res. 2011;29(11):1675–81.
25. Hollister A, Buford WL, Myers LM, Giurintano DJ, Novick A. The axes of rotation of the
thumb carpometacarpal joint. J Orthop Res. 1992;10(3):454–60.
26. Van Heest AE, Kallemeier P. Thumb carpal metacarpal arthritis. J Am Acad Orthop Surg.
2008;16(3):140–51.
27. Abboud JA, Beredjiklian PK, Bozentka DJ. Metacarpophalangeal joint arthroplasty in rheu-
matoid arthritis. J Am Acad Orthop Surg. 2003;11(3):184–91.
28. Bain GI, Polites N, Higgs BG, Heptinstall RJ, McGrath AM. The functional range of motion
of the finger joints. J Hand Surg Eur Vol. 2015;40(4):406–11.
29. Allison DM. Anatomy of the collateral ligaments of the proximal interphalangeal joint. J Hand
Surg Am. 2005;30(5):1026–31.
30. Chen J, Tan J, Zhang AX. In vivo length changes of the proximal interphalangeal joint proper
and accessory collateral ligaments during flexion. J Hand Surg Am. 2015;40(6):1130–7.
31. Halilaj E, Rainbow MJ, Moore DC, Laidlaw DH, Weiss AP, Ladd AL, Crisco JJ. In vivo
recruitment patterns in the anterior oblique and dorsoradial ligaments of the first carpometa-
carpal joint. J Biomech. 2015;48(10):1893–8.
32. Barron OA, Glickel SZ, Eaton RG. Basal joint arthritis of the thumb. J Am Acad Orthop Surg.
2000;8(5):314–23.
33. Gehrmann SV, Tang J, Li ZM, Goitz RJ, Windolf J, Kaufmann RA. Motion deficit of the thumb
in CMC joint arthritis. J Hand Surg Am. 2010;35(9):1449–53.
104 S. Regal et al.
34. Koff MF, Shrivastava N, Gardner TR, Rosenwasser MP, Mow VC, Strauch RJ. An in vitro
analysis of ligament reconstruction or extension osteotomy on trapeziometacarpal joint stabil-
ity and contact area. J Hand Surg Am. 2006;31(3):429–39.
35. Ladd AL, Crisco JJ, Hagert E, Rose J, Weiss AP. The 2014 ABJS Nicolas Andry Award: The
puzzle of the thumb: mobility, stability, and demands in opposition. Clin Orthop Relat Res.
2014;472(12):3605–22.
36. Imaeda T, Cooney WP, Niebur GL, Linscheid RL, An KN. Kinematics of the trapeziometacar-
pal joint: a biomechanical analysis comparing tendon interposition arthroplasty and total-joint
arthroplasty. J Hand Surg Am. 1996;21(4):544–53.
37. Bu J, Patterson RM, Morris R, Yang J, Viegas SF. The effect of radial shortening on wrist joint
mechanics in cadaver specimens with inherent differences in ulnar variance. J Hand Surg Am.
2006;31(10):1594–600.
38. Palmer AK, Werner FW. Biomechanics of the distal radiulnar joint. Clin Orthop.
1984;187:26–35.
39. Pogue DJ, Viegas SF, Patterson RM, Peterson PD, Jenkins DK, Sweo TD, Hokanson
JA. Effects of distal radius fracture malunion on wrist joint mechanics. J Hand Surg Am.
1990;15(5):721–7.
40. Fernandez DL. Correction of post-traumatic wrist deformity in adults by osteotomy, bone-
grafting, and internal fixation. J Bone Joint Surg Am. 1982;64(8):1164–78.
41. Prommersberger KJ, Pillukat T, Muhldorfer M, van Schoonhoven J. Malunion of the distal
radius. Arch Orthop Trauma Surg. 2012;132(5):693–702.
42. Cranford CS, Ho JY, Kalainov DM, Hartigan BJ. Carpal tunnel syndrome. J Am Acad Orthop
Surg. 2007;15(9):537–48.
43. Brown RK, Peimer CA. Changes in digital flexor tendon mechanics after endoscopic and open
carpal tunnel releases in cadaver wrists. J Hand Surg Am. 2000;25(1):112–9.
44. Kiritsis PG, Kline SC. Biomechanical changes after carpal tunnel release: a cadaveric model
for comparing open, endoscopic, and step-cut lengthening techniques. J Hand Surg Am.
1995;20(2):173–80.
45. Netscher D, Mosharrafa A, Lee M, Polsen C, Choi H, Steadman AK, Thornby J. Transverse
carpal ligament: its effect on flexor tendon excursion, morphologic changes of the carpal
canal, and on pinch and grip strengths after open carpal tunnel release. Plast Reconstr Surg.
1997;100(3):636–42.
46. Seitz, WH, A. Lallb Open carpal tunnel release with median neurolysis and Z-plasty recon-
struction of the transverse carpal ligament. Current Orthopaedic Practice. 2013;24(1):53–7.
47. Kwon YE, Gong HS, Shin HS, Lee HR, Kim KH, Baek GH. Evaluation of carpal arch widen-
ing and outcomes after carpal tunnel release. J Hand Surg Am. 2017;42(2):113–7.
48. Guo X, Fan Y, Li ZM. Effects of dividing the transverse carpal ligament on the mechanical
behavior of the carpal bones under axial compressive load: a finite element study. Med Eng
Phys. 2009;31(2):188–94.
49. Li ZM, Tang J, Chakan M, Kaz R. Carpal tunnel expansion by palmarly directed forces to the
transverse carpal ligament. J Biomech Eng. 2009;131(8):081011.
50. Li ZM, Gabra JN, Marquardt TL, Kim DH. Narrowing carpal arch width to increase cross
sectional area of carpal tunnel—a cadaveric study. Clin Biomech. 2013;28(4):402–7.
51. Marquardt TL, Evans PJ, Seitz WH Jr, Li ZM. Carpal arch and median nerve changes
during radioulnar wrist compression in carpal tunnel syndrome patients. J Orthop Res.
2016;34(7):1234–40.
Chapter 5
Biomechanics of the Elbow
1 Anatomy
The elbow, a complex joint, is composed of the ulna, the radial, and the distal
humerus. The elbow joint is formed by the three articulations, which include ulno-
humeral, radiocapitellar, and proximal radioulnar joint in the elbow (Fig. 5.1a, b).
The distal humerus has several important anatomical landmarks including trochlea,
capitellum, medial epicondyle, lateral epicondyle, coronoid fossa, radial fossa, and
coronoid process in the anterior view (Fig. 5.1c) and coronoid process in the poste-
rior view. These anatomical landmarks in the distal humerus play an important role
Lateral epicondyle
Medial epicondyle
Capitellum
Radial head
Trochlea
Coronoid process
Trochlea
Medial epicondyle
Lateral epicondyle
Olecranon
Radial head
Capitellum
Trochlea
Fig. 5.1 (a) Anterior view of elbow joint. (b) Posterior view of elbow joint. (c) Anterior view of
distal humerus
5 Biomechanics of the Elbow 107
to stabilize the elbow joint during the elbow extension and flexion with a wide range
of motion. The coronoid fossa and radial fossa provide the osseous stability and
prevent anterior impingement during flexion of the elbow joint. The olecranon fossa
also provides the osseous stability and prevent posterior impingement during exten-
sion of the elbow joint.
The ulnohumeral joint is formed by the trochlea of the humerus and olecranon of
the ulna (Fig. 5.2a) to stabilize the elbow joint during extension and prevent poste-
rior translation. The function of the ulnohumeral joint, hinge joint, is the movement
of flexion and extension of the arm. The radiohumeral joint, ball-and-socket joint, is
formed by the convex structure of capitellum and concave structure of the radial
head (Fig. 5.2b) to stabilize the elbow joint during flexion and be a valgus stabilizer
of the elbow joint. The proximal radioulnar joint is formed by the lesser sigmoid
notch of ulna and the margin of the radial head. The function of the proximal radio-
ulnar joint and the radiocapitellar joint is the movement of supination and pronation
of the forearm. The radial head is covered with the large area of articular cartilage
which is nearly 270–280° along the proximal radioulnar joint and the radiocapitel-
lar joint.
a Olecranon
Sagittal ridge
Greater sigmoid notch
Sublime tubercle
Lesser sigmoid notch
Coronoid process
b Radial head
Radial neck
Radial tuberosity
Fig. 5.2 (a) Anterior view of ulna. (b) Anterior view of radial
108 S.-Y. Lee and F.-C. Su
Carrying angle, a small degree of cubitus valgus, occurred at the forearm full
extension and supination due to the noncollinearity between the long axis of ulnar
and long axis of humerus. The average carrying angle is 13–16° for women and
11–14° for men. During the movement of elbow joint flexion, the carrying angle
will decrease and disappear due to the colinearity between the long axis of ulnar and
long axis of humerus.
According to movement functions of muscles around the elbow, they can be classi-
fied into the elbow extensor muscles, elbow flexor muscles, wrist flexor muscles,
and wrist extensor muscles. These muscles play an important role in the dynamic
stabilizers to prevent the elbow subluxation and reinforce the osseous stability of the
elbow. These muscles are innervated by the median nerve, radial nerve, ulnar nerve,
and musculocutaneous nerve (C5–C7), respectively.
The elbow extensor muscles are triceps brachii muscles and anconeus muscle.
The triceps brachii muscle is the main elbow flexor muscle and its maximal muscle
is efficient when elbow is flexed at 20–30°. The anconeus muscle has an assistant
elbow extensor role during the elbow extension and a major dynamic stabilizer to
restrain the posterolateral and varus force on the elbow joint. The elbow flexors are
brachialis, brachioradialis, biceps brachii, and pronator teres. The short head of
biceps brachii is a main elbow flexor muscle and a stronger elbow flexor muscle
compared to the brachialis muscle. Brachioradialis has a great mechanical advantage
of any elbow flexor muscles; however, brachialis has a poor mechanical advantage
as it is near the axis of rotation. The common extensor tendon, which is the origin of
the wrist extensor muscles, consists of the extensor carpi radialis brevis, extensor
carpi ulnaris, extensor digitorum communis, and extensor digiti minimi. The wrist
flexor muscles in the medial side of elbow are flexor carpi radialis, flexor carpi ulna-
ris, flexor digitorum superficialis, and flexor digitorum profundus. The common
extensor tendon, which is the major wrist extensor muscles, consists of the flexor
carpi radialis, flexor carpi ulnaris, flexor digitorum superficialis, pronator teres, and
palmaris longus. The wrist extensor muscles in the lateral side of elbow are extensor
carpi radialis longus, extensor carpi radialis brevis, extensor carpi ulnaris, extensor
digitorum communis, extensor digiti minimi, and anconeus. The forearm pronation
muscle is pronator teres and the forearm supination muscles are supinator and long
head of biceps brachii. When the arm is the extreme supination posture, brachiora-
dialis can assist in the pronation of the forearm.
All of wrist flexor and pronator teres are innervated by the median nerve, except
for the flexor carpi ulnaris and little and ring finger flexor digitorum profundus,
which are innervated by the ulnar nerve (Fig. 5.3a). The elbow extensor muscles
include triceps brachii in the lateral and medial heads and anconeus. All of wrist
extensor muscles, brachioradialis and supinator are innervated by the radial nerve
(Fig. 5.3b). Biceps brachii is innerved by the musculocutaneous nerve (C5–C7)
and brachialis is innerved by the musculocutaneous nerve (C5–C6).
5 Biomechanics of the Elbow 109
Ulnar nerve
Medial nerve
Pronator teres
b Radial nerve
Brachioradialis muscle
The elbow joint is enclosed by the joint capsule, which is surrounded around the
olecranon, the coronoid fossa, and the radial fossa, except for the humeral epicon-
dyle (Fig. 5.4). The joint capsule, a secondary static stabilizer, has a laxity on the
anterior and posterior side of elbow joint to provide the appropriate movement of
elbow joint during the flexion and extension. The brachialis muscle and triceps bra-
chialis muscles can prevent the joint capsule to pinching in the joint and maintain
the sufficient tension on the capsule during extension and flexion.
The collateral ligaments are fused with the joint capsules and can be divided into
the lateral collateral ligament and medial collateral ligament. The collateral liga-
ments, a primary static stabilizer, cross the articular joints of elbow. The collateral
ligaments have the tension to restrict the varus motion, the valgus motion, and the
axial rotation motion. The function of lateral collateral ligament complex is to
maintain the stability during the posterolateral force on the elbow joint and hinder
110 S.-Y. Lee and F.-C. Su
varus stress on the elbow joint. The function of medial collateral ligament is to
maintain the stability during the posteromedial force on the elbow joint and hinder
valgus stress on the elbow joint. The lateral collateral ligament complex is com-
posed of lateral ulnar ligament, the radial collateral ligament, and the annular liga-
ment (Fig. 5.5). The annular ligament, one of the lateral collateral ligament complex,
Annular ligament
Anterior bundle
Posterior bundle
Transverse bundle
enclosed the radial head, and the insertion/origin of the annular ligament is located
at the ulna to maintain the stability of forearm rotation.
The medial collateral ligament complex is formed by the anterior bundle, poste-
rior bundle, and transverse ligament (Cooper ligament) (Fig. 5.6). The anterior
bundle of the medial collateral ligament is formed by thick parallel fibers [1], the
posterior bundle of the medial collateral ligament is formed by the fan-shaped cap-
sular fibers [2], and the transverse ligament is formed by the horizontal capsular
fibers [3]. The anterior bundle of the medial collateral ligament is attached to the
antero-inferior aspect of the medial epicondyle and provide the constant tension
during the elbow flexion and extension. The anterior bundle can be further separated
to the anterior band and posterior band. The anterior band of the anterior bundle of
medial collateral ligament shows an isometric strain pattern during the elbow flex-
ion and the posterior band of the anterior bundle of medial collateral ligament has
increased its strain value with the increase of elbow flexion [4]. The difference in
strain pattern between the anterior and posterior bands of the anterior bundle of the
medial collateral ligament is due to the insertion point in the elbow joint [4]. The
posterior bundle of the medial collateral ligament has a tension during the elbow
flexion to restrain the valgus stress [5].
2 Function
of motion for the elbow joint consists of 0° full extension and 146° extension. The
functional range of motion for the elbow joint consists of 30° flexion and 130° exten-
sion. Recently, researchers have investigated the kinematics of elbow joint by nonin-
vasive three-dimensional technique [6–9]. The instant screw axis of rotation of
elbow flexion-extension is a changeable circular path with the elbow flexion. During
the movement of elbow flexion, the path of axis of rotation showed a counterclock-
wise pattern and a conical shape on the lateral condyle and this path of axis of rota-
tion crossed and converged at the medial facet of the trochlea [6]. The average axis
of elbow flexion-extension is a line from the anterior aspect of the medial epicondyle
to the center of trochlea and the capitellum which has 4–8° valgus relative to the long
axis of the humerus. During the movement of flexion, ulna changes its position with
the anterior glide, lateral shift, and varus rotation relative to the humerus [7, 8]. The
ulna has the internal rotation from the full extension to 90° flexion and external rota-
tion from the 90° flexion to the full flexion relative to the humerus [7, 8]. During the
movement of extension, particularly in the last 5–10°, the ulna has the posterior glide
relative to the humerus. The humerus has significant compression force on the ulna
with the elbow 90° flexion because the greater sigmoid notch has a nearly posterior
30° respective to the axis of the ulna (Fig. 5.7) and the capitellum/trochlea has a near
anterior 30° respective to the axis of the humerus [10, 11].
The proximal radioulnar and radiocapitellar articulations allow motion of fore-
arm pronation and supination in the transverse plane. The normal range of motion
for the forearm pronation-supination consists of 71° pronation and 81° supination.
The axis of forearm rotation is a line from the center of the radial head to the distal
ulna. The forearm pronation-supination plays an important role in the daily activ-
ity which required 50° pronation and 50° supination to perform the functional
movement. Previous studies investigated the motion of radius and ulna during the
movement of forearm pronation-supination via two-dimensional computed tomog-
raphy [12] and three-dimensional computational model [9]. From supination to
pronation, the radius showed a varus rotation, internal rotation, and extension rela-
tive to humerus and ulna showed a valgus rotation [9]. The center of radial head
has anterior, proximal, and lateral translation with the forearm pronation. The con-
30°
Fig. 5.7 Angle between greater sigmoid notch and axis of the ulna
5 Biomechanics of the Elbow 113
tact area of radiocapitellar has a large congruency in the forearm pronation and the
contact area of proximal radioulnar joint has a large congruency in the forearm
supination [9].
The elbow joint plays an important role in the daily activity of the upper extremity.
Understanding the force distribution in the elbow joint is essential to design joint
replacement and investigate the effect of osteotomy. With the 15° elbow flexion with
full pronation and wrist extension, the axial compressive load of 100 N transmitted to
the radiocapitellar joint is approximately 61% loading distribution [13]. With the
increasing weight of axial loading, the axial load distribution through the radiocapitel-
lar joint has decreased to 55% [13]. When the axial compressive load of 160 N is
transmitted to the radiocapitellar, there is nearly 90 mm2 contact area at the radiocapi-
tellar joint at 0° of flexion [14]. The contact area at the ulnohumeral joint increased
from the elbow extension to elbow flexion [15, 16]. The contact area at the radiocapitel-
lar joint increased with the increase of angle of elbow flexion [17]. In general, the peak
contact pressure of radial head does not exceed 5 MPa and the daily activities applied
approximately 100 N force at the radiocapitellar articulation [18]. The contact pressure
of the humeroulnar joint increased from the elbow extension to elbow flexion [15, 16].
The free-body diagram is used to estimate the joint reaction force by the equilibrium
equations. The equilibrium equations assume that the sum of forces and sum of
moments acting on the elbow joint are zero. The calculation of elbow joint reaction
force needs to consider the weight of external object, weight of forearm and arm, the
distance between center of mass of forearm/arm and the center of rotation of the joint,
the line of action of muscle, and the angle of forearm and arm in the free-body dia-
gram. The biceps and brachialis muscle was considered as the primary elbow flexors
and the triceps is considered as the primary elbow extensor [10, 19]. Regarding the
dynamic movement that generates the acceleration in the limbs, the joint force and
joint moment at the elbow can be calculated by the inverse Newton-Euler method.
Recently, most of the studies investigated the joint force and moment at the elbow
joint during performing the daily activities of the upper extremity [20, 21]. The block
to head height generates the flexion moment and external rotation moment of the
elbow joint and the elbow joint force along the humeral longitudinal axis and rotation
axis that is perpendicular to the humeral longitudinal axis than other upper extremity
activities [21]. The reach to head side and back are the high demand of elbow flexion,
forearm pronation, elbow extension moment, and elbow internal rotation moment [21].
114 S.-Y. Lee and F.-C. Su
A stable joint provides the normal functional activity of the elbow. The dynamic and
static factors contribute to the joint stability by anatomy of the joint, surrounding liga-
ment, joint capsule, and muscles. The bony geometry of elbow joint contributes much
to the joint stability due to its congruous articulation. The radial head and coronoid
process are the major articulation stabilizers to prevent the posterior subluxation or
dislocation. A previous study showed that at least 50% coronoid height is needed to
provide the stability of elbow joint under the valgus force and foramen supination
[22]. The radial head is the minor articulation stabilizer relative to the coronoid pro-
cess [11]. The ligament, joint capsule, and articulation form the static stabilizer of the
elbow joint. The primary static stabilizer consists of ulnohumeral joint and anterior
bundle of medial collateral ligament and the secondary stabilizer consists of radio-
capitellar joint, common flexor tendon, common extensor tendon, and the joint cap-
sule. The anterior joint capsule can be against the joint distraction, joint hyperextension,
valgus stress, and the posterior joint capsule can against the joint hyperflexion and
posterior directed force [19]. Muscles are the dynamic stabilizer across the elbow
joint because muscle contraction can generate the compressive force. The elbow flex-
ors can generate the varus moment to resist the valgus force and elbow extensors can
resist the varus force of elbow joint [23]. The muscles contributing for the valgus
stability are flexor carpi ulnaris and flexor digitorum superficialis muscle [24].
Collateral ligaments are the important soft tissue of the elbow joint to act as the
static stabilizer of elbow joint. In the medial side of elbow joint, the medial collat-
eral ligament is composed of multiple bundles against the valgus force and postero-
medial (internal) rotatory instability. The anterior bundle of the medial collateral
ligament is the strongest bundle of the medial collateral ligament to prevent the
valgus and posteromedial rotatory instability [5, 25]. Additionally, the anterior bun-
dle of the medial collateral ligament is the priority reconstruction of the medial
collateral ligament in clinics because it is the primary restrain and stabilizer [25,
26]. The anterior bundle of medial collateral ligament is composed of the anterior
and posterior bands. The anterior band of anterior bundle of the medial collateral
ligament is the primary restraint to provide the stability in the valgus force [2]. At
120° elbow flexion, the anterior and posterior bands of the anterior bundle of medial
collateral ligament are the co-primary restraints when applying the valgus force [5].
Therefore, the anterior band of anterior bundle of medial collateral ligament may
have isolated injury between 0° and 90° elbow flexion. When the elbow flexion is
greater than 90°, both the anterior and posterior bands of anterior bundle of medial
collateral ligament may be injured at the same time [5]. Recently, the poster bundle
of medial collateral ligament has been considered as a stabilizer, independent of the
integrity of anterior bundle of medial collateral ligament, in the posteromedial
elbow instability. After transection of posterior bundle of medial collateral ligament,
the proximal ulnohumeral joint gapping and torsion angle of ulnohumeral joint
increase as the elbow flexion increases [27] with the intact anterior bundle of medial
collateral ligament.
5 Biomechanics of the Elbow 115
In the lateral side of elbow joint, the lateral ulnar collateral ligament is attached
to the superior crest of the ulna, which is blended with the distal annular ligament at
the proximal of ulna [3]. The lateral ulnar collateral ligament is the primary con-
straint in the lateral collateral ligaments to provide the stability of the ulnohumeral
joint during the varus force and external rotated force [28]. A previous study showed
that one of the distal sections of lateral ulnar collateral ligament or annular ligament
was dissected which generates minor laxity of the elbow joint during the applied
varus and external rotated forces [29]. Both the lateral collateral ligament and lateral
radial collateral ligament were dissected which result in the significant rotatory lax-
ity [29]. The entire transection of the lateral collateral ligament at the insertion or
origin causes the maximal laxity [29]. The maximal external rotatory laxity was
20.6° at the 110° elbow flexion [29]. According to a previous study, the anterior
portion of the lateral collateral ligament causes the significant laxity when applied
the varus and external rotational forces at 80° elbow flexion [30].
To sum up, previous studies showed that articulation is the primary stabilizer
under varus stress and the medial collateral ligament is the primary stabilizer under
valgus stress in elbow flexion [11, 19, 26]. The lateral collateral ligament, capsule,
and osseous structure provide 9%, 13%, and 75%, respectively, for resisting the
varus force in the 90° flexion of elbow joint [31]. The medial collateral ligament,
anterior capsule, and articulation provide 54%, 10%, and 36%, respectively, for
resisting the valgus force in the 90° flexion of elbow joint [31]. The anterior bundle
of medial collateral ligament is the primary stabilizer and the radial head is a sec-
ondary stabilizer of the elbow to resist the valgus force [26]. In the elbow extension,
articulation is the primary stabilizer during the applied varus force of the elbow
joint, and medial collateral ligament, capsule, and articulation have a similar contri-
bution to stabilize the elbow joint during the applied valgus force of the elbow joint
[11, 19, 26]. The medial collateral ligament, anterior capsule, and articulation pro-
vide 31%, 38%, and 31%, respectively, for resisting the valgus force in the full
extension of elbow joint [31]. The lateral collateral ligament, capsule, and osseous
structure provide 14%, 32%, and 55%, respectively, for resisting the varus force in
the full extension of elbow joint [31].
3 Pathomechanics of Elbow
The valgus and varus loads influence the pattern of contact in the elbow joint, which
is importance for the valgus instability, posterolateral instability, and varus postero-
medial instability in the clinics. Elbow instability pattern is based on the injury
duration, involvement of articulation, the direction of laxity, the extent of laxity, and
116 S.-Y. Lee and F.-C. Su
with or without fracture of elbow joint [32]. The injury of medial collateral liga-
ment, especially anterior bundle, causes the valgus instability after the overuse
injury (chronic injury) [33] or single acute traumatic injury [34]. The disruption of
lateral collateral ligament is the main factor to cause the posterolateral rotatory
instability under an axial load combining valgus load and the forearm supination
after a traumatic injury [33]. The resultant force transmits from the lateral side to
medial side causing the progressive structure injury of the elbow joint. The postero-
lateral rotatory instability is the frequent recurrent instability of the elbow [33]. The
varus posteromedial rotatory instability is associated with the anteromedial facet of
the coronoid fracture, a rupture of the lateral collateral ligament and posterior bun-
dle of the medial collateral ligament under a varus load with the forearm pronation
[35, 36]. Therefore, it is important to understand the contributions of soft tissues and
bony geometry in the stability of elbow joint.
Previous studies have used the cadaver to investigate the extent of valgus laxity
at the specific joint angle by sectioning the different degree of medial collateral liga-
ment and articulation of elbow [2, 26]. Table 5.1 shows the relationship between
valgus laxity and medial collateral ligament, radial head, and muscle force of the
elbow joint. The maximum valgus laxity occurred at the 70–90° elbow flexion with
anterior bundle of medial collateral ligament release and radial head intact [37]. The
entire medial collateral ligament release with the radial head intact causes the 31.2
valgus laxity at the 90° elbow flexion [2]. Additionally, the entire medial collateral
ligament release with radial head removal results in the gross valgus and internal
rotational instability [26].
Posterolateral rotatory instability is a recurrent injury and the most common pat-
tern of elbow instability [32, 38, 39]. Posterolateral rotatory instability commonly
occurs at the tear or disruption from the lateral collateral ligament to the ulnar col-
lateral ligament leading to the elbow dislocation after a traumatic injury [30]. The
tennis elbow or iatrogenic injury from prior lateral elbow surgery may be factors to
cause the posterolateral rotatory instability. Posterolateral rotatory instability is
associated with the posterolateral subluxation or dislocation of the radial head and
proximal ulna away from the humerus due to the laxity of the lateral collateral liga-
ment [40]. Posterolateral rotatory instability can be further divided into three stages
according to the degree of soft tissue disruption and displacement of elbow joint
(Table 5.2).
Posteromedial rotatory instability occurred under the varus load and is associ-
ated with the subluxation and dislocation of the elbow joint [41]. The posteromedial
rotatory instability causes the articular incongruity because the radius and ulnar
rotate posteromedially which is off the articulation of humerus and this injury can
in turn cause the increase of contact pressure in the coronoid surface [41]. The
anteromedial facet fracture [42–44], tear of the lateral ulnar collateral ligament
injury [41], and posterior bundle of the medial collateral ligament injury [27] are
associated with the posteromedial rotatory instability. A previous study indicated
that the isolated sectioning of the posterior bundle of the medial collateral ligament
can cause the increase of 4.5 and 2.7 ulnar rotations at 60° and 90° elbow flexion,
respectively, to generate the posteromedial instability of the elbow joint [27].
5 Biomechanics of the Elbow 117
Table 5.1 The relationship between valgus laxity and medial collateral ligament, radial head, and
muscle force of the elbow joint
UCL intact Radial head Without 5 Normal valgus laxity 10–20° elbow flexion
intact muscle [26]
loading
UCL intact Radial head With 2.6 Valgus laxity [26]
intact muscle
loading
AOL release Radial head • 14 Valgus laxity • 70° elbow flexion [26]
intact • Maximum internal • 60° elbow flexion [2]
rotational laxity
AOL release Radial head 50 Valgus laxity [26]
removal
AB of AOL Radial head • 11.7 Valgus laxity • 30° elbow flexion [2]
release intact • Maximal internal • 40° elbow flexion [2]
rotational laxity
PB of AOL Radial head Little change in valgus [2]
release intact and internal rotation
UCL intact Radial head Little change in valgus
removal and internal rotation
POL release Radial head Little change in valgus [26]
removal or and internal rotation
not
AB Radial head Did not induce more [2]
release + POL intact laxity
release
AOL Radial head • Completely unstable 90° elbow flexion (full
release + POL intact • 31.2 Valgus laxity flexion and full extension
release can confer some stability
with joint surface) [2]
Entire UCL Radial head Gross valgus and • Increase of internal
release removal internal rotational rotational laxity
instability • Increase of elbow
flexion [26]
Entire UCL Radial head Muscle Restore some stability [26]
release removal force (compared to a
UCL-deficient elbow)
Besides, the ulnohumeral joint gapping increase with the increase of elbow flexion
after the sectioning of the posterior bundle of the medial collateral ligament.
According to the findings of contact pressure of the coronoid surface, the anterome-
dial fracture with the lesion of lateral collateral ligament which is the involvement
of posteromedial rotatory instability has the high contact pressure value at the edge
of coronoid fracture with lower contact area compared to the intact elbow [41]. The
anteromedial fracture with the posteromedial rotatory instability and tear of lateral
collateral ligament causes a significant subluxation and decreases of elbow flexion
compared to the coronoid fracture with lesion of posterior medial collateral liga-
ment [41]. Therefore, the lateral collateral ligament is an important factor for pre-
venting the subluxation in the posteromedial rotatory instability. According to
previous studies, posterior medial collateral ligament can contribute part of the sta-
bility under the resistance of varus and internal rotational instability (posteromedial
elbow instability) [27, 45].
3.1.2 Elbow Fracture
Elbow is the second dislocated major joint of the upper extremity in adults [46] after
a traumatic injury. Posterior dislocation, a common disorder in the elbow joint,
causes the injury of soft tissues including tear of lateral collateral ligament, the dif-
ferent extent injury of the ulnar collateral ligament and flexor-pronator muscles of
elbow joint [42]. The valgus posterolateral rotatory load is a more common mecha-
nism to cause the elbow dislocation and fracture than varus posteromedial rotatory
load [47]. When the greater sigmoid notch has posterior and inferior subluxation
under a valgus posterolateral rotatory load, a shearing force transmits to the tip of
coronoid to induce the coronoid fracture [48, 49]. The severe valgus posterolateral
rotatory injury eventually results in the terrible triad injury, which causes the elbow
dislocation with radial head and coronoid fractures and ruptures the medial collat-
eral ligament.
Radial head fracture (Fig. 5.8) is associated with the posterior dislocation of the
elbow joint. The 10–15% patients with posterior dislocation have combined with
the radial head fracture or coronoid fracture [50]. Coronoid fracture which is a fre-
quent fracture type of the elbow joint [51] can be divided into three types including
transverse fractures of the tip, fracture involving the anteromedial facet, and frac-
tures at the coronoid base [43] (Fig. 5.9). In the transverse coronoid fracture with tip
subtype II and 50% coronoid height of the elbow joint, the transection of posterior
bundle of the medial collateral ligament causes the significant increase of ulnohu-
meral joint gapping during the elbow flexion [52]. However, the reconstruction of
the posterior bundle of the medial collateral ligament only can decrease the ulnohu-
meral joint gapping and forearm rotation at 90° elbow flexion to restore posterome-
dial stability [52]. It indicated that the intact posterior bundle of the medial collateral
ligament can provide some of the posteromedial stability after coronoid fracture
without fixing [52]. The anteromedial fracture of the coronoid can be further subdi-
vide to three subtypes according to the injury site of the anteromedial coronoid face
5 Biomechanics of the Elbow 119
Tip
Anteromedial
Radial head
Coronoid process
Basal
Proximal ulna
including (1) injury of the anteromedial rim, (2) injury of the anteromedial rim and
tip, (3) injury of the anteromedial rim, tip, and the whole sublime tubercle [43]
(Fig. 5.6). Both the lateral collateral ligament and the posterior bundle of the medial
collateral ligament have tear associated with the anteromedial fractures [43, 44, 53].
The coronoid fracture can increase the forearm rotation angle, which is associated
with the posteromedial instability of the elbow joint [43, 44, 52]. Recently, many
researchers have investigated the effect of extent injury of anteromedial fractures
with lesions of lateral collateral ligament or the posterior bundle of the medial col-
lateral ligament on the posteromedial elbow instability [43, 44].
In clinics, the appropriate reconstruction and fixation can restore the range of
motion and stability of elbow joint, increase the functional outcomes, and reduce
the pain. The valgus posterolateral rotatory injury, terrible triad injury, causes the
elbow dislocation combined with the lateral collateral ligament rupture and radial
head and coronoid process fraction. The management of the valgus posterolateral
rotatory includes fixation of the coronoid, fixation or replacement of the radial
head, repair of the lateral ulnar collateral ligament, possible repair of the medial
120 S.-Y. Lee and F.-C. Su
collateral ligament, and external fixation of elbow as elbow remains unstable. The
coronoid fracture can be repaired by the suture lasso technique for the small frag-
ment of coronoid and by the lag screws with/without mini fragment plate for the
large fragments of the coronoid [49]. The partial radial head fracture can be fixed
with the screws [54, 55] or mini fragment plate [56]. The radial head replacement
can be used to fix the radial head for more than three fragments and the prosthesis
of radial head needs 2 mm distal to the coronoid [57]. In clinics, the lateral collat-
eral ligament was commonly reconstructed by grasping type stitch to restore the
elbow stability and prevent the subluxation under a valgus force [58] because the
valgus posterolateral rotatory injury causes the detachment and avulsion of lateral
collateral ligament from its attachment in the humerus. Recently, some researchers
have indicated that the soft tissue injury may occur initially from the medial side of
elbow joint [47, 59]. According to the MRI findings, posterolateral direction results
in distractive injury to cause the displacement of the soft tissue in the ulnar side
from the insertion point and detachment of soft tissue in the lateral side near the
original point [47]. Therefore, the anterior bundle of medial collateral ligament
may be the initial injury site at some acute injury situation with posterolateral dis-
location [59].
The varus posteromedial rotatory injury causes the avulsion of the lateral col-
lateral ligament and fracture of anteromedial facet of the coronoid. The surgical
fixation was commonly used to repair the anteromedial facet fracture by internal
fixation [44] through buttress plate and screw to fix the anteromedial facet fragment.
There are many surgical approaches including flexor carpi ulnaris-splitting approach,
over-the-top approach [60], posterior incision following the development of a
medial skin flap [61], and the floor of the cubital tunnel approach following trans-
portation of the ulnar nerve to repair the anteromedial facet fracture [62].
Anterior olecranon fracture dislocation is a high-energy injury in a dorsal aspect
of the forearm at the elbow 90° flexion, causing the anterior dislocation of the
radius and ulnar [63] with a large simple coronoid fragment [35]. Posterior
Monteggia fracture is a low-energy injury causing the fracture at the base of the
coronoid process. About 50% patients with posterior Monteggia fracture has com-
bined with radial head fracture and avulsion of the lateral collateral ligament com-
plex [64].
The osteoarthritic elbow is a degenerative disease that cause osteophytes at the cor-
onoid, olecranon, and radial head, decrease of the joint space of the ulnotrochlear
and radiocapitellar, loose bodies, soft tissue contracture, and synovitis [65, 66]. The
post-traumatic osteoarthritic elbow is in majority of osteoarthritic elbow due to the
elbow dislocation, radial head fracture, and coronoid fracture to cause the cartilage
lesion and post-traumatic deformity [67]. The post-traumatic osteoarthritic elbow
commonly begins from the radiocapitellar joint to ulnohumeral joint with soft tissue
destruction [65, 68, 69]. The osteoarthritic elbow can result in the joint deformity to
5 Biomechanics of the Elbow 121
restrict the joint mobility and elbow stiffness in the advanced stage [66, 68, 69]. The
treatment of osteoarthritic elbow includes the conservative treatment and surgical
treatment. The conservative treatment is the first choice and the surgical treatment
is used when there is failure of nonsurgical treatment [70] and significant impair-
ment of the daily actives (range of motion of elbow less than 30–130°) [68]. The
surgical treatment of osteoarthritic elbow includes arthroscopic approaches [70–72]
and open techniques to restore the joint mobility and relieve the pain. The
arthroscopic debridement focuses on removing the osteophytes, inflamed soft tis-
sue, and loose bodies, releasing the tight capsular, and resecting the radial head [70,
71]. The arthroscopic debridement can restore the functional elbow mobility,
improve the clinical scores, decrease the severity of elbow stiffness, and relieve the
pain for primary and post-traumatic osteoarthritic elbow [71]. According to previ-
ous studies, the early and mild osteoarthritic elbow with the joint congruence can
use the arthroscopy treatment and this treatment can provide the good postoperative
functional and clinical outcomes at follow-up 24 months [70, 71, 73]. The radial
head excision is commonly used in the patients with osteoarthritic elbow involved
in the radiocapitellar arthritis. Patients who underwent radial head excision alone
had a greater return of range of motion and clinical outcomes after mean follow-up
52 months [74]. However, the radial head plays an important role in the valgus sta-
bility. The resection of the radial head decrease valgus instability and increase ulno-
humeral joint pressure under an axial load, eventually leading to the degenerative
changes [13, 75, 76]. The effect of radial head excision has recently been investi-
gated on the progression of ulnohumeral articular surface damage in the arthroscopic
debridement [77]. A previous study showed that the patients with moderate and
severe radiocapitellar chondral loss can improve elbow motion (flexion, extension,
pronation, and supination) and relieve the pain after arthroscopic debridement of
the arthritic elbow without radial head excision [77]. Additionally, a previous study
showed that the shortening osteotomy of proximal radial of 2.5 mm, in the
radiocapitellar osteoarthritis, can preserve the intact radial head to decrease the
radiocapitellar contact pressure under a 250 N axial loading and remain the valgus
stability during the elbow movement [13]. The shortening osteotomy of radial head
might delay the ulnohumeral degeneration [13].
The prosthetic replacement may be considered in patients with severe osteoar-
thritic elbow and joint incongruence or elbow fracture-dislocation. Radiocapitellar
replacement is used as the failure of repairing the radial head by the internal fixa-
tion. The radialcapitellar replacement has the metallic monopolar and bipolar
flexed-neck prosthesis, which have demonstrated good clinical outcomes in repair-
ing the elbow fracture dislocation [78–81]. In regard to the stability of elbow joint,
monopolar implant significantly improves the ulnohumeral laxity and radiocapitel-
lar joint subluxation compared to the bipolar implant in the terrible triad models
[82, 83]. The lateral collateral ligament of elbow is the varus stabilizer; therefore,
the integrity of the lateral collateral is an important factor to affect the elbow stabil-
ity of the radialcapitellar replacement [84]. A previous study indicated that both
monopolar and bipolar prosthesis would improve the stability of the elbow joint in
the coronal plan and axial rotation loading as the adequate lateral collateral liga-
122 S.-Y. Lee and F.-C. Su
ment construction and intact medial collateral ligament [85]. The radiocapitellar
replacement has been used in patients with osteoarthritic elbow who have the asym-
metry elbow joint [86]. The degenerative and inflamed condition of osteoarthritic
elbow commonly involved the lateral side [87]. The radiocapitellar replacement can
restore the joint mobility, improve the clinical outcomes, and relieve the pain in
patients with primary and post-traumatic arthritis after postoperative 22 months
[86]. Although the orientation of the radial head cannot affect the kinematics of
ulnohumeral joint, it can affect the force transmission in the radiocapitellar joint
[88]. Therefore, the design of accurate orientation of radial head is necessary. Total
elbow arthroplasty has been developed from the constrained articulated prosthesis
to the semiconstrained and nonconstrained prostheses to reduce loosing [89]. Total
elbow arthroplasty is indicated in the rheumatoid arthritis patients, patients with
highly comminuted fracture of the distal humerus [90], or primary and post-
traumatic osteoarthritis with the failure reconstruction [91, 92]. The limitation of
total arthroplasty is the lifting weight restriction (5-lb) during a lifetime to ensure
the durability and ultimate satisfactory function [89]. Additionally, the semicon-
strained total elbow arthroplasty cause the impaired proprioception of elbow joint
[93]. The correct positioning of the prostheses [94], ligament integrity, and prosthe-
sis design remains challenging to improve the stability of elbow joint after total
elbow arthroplasty.
3.2.1 Tendinopathy of Elbow
Tendinopathy of elbow can occur on the lateral side and medial side of the elbow.
The lateral tendinopathy of elbow, tennis elbow, causes the tenderness of the origin
of common extensor tendon and pain at the lateral side of the elbow [95]. The
repetitive wrist extension and supination is the major pathomechanics to cause the
degenerative condition of the common extensor tendon [96, 97]. According to previ-
ous studies, the contact pressure between the extensor carpi radialis brevis and capi-
tellum increases with the wrist supination and elbow extension during backhand
strokes [98–100]. Therefore, the repetitive loading of common tendon is a factor to
cause the lateral tendinopathy of elbow. In regard to functional outcomes, patient
with tennis elbow decreases the grip force and isokinetic performances of wrist
extensors [101]. The primary treatment in patients with the tennis elbow is the non-
surgical management by physical therapy, activity modification, nonsteroidal anti-
inflammatory drugs, and injections for the relief of lateral-sided elbow pain [96,
102, 103]. Surgical management is indicated in patients with tennis elbow when
nonsurgical management fails [104]. Surgical management includes percutaneous,
arthroscopic, and open approaches to release of the affected extensor muscles
5 Biomechanics of the Elbow 123
[104, 105]. Recently, ultrasound has been used to diagnosis tennis elbow [106, 107].
Previous studies found that patients with tennis elbow has not only focal hypoechoic
areas and calcification but also alternation of tendon stiffness by sonoelastography
measurement [108, 109].
The medial tendinopathy of elbow, golfer’s elbow, causes the tenderness of the
origin of common flexor tendon and pain at the medial side of the elbow [110, 111].
The repetitive eccentric loading of the flexor muscles is a main pathomechanism to
cause the golfer’s elbow, especially in the wrist flexion and forearm pronation with
valgus stress at the elbow joint [112]. This long-term repetitive loading cause the
degeneration and microtrauma of tendon to result in the fibrosis and calcification
within the tendon [113]. The sonoelastography has been used to evaluate the com-
mon flexor tendon and sonoelastography is correlated with the histological findings
of the common flexor tendon [113]. As in the tennis elbow, nonsurgical management
is a primary treatment in patients with the golfer’s elbow for the relief of medial-
sided elbow pain [112, 114]. Surgical management is indicated after patients receive
nonsurgical therapy for 4–6 months and remain persistent symptoms [115, 116].
References
Cage DJ, Abrams RA, Callahan JJ, Botte MJ. Soft tissue attachments of the ulnar coro-
noid process. An anatomic study with radiographic correlation. Clin Orthop Relat Res.
1995;320:154–8.
Floris S, Olsen BS, Dalstra M, Sojbjerg JO, Sneppen O. The medial collateral ligament of the
elbow joint: anatomy and kinematics. J Shoulder Elbow Surg. 1998;7(4):345–51.
Morrey BF, An KN. Functional anatomy of the ligaments of the elbow. Clin Orthop Relat
Res. 1985;201:84–90.
Jackson TJ, Jarrell SE, Adamson GJ, Chung KC, Lee TQ. Biomechanical differences of the
anterior and posterior bands of the ulnar collateral ligament of the elbow. Knee Surg Sports
Traumatol Arthrosc. 2016;24(7):2319–23. https://doi.org/10.1007/s00167-014-3482-7.
Callaway GH, Field LD, Deng XH, Torzilli PA, O’Brien SJ, Altchek DW, Warren
RF. Biomechanical evaluation of the medial collateral ligament of the elbow. J Bone Joint
Surg Am. 1997;79(8):1223–31.
Goto A, Moritomo H, Murase T, Oka K, Sugamoto K, Arimura T, et al. In vivo elbow bio-
mechanical analysis during flexion: three-dimensional motion analysis using magnetic
resonance imaging. J Shoulder Elbow Surg. 2004;13(4):441–7. https://doi.org/10.1016/
S1058274604000394.
Goto A, Murase T, Moritomo H, Oka K, Sugamoto K, Yoshikawa H. Three-dimensional
in vivo kinematics during elbow flexion in patients with lateral humeral condyle nonunion
by an image-matching technique. J Shoulder Elbow Surg. 2014;23(3):318–26. https://doi.
org/10.1016/j.jse.2013.11.010.
Miyake J, Shimada K, Moritomo H, Kataoka T, Murase T, Sugamoto K. Kinematic changes
in elbow osteoarthritis: in vivo and 3-dimensional analysis using computed tomographic data.
J Hand Surg Am. 2013;38(5):957–64. https://doi.org/10.1016/j.jhsa.2013.02.006.
Omori S, Miyake J, Oka K, Tanaka H, Yoshikawa H, Murase T. In vivo three-dimensional
elbow biomechanics during forearm rotation. J Shoulder Elbow Surg. 2016;25(1):112–9.
https://doi.org/10.1016/j.jse.2015.07.002.
Bryce CD, Armstrong AD. Anatomy and biomechanics of the elbow. Orthop Clin North Am.
2008;39(2):141–54, v. https://doi.org/10.1016/j.ocl.2007.12.001.
Morrey BF, An KN. Stability of the elbow: osseous constraints. J Shoulder Elbow Surg.
2005;14(1 Suppl S):174S–8S. https://doi.org/10.1016/j.jse.2004.09.031.
Kim HJ, Yi JH, Jung JW, Cho DW, van Riet R, Jeon IH. Influence of forearm rotation on
proximal radioulnar joint congruency and translational motion using computed tomography
and computer-aided design technologies. J Hand Surg Am. 2011;36(5):811–5. https://doi.
org/10.1016/j.jhsa.2011.01.043.
Hackl M, Wegmann K, Kahmann SL, Heinze N, Staat M, Neiss WF, et al. Radial shortening
osteotomy reduces radiocapitellar contact pressures while preserving valgus stability of the
elbow. Knee Surg Sports Traumatol Arthrosc. 2017;25(7):2280–8. https://doi.org/10.1007/
s00167-017-4468-z.
Bachman DR, Thaveepunsan S, Park S, Fitzsimmons JS, An KN, O’Driscoll SW. The
effect of prosthetic radial head geometry on the distribution and magnitude of radiocapi-
tellar joint contact pressures. J Hand Surg Am. 2015;40(2):281–8. https://doi.org/10.1016/j.
jhsa.2014.11.005.
Ahmad CS, Park MC, Elattrache NS. Elbow medial ulnar collateral ligament insufficiency
alters posteromedial olecranon contact. Am J Sports Med. 2004;32(7):1607–12. https://doi.
org/10.1177/0363546503263149.
Hassan SE, Parks BG, Douoguih WA, Osbahr DC. Effect of distal ulnar collateral ligament
tear pattern on contact forces and valgus stability in the posteromedial compartment of the
elbow. Am J Sports Med. 2015;43(2):447–52. https://doi.org/10.1177/0363546514557239.
5 Biomechanics of the Elbow 125
Sahu D, Holmes DM, Fitzsimmons JS, Thoreson AR, Berglund LJ, An KN, O’Driscoll
SW. Influence of radial head prosthetic design on radiocapitellar joint contact mechanics. J
Shoulder Elbow Surg. 2014;23(4):456–62. https://doi.org/10.1016/j.jse.2013.11.028.
Chen CT, Bhargava M, Lin PM, Torzilli PA. Time, stress, and location dependent chon-
drocyte death and collagen damage in cyclically loaded articular cartilage. J Orthop Res.
2003;21(5):888–98. https://doi.org/10.1016/S0736-0266(03)00050-0.
Safran MR, Baillargeon D. Soft-tissue stabilizers of the elbow. J Shoulder Elbow Surg.
2005;14(1 Suppl S):179S–85S. https://doi.org/10.1016/j.jse.2004.09.032.
Buckley MA, Yardley A, Johnson GR, Carus DA. Dynamics of the upper limb during per-
formance of the tasks of everyday living—a review of the current knowledge base. Proc Inst
Mech Eng H. 1996;210(4):241–7. https://doi.org/10.1243/PIME_PROC_1996_210_420_02.
Murray IA, Johnson GR. A study of the external forces and moments at the shoulder and
elbow while performing every day tasks. Clin Biomech (Bristol, Avon). 2004;19(6):586–94.
https://doi.org/10.1016/j.clinbiomech.2004.03.004.
Closkey RF, Goode JR, Kirschenbaum D, Cody RP. The role of the coronoid process
in elbow stability. A biomechanical analysis of axial loading. J Bone Joint Surg Am.
2000;82-A(12):1749–53.
An KN, Hui FC, Morrey BF, Linscheid RL, Chao EY. Muscles across the elbow joint: a bio-
mechanical analysis. J Biomech. 1981;14(10):659–69.
Park MC, Ahmad CS. Dynamic contributions of the flexor-pronator mass to elbow valgus
stability. J Bone Joint Surg Am. 2004;86-A(10):2268–74.
Regan WD, Korinek SL, Morrey BF, An KN. Biomechanical study of ligaments around the
elbow joint. Clin Orthop Relat Res. 1991;271:170–9.
Morrey BF, Tanaka S, An KN. Valgus stability of the elbow. A definition of primary and
secondary constraints. Clin Orthop Relat Res. 1991;265:187–95.
Golan EJ, Shukla DR, Nasser P, Hausman M. Isolated ligamentous injury can cause postero-
medial elbow instability: a cadaveric study. J Shoulder Elbow Surg. 2016;25(12):2019–24.
https://doi.org/10.1016/j.jse.2016.04.022.
Hannouche D, Begue T. Functional anatomy of the lateral collateral ligament complex of the
elbow. Surg Radiol Anat. 1999;21(3):187–91.
Olsen BS, Sojbjerg JO, Dalstra M, Sneppen O. Kinematics of the lateral ligamentous con-
straints of the elbow joint. J Shoulder Elbow Surg. 1996;5(5):333–41.
Seki A, Olsen BS, Jensen SL, Eygendaal D, Sojbjerg JO. Functional anatomy of the lateral
collateral ligament complex of the elbow: configuration of Y and its role. J Shoulder Elbow
Surg. 2002;11(1):53–9. https://doi.org/10.1067/mse.2002.119389.
Morrey BF, An KN. Articular and ligamentous contributions to the stability of the elbow
joint. Am J Sports Med. 1983;11(5):315–9. https://doi.org/10.1177/036354658301100506.
O’Driscoll SW. Classification and evaluation of recurrent instability of the elbow. Clin
Orthop Relat Res. 2000;370:34–43.
Karbach LE, Elfar J. Elbow instability: anatomy, biomechanics, diagnostic maneuvers, and
testing. J Hand Surg Am. 2017;42(2):118–26. https://doi.org/10.1016/j.jhsa.2016.11.025.
Chung CB, Stanley AJ, Gentili A. Magnetic resonance imaging of elbow instability. Semin
Musculoskelet Radiol. 2005;9(1):67–76. https://doi.org/10.1055/s-2005-867,104.
Ebrahimzadeh MH, Amadzadeh-Chabock H, Ring D. Traumatic elbow instability. J Hand
Surg Am. 2010;35(7):1220–5. https://doi.org/10.1016/j.jhsa.2010.05.002.
Wyrick JD, Dailey SK, Gunzenhaeuser JM, Casstevens EC. Management of complex elbow
dislocations: a mechanistic approach. J Am Acad Orthop Surg. 2015;23(5):297–306. https://
doi.org/10.5435/JAAOS-D-14-00023.
Eygendaal D, Olsen BS, Jensen SL, Seki A, Sojbjerg JO. Kinematics of partial and total rup-
tures of the medial collateral ligament of the elbow. J Shoulder Elbow Surg. 1999;8(6):612–6.
O’Driscoll SW, Bell DF, Morrey BF. Posterolateral rotatory instability of the elbow. J Bone
Joint Surg Am. 1991;73(3):440–6.
126 S.-Y. Lee and F.-C. Su
O’Driscoll SW, Morrey BF, Korinek S, An KN. Elbow subluxation and dislocation. A spec-
trum of instability. Clin Orthop Relat Res. 1992;280:186–97.
Camp CL, Smith J, O’Driscoll SW. Posterolateral rotatory instability of the elbow:
part I. Mechanism of injury and the posterolateral rotatory drawer test. Arthrosc Tech.
2017;6(2):e401–5. https://doi.org/10.1016/j.eats.2016.10.016.
Bellato E, Kim Y, Fitzsimmons JS, Hooke AW, Berglund LJ, Bachman DR, O’Driscoll
SW. Role of the lateral collateral ligament in posteromedial rotatory instability of the elbow.
J Shoulder Elbow Surg. 2017;26(9):1636–43. https://doi.org/10.1016/j.jse.2017.04.011.
Mellema JJ, Doornberg JN, Dyer GS, Ring D. Distribution of coronoid fracture lines by spe-
cific patterns of traumatic elbow instability. J Hand Surg Am. 2014;39(10):2041–6. https://
doi.org/10.1016/j.jhsa.2014.06.123.
O’Driscoll SW, Jupiter JB, Cohen MS, Ring D, McKee MD. Difficult elbow fractures: pearls
and pitfalls. Instr Course Lect. 2003;52:113–34.
Pollock JW, Brownhill J, Ferreira L, McDonald CP, Johnson J, King G. The effect of antero-
medial facet fractures of the coronoid and lateral collateral ligament injury on elbow sta-
bility and kinematics. J Bone Joint Surg Am. 2009;91(6):1448–58. https://doi.org/10.2106/
JBJS.H.00222.
Pollock JW, Brownhill J, Ferreira LM, McDonald CP, Johnson JA, King GJ. Effect of the
posterior bundle of the medial collateral ligament on elbow stability. J Hand Surg Am.
2009;34(1):116–23. https://doi.org/10.1016/j.jhsa.2008.09.016.
Stoneback JW, Owens BD, Sykes J, Athwal GS, Pointer L, Wolf JM. Incidence of elbow dis-
locations in the United States population. J Bone Joint Surg Am. 2012;94(3):240–5. https://
doi.org/10.2106/JBJS.J.01663.
Rhyou IH, Kim YS. New mechanism of the posterior elbow dislocation. Knee Surg Sports
Traumatol Arthrosc. 2012;20(12):2535–41. https://doi.org/10.1007/s00167-011-1872-7.
Doornberg JN, Ring D. Coronoid fracture patterns. J Hand Surg Am. 2006;31(1):45–52.
https://doi.org/10.1016/j.jhsa.2005.08.014.
Garrigues GE, Wray WH 3rd, Lindenhovius AL, Ring DC, Ruch DS. Fixation of the coro-
noid process in elbow fracture-dislocations. J Bone Joint Surg Am. 2011;93(20):1873–81.
https://doi.org/10.2106/JBJS.I.01673.
Ring D, Jupiter JB. Fracture-dislocation of the elbow. J Bone Joint Surg Am.
1998;80(4):566–80.
Doornberg JN, van Duijn J, Ring D. Coronoid fracture height in terrible-triad injuries. J Hand
Surg Am. 2006;31(5):794–7. https://doi.org/10.1016/j.jhsa.2006.01.004.
Shukla DR, Golan E, Weiser MC, Nasser P, Choueka J, Hausman M. The posterior bundle’s
effect on posteromedial elbow instability after a transverse coronoid fracture: a biomechani-
cal study. J Hand Surg Am. 2017. https://doi.org/10.1016/j.jhsa.2017.09.018.
Steinmann SP. Coronoid process fracture. J Am Acad Orthop Surg. 2008;16(9):519–29.
54. Leigh WB, Ball CM. Radial head reconstruction versus replacement in the treatment of ter-
rible triad injuries of the elbow. J Shoulder Elbow Surg. 2012;21(10):1336–41. https://doi.
org/10.1016/j.jse.2012.03.005.
Watters TS, Garrigues GE, Ring D, Ruch DS. Fixation versus replacement of radial head in
terrible triad: is there a difference in elbow stability and prognosis? Clin Orthop Relat Res.
2014;472(7):2128–35. https://doi.org/10.1007/s11999-013-3331-x.
Kuhn S, Burkhart KJ, Schneider J, Muelbert BK, Hartmann F, Mueller LP, Rommens
PM. The anatomy of the proximal radius: implications on fracture implant design. J Shoulder
Elbow Surg. 2012;21(9):1247–54. https://doi.org/10.1016/j.jse.2011.11.008.
Rowland AS, Athwal GS, MacDermid JC, King GJ. Lateral ulnohumeral joint space
widening is not diagnostic of radial head arthroplasty overstuffing. J Hand Surg Am.
2007;32(5):637–41. https://doi.org/10.1016/j.jhsa.2007.02.024.
Arrigoni P, D’Ambrosi R, Nicoletti S, Randelli P. Arthroscopic reinsertion of lateral collat-
eral ligament, anterior capsular plication, and coronoid tunneling technique for chronic elbow
5 Biomechanics of the Elbow 127
Harrington IJ, Sekyi-Otu A, Barrington TW, Evans DC, Tuli V. The functional outcome with
metallic radial head implants in the treatment of unstable elbow fractures: a long-term review.
J Trauma. 2001;50(1):46–52.
Popovic N, Lemaire R, Georis P, Gillet P. Midterm results with a bipolar radial head prosthe-
sis: radiographic evidence of loosening at the bone-cement interface. J Bone Joint Surg Am.
2007;89(11):2469–76. https://doi.org/10.2106/JBJS.F.00723.
Zunkiewicz MR, Clemente JS, Miller MC, Baratz ME, Wysocki RW, Cohen MS. Radial head
replacement with a bipolar system: a minimum 2-year follow-up. J Shoulder Elbow Surg.
2012;21(1):98–104. https://doi.org/10.1016/j.jse.2011.05.012.
Pomianowski S, Morrey BF, Neale PG, Park MJ, O’Driscoll SW, An KN. Contribution of
monoblock and bipolar radial head prostheses to valgus stability of the elbow. J Bone Joint
Surg Am. 2001;83-A(12):1829–34.
Schneeberger AG, Sadowski MM, Jacob HA. Coronoid process and radial head as posterolat-
eral rotatory stabilizers of the elbow. J Bone Joint Surg Am. 2004;86-A(5):975–82.
Chanlalit C, Shukla DR, Fitzsimmons JS, Thoreson AR, An KN, O’Driscoll
SW. Radiocapitellar stability: the effect of soft tissue integrity on bipolar versus monopolar
radial head prostheses. J Shoulder Elbow Surg. 2011;20(2):219–25. https://doi.org/10.1016/j.
jse.2010.10.033.
Hartzler RU, Morrey BF, Steinmann SP, Llusa-Perez M, Sanchez-Sotelo J. Radial head
reconstruction in elbow fracture-dislocation: monopolar or bipolar prosthesis? Clin Orthop
Relat Res. 2014;472(7):2144–50. https://doi.org/10.1007/s11999-014-3672-0.
Giannicola G, Angeloni R, Mantovani A, Rebuzzi E, Merolla G, Greco A, et al. Open debride-
ment and radiocapitellar replacement in primary and post-traumatic arthritis of the elbow:
a multicenter study. J Shoulder Elbow Surg. 2012;21(4):456–63. https://doi.org/10.1016/j.
jse.2011.08.071.
Ahrens PM, Redfern DR, Forester AJ. Patterns of articular wear in the cadaveric elbow joint.
J Shoulder Elbow Surg. 2001;10(1):52–6. https://doi.org/10.1067/mse.2001.109382.
van Riet RP, Van Glabbeek F, Baumfeld JA, Neale PG, Morrey BF, O’Driscoll SW, An
KN. The effect of the orientation of the radial head on the kinematics of the ulnohumeral
joint and force transmission through the radiocapitellar joint. Clin Biomech (Bristol, Avon).
2006;21(6):554–9. https://doi.org/10.1016/j.clinbiomech.2006.01.006.
Bennett JB, Mehlhoff TL. Total elbow arthroplasty: surgical technique. J Hand Surg Am.
2009;34(5):933–9. https://doi.org/10.1016/j.jhsa.2009.02.021.
Kamineni S, Morrey BF. Distal humeral fractures treated with noncustom total elbow replace-
ment. Surgical technique. J Bone Joint Surg Am. 2005;87 Suppl 1(Pt 1):41–50. https://doi.
org/10.2106/JBJS.D.02871.
Antuna SA, Morrey BF, Adams RA, O’Driscoll SW. Ulnohumeral arthroplasty for primary
degenerative arthritis of the elbow: long-term outcome and complications. J Bone Joint Surg
Am. 2002;84-A(12):2168–73.
Cheung EV, Adams R, Morrey BF. Primary osteoarthritis of the elbow: current treatment
options. J Am Acad Orthop Surg. 2008;16(2):77–87.
Siqueira GSL, Amaral MVG, Schiefer M, Schlee G, Schultz-Wenk TF, de Almeida MN, et al.
Proprioceptive deficit after total elbow arthroplasty: an observational study. J Shoulder Elbow
Surg. 2017;26(11):2017–22. https://doi.org/10.1016/j.jse.2017.07.003.
King GJ, Glauser SJ, Westreich A, Morrey BF, An KN. In vitro stability of an unconstrained
total elbow prosthesis. Influence of axial loading and joint flexion angle. J Arthroplasty.
1993;8(3):291–8.
Walz DM, Newman JS, Konin GP, Ross G. Epicondylitis: pathogenesis, imaging, and treat-
ment. Radiographics. 2010;30(1):167–84. https://doi.org/10.1148/rg.301095078.
Calfee RP, Patel A, DaSilva MF, Akelman E. Management of lateral epicondylitis: current
concepts. J Am Acad Orthop Surg. 2008;16(1):19–29.
Lee SY, Chieh HF, Lin CJ, Jou IM, Kuo LC, Su FC. The potential risk factors relevant to
lateral epicondylitis by wrist coupling posture. PLoS One. 2016;11(5):e0155379. https://doi.
org/10.1371/journal.pone.0155379.
5 Biomechanics of the Elbow 129
Bunata RE, Brown DS, Capelo R. Anatomic factors related to the cause of tennis elbow. J
Bone Joint Surg Am. 2007;89(9):1955–63. https://doi.org/10.2106/JBJS.F.00727.
Orchard J, Kountouris A. The management of tennis elbow. BMJ. 2011;342:d2687. https://
doi.org/10.1136/bmj.d2687.
Tanaka Y, Aoki M, Izumi T, Wada T, Fujimiya M, Yamashita T. Effect of elbow and forearm
position on contact pressure between the extensor origin and the lateral side of the capitellum.
J Hand Surg Am. 2011;36(1):81–8. https://doi.org/10.1016/j.jhsa.2010.10.005.
Unyo C, Chaler J, Rojas-Martinez M, Pujol E, Muller B, Garreta R, Mananas MA. A cross-
sectional study comparing strength profile of dorsal and palmar flexor muscles of the wrist in
epicondylitis and healthy men. Eur J Phys Rehabil Med. 2013;49(4):507–15.
Gosens T, Peerbooms JC, van Laar W, den Oudsten BL. Ongoing positive effect of platelet-
rich plasma versus corticosteroid injection in lateral epicondylitis: a double-blind random-
ized controlled trial with 2-year follow-up. Am J Sports Med. 2011;39(6):1200–8. https://doi.
org/10.1177/0363546510397173.
Tyler TF, Thomas GC, Nicholas SJ, McHugh MP. Addition of isolated wrist extensor eccen-
tric exercise to standard treatment for chronic lateral epicondylosis: a prospective randomized
trial. J Shoulder Elbow Surg. 2010;19(6):917–22. https://doi.org/10.1016/j.jse.2010.04.041.
Othman AM. Arthroscopic versus percutaneous release of common extensor origin for treat-
ment of chronic tennis elbow. Arch Orthop Trauma Surg. 2011;131(3):383–8. https://doi.
org/10.1007/s00402-011-1260-2.
Dunkow PD, Jatti M, Muddu BN. A comparison of open and percutaneous techniques in the
surgical treatment of tennis elbow. J Bone Joint Surg Br. 2004;86(5):701–4.
Connell D, Burke F, Coombes P, McNealy S, Freeman D, Pryde D, Hoy G. Sonographic
examination of lateral epicondylitis. AJR Am J Roentgenol. 2001;176(3):777–82. https://doi.
org/10.2214/ajr.176.3.1760777.
Jaen-Diaz JI, Cerezo-Lopez E, Lopez-de Castro F, Mata-Castrillo M, Barcelo-Galindez JP,
De la Fuente J, Balius-Mata R. Sonographic findings for the common extensor tendon of the
elbow in the general population. J Ultrasound Med. 2010;29(12):1717–24.
Galletti S, Oliva F, Masiero S, Frizziero A, Galletti R, Schiavone C, et al. Sonoelastography
in the diagnosis of tendinopathies: an added value. Muscles Ligaments Tendons
J. 2015;5(4):325–30. https://doi.org/10.11138/mltj/2015.5.4.325.
Klauser AS, Pamminger M, Halpern EJ, Abd Ellah MMH, Moriggl B, Taljanovic MS, et al.
Extensor tendinopathy of the elbow assessed with sonoelastography: histologic correlation.
Eur Radiol. 2017;27(8):3460–6. https://doi.org/10.1007/s00330-016-4711-x.
Shiri R, Viikari-Juntura E, Varonen H, Heliovaara M. Prevalence and determinants of lat-
eral and medial epicondylitis: a population study. Am J Epidemiol. 2006;164(11):1065–74.
https://doi.org/10.1093/aje/kwj325.
Wolf JM, Mountcastle S, Burks R, Sturdivant RX, Owens BD. Epidemiology of lateral and
medial epicondylitis in a military population. Mil Med. 2010;175(5):336–9.
Ciccotti MG, Ramani MN. Medial epicondylitis. Tech Hand Up Extrem Surg. 2003;7(4):190–6.
Klauser AS, Pamminger MJ, Halpern EJ, Abd Ellah MMH, Moriggl B, Taljanovic MS, et al.
Sonoelastography of the common flexor tendon of the elbow with histologic agreement: a
cadaveric study. Radiology. 2017;283(2):486–91. https://doi.org/10.1148/radiol.2016160139.
Harada M, Takahara M, Hirayama T, Sasaki J, Mura N, Ogino T. Outcome of nonop-
erative treatment for humeral medial epicondylar fragmentation before epiphyseal clo-
sure in young baseball players. Am J Sports Med. 2012;40(7):1583–90. https://doi.
org/10.1177/0363546512443807.
Amin NH, Kumar NS, Schickendantz MS. Medial epicondylitis: evaluation and management.
J Am Acad Orthop Surg. 2015;23(6):348–55. https://doi.org/10.5435/JAAOS-D-14-00145.
Mishra A, Pirolo JM, Gosens T. Treatment of medial epicondylar tendinopathy in athletes. Sports
Med Arthrosc Rev. 2014;22(3):164–8. https://doi.org/10.1097/JSA.0000000000000031.
Macchi V, Tiengo C, Porzionato A, Stecco C, Sarasin G, Tubbs S, et al. The cubital tunnel:
a radiologic and histotopographic study. J Anat. 2014;225(2):262–9. https://doi.org/10.1111/
joa.12206.
130 S.-Y. Lee and F.-C. Su
Wright TW, Glowczewskie F Jr, Cowin D, Wheeler DL. Ulnar nerve excursion and
strain at the elbow and wrist associated with upper extremity motion. J Hand Surg Am.
2001;26(4):655–62. https://doi.org/10.1053/jhsu.2001.26140.
Gelberman RH, Yamaguchi K, Hollstien SB, Winn SS, Heidenreich FP Jr, Bindra RR, et al.
Changes in interstitial pressure and cross-sectional area of the cubital tunnel and of the ulnar
nerve with flexion of the elbow. An experimental study in human cadavera. J Bone Joint Surg
Am. 1998;80(4):492–501.
Buehler MJ, Thayer DT. The elbow flexion test. A clinical test for the cubital tunnel syn-
drome. Clin Orthop Relat Res. 1988;233:213–6.
Mehlhoff TL, Noble PC, Bennett JB, Tullos HS. Simple dislocation of the elbow in the adult.
Results after closed treatment. J Bone Joint Surg Am. 1988;70(2):244–9.
Robinson CM, Hill RM, Jacobs N, Dall G, Court-Brown CM. Adult distal humeral metaphy-
seal fractures: epidemiology and results of treatment. J Orthop Trauma. 2003;17(1):38–47.
Shah CM, Calfee RP, Gelberman RH, Goldfarb CA. Outcomes of rigid night splinting
and activity modification in the treatment of cubital tunnel syndrome. J Hand Surg Am.
2013;38(6):1125–30, e1121. https://doi.org/10.1016/j.jhsa.2013.02.039.
Svernlov B, Larsson M, Rehn K, Adolfsson L. Conservative treatment of the cubital tunnel syn-
drome. J Hand Surg Eur Vol. 2009;34(2):201–7. https://doi.org/10.1177/1753193408098480.
Krogue JD, Aleem AW, Osei DA, Goldfarb CA, Calfee RP. Predictors of surgical revision
after in situ decompression of the ulnar nerve. J Shoulder Elbow Surg. 2015;24(4):634–9.
https://doi.org/10.1016/j.jse.2014.12.015.
Osei DA, Padegimas EM, Calfee RP, Gelberman RH. Outcomes following modified oblique
medial epicondylectomy for treatment of cubital tunnel syndrome. J Hand Surg Am.
2013;38(2):336–43. https://doi.org/10.1016/j.jhsa.2012.11.006.
Bartels RH, Verhagen WI, van der Wilt GJ, Meulstee J, van Rossum LG, Grotenhuis
JA. Prospective randomized controlled study comparing simple decompression versus ante-
rior subcutaneous transposition for idiopathic neuropathy of the ulnar nerve at the elbow: part
1. Neurosurgery. 2005;56(3):522–30; discussion 522–30.
Chapter 6
Biomechanics of the Shoulder
Abstract The glenohumeral (GH) joint is a complex and unstable articulation. The
interaction between various structures of the shoulder caused by mechanical stimuli
and motion provides multiple degrees of shoulder motion. The static stability of the
shoulder is supported by the articulation of the humeral head and the glenoid with
additional GH ligaments, capsule, and labrum. The rotator cuff muscles surrounding
the shoulder joint provide dynamic stability. The combination of these factors forms
the biomechanical system that can respond in accordance with the arm movement.
Different pathological processes and injuries may result in similar clinical manifes-
tations. It is crucial to know the etiology of these different pathological factors from
the viewpoint of the shoulder joint biomechanics to provide the most effective and
curable treatments for patients suffering from shoulder diseases and disorders.
1 Introduction
The shoulder motion is subject to the reaction to the shoulder structure stimuli. Due
to the mismatch between the humeral head and the articular surface of the glenoid,
the stabilization of the shoulder is compensated by static and dynamic stabilizers.
M. Zhang
Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University,
Beijing, China
Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education,
School of Biological Science and Medical Engineering, Beihang University, Beijing, China
C.-H. Chen (*)
Department of Orthopedics, Taipei Medical University—Shuang Ho Hospital, Taipei, Taiwan
School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical
University, Taipei, Taiwan
Research Center of Biomedical Devices, Taipei Medical University, Taipei, Taiwan
The static stabilizers include the glenohumeral (GH) ligaments, the labrum, and the
constrained capsule. The dynamic stability is provided by the r otator cuff (RC) mus-
cles surrounding the shoulder joint. The combined effect of the stabilizers is to sup-
port the multiple degrees of motion within the GH joint. The scapulothoracic (ST)
joint provides additional mobility and stability to the shoulder.
The skeletal structure of the shoulder includes the humerus, the scapula, and the
clavicle.
2.1.1 Humerus
The humerus is located on the upper limb and is articulated with the glenoid fossa
at the glenohumeral joint. The head of the humerus is elliptical and inclines between
120° and 145° [1] as well as a retroversion within a range of 60° [2]. There are two
bulges on the lateral and anterior sides of the humeral head. They are greater and
lesser tubercles. These two tubercles are separated by intertubercular groove and are
origins to subscapularis, teres minor, infraspinatus, and supraspinatus muscles. The
humerus has two necks: surgical and anatomical necks. The surgical neck is a con-
striction below the tubercles of the greater tubercle and lesser tubercle, and above
the deltoid tuberosity. The anatomical neck is obliquely directed, forming an obtuse
angle with the body. It provides the attachment to the articular capsule.
2.1.2 Scapula
The scapula is located on the posterior chest wall and is in an inverted triangular
shape. It inclines approximately 40° anteriorly to the coronal plane [3]. There is a
broad concavity on the anterior surface of the scapula and it forms the origin of the
subscapularis muscle. The posterior surface of the scapula consists of the infraspi-
natus and supraspinal fossae, which is separated by the scapular spine provide the
origins to the infraspinatus and supraspinatus muscles. Further, the scapula is com-
6 Biomechanics of the Shoulder 133
posed of three corners and three sides, namely the lateral, superior, and inferior
angles, and the medial, superior, and lateral borders, respectively. Next to the lateral
angle, the glenoid is a shallow and pear-shape cavity. It inclines 3–5° superiorly [4]
and 2.5–12.5° posteriorly [5].
2.1.3 Clavicle
The clavicle is an elongated bone in an S-shaped and connects the upper limb to the
torso. The lateral and medial end of the clavicle is flat and thick, respectively. The
clavicle transmits forces from the trunk to the upper limb. The articulation between
the clavicle and the manubrium of the sternum is a sternoclavicular (SC) joint. The
articulation between the clavicle and the acromion of the scapula is the acromiocla-
vicular (AC) joint.
The shoulder joint is composed of four joints (SC joint, AC joint, GH joint, and ST
articulation) with bony and soft-tissue structures. These articulations provide a high
degree of shoulder motion. The arm can reach an elevation angle of 180°, internal
and external rotations of approximately 150°, and flexion and extension of 170° [6].
2.2.1 Sternoclavicular Joint
The articulation between the manubrium sterni and the medial end of the clavicle is
the SC joint. It connects the thorax to the upper limb and allows approximately 35°
of elevation and 50° of axial rotation [6]. The intrinsic bony stability of the SC joint
is mainly supported by the anterior and posterior SC ligaments, the interclavicular
ligament, the costoclavicular ligament, and the articular disc.
2.2.2 Acromioclavicular Joint
The AC joint is composed of the acromion and the clavicle surfaces, which trans-
mits forces from the upper extremity to the chest musculature. The AC articulation
is stabilized by the coracoclavicular and the AC ligaments. The coracoacromial liga-
ment spans between the lateral aspect of the coracoid process and the anterior facet
of the acromion. It is to restraint the superior-inferior migration of the clavicle [7,
8]. The AC ligament is to restrain the anterior-posterior translation of the clavicle [7,
8] and provides scapular and synchronous clavicular rotation [9].
134 M. Zhang and C.-H. Chen
2.2.3 Glenohumeral Joint
The GH joint is a flexible and unstable articulation composed of the scapula glenoid
fossa and humeral head. The glenoid cavity accounts for 25–30% of the humeral
head and can move relatively to the humerus [9]. The glenoid-humerus contact area
varies in degrees during the shoulder motion. Its stability is mainly supported by the
articular capsule, the labrum, and the surrounding muscles [10]. The capsular-liga-
mentous complex includes the superior, the medial, and the inferior GH ligaments.
The superior and the medial GH ligaments are both single-band structures and reach
the lesser tubercle and the humeral neck, respectively. The inferior GH ligament
complex consists of anterior and posterior bands and inserts on the humerus beyond
the lesser tuberosity [11]. The glenoid labrum is a circumferential and fibrocartilagi-
nous ring attaching to the glenoid rim. The glenoid labrum serves as a bumper dur-
ing the shoulder motion and an attachment site of the GH ligaments. The muscles
supplying the stability of the GH joint refers to the RC, which will be described in
Sect. 2.3.
2.2.4 Scapulothoracic Articulation
The ST articulation is composed of the anterior surface of the scapula and the pos-
terior surface of the thoracic [12]. The neurovascular, muscular, and bursal struc-
tures allow smooth motion of the scapula on the thorax. The ST articulation allows
30° of abduction internal rotation, respectively. It increases the shoulder movement
after the initial 120°, which is supplied by the GH joint [12].
The shoulder muscles supply athletic ability and dynamic stability. The function of
muscles are subjected to its origin and endpoint. The muscles and their functions on
the shoulder complex are described below.
The anterior outer layer muscles on the shoulder complex involve the pectoralis
major and the deltoid muscles. The pectoralis major muscle has two heads (sterno-
costal and clavicular heads) and is located above the anterior chest wall. The sterno-
costal head originates from the anterior sternum surface, the superior six costal
cartilages, and the external oblique muscle aponeurosis. In contrast, the clavicular
head originates from the medial clavicle anterior surface. Both the sternocostal and
clavicular heads insert to the crest of the greater tubercle of the humerus and serve
the elevation and adduction of the arm. The deltoid muscle is composed of the ante-
rior, the intermediate, and the posterior fibers. The anterior fibers originate from the
upper surface and the anterior border of the lateral third of the clavicle and serve the
6 Biomechanics of the Shoulder 135
flexion and internal rotation of the arm. The intermediate fibers originate from the
acromion process and the spine of the scapula and serve the abduction of the arm
after the initial 15° of arm rotation [6]. The posterior section of the deltoid origi-
nates from the spine of the scapula. It serves the external rotation and the extension
of the humerus. All the fibers of the deltoid muscle insert into the deltoid tuberosity
of the humerus.
The inner muscles involve the pectoralis minor, the subclavius, and the subscap-
ularis muscles. The pectoralis minor plays an important stabilizing role on the scap-
ula. The subclavian muscle is located underneath the clavicle and contributes to the
clavicular movement. The subscapularis muscle, which is located in the anterior
scapula, functions as the arm rotator.
The posterior outer layer muscles involve the latissimus dorsi, the trapezius, the
serratus anterior, and the posterior deltoid muscles. The latissimus dorsi muscle
arises from the thoracic vertebrae and inserts into the intertubercular groove of the
humerus. It works in collaboration with the pectoralis major to contribute to the
adduction and medial rotation of the humerus. The trapezius muscle is one of the
broadest back muscles. It arises from the occipital bone, the ligamentum nuchae,
and the spinous processes of T01–T12 and inserts into the third clavicle lateral, as
well as the acromion and scapular spine. The trapezius muscle contributes to the
shoulder elevation and rotation and also acts in head/neck extension [13]. The ser-
ratus anterior muscle arises from the anterior surfaces of the eighth upper ribs and
inserts into the inner medial border of the scapula. The serratus anterior muscle
allows the forward rotation of the arm and to pull the scapula forward and around
the rib cage.
The posterior inner layer muscle below the superficial muscles (the trapezius and
deltoid muscles) includes the supraspinatus, infraspinatus, teres minor, and teres
major muscles. The origin of the supraspinatus muscle is the supraspinatus fossa.
The muscle tracks laterally underneath the acromion and goes on the insertion at the
greater tuberosity [14]. Thus, the supraspinatus muscle assists in the arm abduction
and humerus stabilization [6]. The infraspinatus muscle originates from the infra-
spinous fossa of the scapula and inserts at the greater tuberosity. The origin of the
teres minor muscle is at the lateral margin of the scapula. The muscle inserts at the
most posterior and inferior facet of the greater tuberosity. Both the teres minor and
the infraspinatus muscles assist the stability and rotation of the humerus. The sub-
scapularis muscle is trapezoidal and origins from the anterior scapula aspect and
inserts at the lesser tuberosity [15]. The teres major muscle arises from the inferior
angle of the scapula and passes laterally and superiorly to the bicipital groove. It
contributes to the extension and rotation of the humerus.
The RC is formed by the subscapularis, the infraspinatus, the teres minor, the
supraspinatus muscles, and their associated tendons. The RC muscles contribute to
the abduction and rotation of the humerus and also provide a compressive force to
centralize the humeral head on the glenoid. In the case of a massive rotator cuff tear,
the loss of enough passive muscle tension and dynamic contraction leads to exces-
136 M. Zhang and C.-H. Chen
sive superior translation of the humerus to the glenoid cavity. The translation can
reach 12 mm in some cases [9]. It may result in subacromial impingement and the
erosions of surrounding bone and articulations (i.e., the glenoid, the acromiocla-
vicular joint, and the anterior acromion) (the severe RC tear is described further in
Sect. 4.2.1).
2.4 Summary
The GH joint is an enarthrodial socket-to-ball joint and supports the polyaxial arm
motions. It includes abduction/adduction around the sagittal axis, flexion/extension
around the frontal axis, and external/internal rotation around a longitudinal-
humeral axis.
GH joint kinematics is not precisely equivalent to enarthrodial kinematics.
Numerous studies have reported the exact determination of the GH joint kinematics
and the founding is controversial. Due to different methodological approaches in
different studies, it is difficult to compare the results. In general, the humeral head
translates approximately 1.1 mm inferiorly during the whole abduction and 2.4 mm
anteriorly before the abduction of 90° and 1.4 mm posteriorly during the abduction
of 90–150° [16].
The GH joint maintains stability in utilizing static and dynamic restraints.
These static stabilizers dynamic restraint involve ligamentous, capsular, cartilagi-
nous, and bony structures, as well as musculature structure of the shoulder,
respectively. The GH ligament stabilizes the GH joint by preventing excessive
movements of the humeral head relative to the glenoid cavity in the extremes of
motion [17]. A competent sealed capsule of appropriate volume, minimal joint
fluid, and an intact congruent glenoid labrum provides the stability of the GH
articulation [18]. Neuromuscular control primarily provides dynamic stability
between the RC muscles and ST musculature. The functional ST musculature
can ideally release the instability of the shoulder joint and the neural feedback
from the GH ligaments and RC muscles, which are used to prevent pathologic
translation of the GH joint.
Dynamic stabilizers may contribute to joint stability by the muscle contraction,
which leads to compression of the articular surfaces, and the passive muscle ten-
sion from the bulk effect of the muscles [17]. The contraction of the RC muscles
compresses the humeral head on the glenoid cavity and the asymmetric contrac-
tion leads to the humeral head rotation during the shoulder motion. The interac-
tion of the RC muscles works in conjunction with other muscles in the shoulder
girdle [19].
The shoulder joint kinematics relies on the ST and deltoid muscles, as well as the
RC muscles interaction. The subscapularis and infraspinatus muscles act as a trans-
verse force couple generating compressive forces. Also, the supraspinatus muscle
plays a significant role in the concavity compression during early abduction [20].
6 Biomechanics of the Shoulder 137
a b
Fig. 6.1 The glenoid bone (a) sagittal view; (b) transverse view. L lateral, M medial, P posterior,
A anterior, I inferior, S superior. (Adapted from Frich et al., 1998)
138 M. Zhang and C.-H. Chen
the strength ranges from 26 to 110 MPa [21, 23, 25]. Young’s modulus and strength
values are related to many factors such as health conditions, gender, and ages [23,
27, 28].
Shoulder joint arthroplasty is a surgical way to alleviate shoulder pain and restore
the shoulder function. This section introduces the shoulder joint replacements from
the following sections: (1) the types of shoulder joint arthroplasty; (2) clinical indi-
cations of shoulder joint arthroplasty; and (3) knowledge-based related to the shoul-
der joint arthroplasty.
The artificial shoulder joint arthroplasty can be classified into (1) anatomical and
reverse shoulder arthroplasties, (2) total shoulder arthroplasty (TSA) and prosthetic
humeral hemiarthroplasty (HA), and (3) stemmed and resurfacing shoulder
arthroplasties.
In humeral HA (Fig. 6.2a), only the proximal humerus was replaced by an artificial
device, while in the TSA, both the humerus and the glenoid are reconstructed. In the
surgical technique of HA, the entire glenoid can be preserved and the complications
associating with the glenoid component in the TSA can be avoided. HA is used for
patients with intact articular cartilage surfaces. When the cartilage is damaged or the
glenoid erosion is severe, TSA is usually preferred.
6 Biomechanics of the Shoulder 139
a b
Prosthesis resurfacing (Fig. 6.2b) refers to the surgical technique in which only the
damaged surface of the humeral head is reconstructed. This technique can preserve
the native inclination and offset of the humerus as well as the head shaft angle.
Prosthesis resurfacing is usually used for the treatments of young and active
patients [30, 31]. The contact between the glenoid bone and the stiff metallic
humeral resurfacing device may lead to late glenoid arthrosis and need a revision
to a TSA [30].
The indications for shoulder joint replacements involve cuff tear arthropathy (CTA),
primary OA, rheumatoid arthritis, humeral fracture, and the failed shoulder joint
arthroplasty [29].
a b
4.2.2 Osteoarthritis
Humerus
Centre of rotation
RSA
Scapula
Humerus
Glenoid socket Humerus
Fig. 6.4 Schematic diagram of the CTA in (a) the anatomical shoulder; (b) the TSA and
(c) the RSA
[39]. The mild OA is usually treated with a nonsurgical method (i.e., local injec-
tions, medications, and physical therapy [40]). When OA is severe, HA or TSA is
usually recommended [41–43].
4.2.3 Rheumatoid Arthritis
There are four types of proximal humeral fractures (Fig. 6.5). Type 1 (Fig. 6.5a)
refers to the fracture with collapse or necrosis on the humeral head; Type 2
142 M. Zhang and C.-H. Chen
(Fig. 6.5b) refers to the fracture with irreducible dislocations; Type 3 (Fig. 6.5c)
refers to the facture with completely broken surgical neck; Type 4 (Fig. 6.5d) is
the fracture with severe tuberosity malunions. For Type 1 and Type 2, TSA treat-
ment is recommended. For Types 3 and 4, the treatment depends on the degree of
RC deficiency. HA treatment is recommended for patients with intact RC, and
RSA treatment is used for patients over the age of 65-years-old with massive RC
tear [29].
4.2.5 Revision
The revision of the shoulder arthroplasty is usually recommended for the treatments
of implant problems (i.e., loosening, wearing, improper sizing, and malposition),
osseous problem (i.e., bone loss, glenoid arthrosis), and soft-tissue deficiency. It is
reported that the application of RSA for the failed HA can increase the American
Shoulder and Elbow Surgeons (ASES) score to 30, and improve the forward eleva-
tion from 38.1° to 72.7° [45].
a b
c d
Fig. 6.5 (a) Type 1 proximal humeral fracture; (b) Type 2 proximal humeral fracture; (c) Type 3
proximal humeral fracture; (d) Type 4 proximal humeral fracture
6 Biomechanics of the Shoulder 143
5 Conclusions
References
Iannotti JP, Gabriel JP, Schneck S, Evans B, Misra S. The normal glenohumeral relationships.
An anatomical study of one hundred and forty shoulders. JBJS. 1992;74(4):491–500.
Roberts S, Foley A, Swallow H, Wallace W, Coughlan D. The geometry of the humeral head
and the design of prostheses. Bone Joint J. 1991;73(4):647–50.
Saha A, Das A, Dutta S. Mechanism of shoulder movements and a plea for the recognition of
“zero position” of glenohumeral joint. Clin Orthop Relat Res. 1983;173:3–10.
Basmajian JV, Bazant FJ. Factors preventing downward dislocation of the adducted shoulder
joint: an electromyographic and morphological study. JBJS. 1959;41(7):1182–6.
Friedman RJ, Hawthorne K, Genez B. The use of computerized tomography in the measure-
ment of glenoid version. JBJS. 1992;74(7):1032–7.
Inman VT, Abbott LC. Observations on the function of the shoulder joint. JBJS. 1944;
26(1):1–30.
Su WR, Budoff JE, Luo ZP. The effect of coracoacromial ligament excision and acromioplasty
on superior and anterosuperior glenohumeral stability. Arthroscopy. 2009;25(1):13–8.
Lee KW, Debski RE, Chen CH, Woo SL, Fu FH. Functional evaluation of the ligaments at
the acromioclavicular joint during anteroposterior and superoinferior translation. Am J Sports
Med. 1997;25(6):858–62.
Anthony AS, Brian DB. Sports medicine. Philadelphia, PA: Lippincott Williams &
Wilkins; 2006.
Lucas DB. Biomechanics of the shoulder joint. Arch Surg. 1973;107(3):425–32.
O'Brien SJ, Neves MC, Arnoczky SP, Rozbruck SR, Dicarlo EF, Warren RF, et al. The anatomy
and histology of the inferior glenohumeral ligament complex of the shoulder. Am J Sports
Med. 1990;18(5):449–56.
Terry GC, Chopp TM. Functional anatomy of the shoulder. J Athl Train. 2000;35(3):248.
Martini FH, Nath JL, Bartholomew EF. Fundamentals of anatomy & physiology. New York:
Pearson Education, Inc.; 2006.
Mochizuki T, Sugaya H, Uomizu M, Maeda K, Matsuki K, Sekiya I, et al. Humeral insertion
of the supraspinatus and infraspinatus: new anatomical findings regarding the footprint of the
rotator cuff. JBJS. 2008;90(5):962–9.
Richards DP, Burkhart SS, Tehrany AM, Wirth MA. The subscapularis footprint: an anatomic
description of its insertion site. Arthroscopy. 2007;23(3):251–4.
Sahara W, Sugamoto K, Murai M, Tanaka H, Yoshikawa H. The three-dimensional motions of
glenohumeral joint under semi-loaded condition during arm abduction using vertically open
MRI. Clin Biomech. 2007;22(3):304–12.
Abboud JA, Soslowsky LJ. Interplay of the static and dynamic restraints in glenohumeral
instability. Clin Orthop Relat Res. 2002;400:48–57.
144 M. Zhang and C.-H. Chen
Soslowsky LJ, Flatow EL, Bigliani LU, Mow VC. Articular geometry of the glenohumeral
joint. Clin Orthop Relat Res. 1992;285:181–90.
Frankle MA, Teramoto A, Luo ZP, Levy JC, Pupello D. Glenoid morphology in reverse
shoulder arthroplasty: classification and surgical implications. J Shoulder Elbow Surg.
2009;18(6):874–85.
Apreleva M, Parsons I IV, Warner JJ, Fu FH, Woo SL-Y. Experimental investigation of
reaction forces at the glenohumeral joint during active abduction. J Shoulder Elbow Surg.
2000;9(5):409–17.
Mimar R, Limb D, Hall RM. Evaluation of the mechanical and architectural properties of
glenoid bone. J Shoulder Elbow Surg. 2008;17(2):336–41.
Mansat P, Barea C, Hobatho MC, Darmana R, Mansat M. Anatomic variation of the mechani-
cal properties of the glenoid. J Shoulder Elbow Surg. 1998;7(2):109–15.
Anglin C, Tolhurst P, Wyss UP, Pichora DR. Glenoid cancellous bone strength and modulus.
J Biomech. 1999;32(10):1091–7.
Frich LH, Odgaard A, Dalstra M. Glenoid bone architecture. J Shoulder Elbow Surg.
1998;7(4):356–61.
Frich LH, Jensen NC, Odgaard A, Pedersen CM, Søjbjerg JO, Dalstra M. Bone strength and
material properties of the glenoid. J Shoulder Elbow Surg. 1997;6(2):97–104.
Lehtinen JT, Tingart MJ, Apreleva M, Warner JJP. Total, trabecular, and cortical bone mineral
density in different regions of the glenoid. J Shoulder Elbow Surg. 2004;13(3):344–8.
Kalouche I, Abdelmoumen S, Crepin J, Mitton D, Guillot G, Gagey O. Mechanical properties
of glenoid cancellous bone. In: 16th ESB congress, London, UK; 2008.
Keaveny TM, Hayes WC. A 20-year perspective on the mechanical-properties of trabecular
bone. J Biomech Eng T ASME. 1993;115(4):534–42.
Wiater MJ, Fabing MH. Shoulder arthroplasty: prosthetic options and indications.
JAAOS. 2009;17(7):415–25.
Bailie DS, Llinas PJ, Ellenbecker TS. Cementless humeral resurfacing arthroplasty in active
patients less than fifty-five years of age. JBJS. 2008;90(1):110–7.
Levy O, Copeland SA. Cementless surface replacement arthroplasty (Copeland CSRA) for
osteoarthritis of the shoulder. J Shoulder Elbow Surg. 2004;13(3):266–71.
Craig E, Fukuda H. Cuff-tear arthropathy. JBJS. 1983;65(9):1232–44.
Joshi D. Reverse shoulder replacement soaring popularity. Orthopedics. 2009;5(22):23–6.
Sanchez-Sotelo J, Cofield R, Rowland C. Shoulder hemiarthroplasty for glenohumeral arthritis
associated with severe rotator cuff deficiency. JBJS. 2001;83-A:1814–22.
Williams GR Jr, Rockwood CA Jr. Hemiarthroplasty in rotator cuff-deficient shoulders.
J Shoulder Elbow Surg. 1996;5(5):362–7.
Boileau P. Neer award 2005: the Grammont reverse shoulder prosthesis: results in cuff tear
arthritis, fracture sequelae, and revision arthroplasty. J Shoulder Elbow Surg. 2006;15(5):
527–40.
Guery J, Favard L, Sirveaux F, Oudet D, Mole D, Walch G. Reverse total shoulder arthro-
plasty: survivorship analysis of eighty replacements followed for five to ten years.
JBJS. 2006;88:1742–7.
Sirveaux F, Favard L, Oudet D, Huquet D, Walch G, Mole D. Gammont inverted total shoulder
arthroplasty in the treatment of glenohumeral osteoarthritis with massive rupture of the cuff:
results of a multicentre study of 80 shoulders. Bone Joint J. 2004;86(3):388–95.
Susan BH. Basic biomechanics. Boston, MA: McGraw-Hill; 2007.
Michael L, Kelley T. Nonsurgical management of osteoarthritis of the knee.
JAAPA. 2006;19(1):26–32.
Edwards TB, Kadakia NR, Boulahia A, Kempf J-F, Boileau P, Némoz C, et al. A comparison
of hemiarthroplasty and total shoulder arthroplasty in the treatment of primary glenohumeral
osteoarthritis: results of a multicenter study. J Shoulder Elbow Surg. 2003;12(3):207–13.
Lo IK, Litchfield RB, Griffin S, Faber K, Patterson SD, Kirkley A. Quality-of-life outcome
following hemiarthroplasty or total shoulder arthroplasty in patients with osteoarthritis: a pro-
spective, randomized trial. JBJS. 2005;87(10):2178–85.
6 Biomechanics of the Shoulder 145
Radnay CS, Setter KJ, Chambers L, Levine WN, Bigliani LU, Ahmad CS. Total shoulder
replacement compared with humeral head replacement for the treatment of primary glenohu-
meral osteoarthritis: a systematic review. J Shoulder Elbow Surg. 2007;16(4):396–402.
Hedtmann A, Werner A. Shoulder arthroplasty in rheumatoid arthritis. Der Orthopade.
2007;36(11):1050–61.
Levy JC, Virani N, Pupello D, Frankle M. Use of the reverse shoulder prosthesis for the treat-
ment of failed hemiarthroplasty in patients with glenohumeral arthritis and rotator cuff defi-
ciency. Bone Joint J. 2007;89(2):189–95.
Chapter 7
Biomechanics of Spine
Lizhen Wang, Zhongjun Mo, Yuanjun Zhu, Enze Zhou, and Yubo Fan
Abstract Spinal disease including primary and secondary tumors occurs in various
tissues such as nerve root, blood vessel, spinal cord, and so on. There was about 9.7
patient with spinal tumor in every 1 million. Spine laminectomy has usually been
applied in treating the spinal cord tumor. Spinal instability after spine surgery was
observed with high occurrence rate, due to excision of posterior structures. Total
disc replacement (TDR) is an effective surgical method as well as anterior cervical
discectomy and fusion (ACDF) to decompressing neural elements and restore disc
height. With regard to keeping cervical spine’s normal kinematics, TDR is more
feasible and widely accepted than ACDF for treating degenerative cervical disc dis-
ease. It is important to investigate the biomechanical performances of ligament and
vertebral on the spine stability in repair surgery, which would provide the theoretical
suggestion to disc or cage design. In this chapter, the cervical spine (C2–C7) with
lamina repair surgery was studied at C3–C6 segments. The spinal stability in the
adjacent segments with posterior ligaments excised was worse than the one with
posterior ligament repaired. Repairing or preserving the posterior ligaments in the
lamina repair surgery is benefit to spinal integrity and stability. On the other hand,
the design of disc and the placement location of it were important factors for the
postoperative rehabilitation, especially for the ROM (range of motion) of flexion/
extension and the stress of implant. Therefore, reserving the range of motion and
avoiding adverse problems should be taken into consideration, and more attentions
also should be paid to a proper implant position along the anterior-posterior direc-
tion in ADR surgery.
Spinal tumors including primary and secondary tumors occurs in various tissues
such as nerve root, blood vessel, spinal cord, and so on. The incidence rate of spinal
tumor was 9.7 in 1 million [1]. Cervical laminectomy has usually been applied in
treating cervical spinal cord tumor [2]. However, the occurrence rate of spinal insta-
bility after laminectomy was reportedly 20% in adults [3] and up to 45% in children
[4], since the posterior structures of vertebrae, including interspinous ligament,
supraspinous ligament, flaval ligament, and spinous process, also was excised with
laminectomy [5]. Excision of posterior structures causes movement of the anterior
vertebrae body, which induces declination and torsion of vertebral body, resulting in
spinal instability [6, 7]. Previous studies reported the increase in ROM after lami-
nectomies, which was likely to result in developing spinal kyphosis or other defor-
mity [8–16]. Spinal deformity following laminectomy would oppress spinal cord or
nerve root, and subsequently result in neurological symptoms as back pain and
radiculopathy.
In order to avoid the complications caused by laminectomy, lamina repairment
was firstly applied in clinics to treat intraspinal lesions in 1976 [17]. Since then,
repairing of the excised lamina simultaneously with excision of the neoplasm was
advocated to maintaining spinal stability and preserving daily activities of the spine
[2, 18]. Good clinical results were found in the follow-up of the patients after lamina
repairment surgery, which can maintain spinal integrity, and prevent postoperative
instability, subluxation, and kyphotic deformities [19–22].
7 Biomechanics of Spine 149
To develop the finite element model of the intact cervical spine (C2–C7), computer-
ized tomographies (CT) of a healthy volunteer (male, 28 years, 60 kg, 173 cm) were
achieved by CT scanner (Brilliance iCT, Philips, the Netherlands). The corresponding
ethical committee has approved this research plan (No. IRB00006761-L2010021),
and the participant signed the informed consent form. The CT scan images were
imported into medical image-processing software (Mimics 15.1, Materialise Inc.,
Belgium) to rebuild the vertebrae’s geometry. The intervertebral space was filled with
a solid block as disc, which was divided into nucleus pulposus and annulus ground
substance in a ratio of 4.3:5.7 [27, 28] by CAD software (Solidworks 2012, Dassault
Inc., France). The vertebrae and intervertebral disc were meshed into tetrahedral ele-
ments and hexahedral elements, respectively, by using Hypermesh (12.0, Altair Inc.,
America). Finally, all the meshed vertebrae and discs were imported into finite ele-
ment software (ABAQUS 6.14, Simulia Inc., USA) as separated solid volumes. A
layer of shell with a thickness of 0.4 mm was generated by sharing the common nodes
with the solid vertebrae volume (cancellous bone), and partitioned into cortical shell
and endplate region. A layer of netlike truss elements (annulus fiber) were attached on
the circumferential surface of the ground substance to simulate annulus fibrosus by
sharing the common nodes. Intervertebral ligaments, including anterior longitudinal
ligament (ALL), capsular ligament (CL), posterior longitudinal ligament (PLL), liga-
mentum flavum (LF), and interspinous ligaments (ISL), were simulated using tension-
only nonlinear truss element (Fig. 7.1). In the healthy model of cervical spine, the total
numbers of nodes and elements are 47,068 and 176,373, respectively. The material
properties and mesh type of cervical components [29, 30] are listed in Table 7.1.
150 L. Wang et al.
Table 7.1 Mesh types and material properties of the cervical spine components
Young modulus Poisson Cross section
Components Element type (MPa) ratio area (mm2)
Cortical bone S3 12,000.0 0.30 –
Cancellous bone C3D4 450.0 0.30 –
Endplate S3 1000.0 0.40 –
Articular cartilage C3D6 10.0 0.30 –
Annulus fiber T3D2 450.0 0.30 –
Annulus ground C3D8 3.4 0.40 –
substance
Nucleus pulposus C3D8 1.0 0.49 –
Mini-plate and screw
C2–C5 ALL T3D2 26.3 0.40 11
PLL T3D2 22.2 0.40 11
FL T3D2 3.1 0.40 46
CL T3D2 3.3 0.40 42
ISL T3D2 4.9 0.40 13
C5–C7 ALL T3D2 28.2 0.40 12
PLL T3D2 23.0 0.40 14
FL T3D2 3.5 0.40 49
CL T3D2 4.8 0.40 50
ISL T3D2 5.0 0.40 13
7 Biomechanics of Spine 151
Based on the healthy model, all the FL and ISL at C2–C3 and C6–C7 levels were
removed, and both the left and right sides of the laminas at C3–C6 were cut by gaps
with 2 mm width, and then titanium alloy mini-plate is used to fix the lamina and
spinous process to simulate the lamina repairment surgery (Fig. 7.2). The lamina
ligament repair model (LLRM) was based on the lamina repair model (LRM) by
reconstructing the FL and ISL at C2–C3 and C6–C7 segments. The metal compo-
nents in the titanium mini-plate and screw were made of Ti6Al4V with the sizes of
the mini-plate as 10 × 2 × 0.5 mm (length × width × height) and the screw as
4 × 1.6 mm (length × diameter).
The ligaments were inserted in corresponding position and attached on the adjacent
vertebrae, to which the intervertebral disc was also adhered. The interaction between
the facet joint cartilages was defined as surface-to-surface contact formulation with
0.1 friction coefficient. The bone-implant interface was assigned with a tie con-
straint to simulate thorough osseointegration. All models were immobilized at the
The ROM of each motion segment in the healthy model subjected to the pure
moments of 1.5 Nm is shown in Fig. 7.3. The ROM was in agreement with the
in vitro experimental data from the literature [31–33].
Compared with the healthy model, in the LRM, the ROM of C2–C3 in flexion, lateral
bending, and axial rotation increased by 113.6%, 23.6%, and 28.5%, while that of
C6–C7 increased by 88.9%, 12.8%, and 20.7%, and change of ROM in other seg-
ments under different conditions was less than 9.2%. The change of ROM at the cor-
responding levels in LLRM was lower than 7.2% compared to the healthy model
(Fig. 7.4).
18
Healthy
16 Moroney (1988)
14 Panjabi (2001)
Finn (2009)
Rotation (degree)
12
10
8
6
4
2
0
C2/C3
C3/C4
C4/C5
C5/C6
C6/C7
C2/C3
C3/C4
C4/C5
C5/C6
C6/C7
C2/C3
C3/C4
C4/C5
C5/C6
C6/C7
C2/C3
C3/C4
C4/C5
C5/C6
C6/C7
Fig. 7.3 Comparison of ROM in healthy cervical spine and data reported in the literature
7 Biomechanics of Spine 153
Healthy
12 Lamina repair
Lamina and ligament repair
10
Rotation (degree)
0
C2/C3
C3/C4
C4/C5
C5/C6
C6/C7
C2/C3
C3/C4
C4/C5
C5/C6
C6/C7
C2/C3
C3/C4
C4/C5
C5/C6
C6/C7
C2/C3
C3/C4
C4/C5
C5/C6
C6/C7
Flexion Extension Lateral Bending Axial Rotation
Fig. 7.4 ROM comparison of the healthy cervical spine and surgical models
9
Healthy
8 Lamina repair
Lamina and ligament repair
Facet Joint Contact Stress (MPa)
0
C2-C3(RB) C3-C4(Ex) C4-C5(Ex) C5-C6(RB) C6-C7(LB)
Fig. 7.5 Contact stress in facet joint in the healthy cervical spine and surgical models
The maximal contact stress in the facet joint in each motion segment is shown in
Fig. 7.5. In the LRM, the facet cartilage at C2–C3 was in a zero-stress state in flex-
ion. Compared to healthy model, the maximal contact stresses of C2–C3 decreased
by 31.0% in right bending. Compared to healthy model, the change of the maximal
contact stresses in the LLRM was less than 3.0% in each motion segment.
154 L. Wang et al.
0.6
Healthy
Lamina repair
Intervertebral Pressure (Mpa)
0.4
0.3
0.2
0.1
0
Fl Ex LB RB LR RR Fl Ex LB RB LR RR
C2-C3 C6-C7
Fig. 7.6 Intradiscal pressure in the healthy cervical spine and surgical models
1.4.3 Intradiscal Pressure
In the LRM, the stress of capsular ligament under different loading conditions
increased by 4.5–168.6% at C2–C3 and 9.5–115.6% at C6–C7 compared with the
healthy model. However, in the LLRM, the change of the stresses (C2–C3, C6–C7)
in all conditions was lower than 4.4% as shown in Fig. 7.7.
The stresses on the bone-screw interface and in the screw-plate system of C3–C6
are listed in Table 7.2. In the two kinds of surgical models, the differences of maxi-
mum stresses on screw and bone-screw interface was less than 6 MPa (C4 and C5)
and 106 MPa (C3 and C6), respectively.
4.5
Healthy
4 Lamina repair
3.5 Lamina and ligament repair
3
Stress (MPa)
2.5
2
1.5
1
0.5
0
Fl Ex LB RB LR RR Fl Ex LB RB LR RR
C2-C3 C6-C7
Fig. 7.7 Stress in the CL in the healthy cervical spine and surgical models
Table 7.2 Maximum Von Mises stress on bone-screw interface and screw-plate system (MPa)
Model C3 C4 C5 C6
Screw LRM 144.6 115.9 130.4 116.2
LLRM 112.9 115.1 133.9 164.8
Titanium plate LRM 259.3 166.5 220.5 257.4
LLRM 153.6 173.8 214.6 359.8
Bone-screw interface LRM 1.458 1.45 2.085 1.844
LLRM 1.69 1.389 2.105 1.654
prevent the development of spinal instability and kyphosis [20–23]. Nong et al. [34]
performed a cadaveric study and proved that the lamina repairment surgery brought
more stability than laminectomy. Healy et al. [35] also performed a cadaveric study
to explore the effect of ISL on the spinal stability, and found that after excision of
the ISL between C2–C3 and C7–T1, the ROM of C2–T1 increased by 7.9%, 2.4%,
and 5.6% in flexion, bending, and rotation, while the ROM of C2–C3 and C7–T1
increased by 36.5% and 25.4% in flexion. Other studies demonstrated the impor-
tance of the ISL in the enhancement of spinal flexion stability, and they proposed
methods to conserve or repair the posterior ligament during laminectomy [23–26].
In the present study, a healthy cervical model (C2–C7), a lamina repair model,
and a lamina ligament repair model were built. The ROM of C2–C3 and C6–C7 in
LRM had an obvious increase which range from 12.8% to 113.6% compared to the
healthy model, whereas the ROM of C2–C3 and C6–C7 in LLRM were close to the
healthy model (<7.2%). The significant difference of ROM in the adjacent segments
(C2–C3 and C6–C7) in LRM and LLRM was due to whether the posterior liga-
ments existing. Since the physiologic role of ISL and FL include a tethering or ten-
sion constraint during anterior flexion [21], without ISL and FL can lead to a
significant increase of the ROM, which was consistent with the experimental results
156 L. Wang et al.
by Healy [35]. However, the changes of ROM at C3–C4, C4–C5, and C5–C6 in
LRM and LLRM were not apparent in comparison with healthy model (<9.2%).
In LRM, the maximal contact stress in facet joint at C2–C3 was 0 MPa in flexion
due to the absence of posterior ligaments, in comparison with healthy model, but the
maximal stress of C2–C3 decreased by 31.0% in right bending. Nevertheless, there’s
no obvious change of maximal facet contact stress at each adjacent segments in the
LLRM (<2.9%).
There is an evident increase of intradiscal pressure at C2–C3 and C6–C7 up to
73.7% in LRM, while the change at the corresponding segments in LLRM was not
obvious (<11.5%). In LRM, the intradiscal pressure at C2–C3 and C6–C7 both
increased by 73.7% in flexion, because the excision of posterior structures causes
movement of the anterior part of vertebrae around the nucleus pulposus [6, 7], and
the posterior ligaments had not been repaired in LRM; therefore, the disks in C2–C3
and C6–C7 have suffered much higher intradiscal pressure in flexion. The increase
in intradiscal pressure is related to adjacent segment degenerations.
Compared to the healthy model, the maximal stresses in capsular ligament at
C2–C3, C6–C7 in LRM increased obviously by up to 168.6%. Exaggerated stresses
do harm to the ligaments and may affect long-term clinical results. However, the
change of stresses at each motion segments in LLRM was small (<4.4%).
In LRM and LLRM, the difference of stresses on bone-screw interface and
screw-plate system in C4, C5 was less than 6 MPa (2.7%), but the difference in C3,
C6 was up to 105.7 MPa (41.8%).
Compared with the healthy model, the ROM, intervertebral pressure, facet joint
contact stress, stresses of capsular ligament at C2–C3, C6–C7 had significant
changes under different loading conditions in LRM. However, those parameters in
LLRM were close to healthy cervical spine. In addition, the stresses on bone-screw
interface and screw-plate system (C3, C6) between LRM and LLRM had great dif-
ferences, because of the integrity of posterior ligaments. Besides, the variation ten-
dency between LRM and LLRM was almost same in terms of ROM, intervertebral
pressure, contact stress in facet joint, stresses in capsular ligament, and stresses on
bone-screw interface and in screw-plate system (C3–C4, C4–C5, C5–C6). It meant
that lamina repairment with posterior structure preserved can maintain stability of
the cervical segments.
The study focused on the effect of with and without posterior ligaments in lamina
repair surgery, and explored the biomechanical properties of cervical spine such as
ROM, intervertebral pressure, facet joint contact stress, stresses of capsular liga-
ment, etc. However, the soft tissues in maintaining the cervical spine stability is
important; therefore, as part of this study to improve the accuracy of the cervical
spine model, a more complete model with soft tissues (muscles) will be built in
future study.
The spinal stability in the adjacent segments with posterior ligaments excised
was worse than the one with posterior ligament repaired. Repairing or preserving
the posterior ligaments in the lamina repair surgery is benefit to spinal integrity and
stability. It is suggested to conserve or reconstruct the posterior ligaments in lamina
repair surgery if the operation technology is realizable.
7 Biomechanics of Spine 157
The geometry model of the vertebrae was reconstructed based on the computed
tomography (CT) scan images with the slice of 0.625 mm from a healthy male
(32 years, 68 kg, 170 cm) through a commercial software MIMICS (Materialise,
Belgium). Then a 3D nonlinear finite element model of the cervical spine was estab-
lished using software ABAQUS 6.11 (Simulia Inc.). The intact FE model consists of
intervertebral disc, endplate, and five groups of ligaments (anterior longitudinal, pos-
terior longitudinal, capsular, ligamentum flavum, and interspinous ligaments). A
layer of 0.4 mm thick shell was used to model the cortical bone surrounding the
cancellous core. A nonlinear 3D contact was set to the facet joint. The intervertebral
disc consists of the nucleus pulposus and the annulus fibrosus. The nucleus pulposus
accounts for 33% of the total disc volume surrounded by the annulus fibrosus. Three-
node link elements were used to simulate the annulus fibers, which were placed
approximately 30 from the horizontal plane. Truss elements were used to reconstruct
the ligaments and resisted tension only [40]. The detailed information about material
properties and element types used to reconstruct the FE model of the cervical spine
is described below (Table 7.1).
The basic ball-and-socket disc prostheses were constructed in ABAQUS 6.11
(Simulia Inc.). The geometry parameters were obtained from the product called
Prodisc-C (Synthes Inc., West Chester, PA, USA). The Prodisc size M (width
15 mm, depth 12 mm, height 5 mm) with 5 mm radius of curvature was chosen to
fit the specimen properly, and which was named as ProC-R5 [40].
Similarly, ProC-R4 with 4 mm radius of curvature and ProC-R6 with 6 mm
radius of curvature were generated. The heights of the three models are consistent,
and the superior endplate “socket” was altered for fitting the “ball” component with
different radius of curvature (Fig. 7.8). The material properties of the cobalt-
chromium- molybdenum endplates (Alloy of CoCrMo) and polyethylene core
(UHMWPE) were set as linearly elastic materials (E = 220,000 MPa, υ = 0.32;
E = 1000 MPa, υ = 0.49). The spherical joint between the two components was set
as face-to-face contact with no friction to model the kinematical behavior. Then
three FE models of different radiuses of curvature were generated and meshed into
tetrahedron elements [40].
To simulating the anterior arthroplasty, the anterior longitudinal ligaments and
intervertebral disc at C5–6 level were deleted. Each prostheses of intervertebral disc
had 5 implantation sites that vary along the anteroposterior direction and lateral direc-
tion. And then these three types of prostheses were implanted at C5–6 level commonly
7 Biomechanics of Spine 159
b
5mm
c
6mm
applied in surgery. As some studies had demonstrated the finite axes of rotation were
sited below the disc and posterior to the endplate center [40, 42]. The center of the
prostheses was set at a position called neutral position, and we named them N4, N5,
N6 (4, 5, 6 for radius of curvature, N for neutral), respectively. The models implanted
2 mm anterior to the neutral position were named A4, A5, A6 (A means anterior). In
the same way, other implanted models were built named P4, P5, P6 (1.2 mm Posterior
to the neutral position; note that, the moving distance was different because the neutral
position wasn’t at central location of endplate), L4, L5, L6 (1 mm Left to the neutral
position), and R4, R5, R6 (1 mm Right to the neutral position). Apart from the anterior
position (15° from the horizontal plane), all prostheses were implanted at an angle of
12° to the horizontal for fitting the super-inferior endplate of this model well [40].
The ROM of the C5–6 intact model in flexion//extension, lateral bending, and axial
rotation was 7.41°, 4.23°, and 5.49°, respectively [40]. Compared with the previous
studies [42], the ROM of the intact model was consistent with the published data
160 L. Wang et al.
above. In addition to the validation, Lee et al. reviewed and summed up the ROM
data from previous FE researches and in vitro experiments, finding that ROM of
C5–6 segment ranged from 7.2° to 9.9° for flexion/extension in sagittal plane, ROM
of C5–6 segment ranged from 3.1° to 15.4° for bilateral bending in coronal plane,
and ROM of C5–6 segment ranged from 2.3° to 13.3° for axial rotation in transverse
plane. Based on the fact that all the ROMs of C5–6 segment were within the range
of results, we considered the motion unit of the 3D finite element model was valid.
7.41°, 4.23°, and 5.49° were observed in the ROM of the intact model in sagittal
plane, coronal plane, and transverse plane, respectively. Compared with the intact
FE model, both the ROM of the FE models implanted with different Prodisc-C
prostheses and positions were all rose under the same external loading conditions.
In flexion/extension load, the ROMs of ProC-R4 and ProC-R5 were approximately
the same (difference <3%), but a remarkable increase was observed in ROM com-
pared with ProC-R4 and ProC-R5 in any position. In lateral bending and axial rota-
tion load, no apparent differences were seen among ProC-R4, ProC-R5, and
ProC-R6 (<2% change). We can also find that there was a trend that the posterior
position caused higher level of ROM to the neutral, middle, left, and right positions
under every loading condition. The closest ROM was observed in N5 (ProC-R5
model with neutral position) compared with intact model. The highest ROM was
observed in P6 model (ProC-R6 with posterior position), which was 25.2% and
23.9% higher than P4 and P5, respectively. The ROM of the intact model and mod-
els implanted with different Prodisc-C prostheses is described in Table 7.3.
In flexion, the force of facet joint was observed in all positions for the prosthesis
with 4 mm radius of curvature (ProC-R4), and second one was ProC-R5, and the
largest force of facet joint occurred in the ProC-R6. In terms of the positions, facet
force was highest in posterior among all positions. P6 model showed the maximum
facet force (47.1 N), increased by 106.9% to the intact model. Compared with the
Table 7.3 The ROM of the intact model and models implanted with different Prodisc-C prostheses
Flexion&Extension (°) Left&Right bending (°) Axial torsion (°)
Intact model 7.41 4.23 5.49
Pro-C-M 9.14 4.58 6.13
Pro-C-A 9.07 4.67 6.54
Pro-C-P 12.44 6.06 8.36
Pro-C-L 11.46 5.73 8.05
Pro-C-R 11.80 5.93 8.20
7 Biomechanics of Spine 161
intact model, an increase was observed in force transmitted through the facet joints
in all cases under extension, left-right lateral bending, and right axial torsion.
Similar facet joint force was also observed in prostheses with different radius size
(4, 5, 6 mm) in all the conditions. However, the changes were significant with the
variation of the positions of implant. In left axial torsion, all models except for the
anterior position showed the decrease of the facet force, with the increase of the A4,
A5, and A6 by 2.69%, 7.23%, and 2.34%, respectively.
In flexion, the stress of the posterior longitudinal ligament (PLL) tensile in the intact
model was 2.577 MPa as shown in Table 7.4. The increased percentage was 10.48%,
20.14%, 34.69%, 25.81%, and 30.77% for the ProC-R4 models with neutral, ante-
rior, posterior, left, and right positions, respectively; 5.63%, 10.48%, 45.67%,
33.14%, and 37.10% for the ProC-R4 models; and 42.18%, 45.36%, 80.05%,79.71%,
and 86.03% for the ProC-R6. The stress of the capsular ligament (CL) was
Table 7.4 Ligaments tensile stress in flexion, extension, left bending, right bending, left torsion,
and right torsion for intact model, ProC-R4, ProC-R5, and ProC-R6
PLL CL FL SL
Flexion Intact model 2.577 2.698 0.4802 0.4288
ProC-R4 2.858 3.003 0.5233 0.4918
ProC-R5 2.722 2.765 0.4792 0.4387
ProC-R6 3.693 3.713 0.6099 0.5681
Extension Intact model 0 0.7841 0 0
ProC-R4 0.5302 1.882 0 0
ProC-R5 0.585 1.987 0 0
ProC-R6 0.5522 1.923 0 0
Left bending Intact model 0.8529 2.297 0.1217 0.07164
ProC-R4 1.133 2.897 0.1438 0.08785
ProC-R5 1.166 2.908 0.1436 0.0894
ProC-R6 1.175 2.914 0.1373 0.0894
Right bending Intact model 0.8197 2.117 0.1761 0.09752
ProC-R4 1.137 2.802 0.1957 0.1143
ProC-R5 1.147 2.659 0.1944 0.1115
ProC-R6 1.206 2.794 0.1838 0.1078
Left torsion Intact model 0.5332 2.928 0.1151 0.03631
ProC-R4 0.8837 3.448 0.1341 0.0475
ProC-R5 0.8977 3.409 0.1313 0.0468
ProC-R6 0.9331 3.466 0.1302 0.051
Right torsion Intact model 0.6196 2.316 0.1768 0.09282
ProC-R4 1.003 3.225 0.1983 0.1106
ProC-R5 1.008 3.014 0.1988 0.107
ProC-R6 1.034 3.213 0.1835 0.1018
162 L. Wang et al.
2.698 MPa in flexion. The increased percentage was 5.74%, 18.35%, 34.40%,
14.53%, and 36.77% for the ProC-R4 models with neutral, anterior, posterior, left,
and right positions, respectively; 2.48%, 7.15%, 43.66%, 19.46%, and 40.96% for
the ProC-R4 models; and 39.66%, 42.18%, 87.88%, 67.27%, and 95.48% for the
ProC-R6.
In extension, the tensile stress of PLL was zero, 0.438, 0.637, 0.987, and
0.874 MPa in the ProC-R4 models in five positions, respectively; 0.585, 0.864,
1.171, 1.008, and 1.334 MPa for the ProC-R5 models; and 0.519, 0.7662, 1.050,
0.969, and 1.292 MPa for the ProC-R6 models. Also, for the tensile stress of CL
apparently increasement was seen in all models, which was up to 114.39–305.31%
compared with the intact model.
In lateral bending and axial rotation, compared with the intact model, increase-
ment was observed in all models for the tensile stress of PLL and CL. However, the
ligament tensile stress of different radiuses and position models were similar, no
marked changes observed.
The stresses on the polyethylene (PE) ball component of different designs were
measured under flexion, extension, bilateral bending, and axial rotation conditions,
and the stresses were maximal in flexion compared with other loading conditions.
In flexion, the stress of 93.96, 54.63, 127, 128.6, and 130.7 MPa were seen on poly-
ethylene ball of ProC-R4 models with neutral, anterior, posterior, left, and right
positions, respectively; 75.61, 84.75, 97.66, 87.91, and 111.7 MPa were seen on
polyethylene ball of the ProC-R5 models; and 16.18, 16.77, 15.97, 18.3, and
19.55 MPa were seen on polyethylene ball of the ProC-R6 models, which decreased
markedly. In all positions, the stresses distributions of ProC-R4 and ProC-R5 were
concentrated on the part anterior to the PE ball and the maximum stresses—130.7
and 111.7 MPa—were observed in right position. As for the ProC-R6, the stress was
distributed at the edge of the joint between the hemisphere and plate, and the maxi-
mum stress—19.55 MPa—was also observed in right position as shown in Table 7.5.
In this part, we investigated and quantified the theoretical effects on the spine
biomechanics of the ball-and-socket TDR at the C5–6 level with prostheses of 3
designs and 5 implanted positions. These variations were related to not only design
parameters (radius of curvature) but also surgical procedure (the positioning and
insertion trajectory varied along the anteroposterior direction and lateral direction).
Table 7.5 Stress on the ball component in flexion, extension, left bending, right bending, left
torsion, and right torsion for intact model, ProC-R4, ProC-R5, and ProC-R6
Flexion Extension Left bending Right bending Left torsion Right torsion
ProC-R4 144.2 29.52 16.72 16.93 17.2 16.59
ProC-R5 75.61 18.95 13.96 13.65 14.47 13.95
ProC-R6 12.06 17.39 4.932 5.144 5.661 4.885
7 Biomechanics of Spine 163
We observed the major changes in ROM, stress on polyethylene ball, the force of
the facet joint, and the tensile stress of ligament, which were influenced by the sizes
of implant and the implanted position.
After the implantation of prostheses, the ROM data of C5–C6 finite element
cervical model in this study presented an increase compared with the intact model.
This result was in accordance with the previous study, in which ROM increased
after replacing the disc with the semi-constrained ProDisc-C at the treated segment.
Meanwhile, in flexion/extension there was increasement observed in the ROMs of
ProC-R4, ProC-R5, and ProC-R6 and the increasement was lager in ProC-R6.
However, we do not find the same phenomenon in lateral bending and axial rotation.
These suggested that the influence of different radius of curvature on the ROM was
not obvious apart from flexion/extension. Rousseau et al. [39] verified our results
through the finite element analysis of three types of ball-and-socket designs (PL,
PS, and CL), in which they found that the ROMs increased in flexion/extension but
was consistent with the intact model in axial rotation and bilateral bending.
Most studies found the ROMs can preserve the general motion and similar to the
physiological values, regardless of the various prosthesis designs, which mean pros-
thesis design more strongly related to the quality of motion, than to the quantity
[41]. Some similar researches have indicated that the typical motion quality was the
location of rotation axis [43, 44]. The mean center of rotation (MCR) was analyzed,
finding that the intact model’s MCR was at the posterior one-third of the lower ver-
tebra endplate and varied in a certain area under flexion/extension [39]. With regard
to different types of prostheses, MCR presented a trend to match the prosthesis
center. It is a matter of course that the variation of the position of the implant accom-
panied by that of MCR based on this fact. Therefore, the larger ROM was seen in
posterior position models, the rotation axis of which was adjacent to the actual
center of rotation axis. Take the radius of curvature into account; in order to keep the
height of the disc, the MCR of ProC-R4, ProC-R5, and ProC-R6 would change as
the hemisphere moves up and down. Probabilistic variables were investigated in the
center of rotation (COR). The intervertebral rotation was affected by the anteropos-
terior position of COR only in flexion/extension, but not being affected by the lat-
eral position of the COR. In this part, it is really no apparent difference from left to
right positions, but they all differed greatly from the neutral position. After the pros-
thesis being implanted, the force of the facet joint was observed to be less than that
of ProC-R5 and ProC-R6 in most kinetic situations, and there were significant dis-
tinctions in flexion, with the value of P6 being the largest. It was also found that the
maximum ROM was seen in ProC-R6 model with posterior position (P6) in flexion/
extension. It seemed that the larger force of facet joint was caused by the lager
ROM. Although some researches have pointed out this phenomenon [44], the rele-
vance between ROM and force of facet joint still remained to be figured out.
Womack et al. employed ProDisc-C models varying with 5-, 6-, and 7-mm implant
heights to explore the effects of the size of implant, founding that the size of implant
influenced contact force of facet without influencing the ROM [45]. It was demon-
strated that the force of facet joint was vulnerable to the variation of implanted posi-
tions, but no tendency was observed.
164 L. Wang et al.
The capsular ligament (CL) was associated with the force of facet joint and the
related pain, so it was paid main attention in preview studies. In the part, increased
tensile stress of CL was observed in all cases after implantation. And the ligament
tensile stress of larger radius model with posterior position is the largest, while cap-
sular ligament tension of model with neutral and anterior positions slightly reduced.
The overmuch tensile stress companied by the force of facet joint may cause fatigue
damage, and partially affected the adjacent ligament, finally had a bad impact on the
long-term clinical results [40].
For each prosthesis implanted, the stress distribution on the core of PE is
approximately similar, no matter how the position of implant varied, and the max-
imum value of stress was witnessed in flexion. The stress of ProC-R4 and ProC-R5
concentrated on the part anterior to the PE ball. In contrast, the pressure of
ProC-R6 was lower and spread over the joint edge between plate and hemisphere.
In view of this point, ProC-R6, a prosthesis having a larger curvature, seemingly
could lower the stress of implant and avoid the concentration of stress. In the long
run, this (large curvature) design may delay spherical bearing wear on surfaces,
thereby avoiding the inflammatory reactions caused which wear debris led to.
Speaking of wear debris, most studies primarily focused on osseointegration and
biological response (to wear debris) at the interface between prosthesis and bone.
A caprine model was used to assess the biomechanical characteristic. No evi-
dence of wear particles or loosening implant (subluxation) was found, while it
was reported that one patient implanted the PCM disc had a large amount of wear
debris [40, 46].
Based on the observation, the ROM, force of facet joint and tension of capsule
ligament were affected by the radius of implanted prosthesis, while the position
had an influence on all these aspects under all loading conditions. These aspects
(the ROM, force of facet joint and tension of capsule ligament) are sensitive to the
position in anteroposterior direction. The disc having larger curvature has less
stress on the polyethylene, which can avoid stress concentration, but bring adverse
outcomes such as increased force of facet joint and tension of ligament. Therefore,
the design of disc prosthesis should be considered retaining the range of motion as
well as avoiding the adverse problems, so as not to affect the long-term clinical
results [40].
In summary, surgical procedure such as repairing or preserving the posterior
ligaments in the lamina repair surgery and better implant design are important fac-
tors for the postoperative rehabilitation, especially the range of motion (ROM) of
flexion/extension and implant stress, and spinal integrity and stability. Thus, a
proper implant design should consider reserving the range of motion as well as
avoiding adverse problems, and a proper surgical procedure and implant position
along the anterior-posterior direction ought to be paid more attentions in spine
biomechanics.
Acknowledgement This work was supported by Beijing Municipal Science and Technology
Project (No. 161100001016013).
7 Biomechanics of Spine 165
References
1. Duetzmann S, Cole T, Ratliff JK. Cervical laminoplasty developments and trends, 2003–2013:
a systematic review. J Neurosurg Spine. 2015;23:24–34.
2. Hirabayashi K, Watanabe K, Wakana KR, et al. Expansive open-door laminoplasty for cervical
spinal stenotic myelopathy. Spine. 1983;8:693–9.
3. Katsum Y, Honma T, Nakamura T. Analysis of cervical instability resulting from laminecto-
mies for removal of spinal cord tumor. Spine. 1989;14:1171–6.
4. Yeh JS, Sgouros S, Walsh AR, et al. Spinal sagittal malalignment following surgery for pri-
mary intramedullary tumours in children. Pediatr Neurosurg. 2001;35:318–24.
5. Haddadi A, Qazvini HR. Outcome after surgery of lumbar spinal stenosis: a randomized com-
parison of bilateral laminotomy, trumpet laminectomy, and conventional laminectomy. Front
Surg. 2016;3:19. https://doi.org/10.3389/fsurg.2016.00019.
6. Kotani Y, Cunningham BW, Abumi K, et al. Biomechanical analysis of cervical stabiliza-
tion systems: an assessment of transpedicular screw fixation in the cervical spine. Spine.
1994;19:2529–39.
7. McGirt MJ, Garces-Ambrossi GL, Parker SL, et al. Short-term progressive spinal defor-
mity following laminoplasty versus laminectomy for resection of intradural spinal tumors:
analysis of 238 patients. Neurosurgery. 2010;66:1005–12. https://doi.org/10.1227/01.
NEU.0000367721.73220.C9.
8. Goel VK, Clark CR, Harris KG, et al. Kinematics of the cervical spine: effects of multiple total
laminectomy and facet wiring. J Orthop Res. 1988;6:611–9.
9. Kode S, Kallemeyn NA, Smucker JD, et al. The effect of multi-level laminoplasty and lami-
nectomy on the biomechanics of the cervical spine: a finite element study. Iowa Orthop
J. 2014;34:150–7.
10. Tadepalli SC, Gandhi AA, Fredericks DC, et al. Cervical laminoplasty construct stability: an
experimental and finite element investigation. Iowa Orthop J. 2011;31:207–14.
11. Xie T, Qian J, Lu Y, et al. Biomechanical comparison of laminectomy, hemilaminectomy and
a new minimally invasive approach in the surgical treatment of multilevel cervical intradu-
ral tumour: a finite element analysis. Eur Spine J. 2013;22:2719–30. https://doi.org/10.1007/
s00586-013-2992-1.
12. Saito T, Yamamuro T, Shikata J, et al. Analysis and prevention of spinal column deformity fol-
lowing cervical laminectomy. I. Pathogenetic analysis of postlaminectomy deformities. Spine.
1991;16:494–502.
13. Ng HW, Teo EC, Zhang QH. Influence of laminotomies and laminectomies on cervical spine
biomechanics under combined flexion-extension. J Appl Biomech. 2004;20:243–59.
14. Ng HW, Teo EC, Zhang QH. Biomechanical effects of C2-C7 intersegmental stability due to
laminectomy with unilateral and bilateral facetectomy. Spine. 2004;29:1737–45.
15. Ogden AT, Bresnahan L, Smith JS, et al. Biomechanical comparison of traditional and minimally
invasive intradural tumor exposures using finite element analysis. Clin Biomech. 2009;24:143–7.
16. Ahmed R, Menezes AH, Awe OO, et al. Long-term incidence and risk factors for develop-
ment of spinal deformity following resection of pediatric intramedullary spinal cord tumors. J
Neurosurg Pediatr. 2014;13:613–21.
17. Raimondi AJ, Gutierrez FA, Di RC. Laminotomy and total reconstruction of the posterior
spinal arch for spinal canal surgery in childhood. J Neurosurg. 1976;45:555–60.
18. Wiedemayer H, Schoch B, Stolke D. Osteoplastic laminotomy using titanium microplates for
reconstruction of the laminar roof: a technical note. Neurosurg Rev. 1998;21:93–7.
19. Jeon SC, Chough CK, Park HK, et al. Surgical technique and long-term follow-up of cervical
laminoplasty using titanium miniplates. J Korean Neurosurg. 2004;36:369–74.
20. Hida S, Naito M, Arimizu J, et al. The transverse placement laminoplasty using titanium
miniplates for the reconstruction of the laminae in thoracic and lumbar lesion. Eur Spine
J. 2006;15:1292–7. https://doi.org/10.1007/s00586-006-0099-7.
166 L. Wang et al.
21. Menku A, Koc RK, Oktem IS, et al. Laminoplasty with miniplates for posterior approach in
thoracic and lumbar intraspinal surgery. Turk Neurosurg. 2010;20:27–32.
22. Zhou D, Nong L, Gao G, et al. Application of lamina replantation with ARCH plate fixa-
tion in thoracic and lumbar intraspinal tumors. Exp Therap Med. 2013;6:596–600. https://doi.
org/10.3892/etm.2013.1170.
23. Hotta H. An experimental study on stability of human spine, especially the role of the lumbar
ligaments. J Jpn Orthop Assoc. 1976;50:1–14.
24. Sano S, Masuda A, Kabata K, et al. Laminectomy with spinous process reattachment—pre-
liminary report. Orthop Surg Traumatol. 1983;26:1227–30.
25. Joson RM, McCormick KJ. Preservation of the supraspinous ligament for spinal stenosis: a
technical note. Neurosurgery. 1987;21:420–2.
26. Hirofuji E, Tanaka K, Nakano A. Ligamentous reconstruction with artificial ligament to pre-
vent the unstable lumbar spine. Clin Orthop Surg. 1990;25:501–6.
27. Zhao Y, Li Q, Mo Z, et al. Finite element analysis of cervical arthroplasty combined with
fusion against 2-level fusion. J Spinal Disord Tech. 2013;26:347–50.
28. Mo Z, Zhao Y, Du C, et al. Does location of rotation center in artificial disc affect cervical
biomechanics? Spine. 2015;40:E469–75.
29. Yoganandan N, Kumaresan S, Pintar FA. Geometric and mechanical properties of human cer-
vical spine ligaments. J Biomech Eng. 2000;122:623–9.
30. Li Q, Mo Z, Fan Y. Biomechanical comparison of cervical arthroplasty with and without
posterior longitudinal ligament resection: a finite element study. J Med Imaging Health Inf.
2016;6:1559–65. https://doi.org/10.1166/jmihi.2016.1848.
31. Panjabi MM, Crisco JJ, Vasavada A, et al. Mechanical properties of the human cervical spine
as shown by three-dimensional load displacement curves. Spine. 2001;26:2692–700.
32. Finn MA, Brodke DS, Daubs M, et al. Local and global subaxial cervical spine biomechan-
ics after single-level fusion or cervical arthroplasty. Eur Spine J. 2009;18:1520–7. https://doi.
org/10.1007/s00586-009-1085-7.
33. Moroney SP, Schultz AB, Miller JAA, et al. Load-displacement properties of lower cervical
spine motion segments. J Biomech. 1988;21:769–79.
34. Nong L, Zhou D, Xu N, et al. Lamina replacement with titanium plate fixation improves
spinal stability after total lumbar laminectomy. Comput Methods Biomech Biomed Eng.
2014;18:1753–9.
35. Healy AT, Lubelski D, West JL, et al. Biomechanics of open-door laminoplasty with and with-
out preservation of posterior structures. J Neurosurg Spine. 2016;24:1–6.
36. Park JJ, Quirno M, Cunningham MR, et al. Analysis of segmental cervical spine vertebral
motion after prodisc-C cervical disc replacement. Spine. 2010;35(8):E285–9.
37. Galbusera F, Bellini CM, Brayda-Bruno M, et al. Biomechanical studies on cervical total disc
arthroplasty: a literature review. Clin Biomech. 2008;23(9):1095–104.
38. Puttlitz CM, DiAngelo DJ. Cervical spine arthroplasty biomechanics. Neurosurg Clin N Am.
2005;16(4):589–94.
39. Rousseau MA, Bonnet X, Skalli W. Influence of the geometry of a ball-and-socket interverte-
bral prosthesis at the cervical spine: a finite element study. Spine. 2008;33(1):E10–4.
40. Tang Q, Wang L, Mo Z, et al. Biomechanical analysis of different prodisc-C arthroplasty designs
after implantation: a numerical sensitivity study. J Mech Med Biol. 2015;15(1):1550007.
41. Galbusera F, Anasetti F, Bellini CM, et al. The influence of the axial, antero-posterior and lat-
eral positions of the center of rotation of a ball-and-socket disc prosthesis on the cervical spine
biomechanics. Clin Biomech. 2010;25(5):397–401.
42. Bogduk N, Mercer S. Biomechanics of the cervical spine. I: normal kinematics. Clin Biomech.
2000;15(9):633–48.
43. Ha SK. Finite element modeling of multi-level cervical spinal segments (C3–C6) and biome-
chanical analysis of an elastomer-type prosthetic disc. Med Eng Phys. 2006;28(6):534–41.
44. Ahn HS, DiAngelo DJ. A biomechanical study of artificial cervical discs using computer sim-
ulation. Spine. 2008;33(8):883–92.
7 Biomechanics of Spine 167
45. Womack W, Leahy PD, Patel VV, et al. Finite element modeling of kinematic and
load transmission alterations due to cervical intervertebral disc replacement. Spine.
2011;36(17):E1126–33.
46. Pitzen T, Kettler A, Drumm J, et al. Cervical spine disc prosthesis: radiographic, biomechani-
cal and morphological post mortal findings 12 weeks after implantation. A retrieval example.
Eur Spine J. 2007;16(7):1015–20.
Chapter 8
Biomechanics of the Hip
Abstract The hip joint is a crucial support structure for the human body structure,
second only to the knee in the terms of weight-bearing forces placed upon it, and is
an essential joint for walking. A solid understanding of the anatomy and biome-
chanics of the hip joint is necessary for improving treatments for hip disorders and
for the design of hip prostheses. In this chapter, the anatomy of the hip joint is intro-
duced and is then followed by a more in-depth analysis of the kinetic and kinematic
movements of the joint.
The femoral head and acetabulum form a ball-and-socket joint (Fig. 8.1), which is
surrounded and protected by powerful muscles, tendons, and a ligamentous capsule.
Both the femoral head and acetabulum are covered with articular cartilage and sepa-
rated by a small volume of synovial fluid which is produced by the synovial mem-
brane. The synovial fluid keeps the cartilage nourished and provides lubrication for
the joint. The bones, muscles, tendons, ligaments, and synovial fluid work together
to make the hip joint flexible, stable, and strong.
B. Liu
School of Biological Science and Medical Engineering, Beihang University, Beijing, China
J. Hua
Department of Nature Science, Faculty of Science and Technology, Middlesex University,
London, UK
C.-K. Cheng (*)
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
e-mail: ckcheng2020@sjtu.edu.cn
Femoral Acetabulum
head
Labrum
Femoral
neck
Capsule
Femur
1.1 Bony Structure
1.1.1 Acetabulum
Sharp’s
angle
teardrop line
is formed between a line connecting the medial sourcil and lateral rim of the acetab-
ulum and a horizontal line between the lowest points of two teardrops (Fig. 8.3).
The acetabular roof angle typically ranges from 0° to 10°, while greater than 10°
can be considered as hip dysplasia. A larger angle generally means a more unstable
hip joint, while an angle of less than 0° indicates that the acetabulum tends to over-
cover the femoral head [7].
The acetabular development can be evaluated by the acetabular depth-to-width
ratio (ADR) [8], which is calculated by depth/width × 1000. The width of the ace-
tabulum is measured as the distance between the tip of the inferior teardrop to the
lateral rim of the acetabulum on the coronal plane (Fig. 8.3). The depth is the dis-
tance between the deepest point of the acetabulum to a line connecting the medial
and lateral rim of the acetabulum [9]. A depth-to-width ratio of less than 250 is
considered hip dysplasia [10].
The bony coverage of the femoral head by the acetabulum provides inherent
stability for the hip joint. The center-edge angle (CE angle) was first proposed by
Wiberg [11] to describe the relationship between the femoral head and acetabulum.
The CE angle is formed by the intersection of a line through the center of femoral
head and superolateral rim of acetabulum and a vertical line passing through the
center of the femoral head (Fig. 8.4a). A CE angle of over 25° is normal in adults,
lower than 20° is associated with DDH [11, 12], and an angle greater than 39° is
considered as acetabular over-coverage, which often causes impingement.
Another index for measuring the bony coverage of the femoral head by the
acetabulum is the femoral head extrusion index (FHEI). This index is calculated as
the percentage of the uncovered portion of the femoral head to the width of the
femoral head (Fig. 8.4b) and is useful in assessing developmental dysplasia as well
as femoroacetabular impingement. Values between 17 and 27% are considered
within the normal range. An FHEI of greater than 27% may be categorized as
developmental dysplasia, while values lower than 17% can be considered as over-
coverage [13].
a CE Angle b
FHEI=A/(A+B)
A B
Fig. 8.4 Measurement of center-edge (CE) angle (a) and femoral head extrusion index (FHEI) (b)
8 Biomechanics of the Hip 173
1.1.3 Femoral Head
The femoral head, consisting of a dense trabecular bone structure, is located at the
proximal end of the femur and is supported by the femoral neck (Fig. 8.1). The
femoral head is crucial for supporting the body as well as transmitting and absorb-
ing loads generated during daily activities. The diameter of the femoral head is
generally larger in men than in women [20].
The femoral head is approximately spherical with a smooth surface, and about
60–70% of the femoral head is covered by articular cartilage [21]. There is a small
rough depression called the fovea capitis on the posterior and inferior part of the
femoral head which acts as an attachment point for the ligamentum teres femoris. The
strength and stiffness of the femoral head vary from region to region depending on
the thickness of the surface cartilage. The cartilage is thickest around the area anterior
to the zenith region [22]. Since the articular cartilage is viscoelastic, the load-bearing
area of the femoral head changes in accordance with the magnitude of the load. As
the load increases, the load-bearing area changes from the periphery of the femoral
head to the center of the lunate surface and the anterior and posterior horns [23].
1.1.4 Femoral Neck
The femoral Neck-Shaft Angle (NSA) is the angle formed by the long axis of the
femoral neck and the longitudinal axis of the femoral shaft. During infancy, the
NSA has an angle of inclination of about 140–150°, and this decreases progres-
sively as children get older to reach a value of about 125° at puberty (average
125 ± 5°). The NSA does not typically change again after this. When the angle
exceeds 130° after growth has stopped, this condition is known as a coxa valgus
deformity. In cases where the angle is less than 120°, this is known as a coxa varus
deformity. Coxa valga is more likely to be found in patients with hip dysplasia [24,
25]. In contrast, coxa vara is more commonly associated with femoroacetabular
impingement (FAI) [26].
Femoral neck anteversion (FNA) is the angle between the projection of the femo-
ral head-neck axis on the transverse plane and on the coronal plane. If the head-neck
axis is inclined forwards towards the transcondylar plane, then it is termed antever-
sion, and if the same is directed downwards and posterior, it is termed as retrover-
sion. Anteversion reduces the coverage of the femoral head by the acetabulum, and
thus the leg tends to rotate internally to prevent the femoral head from dislocating,
whereas retroversion has the opposite effect. Femoral neck anteversion ranges from
30° to 40° in infancy and decreases progressively as the body grows. For adults, a
normal anteversion angle ranges from 15° to 20°, with males having slightly less
femoral anteversion than females [27]. Abnormal femoral neck anteversion and
structural deformities cause lead to FAI, which is considered a precursor to osteoar-
thritis [28].
1.1.5 Femoral Calcar
1.2.1 Muscles
The muscles of the hip, working together to provide force for movement and stabil-
ity, are crucial for the normal functioning of the hip joint. By contracting various
muscles, the hip joint is capable of moving through 6 degrees of freedom based on
the femoral head center: flexion and extension around a transverse axis, abduction
and adduction around a sagittal axis, external rotation and internal rotation around a
longitudinal axis (Fig. 8.5).
The most powerful flexor of the hip joint is the iliopsoas, which is composed of
the psoas major, iliacus, and psoas minor muscles. The psoas major (superficial
layer) originates at the T12-L4 vertebrae and the iliacus originates at the iliac fossa.
At the other end, both muscles have an insertion point on the lesser trochanter. Only
about half of the population have a psoas minor muscle (deep layer), which origi-
nates from the costal processes of the L1 to L5 vertebrae and inserts at the iliopec-
tineal arch. The rectus femoris, sartorius, and tensor fasciae latae are other flexors
which assist the iliopsoas.
The gluteus maximus, as the largest muscle in the body, acts as the main extensor
of the hip joint. The gluteus maximus mainly arises from the dorsal surface of the
sacrum and gluteal surface of the ilium and then branches out as two fibers. The
upper and lower fibers insert on the iliotibial tract and gluteal tuberosity, respec-
tively. The force of the gluteus maximus also contributes when rotating the hip
externally as well as maintaining the body’s upright posture. Other main extensors
of hip include the biceps femoris, semimembranosus, and semitendinosus.
Extension Flexion
The gluteus medius, gluteus minimus, and tensor fascia latae form the major
abductors of the hip joint, assisted by the piriformis and sartorius. The gluteus
medius and gluteus minimus originate at the gluteal surface of the ilium and insert
at the lateral and anterolateral surface of the greater trochanter, respectively. The
tensor fasciae latae originates from the anterior superior iliac spine and inserts at the
iliotibial tract. The piriformis is located in the middle of the gluteal region; it arises
from the surface of the sacrum and inserts at the greater trochanter.
Hip adduction is primarily accomplished by the adductor longus, brevis, and
magnus muscles with assistance from the gracilis and pectineus muscles. The
adductor longus, brevis, and gracilis also play roles in flexing the hip, whereas the
adductor magnus and pectineus contribute to hip external rotation.
The primarily external rotators of the hip are obturator externus, quadratus femo-
ris, and gemelli with assistance from the gluteus maximus, sartorius, and piriformis.
The obturator externus originates from the external surface of the obturator mem-
brane and the surrounding bony surface and inserts at the intertrochanteric fossa
through the posterior part of the femoral neck. This muscle also plays a role as an
adductor of the hip joint. The quadratus femoris arises from the lateral side of the
ischial tuberosity and ends at the intertrochanteric crest of the femur.
Internal rotation of the hip joint is mainly accomplished by the actions of the
tensor fascia latae, anterior fibers of the gluteus medius, and minimus muscles.
The hip capsule helps maintain the stability of the hip joint. The capsule is com-
posed of a strong and dense fibrous cylindrical sleeve originating from the acetabu-
lar margin 5–6 mm beyond the acetabular labrum and attaching anteriorly at the
intertrochanteric line [32]. The femoral head and most of the femoral neck are
enclosed in the joint capsule, whereas the lateral half of the femoral neck as well as
the great trochanter and lesser trochanter are not enclosed in capsule. Because of the
need to bear greater loads, the anterosuperior part of the hip joint capsule is thicker,
whereas the posteroinferior part is thinner and looser. On the inside, a synovial
membrane lines the inner surface of the capsule and produces synovial fluid to
lubricate the articular surface and provide nutrition to the articular cartilage.
The stability of hip joint is strengthened by four ligaments, three extracapsular
ligaments (iliofemoral ligament, ischiofemoral ligament, and pubofemoral liga-
ment) and one intracapsular ligament (ligamentum teres femoris).
The iliofemoral ligament (also called Y-ligament) lies on the anterior side of the
hip joint and resembles an inverted “Y.” The ligament originates on the lower part
of the AIIS (Anterior inferior iliac spine) and then divides into two branches which
subsequently attach at the distal and proximal part of the intertrochanteric line,
respectively. The iliofemoral ligament is the strongest ligament in the body with a
tensile strength exceeding 350 N [1]. Its lateral branch limits excessive abduction
and adduction of the hip joint. It also limits internal and external rotation when the
hip joint is in extension and limits external rotation when the hip joint is in flexion.
8 Biomechanics of the Hip 177
The medial fibers of the iliofemoral ligament limit abduction and external rotation
of the hip [1, 32].
The ischiofemoral ligament is located on the posterior superior side of the hip
joint. It originates at the ischial portion of the acetabulum and then crosses the
hip capsule as two fibers. The central fibers (superior ischiofemoral ligament)
spiral superiorly around the neck of the femur from the posterior surface of the
ischium to the medial surface of the greater trochanter of the femur. The lateral
and medial inferior ischiofemoral ligaments attach to the neck of the femur and
reinforce the posterior aspect of the joint capsule. The ischiofemoral ligament
contributes over 60% of the restraining force required to limit excessive internal
rotation in flexion and extension [33]. It is helpful for maintaining stability as
well as reducing the amount of muscle energy required to maintain a standing
position.
The pubofemoral ligament (also called pubocapsular ligament), located at the
anterior inferior aspect of the hip joint, plays a role in supporting the joint capsule
and maintaining the anterior and inferior stability of the hip joint. It originates at the
iliopubic eminence, the pubic portion of the acetabular rim, superior aspect of the
pubic ramus and the obturator crest, and inserts at the inferior surface of the femoral
neck. The pubofemoral ligament restricts internal rotation once the hip is flexed
beyond 30°, as well as restricting excessive abduction [34].
The ligamentum teres femoris (also called femoral head ligament) is a flat trian-
gular fibrous band in the hip capsule. The base attaches to both sides of the trans-
verse acetabular ligament and the edges of the acetabular notch, and the tip connects
to the fovea capitis on the head of the femur. The femoral head ligament is covered
by synovium with blood vessels passing through it. It is generally believed that this
ligament has no restrictive effect on the motion of the hip joint [35].
Kinematics is the evaluation of motion and positioning, but does not consider the
physical properties of the object itself or the forces imposed on it. In human motion
analysis, joint angular displacement is the most common method for describing the
relative motion between limbs. A comprehensive understanding of joint kinematics
is important for the diagnosis and treatment of joint diseases.
Because of the congruency of the surface of the hip, almost all movement
between the femoral head and the acetabulum is rotational, without detectable
translation [36]. Usually, hip motion is described by the movement of the femur
relative to the pelvis around the hip joint center. The hip joint can move through 3
degrees of freedom: flexion and extension in the sagittal plane, abduction and
adduction in the frontal plane, and internal and external rotation in the transverse
plane (Fig. 8.5).
178 B. Liu et al.
Hip joint motion is achieved through the action of a series of muscles and con-
nected tendons, and is limited by the bony structure of a hip joint, the acetabular
labrum, and surrounding ligaments. The largest movement occurs in the sagittal plane,
where the flexion angle ranges from 0° to 140° and the extension angle ranges from
0° to 20°. In the frontal plane, the range of abduction is from 0° to 50° and adduction
is from 0° to 30° with the hip joint extended. When the hip joint is flexed to 90°, the
abduction limit increases to 80° whereas the adduction limit decreases to 20°. In the
transverse plane, the limits of external and internal rotation are 50° and 40°, respec-
tively, with the hip flexed to 90°. When in the prone position with the hip extended, the
external rotation limit decreases to 30° because of the restrictive actions of the soft
tissues [37]. The ranges detailed above show the largest angles that the hip joint can
safely reach, but the joint seldom reaches such extremes during daily activities.
Typical joint ranges of motion in daily activities can be used to evaluate a
patient’s functional movement in clinical settings. Common daily activities that
require a large range of motion include trying shoes on the floor, sitting down on a
chair or rising from it, and ascending or descending stairs. A study by Johnston and
Smidt [38] measured the range of motion for 33 males performing these sample
activities. It was found that the greatest motion in the sagittal plane occurred when
squatting on the floor to tie shoes. In the frontal and transverse planes, the two great-
est motions occurred when squatting to tie a shoe on the opposite foot across the
thigh. These values indicate that for carrying out daily activities a flexion angle of
at least 120° and an external rotation angle of at least 20° is necessary.
Walking is one of the most repetitive activities for the hip joint. Understanding
the movement of the hip joint during walking is helpful for further understanding
hip joint function. Compared with other joints, the hip has a larger range of motion
in the frontal and transverse planes during level gait [39]. One single gait cycle is
typically composed of an initial heel strike (0%) through to the second contact of
the same foot (100%) and can be divided into two phases: stance phase and swing
phase (Fig. 8.6). The stance phase refers to the period when the foot contacts with
STANCE SWING
Fig. 8.6 Period of one gait cycle [31] (Reprinted with permission from ‘Orthopaedics and
Trauma’. License Number: 4567661288157)
8 Biomechanics of the Hip 179
a b c
40 15 20
10
10
20
5
10
0
0 0
-10 HLTHY
-10
-5 MOA
-20
SOA
-30 -10 -20
0 50 100 0 50 100 0 50 100
Percent of Gait Cycle Percent of Gait Cycle Percent of Gait Cycle
Fig. 8.7 Hip movements in the (a) sagittal, (b) frontal, and (c) transverse plane for nonexistent
(solid), moderate (dotted), and severe hip osteoarthritis (dashed) groups. Flexion, adduction, and
internal rotation are represented as positive angles. The shaded region represents 1SD above and
below the healthy/non-OA group waveform and the vertical line represents the toe off period of the
gait cycle [40] (Reprinted with permission from ‘Journal of Electromyography and Kinesiology’.
License Number: 4567410168348)
ground and accounts for about 60% of the gait cycle. The swing phase is the period
when the limb is swinging forward and accounts for about 40% of the gait cycle.
The motions of the hip joint during gait are illustrated in Fig. 8.7 [40]. At heel
strike (0% of gait cycle), the hip is flexed to 30–35° in the sagittal plane, before
extending to a neutral position at midstance [39, 40]. The end of the stance phase is
gauged by the “toe off” period at about 60% of the gait cycle, at which point the hip
is extended to 10–15°. In the frontal plane, the hip joint is in an adducted position
during the stance phase, and the maximum adduction angle occurs at about 20% of
the whole gait. In the swing phase, the hip joint reverts to an abducted position,
which continues until the middle of the swing phase. In the transverse plane, the hip
is externally rotated in the stance phase and converts to internal rotation around the
middle of the swing phase. The hip joint remains internally rotated until late in the
following stance phase, at which point it rotates externally again.
Hip joint diseases such as osteoarthritis (OA) often have an influence on the range of
motion of the hip joint. Moderate OA results in a considerable decrease in hip range of
motion in the sagittal plane, but there is only a significant decrease in the range of
motion in the frontal and transverse planes in patients with severe OA [40] (Fig. 8.7).
Total hip arthroplasty (THA) involves replacing a diseased or damaged hip joint
with a prosthesis. THA is often performed in cases of severe joint pain, OA,
avascular necrosis (AVN), and femoral neck fracture. However, numerous studies
found that the range of motion of the hip in the sagittal plane was reduced after THA
[41–44]. The achievable limit of hip extension is reduced after THA [41, 44–46],
but it has been reported that the limit of hip flexion is not significantly impacted by
THA [42, 47]. But, in contrast, Beaulieu et al. did find a reduction in peak flexion
angle after THA [41]. The decrease in the extension limit may be related to the
increased passive resistance of the flexors (i.e., a flexion contracture) [48].
180 B. Liu et al.
Kinetic analysis of the hip joint focuses on the forces acting on the hip that cause
motion. Hip motion is the result of the interaction between internal forces (muscles,
joints, tendons, ligaments, and joint contact forces [49]) and external forces acting
on the system (gravity, ground reaction forces, and inertia [49]). Such kinetic infor-
mation is useful for evaluating hip motion and diseases, as well as developing treat-
ment methods and designing hip prostheses.
2.2.1 Static Loading
The human body is an elaborate structure in which bones and soft tissues interact
under static or dynamic conditions to maintain balance and generate motion. Static
analysis of the hip joint is useful for evaluating changes in the anatomical structure
or different treatment modalities on the hip joint reaction forces. Free-body dia-
grams, as shown in Figs. 8.8 and 8.9, can be used to illustrate contact forces during
various loading conditions, such as single leg stance, double leg stance, and carry-
ing external loads.
Single-Leg Stance
In a single leg stance, the human body can be considered as a lever-like structure
with the femoral head acting as the fulcrum. A simplified free-body diagram can be
used to calculate and illustrate the joint reaction force indirectly (Fig. 8.8). Since the
weight of the non-supporting leg is included in the load acting on the load-bearing
hip, the effective center of gravity of the body moves close to the non-supporting leg.
A B
W
8 Biomechanics of the Hip 181
As the lower extremities account for 1/3 of the full body weight, the non-supporting
limb accounts for half of that, or 1/6 of the full body weight. Therefore, the gravita-
tional force (W) acting on the load-bearing hip is 5/6 body weight (BW). This down-
ward force will create a moment around the center of the femoral head, with the
gravitational moment arm (B) being the distance between the center of the femoral
head and the center of gravity of the body. To maintain balance, this moment is off-
set by a counteracting moment generated by the combined abductor muscle force
(M) acting over a lever arm measured as the distance between the center of the femo-
ral head and the insertion site of the abduct muscles on the greater trochanter.
To calculate the joint reaction force (R) in single leg stance, the sum of all
moments acting on the joint equals zero:
( A × My ) − ( B × 5 / 6W ) = 0.
Usually the ratio between B and A (B:A) is about 2.5.
Thus, My = 2.5W,
Ry = My + W = 2.5W + W = 3.5W
When the hip joint succumbs to disease or trauma, loading the joint can be
extremely painful [21]. Reducing the hip joint reaction force can offer immediate
pain relief. According to the above calculations, the joint reaction force can be
reduced in two ways. One way is to reduce the moment generated by body weight.
Since the moment is calculated as the force multiplied by the moment arm, the
moment can be reduced by either reducing body weight or the length of the moment
arm. For example, tilting the trunk towards the diseased hip will shift the center of
gravity closer to the hip joint (this results in Trendelenburg gait), thereby reducing
the body weight moment arm. Another way is to reduce the required hip abductor
force. This can be achieved by increasing the moment arm of the abductor muscle
182 B. Liu et al.
force. For example, in THA a femoral implant with a greater offset may be chosen
or the acetabular component may be positioned medially. The effectiveness of off-
setting the hip is illustrated by the fact that the reaction force on the hip in patients
with coxa vara is 25% lower than the average hip joint, while the reaction force in
patients with coxa valgus is 25% higher than the average hip joint [50]. This is due
to the moment arm of the abductors decreasing; thus, more abductor muscle force is
need to balance the body weight.
Using a cane in the hand contralateral to the diseased hip is also a way to lower
the joint reaction force by reducing the body weight passing through the affected
hip. The cane effectively creates a moment that counteracts the moments gener-
ated by body weight. Therefore, lower abductor muscle force is required and so
the joint reaction force is reduced, which in turn alleviates pain in the affected
hip. Since the force arm produced by cane is much larger than that of abductor
muscles, even small forces on the cane can generate moments that are large
enough to significantly reduce the force of abductors. It has been shown that
applying about 15% of body weight to a cane can cut the joint reaction force by
50% [50].
Double-Leg Stance
In double leg stance, the upper body weight is distributed equally between the
two femoral heads (WR = WL) with equivalent gravitational moment arms
(DR = DL) (Fig. 8.9). The moment acting on each hip joint is equal in magni-
tude and opposite in direction. The pelvis is in balance in the coronal plane
with negligible assistance from surrounding musculature. Therefore, if abduc-
tor muscles are not required to maintain balance in frontal plane and sagittal
plane, there would be an equal force of 1/2 superimposed body weight (W).
This force will be increased if the abductor muscle acts to maintain balance and
stability.
stresses in the bone [51]. This nonuniform arrangement increases the resistance to
mechanical stress. The primary pathway of force transmission in the proximal
femur is reflected by the appearance of the trabecular system.
2.2.2 Dynamic Loading
As detailed previously, free-body diagrams are a simple method for indirectly esti-
mating hip contact forces. Knowing the real-time hip contact forces during daily
dynamic activities (walking, running, squatting, etc.) is also necessary for better
understanding the relevant muscle function and strength and the wear and stability
of the hip joint. Dynamic hip contact forces (HCFs) may be measured using direct
and indirect means. Direct measurements are carried in vivo using instrumented
telemetric prostheses [52, 53]. This method is considered as the gold standard for
measuring loading on the hip [54] and can be used to verify indirect measurement
models. Indirect measurements may be performed using musculoskeletal models
and inverse static optimization techniques to calculate muscle forces and hence joint
loading [55, 56]. Some studies have claimed that results calculated using this indi-
rect method are comparable to experimental data [53, 57–60].
Numerous studies have measured HCFs during dynamic activities [53, 54, 61–
63], of which walking and running are the most two common activities. It has been
shown that the typical peak force (BW) in gait ranges from 1.6 to 5.7 times body
weight, but is influenced by physiological differences, gender, age, and walking
184 B. Liu et al.
1.5
0.5
0
0 20 40 60 80 100
Gait Cycle (%)
speed. Figure 8.11 details the typical HCF during one gait cycle, showing two peaks
for hip contact force. The initial peak occurs during heel-strike and early midstance,
and the second peak occurs just before toe-off.
The contact force is related to the walking speed, whereby increased speed
results in an increased HCF [53, 54, 64]. A study [54] on musculoskeletal models
calculated the HCFs for 20 young healthy adults and found that the peak HCF
increased from 4.22 body weight to 5.41 body weight when the walking speed
increased from 3 to 6 km/h. The greater force is because (1) the increased dynamic
variation of ground reaction force with the walking speed in the heel-strike stage
[65], and (2) the increased stride length corresponds to the increased velocity, which
generates a greater offset of the ground reaction force from the joint axis and mobi-
lizes more muscle activity to maintain balance. The HCF is also linearly and posi-
tively correlated with BMI, meaning that obese people generate a greater HCF
during most of the stance period [57].
During running, there is only one peak HCF, which happens in the stance phase
at 0–30% of the gait cycle [54]. As with walking, the HCF is positively correlated
with speed.
Bergmann et al. has measured hip contact forces using instrumented total hip
implants [52]. The results of his study are illustrated in Fig. 8.12. The graph demon-
strates that the greatest contact force was measured during jogging, reaching about
4.1 times body weight, and walking down stairs generated the second greatest force.
The lowest joint contact force was produced during cycling.
There are also other unpredictable loads placed on the joints that can generate
substantial force, such as the peak HCF during stumbling being recorded at 7.2–8.7
times body weight [61, 66]. Therefore, it should be emphasized that after THA
patients should pay attention not only to daily activities that generate relatively low
HCFs, but also to unexpected events such as stumbling or unstable periods when
standing on one leg which may produce a resultant force in excess of 8 times
body weight.
Furthermore, it should also be noted that direct measurements using instru-
mented telemetric prostheses can only be performed in patients who need THA. Such
methods cannot evaluate the physiological condition of healthy joints.
8 Biomechanics of the Hip 185
4.5
4.1
4.0
3.5
Hip Contact Force/BW
3.0 3.1
3.0
2.6
2.5 2.3
2.1
2.0 1.8
1.5
1.0
1.0
0.5
0.0
Walking Jogging Stand up Sit Stairs Stairs Knee Cycling
Down Up Down Bend
Daily Activities
Fig. 8.12 Hip contact force (resultant forces) during different activities [52]
3 Summary
The mechanical conditions of the hip joint are determined and reflected by its ana-
tomical features. This chapter provides essential information on a normal hip joint
based on an evaluation of its bony structure and surrounding muscles and ligaments,
in addition to their roles in hip functionality and related static and dynamic condi-
tions of the hip joint. However, when the hip joint undergoes pathological changes
such as osteoarthritis or trauma, these mechanical conditions of the hip may be
altered. This needs to be taken into consideration when considering a suitable clini-
cal treatment such as total hip replacement or other surgery.
References
1. Schünke M, Schulte E, Schumacher U, Ross LM, Lamperti ED. Thieme atlas of anatomy: latin
nomenclature: general anatomy and musculoskeletal system. New York: Thieme; 2006.
2. Moore KL, Dalley AF, Agur AM. Clinically oriented anatomy. Philadelphia, PA: Lippincott
Williams & Wilkins; 2013.
3. Stem ES, O’Connor MI, Kransdorf MJ, Crook J. Computed tomography analysis of acetabular
anteversion and abduction. Skelet Radiol. 2006;35:385–9.
4. Engesæter IØ, Laborie LB, Lehmann TG, Sera F, Fevang J, Pedersen D, et al. Radiological
findings for hip dysplasia at skeletal maturity. Validation of digital and manual measurement
techniques. Skelet Radiol. 2012;41:775–85.
5. Laborie LB, Engesæter IØ, Lehmann TG, Sera F, Dezateux C, Engesæter LB, et al.
Radiographic measurements of hip dysplasia at skeletal maturity—new reference intervals
based on 2,038 19-year-old Norwegians. Skelet Radiol. 2013;42:925–35.
186 B. Liu et al.
6. Mannava S, Geeslin AG, Frangiamore SJ, Cinque ME, Geeslin MG, Chahla J, et al.
Comprehensive clinical evaluation of femoroacetabular impingement: part 2, plain radiogra-
phy. Arthrosc Tech. 2017;6:e2003–9.
7. Tönnis D. Normal Values of the Hip Joint for the Evaluation of X-rays in Children and Adults.
Clin Orthop Relat Res. 1976;119:39–47.
8. Cooperman DR, Wallensten R, Stulberg SD. Acetabular dysplasia in the adult. Clin Orthop
Relat Res. 1983;175:79–85.
9. Zeng Y, Wang Y, Zhu Z, Tang T, Dai K, Qiu S. Differences in acetabular morphology related to
side and sex in a Chinese population. J Anat. 2012;220:256–62.
10. Jacobsen S, Sonne-Holm S, Søballe K, Gebuhr P, Lund B. Hip dysplasia and osteoarthrosis: a
survey of 4 151 subjects from the Osteoarthrosis Substudy of the Copenhagen City heart study.
Acta Orthop. 2005;76:149–58.
11. Wiberg G. Studies on dysplastic acetabulum and congenital subluxation of the hip joint with
special reference to the complications of osteoarthritis. Acta Chir Scand. 1939;83:58.
12. Bouttier R, Morvan J, Mazieres B, Rat A-C, Ziegler LE, Fardellone P, et al. Reproducibility of
radiographic hip measurements in adults. Joint Bone Spine. 2013;80:52–6.
13. Tannast M, Hanke MS, Zheng G, Steppacher SD, Siebenrock KA. What are the radio-
graphic reference values for acetabular under-and overcoverage? Clin Orthop Relat Res.
2015;473:1234–46.
14. Karns MR, Patel SH, Kolaczko J, Liu RW, Mather RC, White BJ, et al. Acetabular rim length:
an anatomical study to determine reasonable graft sizes for labral reconstruction. J Hip Preserv
Surg. 2017;4:106–12.
15. Groh MM, Herrera J. A comprehensive review of hip labral tears. Curr Rev Musculoskelet
Med. 2009;2:105–17.
16. Bsat S, Frei H, Beaulé P. The acetabular labrum: a review of its function. Bone Joint
J. 2016;98:730–5.
17. Philippon MJ, Nepple JJ, Campbell KJ, Dornan GJ, Jansson KS, LaPrade RF, et al. The hip
fluid seal—part I: the effect of an acetabular labral tear, repair, resection, and reconstruction on
hip fluid pressurization. Knee Surg Sports Traumatol Arthrosc. 2014;22:722–9.
18. Henak CR, Ellis BJ, Harris MD, Anderson AE, Peters CL, Weiss JA. Role of the acetabular
labrum in load support across the hip joint. J Biomech. 2011;44:2201–6.
19. Seldes R, Tan V, Hunt J, Katz M, Winiarsky R, Fitzgerald RJ. Anatomy, histologic features,
and vascularity of the adult acetabular labrum. Clin Orthop Relat Res. 2001;382:232–40.
20. Bartoska R. Measurement of femoral head diameter: a clinical study. Acta Chir Orthop
Traumatol Cechoslov. 2009;76:133–6.
21. Byrne DP, Mulhall KJ, Baker JF. Anatomy & biomechanics of the hip. Open Sports Med
J. 2010;4:51–7.
22. Armstrong C, Gardner D. Thickness and distribution of human femoral head articular carti-
lage. Changes with age. Ann Rheum Dis. 1977;36:407–12.
23. von Eisenhart-Rothe R, Eckstein F, Müller-Gerbl M, Landgraf J, Rock C, Putz R. Direct com-
parison of contact areas, contact stress and subchondral mineralization in human hip joint
specimens. Anat Embryol. 1997;195:279–88.
24. Clohisy JC, Nunley RM, Carlisle JC, Schoenecker PL. Incidence and characteristics of femo-
ral deformities in the dysplastic hip. Clin Orthop Relat Res. 2009;467:128–34.
25. Fabry G, Macewen GD, Shands A Jr. Torsion of the femur: a follow-up study in normal and
abnormal conditions. JBJS. 1973;55:1726–38.
26. Beall DP, Sweet CF, Martin HD, Lastine CL, Grayson DE, Ly JQ, et al. Imaging findings of
femoroacetabular impingement syndrome. Skelet Radiol. 2005;34:691–701.
27. Pollard TC, Villar RN, Norton MR, Fern ED, Williams MR, Simpson DJ, et al. Femoroacetabular
impingement and classification of the cam deformity: the reference interval in normal hips.
Acta Orthop. 2010;81:134–41.
28. Maheshwari AV, Malik A, Dorr LD. Impingement of the native hip joint. JBJS. 2007;89:2508–18.
29. Li B, Aspden R. Material properties of bone from the femoral neck and calcar femorale of
patients with osteoporosis or osteoarthritis. Osteoporos Int. 1997;7:450–6.
8 Biomechanics of the Hip 187
30. Zhang Q, Chen W, Liu HJ, Li ZY, Song ZH, Pan JS, et al. The role of the calcar femorale in
stress distribution in the proximal femur. Orthop Surg. 2009;1:311–6.
31. Lunn DE, Lampropoulos A, Stewart TD. Basic biomechanics of the hip. Orthop Trauma.
2016;30:239–46.
32. Gray H, Standring S. Gray’s anatomy. London: Arcturus Publishing; 2008.
33. Martin HD, Savage A, Braly BA, Palmer IJ, Beall DP, Kelly B. The function of the hip capsular
ligaments: a quantitative report. Arthroscopy. 2008;24:188–95.
34. Martin HD, Khoury AN, Schröder R, Johnson E, Gómez-Hoyos J, Campos S, et al.
Contribution of the pubofemoral ligament to hip stability: a biomechanical study. Arthroscopy.
2017;33:305–13.
35. Perumal V, Woodley SJ, Nicholson HD. Ligament of the head of femur: a comprehensive
review of its anatomy, embryology, and potential function. Clin Anat. 2016;29:247–55.
36. Bowman KF Jr, Fox J, Sekiya JK. A clinically relevant review of hip biomechanics.
Arthroscopy. 2010;26:1118–29.
37. Nordin M, Frankel VH. Basic biomechanics of the musculoskeletal system. Philadelphia, PA:
Lippincott Williams & Wilkins; 2001.
38. Johnston RC, Smidt GL. Hip motion measurements for selected activities of daily living. Clin
Orthop Relat Res. 1970;72:205–15.
39. Bovi G, Rabuffetti M, Mazzoleni P, Ferrarin M. A multiple-task gait analysis approach: kine-
matic, kinetic and EMG reference data for healthy young and adult subjects. Gait Posture.
2011;33:6–13.
40. Rutherford DJ, Moreside J, Wong I. Hip joint motion and gluteal muscle activation differences
between healthy controls and those with varying degrees of hip osteoarthritis during walking.
J Electromyogr Kinesiol. 2015;25:944–50.
41. Beaulieu ML, Lamontagne M, Beaulé PE. Lower limb biomechanics during gait do not return
to normal following total hip arthroplasty. Gait Posture. 2010;32:269–73.
42. Kiss RM, Illyés Á. Comparison of gait parameters in patients following total hip arthroplasty
with a direct-lateral or antero-lateral surgical approach. Hum Mov Sci. 2012;31:1302–16.
43. Madsen MS, Ritter MA, Morris HH, Meding JB, Berend ME, Faris PM, et al. The effect of
total hip arthroplasty surgical approach on gait. J Orthop Res. 2004;22:44–50.
44. Varin D, Lamontagne M, Beaulé PE. Does the anterior approach for THA provide closer-to-
normal lower-limb motion? J Arthroplast. 2013;28:1401–7.
45. Perron M, Malouin F, Moffet H, McFadyen BJ. Three-dimensional gait analysis in women
with a total hip arthroplasty. Clin Biomech. 2000;15:504–15.
46. Sander K, Layher F, Babisch J, Roth A. Evaluation of results after total hip replacement using
a minimally invasive and a conventional approach. Clinical scores and gait analysis. Zeitschrift
fur Orthopadie und Unfallchirurgie. 2011;149:191–9.
47. Tateuchi H, Tsukagoshi R, Fukumoto Y, Oda S, Ichihashi N. Dynamic hip joint stiffness in
individuals with total hip arthroplasty: relationships between hip impairments and dynamics
of the other joints. Clin Biomech. 2011;26:598–604.
48. Kolk S, Minten MJ, van Bon GE, Rijnen WH, Geurts AC, Verdonschot N, et al. Gait and gait-
related activities of daily living after total hip arthroplasty: a systematic review. Clin Biomech.
2014;29:705–18.
49. Chao E. Biomechanics of the human gait. In: Frontiers in biomechanics. New York: Springer;
1986. p. 225–44.
50. Pauwels F. Biomechanics of the normal and diseased hip: theoretical foundation, technique
and results of treatment an atlas. Springer Science & Business Media; 2012.
51. Pugh J, Rose R, Radin E. A possible mechanism of Wolff’s law: trabecular microfractures.
Arch Int Physiol Biochim. 1973;81:27–40.
52. Bergmann G, Bender A, Dymke J, Duda G, Damm P. Standardized loads acting in hip implants.
PLoS One. 2016;11:e0155612.
53. Bergmann G, Deuretzbacher G, Heller M, Graichen F, Rohlmann A, Strauss J, et al. Hip con-
tact forces and gait patterns from routine activities. J Biomech. 2001;34:859–71.
188 B. Liu et al.
54. Giarmatzis G, Jonkers I, Wesseling M, Van Rossom S, Verschueren S. Loading of hip mea-
sured by hip contact forces at different speeds of walking and running. J Bone Miner Res.
2015;30:1431–40.
55. Pedersen DR, Brand RA, Cheng C, Arora JS. Direct comparison of muscle force predictions
using linear and nonlinear programming. J Biomech Eng. 1987;109:192–9.
56. Andrews JG, Cheng CK. The joint distribution problem with multiple articular contact forces.
J Biomech Eng. 1990;112:364–6.
57. De Pieri E, Lunn D, Chapman G, Rasmussen K, Ferguson S, Redmond A. Patient characteris-
tics affect hip contact forces during gait. Osteoarthr Cartil. 2019;27:895–905.
58. Heller M, Bergmann G, Deuretzbacher G, Dürselen L, Pohl M, Claes L, et al. Musculo-skeletal
loading conditions at the hip during walking and stair climbing. J Biomech. 2001;34:883–93.
59. Lenaerts G, Mulier M, Spaepen A, Van der Perre G, Jonkers I. Aberrant pelvis and hip kinemat-
ics impair hip loading before and after total hip replacement. Gait Posture. 2009;30:296–302.
60. Stansfield B, Nicol A, Paul J, Kelly I, Graichen F, Bergmann G. Direct comparison of calcu-
lated hip joint contact forces with those measured using instrumented implants. An evaluation
of a three-dimensional mathematical model of the lower limb. J Biomech. 2003;36:929–36.
61. Bergmann G, Graichen F, Rohlmann A. Hip joint loading during walking and running, mea-
sured in two patients. J Biomech. 1993;26:969–90.
62. Bergmann G, Graichen F, Rohlmann A. Is staircase walking a risk for the fixation of hip
implants? J Biomech. 1995;28:535–53.
63. Rydell NW. Forces acting on the femoral head-prosthesis: a study on strain gauge supplied
prostheses in living persons. Acta Orthop Scand. 1966;37:1–132.
64. Weinhandl JT, Irmischer BS, Sievert ZA. Effects of gait speed of femoroacetabular joint
forces. Appl Bionics Biomech. 2017;2017:6432969.
65. Chung M-J, Wang M-JJ. The change of gait parameters during walking at different percentage
of preferred walking speed for healthy adults aged 20–60 years. Gait Posture. 2010;31:131–5.
66. Bergmann G, Graichen F, Rohlmann A. Hip joint contact forces during stumbling. Langenbeck’s
Arch Surg. 2004;389:53–9.
Chapter 9
Biomechanics of the Knee
The tibiofemoral joint and patellofemoral joint are the two articulating surfaces that
constitute the knee joint and both are diarthrodial joints that consist of bones, hya-
line cartilage, ligaments, tendons, as well as the musculature (Fig. 9.1). The tibio-
femoral joint also has medial and lateral menisci between its surface to distribute
H. Wang · B. Liu · X. Qi
School of Biological Science and Medical Engineering, Beihang University, Beijing, China
S. L-Y. Woo
Musculoskeletal Research Center, University of Pittsburgh, Pittsburgh, PA, USA
C.-K. Cheng (*)
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
e-mail: ckcheng2020@sjtu.edu.cn
patella
femur medial collateral
ligament
cartilage
posterior cruciate
lateral collateral
ligament
ligament
lateral
meniscus
tibia
the loads as well as to absorb impact forces on the joint. All these soft tissues work
in synergy to stabilize the knee while the muscles power its motion.
Using the principles of biomechanics, the mechanical behavior and the function
of the biological system, in this case the knee joint, can be analyzed and understood.
The motion between two bony structures in a joint is known as kinematics. Normal
kinematics of the knee is maintained by biomechanical functions of each joint com-
ponent. Changing of any one of these components secondary to injury or diseases
could alter the normal balance and synergy of the knee joint and introduce abnormal
kinematics that could cause increased loading of other soft tissue structures and
induce additional damages.
In this section, the anatomy and function of the knee components, as well as the
joint kinematics, are presented.
1.1 Bones
As the femoral condyles are round while the tibial plateau is relatively flat, the con-
formity is reinforced by the menisci lying between them. In the sagittal view, the
anterior section of the tibia is generally higher than its posterior section. Hashemi
et al. [1] demonstrated that the posterior slope of the tibia ranged from −3° to 14°,
and was steeper in females than the males. Also, in the coronal view, the tibial pla-
teau is oriented upward in a medial-to-lateral direction. The coronal tibial slope
ranged from −1° to 6° and was less steep in females. Besides, there is typically a
valgus angle of 7–10° between the tibia and femur. The medial condyle of the distal
femur projects more distally than the lateral condyle [2].
9 Biomechanics of the Knee 191
The patella is a sesamoid bone and has the appearance of an inverted triangle.
The cartilage of the patella is also the thickest in the human body [3].
1.2 Hyaline Cartilage
The hyaline cartilage in the knee joint is a layer of elastic tissue which covers the
contact surfaces of the bones along which the joint moves. Hyaline cartilage is
mainly composed of a matrix of proteoglycans and collagen traversed with intersti-
tial water. This cartilage layer serves numerous functions, including providing a
smooth surface for joint movement, buffering compressive loads, and protecting the
underlying bone. With a thickness of between 1.69 and 2.55 mm, the hyaline carti-
lage in the knee is significantly thicker than in the hip (1.35–2.00 mm) and ankle
joints (1.00–1.62 mm) [4].
The micro-structure of hyaline cartilage has low permeability, which acts to trap
water within the matrix during loading. This pressurizes the cartilage and increases
the stiffness by up to ten times that of the intrinsic modulus of the solid matrix [5].
The increased stiffness of the matrix means there is very little deformation, and the
cartilage can take up to 90 min to recover after loading [6]. It has been shown that
the thickest regions of the knee cartilage on the femur and tibia are aligned at full
knee extension, suggesting an adaption of the cartilage to high loading such as dur-
ing the heel strike phase of gait [7]. In contrast, under conditions of reduced loading,
such as immobilization following surgery, the hyaline cartilage suffers atrophy.
However, a sudden change in the loading pattern on the knee (for example, a knee
with a ruptured ACL could cause a redistribution of loading on the articular surface)
does not produce a significant increase in cartilage thickness, but may in fact initiate
degeneration of the cartilage [7]. It has been reported that regular cyclic loading
enhances the synthesis of proteoglycans which makes the cartilage stiffer, but con-
tinuous compressive loading hinders the synthesis of proteoglycans, which causes
damage to the cartilage through necrosis [5].
1.3 Menisci
Lateral meniscus
Medial meniscus
As the knee moves, the menisci deform to share and buffer forces in the joint and
indirectly increase the articular contact area, thus stabilizing the knee and decreas-
ing stresses on the hyaline cartilage of the tibia and femur. Studies have demon-
strated that up to 70% of the loading on the lateral compartment of the knee is
carried by the lateral meniscus while 50% of the loading on the medial compartment
is borne by the medial meniscus [8].
The menisci also work with the hyaline cartilage to lubricate the knee joint and
allow smooth motion between the articular contact surfaces. McCann et al. con-
ducted cadaveric testing on bovine knees and demonstrated that removal of the
menisci resulted in an increased friction coefficient on the joint [9].
Undue femoral gliding on the tibia plateau is also restrained by the menisci, mak-
ing them particularly important for a knee with ACL deficiency. Previous studies
have shown greater loading on the menisci following resection of the ACL [10].
1.4 Ligaments
Four major ligaments in the tibiofemoral joint are: the anterior and posterior cruci-
ate ligaments (ACL and PCL), and the medial and lateral collateral ligaments (MCL
and LCL). The ACL and PCL are intra-articular ligaments while the MCL and LCL
are extra-articular (Fig. 9.1).
The bone insertions of the ACL are located at the medial posterior side of the
lateral femoral condyle and the anterior side of the tibial plateau. Both insertion
sites have a considerably larger cross section than the midsubstance of the ligament
[11]. For biomechanical purposes, some researchers [11] have proposed dividing
the ACL into two bundles according to the anatomical location of its tibial insertion:
the anteromedial (AM) and posterolateral (PL) bundles. Length of the AM bundle is
relatively constant through joint flexion angles from 0° to 90°, while the PL bundle
is more stretched in extension [12]. However, pursuing this approach even further,
other studies [13] have proposed dividing the ACL into three bundles (the anterome-
dial, intermediate, and posterolateral bundles).
9 Biomechanics of the Knee 193
The ACL plays an important role in constraining anterior tibial translation, thus
preventing the femur from posterior luxation, while also helping to avoid excessive
internal and valgus rotations of the tibia [14–16]. As the knee flexes, the femur ini-
tially rolls back along the tibial plateau during early flexion, before converting to
anterior sliding due to the drag force from the ACL. Besides, previous studies [17]
also showed an increased ACL force under a combined loading of axial compressive
force and anterior tibial load, comparing with using an anterior tibial load alone,
indicating a biomechanical role of ACL in resisting compressive joint impact.
The PCL inserts at the lateral posterior side of the medial condyle of the femur
and stretches to the inferior posterior side of the tibial plateau. As with the ACL, the
PCL may also be divided into two functional bundles, which are named according
to the location of their femoral attachments [12]. Both the AL bundle and the PM
bundle have increased lengths with flexion [12].
In contrast to the ACL, the PCL prevents the tibia from undue posterior transla-
tion, which decreases the risk of anterior luxation of the femur [18]. During knee
extension, the PCL forces the femur to slide back along the tibial surface as it rolls
forward. Besides, the PCL has been reported to prevent excessive external tibial
rotation, especially for the joint flexion angles beyond 60° [19].
The MCL and LCL are located at the medial and lateral side of the knee, respec-
tively. The LCL inserts at the lateral femoral condyle and the lateral superior side of
the fibular head, while the MCL attaches to the medial femoral condyle and the
inferior medial side of the tibial plateau.
The MCL and LCL function together to constrain the knee joint from excessive
internal-external and varus-valgus rotations. The MCL cooperates with the ACL in
constraining anterior tibial translation. This partnership is clearly evident in cases of
ACL injury, which often result in a decrease in joint stability and an increase in the
MCL force, subsequently placing the MCL at a greater risk of injury [20].
1.5 Muscle System
There are two major muscle groups primarily responsible for the extension and
flexion of the knee: the quadriceps muscles, located at the front of the femur, which
contract to extend the knee; and the hamstring muscles, located the back of the
femur, which contract for knee flexion.
The quadriceps muscles consist of the vastus lateralis, rectus femoris, vastus
intermedius, and vastus medialis muscles, all of which are controlled by the femoral
nerve [21]. The rectus femoris muscle inserts at the hip bone and the tibia, while the
other three muscles insert at the femur and connect to the quadriceps tendon.
The quadriceps muscles, mainly serving as a knee extensor, form a large fleshy
mass which covers the front and sides of the femur. The rectus femoris muscle also
acts as a flexor of the hip due to its attachment to the ilium [22]. As the knee exten-
sor, the quadriceps muscles are crucial for most daily activities (walking, running,
jumping, etc.) and serve to stabilize the knee. Weak quadriceps muscles are reported
194 H. Wang et al.
to compromise knee stability [23]. The lever arm of the quadriceps muscle is
increased by the presence of the patella, which is discussed in more detail in
Sect. 1.7.
The hamstring muscles consist of the semimembranosus, biceps femoris mus-
cles, and semitendinosus, all of which are controlled by the sciatic nerve. The short
head of the biceps femoris inserts at the trochanter of the femur and the lateral side
of the fibular head, while the long head originates at the sciatic tuberosities and has
the same fibular insertion point as the short head. The semitendinosus and semi-
membranosus muscles both originate from the sciatic tuberosities and insert at the
medial side of the tibia.
The hamstring muscles act to flex the knee, while also function to extend the hip
except for the short head of the biceps femoris [24]. The short head of the biceps
femoris crosses only the knee joint and is therefore not involved in hip extension.
The biceps femoris is an external rotator of the tibia, while the semitendinosus and
semimembranosus muscles function to rotate the tibia internally [25]. As an antago-
nist to the quadriceps muscles, the hamstring muscles also function to control the
joint motion during daily activities.
Motion and biomechanics of the tibiofemoral joint are greatly influenced by the
morphology of the articular surfaces, and of course the integrity of all the joint
structures [16]. The posterior tibial slope is considered an important characteristic
for producing anterior shear forces on the tibia. Wang et al. suggested that in knee
arthroplasty surgery, the posterior tibial slope has a significant effect on postopera-
tive kinematics of the joint and thus the slope angle should be highly scrutinized
intraoperatively [26]. Woo et al. found that increasing the posterior tibial slope
caused an increased anterior tibial translation which could stabilize the knee with a
deficient PCL. In contrast, decreasing the posterior slope may be considered for a
knee with ACL deficiency [27]. Females have a deeper posterior tibial slope than
males, resulting in a greater anterior tibial force, especially when in the posture of
weight-bearing, placing females at greater risk of ACL rupture than males [1, 28].
Conversely, the lateral section of the tibial plateau is generally located higher (more
proximal) than the medial section, forming a slope which is larger in males than in
females, but the effect of this coronal slope on joint mechanics is not yet clear [1].
However, while the link between slop angle and knee mechanics has been widely
studied, large differences between individuals means there is a large degree of varia-
tion across the general population [1]. Thus, understanding this variation could be
beneficial for developing more accurate subject-specific implants that could better
replicate normal knee biomechanics.
The tibiofemoral joint has 6 degrees of freedom (DOFs) of motion: three (anterior-
posterior, proximal-distal, and medial-lateral) translations, as well as three (flexion-
extension, varus-valgus, and internal-external) rotations (Fig. 9.3). Because the femoral
9 Biomechanics of the Knee 195
A-P translation
M-L translation
F-E rotation
V-V rotation
P-D transalation
condyles have a circular shape and the tibial plateau is relatively flat, the tibiofemoral
joint has good rotational flexibility in terms of flexion-extension (0–165°) [29].
The flexion movement of the knee joint can be divided into three functional
phases, comprising the screw-home arc, the functional active arc, and the passive
flexion arc [30]. The first phase is defined to represent joint activity through joint
flexions from 0° to 20°, during which the joint motions are mainly determined by
the morphology of the tibial plateau and the femoral condyles. Because of the asym-
metry between the distal regions of the lateral and medial femoral condyles, the
tibia undergoes internal rotation during this phase. The second phase represents
joint activity from 20° to 120° of flexion, during which there is little axial rotation
of the tibia. The final phase is defined as joint flexion over 120°. During this phase,
the medial femoral condyle moves proximally because of its contact with the poste-
rior section of the medial meniscus; this might explain why the posterior horn of the
medial meniscus is typically ruptured during deep joint flexion [31]. Meanwhile,
the lateral femoral condyle continues to move posteriorly during the passive flexion
arc, resulting in a subluxation at the end of this phase.
During normal gait, the knee has a flexion angle of 0–10° at heel strike, then
flexes to 15–20° at 15–20% of the gait cycle, followed by an extension during
20–40% of the gait cycle, and then flexes to around 60° in the swing phase [32].
Meanwhile, the knee undergoes internal-external rotation of up to 5° and varus-
valgus rotation of up to 4°, combined with medial-lateral translation of up to 12 mm,
proximal-distal translation of up to 14 mm, and anterior-posterior tibial translation
of up to 7 mm [33]. As the knee is flexed, the medial condyle of the femur has little
anterior-posterior movement but the lateral condyle rolls backward on the tibia,
resulting in a coupled internal tibial rotation [34].
196 H. Wang et al.
During normal gait, axial compression on the articular contact surface of the
knee ranges from 0 to 3.2 times body weight (BW) [35]. The greatest axial
compression occurs at around 50% of the gait cycle, just before the toe-off phase,
and most of the joint forces are distributed on the medial tibial plateau. Loading in
the A-P and M-L directions is much lower than the axial loading, ranging from 0 to
0.28 BW and 0 to 0.14 BW, respectively. Internal-external moments range from 0 to
0.02 Nm per BW.
As the name suggests, the PF joint mainly consists of the patella and the femur. The
quadriceps tendon inserts at proximal patella which connects the patella to quadri-
ceps femoris muscle. On the distal side, the patellar tendon attaches directly to the
tibial bone. As a gliding joint, the patella slides along the femoral trochlea during
knee flexion/extension. The primary function of the PF joint is to increase the lever
arm of the quadriceps muscles and thus to enhance the efficiency of extending the
knee [36].
During flexion, the patella moves posteriorly and distally with respect to the
femur condyles, following a “C”–shaped pattern (Fig. 9.4). Movement of the patella
outside the sagittal plane is minor by comparison, tilting and twisting by less than
15° [37]. The lateral facet of the patella has been reported to bear greater contact
forces than the medial facet during knee flexion [37]. Contact between the articular
surfaces of the PF joint reach a peak when the joint flexes to 90°. Past 90°, the
patella starts to rotate laterally and contacts the femur at the medial side. During
deep flexion (around 140°), the patella falls into the femoral notch and the contact
area is greatly reduced (termed the “odd facet”), resulting in high localized stresses.
Therefore, patellar chondromalacia and patellofemoral pain syndrome are often
experienced during this “odd facet” stage [38].
To have a better understanding on the biomechanics of the PF joint, the quadri-
ceps angle (Q angle) needs to be introduced first. As shown in Fig. 9.5, the Q angle
Lateral Medial
epicondyle epicondyle
90º
135º
9 Biomechanics of the Knee 197
Q angle
tibial tuberosity
is the angle between a line that connects the center of the patella and the superior
anterior iliac and another line that goes through the center of the patella and the
tuberosity of the tibia. However, there is a significant difference in Q angle between
males and females. For males, the Q angle at full knee extension ranges from 0° to
19°, while that for females is larger, ranging from 6° to 27° [39]. This is mainly due
to the larger pelvis in females. Changes in the Q angle can help with understanding
the mechanics of knee motions and patellar traction. During knee extension, the
tibia gradually rotates externally and as a result the Q angle increases. Inversely, the
Q angle decreases during knee flexion when the patella is pulled into the trochlea of
the femur by a traction of horizontal forces from the oblique muscle. Thus, stability
of the patella is largely dependent on the value of the Q angle and strength of the
oblique muscle. Loss of stability may result in the patella tilting externally, resulting
in wearing of the lateral patellar cartilage. The size of the Q angle can be affected
198 H. Wang et al.
by some factors, such as the size of the pelvic bone and connection between joint
structures [40]. Abnormal joint structure such as excessive anterior tilt of the femoral
head, excessive knee abduction, and other irregular rotations of the tibiofemoral
joint could all result in an increased Q angle.
2.1.1 ACL Injury
The ACL is frequently injured because of its critical role in constraining joint
motion [41]. Most ACL injuries occurred in the absence of physical contact, and
frequently during sport activities such as skiing, football, and basketball [41–43].
Females also have a higher incidence of injury than males [28]. Serious damage to
the ACL can have a considerable impact on absence from work and the life quality.
Long-term complications of ACL injury include secondary injuries to other knee
structures such as meniscal tear, MCL injury that could induce degenerative changes
to the cartilage both without and with surgical reconstruction [44, 45].
More than 75% of ACL tears are a complete rupture of the ligament, while the
remaining 25% are partial tears [46]. Generally speaking, 1/3 of the patients had
ACL reconstruction surgery, while a second 1/3 had delayed surgery. The remaining
1/3 would not require surgery. For the latter group, the ACL healed and the knee
functions as well as those following surgery.
2.1.2 Conservative Treatment
Conservative treatments for ACL injury include plaster fixation, cryotherapy, mus-
cle training, and strengthening of coordination [47]. With limited ACL function,
anterior translation of the tibia may be restricted if there is adequate proprioception
to contract the hamstring muscles [47]. However, although such conservative treat-
ments are noninvasive and less stressful for the patient, the clinical outcome is rela-
tively unpredictable and remains controversial. In a clinical report at 10–13 years
after surgery, the ACL reconstruction group was reported to perform significantly
better than the nonoperative group (conservative treatments) in maintaining involve-
ment in sports [48]. In contrast, other studies have shown good patient satisfaction
after 20 years following conservative treatments, though the objective measures
indicated increasing knee degeneration [47]. When treated by conservative means
alone, Noyes [49] reported that tears across one-quarter of the ACL body did not
usually progress further, while one-half tears progressed in 50% of people and
three-quarter tears progressed in 86% of people, eventually becoming a complete
deficiency 7 years after treatment. Thus, the initial size of the ACL tear might be
important for evaluating the expected clinical outcome of conservative treatment.
9 Biomechanics of the Knee 199
2.1.3 ACL Repair
The first attempts at repairing ruptured ACLs involved reconnecting the ligament
remnants by sutures (termed a primary repair) to facilitate healing. However, in a
5-year clinical follow-up study on ACL primary repair, 53% of the patients had a
reinjury, 94% suffered instability, and 71% had pain [50]. It was generally con-
cluded that the poor results of primary repair could be attributed to the poor healing
capacity of the ACL [51, 52].
Functional tissue engineering was introduced in the 1980s as a novel method to
“grow tissues or organs from a single cell taken from an individual” [53]. With
developments in functional tissue engineering, biological scaffolds such as extracel-
lular matrix (ECM) have been used to reinforce ACL healing [54, 55]. ECM bioscaf-
folds serve to bridge the gap between the ACL remnants to facilitate the migration
and proliferation of the cells and formation of the blood vessels, as well as the
transportation of wastes, and thus consequently accelerate tissue formation and
improve the healing of the ACL. Woo et al. [55] used ECM in sheet and gel forms
to repair a transected goat ACL. The results showed continuous formation of neo-
tissue in the ECM group while there was limited tissue growth in the control group
(suture only). Also, the stiffness of the femur-ACL-tibia complex for the group
using ECM was 2–3 times that of the group using suture repair alone.
Mechanical augmentation for ACL repair has also shown positive results for
improving ACL healing. Fleming et al. introduced a form of suture augmentation
where the sutures were fixed directly to the bone to reinforce the repaired ACL and
reduce anterior laxity of the joint [56]. Fisher et al. [10] showed that, in comparison
to traditional suture repair, mechanical augmentation of ruptured ACLs with sutures
resulted in improved joint stability, higher load in the repaired ACL, and lower load
in the medial meniscus, demonstrating the effectiveness of suture augmentation for
restoring joint stability and protecting the medial meniscus. The subsequent intro-
duction of biodegradable ACL grafts allowed for early reinforcement of the repaired
ACL and gradual transferring of loading to the ACL as it heals, along with the con-
current dissolution of the graft [57]. Similarly, a degradable magnesium ring device
was designed and used by Farraro et al. as mechanical reinforcement which was
combined with biological reinforcement (ECM scaffold) for ACL repair to restore
joint stability and to load the femoral insertion site to prevent disuse atrophy
throughout the ligament healing [58].
2.1.4 ACL Reconstruction
Because of the bad capacity of healing of the ACL and the unsatisfying clinical
outcomes of conservative treatments and repair surgeries, ACL reconstruction has
remained the gold standard for treating ACL ruptures [59]. This involves replace-
ment of the original ACL with a graft that is fixed into femoral and tibial tunnels to
restore joint stability. There are a variety of sources for the graft, with the choice of
using an allograft, autograft, or artificial graft. The bone-patellar tendon-bone
complex (BPTB) and four-strand hamstring tendon (HS) have become the two most
200 H. Wang et al.
commonly used autografts, and the LARS artificial graft (LARS Company, France)
is the most popular synthetic graft, with a market history of over 20 years [60, 61].
Satisfactory outcomes have been reported in the short term following ACL
reconstruction. Previous study displayed no loss in range of motion of the knee
2 years after the surgery and limited postsurgical complications [62]. Another recent
study [63] reported that 1 year following ACL reconstruction, the rate of graft fail-
ure was 6.5% for HS and 2.1% for BPTB, while the rate of returning to sports was
71% when using a HS graft and 78% when using a BPTB graft.
However, although ACL reconstruction shows positive results in the short term,
long-term complications have been reported. First, the complex anatomy of the
ACL insertions cannot be restored, and thus the rotational joint stability cannot be
totally restored. Tashman et al. [64] measured knee kinematics during downhill run-
ning 4–12 months following ACL reconstruction and found that the anterior tibial
translation for the reconstructed knee had no significant difference with the unin-
jured knee, but the reconstructed knee had larger external rotation (3.8° larger on
average) and varus rotation (2.8° larger on average). Second, there could be donor
site morbidity when using autografts. In a study by Salmon et al. [65], donor site
related complications were reported to appear in 42% of the patients 13 years after
ACL reconstruction, while 45% of patients suffered pain when kneeling. Third,
there have been reports of enlargement of the bone tunnels and rupture of the grafts,
which have an incidence of up to 72% and 22%, respectively [66, 67]. Bone tunnel
enlargement can cause graft loosening, fixation failure, knee instability, and osteo-
arthritis, while a graft rupture directly calls for follow-up surgery. Other potential
complications include loss of proprioception, graft wear, graft laxity [68], extension
loss, and long-term osteoarthritis [69, 70]. A recent study reported an incidence of
knee osteoarthritis of nearly 20% at 12 years following ACL reconstruction [45].
2.2.1 Meniscal Tear
A tear in the meniscus is a relatively common soft-tissue injury. A meniscal tear can
cause pain, swelling, and joint instability, and long term could induce knee osteoar-
thritis due to increased stresses on the hyaline cartilage. Englund et al. [71] assessed
the integrity of the menisci in 991 subjects using a 1.5 T MRI and found that 19%
of females and 56% of males had some level of tearing, and over 60% of subjects
with osteoarthritis had a meniscal tear. Among the subjects without any symptoms
of knee pain, aching, or stiffness, 60% were found to have a meniscal tear.
There are a variety of types of meniscal tear which may be characterized based
on the location and appearance: horizontal tear, flap tear, radial tear, root tear, verti-
cal tear, oblique tear, and complex tear.
Horizontal tears are defined as occurring parallel to the tibial plateau in the
region that divides the meniscus into superior and inferior halves. A flap tear occurs
9 Biomechanics of the Knee 201
where there are displacements of torn fragments toward the articular space. A radial
tear is vertical to the tibial plateau and transects the longitudinal collagen bundle.
This type of meniscal tear impairs the axial strength of meniscus and might increase
the risk of meniscal extrusion [72]. A meniscal root tear is defined when a radial tear
happens within 1 cm of the bony tibial attachment. A vertical tear runs parallel to
the longitudinal collagen bundle and divides the meniscus into peripheral and cen-
tral portions. Oblique tears are oriented oblique to the longitudinal collagen fibers.
Complex tears are defined as a combination of two or more types of tears occurring
simultaneously. According to Englund et al. [71], among 308 subjects with menis-
cal tear, 40% were horizontal tears, 37% were complex tears, 15% were radial tears,
12% were oblique tears, 7% were longitudinal (vertical) tears, and 1% were
root tears.
2.2.2 Meniscal Repair
Where appropriate, repairing the meniscus with sutures is often a good option for
restoring normal mechanical function while maintaining proprioception and vascu-
larity. In a 2-year follow-up study on 280 meniscal repairs [73], successful results
were obtained in 252 knees, with a total failure rate of 10%. Of those patients that
were deemed successful, there was an absence of any tenderness of the joint line,
blocking or swelling. However, whether to perform a meniscal repair could be
largely dependent on the tear type and the surgeon’s individual ability and experi-
ence. Vascularity is only provided at the periphery of the menisci (up to 25% of the
width, termed the red-red zone) (Fig. 9.6) as well as at its horn attachments [74], and
thus the capacity of healing of the meniscus is rather poor in the middle portion
(termed the white-white zone) (Fig. 9.6). Previous studies demonstrated how tears
on the posterior root of the medial meniscus could be repaired to restore the normal
joint loading profile [75]. If a vertical tear is located peripherally, meniscal repair
using sutures is usually the preferred treatment [76]. Other types of meniscal tears,
particularly radial tears, are more difficult to repair and heal. For these difficult
white-white zone
202 H. Wang et al.
2.2.3 Meniscectomy
2.2.4 Meniscal Replacement
If a patient develops complications such as joint pain from early degeneration after
meniscectomy, they may be considered as a candidate for meniscal replacement.
Allograft
Meniscal allografts are obtained from donors and should contain no living cells
[77]. If donor cellular material remains in the graft, a dense cartilage matrix would
be used to lock the materials so that the allograft does not elicit any immune
response.
Meniscal replacement was first introduced in the 1980s [83]. A previous study
[84] reported a total failure rate of 29% for allograft transplants 13 years after the
replacement surgery, while the overall Lysholm score improved from 36 to 61
9 Biomechanics of the Knee 203
Meniscal Bioscaffold
The red-red zone of the meniscus has a vascular supply while the middle section
(white-white zone) does not. In a partial meniscectomy, if the red-red zone is well
preserved, an artificial scaffold can be sutured to replace the missing tissue and
restore the mechanical function of the meniscus.
The Menaflex Collagen Meniscal Scaffold (Regen Biologics Inc., New Jersey,
USA) and the Actifit Scaffold (Orteq, London, UK) are two of the most commonly
used meniscal scaffolds on the market. The former is made of Achilles tendon of
bovine that is pressure heat molded into the shape of a meniscus. A clinical follow-
up study demonstrated significantly increased tissue formation with the use of this
scaffold over a partial meniscectomy [87], and more activity was regained postop-
eratively in the scaffold group than the meniscectomy group. The Actifit scaffold is
a porous biodegradable scaffold made of polyurethane that allows neo-tissue to
gradually replace the scaffold material over time. A 1-year biopsy study showed
viable “meniscus-like” tissue replacing the scaffold material and a considerable
reduction in patient pain [77].
for surgical knee osteotomy. This may be a high tibial osteotomy or fibular osteot-
omy. However, under severe loss of cartilage and/or bone and consequent intolera-
ble pain and dysfunction of the knee joint, the only viable option is to undergo knee
arthroplasty, during which the joint surfaces are replaced by prosthetic components.
The ultimate goal of knee arthroplasty is to replace the problematic knee joint with
a prosthetic knee in order to restore the joint functionality and relieve the pain.
Although osteoarthritis can present in any of the knee compartments, a large portion
of the cases reported occur in the medial compartment of the tibiofemoral joint [92],
with fewer cases being reported for the lateral side or the patellofemoral joint [93].
When osteoarthritis appears in the medial side, the medial joint space is usually nar-
rowed because of the loss of cartilage and bone, resulting in abnormal force trans-
mission along the lower limb.
For patients who are younger and more active, high tibial osteotomy (HTO) is
often chosen for early and moderate osteoarthritis. In this case, part of the tibial
bone is removed to relocate the joint surface and to restore joint alignment. For
medial arthritis, more bone would be removed at the medial side, changing the joint
alignment from varus to be slightly valgus, thus decreasing medial joint pressure
and slowing down the degeneration of the medial cartilage. This may postpone or
avoid the necessity for knee arthroplasty. Previous studies comparing clinical results
of HTO and unicompartmental knee arthroplasty (UKA) concluded that UKA pres-
ents lower revision rates, less postoperative complications, and less postoperative
pain. However, HTO allowed for a better range of motion, which could be more
satisfactory for highly active patients [94].
2.3.3 Fibular Osteotomy
The fibula bone is generally considered to be less critical for joint functionality than
the other long bones of the lower limb. The fibula provides an origin for several
muscles of the knee joint and carries axial weight-bearing loads ranging from 6.4%
[95] to 16.7% [96]. It has been freely used as a source for grafts and as a vascular-
ized transplant to bridge large bony defects for conditions such as congenital pseud-
arthrosis of the tibia [97], tumor resection [98], nonunion [99] and grafting
operations in case of femoral head necrosis [100].
Proximal fibulectomy (fibular osteotomy) has been demonstrated by several
studies to improve the restoration of joint functionality and reduce pain in
patients with OA. It is a simple but effective procedure, with the number of
patients with a fibulectomy in China surpassing 1000 in 2015 [101]. To perform
a fibular osteotomy, the proximal part of the fibula is first exposed, followed by
an osteotomy with a length of 2–4 cm at 7–8 cm from the proximal fibula head.
In a follow-up study on 110 patients over a 2-year period, Yang et al. [102]
9 Biomechanics of the Knee 205
a b
reported significant improvement in alignment (Figs. 9.7 and 9.8) and function
of the knee, as well as relief of pain following the fibular osteotomy. Zou et al.
[103] investigated 92 patients with varus knee OA treated by either fibular oste-
otomy or HTO and found that the outcomes of fibular osteotomy were superior
to HTO in either short term or long term. Nie et al. [104] reported on 16 patients
with knee OA in the medial compartment who underwent proximal fibulectomy
and found that there were significant improvement in VAS pain and HSS score 1
day and 6 months after surgery. In addition, the hip-knee-angle (HKA) improved
and remained stable for 3 months and there was an inverse relationship between
the overall peak knee adduction moment (KAM) and HKA. Yazdi et al. [105]
evaluated the effect of partial fibulectomy on the articular contact pressure in
cadaver knees and it was demonstrated that partial fibulectomy decreased the
medial articular pressure and increased that in the lateral compartment. Although
the procedure is relatively simple and there are good clinical results, it may
206 H. Wang et al.
a b
Fig. 9.8 Immediately postoperative radiographs; anteroposterior (a) and lateral (b)
cause adjacent peroneal nerve injury. Sandoval et al. [106] followed up 116
patients with proximal fibulectomy for 2 years and reported that only 9.4% suf-
fered complications, mainly for neuropraxia of the peroneal nerve, hematoma of
the wound and infection. While fibulectomy has generally been shown to be
effective, it is not clear exactly how the procedure relieves knee pain. Zhang
et al. [107] stated that nonuniform settlement and bilateral degeneration of the
plateau leads to varus knee, while with an osteotomy of the fibula can modify
this situation. Weakened support from the lateral fibula results in a correction of
the varus deformity, and consequently shifted the loading from the medial com-
partment to the lateral, which eventually leads to relieved pain and restored joint
functionality.
Qi et al. [108] hypothesized that a reduction in lateral muscle contraction after
fibulectomy may cause a rebalance in the resultant joint moment, making the con-
tact forces in the medial compartment shift laterally and decrease in magnitude.
According to Qi [108], the balance of joint forces in the coronal plane of a varus
knee may be gauged by the following equations (Fig. 9.9):
FLM + FB + FMM = FJL + FJM, where FJM is the medial joint contact force, FJL is the
lateral joint contact force, FLM is the lateral resultant muscle force, FB is body weight
due to the gravity, FMM is the medial resultant muscle force, and FJM can be ignored
due to varus knee.
9 Biomechanics of the Knee 207
F
B FJM FJL
FLM
FMM
dB dLM
dLM × FLM = dB × FB + dMM × FMM, where dLM is the moment arm of lateral resul-
tant muscle force to knee joint center, dB is the moment arm of body weight to the
knee joint center, and dMM is the moment arm of medial resultant muscle force to the
knee joint.
′
After removing a portion of the fibula (Fig. 9.10), the lateral muscle ( FLM ) is
′
released which decreases the valgus moment ( d LM × FLM < d B × FB ). To compen-
sate, the varus resultant moment is reduced by shifting the body weight laterally
which subsequently decreases the moment arm of body weight ( d B′ < d B ).
′
Meanwhile due to the decrease in the lateral muscle force ( FLM ), the overall knee
′ ′ ′
joint forces also decrease ( FLM + FB = FJL + FJM ).
In summary, proximal fibulectomy is an innovative procedure to treat medial
knee OA without the need for resecting the knee joint. Studies performed to date
lack sufficient quantitative biomechanical evidence to recommend using this proce-
dure as standard practice. Further clinical follow-up studies and comparative analy-
ses to other common treatment methods are required.
208 H. Wang et al.
FB
F′JM F′JL
F′LM
FMM
d′B dLM
2.3.4 Knee Arthroplasty
Knee arthroplasty is often considered when conservative treatments and less inva-
sive bone osteotomies are either unsuccessful or are not considered a viable option
because the natural cartilage has reached an irreparable state. Knee arthroplasty can
offer pain relief and help regain normal joint function. The knee prosthesis gener-
ally consists of the femoral, tibial, and patellar components. The tibial component
usually contains a polyethylene liner inserted into a metal baseplate.
Knee arthroplasty may be performed as a unicompartmental knee arthroplasty
(UKA) or a total knee arthroplasty (TKA). For TKA, there are generally two types
of knee prosthesis in clinical use: cruciate-retaining type (CR type) and posterior-
stabilized type (PS type).
compartments at the same time. In theory, because UKA retains more soft tissues, it
can better restore knee functionality and stability than TKA and would allow the
patient to retain a better ontological feel. In the early years following the introduc-
tion of unicompartmental procedures, the failure rate was quite high due to poor
surgical techniques and poorly designed components. The average revision rate of
UKA at 2 and 6 years has been reported at 20% and 28%, respectively, in 1970s
[109, 110]. With successive improvements in the design of components, mature
clinical techniques using minimally invasive surgery have significantly improved
the clinical success rate with appropriate patient selection [111]. The survival rate
of UKA at 2 and 5 year is 98.7% and 95.5%, respectively, and the knee function
score remains satisfactory [112]. However, the long-term survival rate is signifi-
cantly lower than that of TKA [113]. Failure rates of UKA vary across literature and
for different regions [114–117]; 27% (10 years) in Finland, 12.1% (8 years) in UK,
and 4.6% (10 years) in Japan. The success of UKA is dependent on many factors,
including the age and activity of the patient, surgeon’s experience, and the type of
implant used.
UKA has been used in clinic for over 60 years. In the early 1950s, McKeever
[118] inserted metal plates into a patient to replace the damaged articular cartilage
surface, becoming one of the first documented cases of UKA. By the 1960s the
procedure had reached mainstream practice thanks to improved designs and the use
of alternative materials. Gunston [119] developed the first unilateral metal-on-PE
bearing, which is still the most common material combination in use today. The
1970s saw the introduction of non-constrained prostheses that increased the activity
of the joint and could achieve a greater joint range of motion than TKA prostheses
[120]. In the 1980s, Goodfellow introduced the idea of an Oxford active platform
into the design of single condyle prostheses (Oxford unicompartmental knee
replacement, OUKR) which increased the articular contact area and reduced the
contact stress [121]. In recent years, custom-made UKA prostheses (for example,
iUni by ConforMIS Inc.) developed from CT scans and manufactured by 3D recon-
struction and printing have been introduced. As the shape of the prosthesis is
designed according to the individual anatomy of the femoral condyles, custom pros-
theses can provide better alignment and more natural movement. However, given
that custom implants are a recent development, the long-term outcome is relatively
unknown. According to the American Joint Replacement Registry [122], UKA
accounted for about 3.2% of primary knee replacement procedures from 2012 to
2017, and there was a downward trend year on year. This same trend has also been
observed in Australia [123] and Sweden [124], with the former showing a decrease
in UKA from 14.5% in 2003 to 5.1% in 2016. However, since 2017 the trend has
reversed and there has been a slight uptake in UKA procedures in Australia [123],
Sweden [124], and Britain [125].
There are a number of potential causes of failure of unilateral knee prostheses,
including degeneration of the adjacent tibiofemoral joint and patellofemoral joint,
implant loosening, mechanical injury, malposition, and infection. Among them,
aseptic loosening and joint disease (including contralateral arthritis) are the most
common causes of failure [123–125]. Typical factors leading to loosening of the
210 H. Wang et al.
In the operation of TKA, all of the articular surfaces in the knee are replaced with
prosthetic components. The two most common types of total knee prostheses in
clinical use today are cruciate-retaining (CR) and posterior-stabilized (PS). Both
types require the removing of the ACL, but with CR prostheses the posterior cruci-
ate ligament (PCL) is retained, while PS prostheses also require a resection of the
PCL. With the PS knee, a femoral cam and a tibial post interact as a functional
replacement for the PCL. When the femoral component slides anteriorly, the cam
contacts the tibial post, forcing the femoral component to roll back along the tibia.
This mechanism is designed to prevent excessive anterior femoral translation and
help to maintain joint stability.
The anterior region of the tibial liner in TKA is often designed to be higher than
the posterior part in order to avoid excessive anterior femoral translation and
increase the contact area of the tibiofemoral joint. This stabilizes the knee and
decreases contact stress on the components [128].
The polyethylene liner in the tibial component may be anatomically shaped
(“anatomical design”) (Fig. 9.11b) or be a simpler nonanatomical design (Fig. 9.11a).
For the nonanatomical design, the medial and lateral part of the polyethylene liner
are often symmetrical, concave, and have the same curvature radius [128]. In this
case, the curvature of the femoral component may be also designed to conform to
9 Biomechanics of the Knee 211
a b
proximal
distal
Fig. 9.11 Different designs of the polyethylene liner (a) nonanatomical design and (b) anatomical
design [129] (Reprinted with permission from Elsevier, license number: 4687410127428)
the tibial liner in order to maintain the knee stability and prevent component dislo-
cation [128]. However, the normal knee shape is asymmetrical and this nonanatomi-
cal design could induce abnormal knee kinematics such as inadequate rollback of
the femur and insufficient internal rotation of the tibia. Thus, anatomical prosthesis
designs aim to mimic the morphology of the intact knee to restore the kinematics of
the knee. Liu et al. found that [129] changing the posterolateral region of the tibial
liner to be convex could better restore the posterior translation of the lateral femoral
condyle and the internal rotation of the tibia through joint flexion from 0° to 140°.
A study by Moonot et al. [130] demonstrated that asymmetrical tibial liners with a
highly conforming medial aspect and a moderately conforming lateral aspect could
adequately restore the internal tibial rotation and reduce excessive anterior move-
ment of the lateral femoral condyle, thus reducing wear on the liner and increasing
implant longevity.
When considering the type of TKA implant to be used, it is still controversial
whether the PCL needs to be retained for proper knee functionality [131]. CR knees
require an intact and functioning PCL, and so this must be carefully evaluated
beforehand. Otherwise, if the PCL is compromised then joint stability could be at
risk. In this case a PS knee is more desirable. However, when using a PS design,
joint stability is greatly affected by the design of the tibial post and the femoral cam,
and this would also have a considerable effect on joint kinematics and kinetics.
If the cartilage on the patella is degraded and may compromise knee functional-
ity, then there is the option of replacing the cartilage with a smooth p olymer surface
(patellar replacement). However, Cadambi et al. reported that replacing the patellar
surface reduces the success rate of TKA [132]. Huang et al. also showed that tensile
stress on the patellar bone increased after the surface was replaced [133]. Rand
[134] stated that introduction of the patellar component could alleviate anterior
knee pain of the patients. However, the revision rate of TKA is still relatively high
because of the patellar component related complications, such as infection, fracture,
212 H. Wang et al.
subluxation, wear, and loosening of the patellar component [135, 136]. Obese
patients are also not recommended to undergo patellar arthroplasty because the
higher loading could increase the risk of loosening and fracture [137].
3 Summary
In this chapter, the basic anatomy and functionality of bony structures, menisci,
cartilage, tendons, and muscles in a normal knee joint were presented, accompanied
by a discussion of the biomechanics of the patellofemoral and tibiofemoral joints.
Pathological changes to the structures of the knee could alter the joint kinematics,
which in long term could induce secondary injuries to the other surrounding tissues.
Clinical treatments should be carefully chosen to best rebuild the normal biome-
chanics and functionality of the knee.
References
1. Hashemi J, Chandrashekar N, Gill B, Beynnon BD, Slauterbeck JR, Schutt RC, Mansouri H,
Dabezies E. The geometry of the tibial plateau and its influence on the biomechanics of the
tibiofemoral joint. J Bone Joint Surg. 2008;90:2724–34.
2. Flandry F, Hommel G. Normal anatomy and biomechanics of the knee. Sports Med Arthrosc
Rev. 2011;19:82–92.
3. Sullivan NP, Robinson PW, Ansari A, Hassaballa M, Robinson JR, Porteous AJ, Eldridge JD,
Murray JR. Bristol index of patellar width to thickness (BIPWiT): a reproducible measure of
patellar thickness from adult MRI. Knee. 2014;21:1058–62.
4. Shepherd D, Seedhom B. Thickness of human articular cartilage in joints of the lower limb.
Ann Rheum Dis. 1999;58:27–34.
5. Arokoski J, Jurvelin J, Väätäinen U, Helminen H. Normal and pathological adaptations of
articular cartilage to joint loading. Scand J Med Sci Sports. 2000;10:186–98.
6. Eckstein F, Hudelmaier M, Putz R. The effects of exercise on human articular cartilage. J
Anat. 2006;208:491–512.
7. Andriacchi TP, Mündermann A, Smith RL, Alexander EJ, Dyrby CO, Koo S. A framework for
the in vivo pathomechanics of osteoarthritis at the knee. Ann Biomed Eng. 2004;32:447–57.
8. Seedhom B, Dowson D, Wright V. Proceedings: functions of the menisci. A preliminary
study. Ann Rheum Dis. 1974;33:111.
9. McCann L, Ingham E, Jin Z, Fisher J. Influence of the meniscus on friction and degradation
of cartilage in the natural knee joint. Osteoarthr Cartil. 2009;17:995–1000.
10. Fisher MB, Jung HJ, McMahon PJ, Woo SLY. Suture augmentation following ACL injury
to restore the function of the ACL, MCL, and medial meniscus in the goat stifle joint. J
Biomech. 2011;44:1530–5.
11. Harner CD, Baek GH, Vogrin TM, Carlin GJ, Kashiwaguchi S, Woo SLY. Quantitative analy-
sis of human cruciate ligament insertions. Arthroscopy. 1999;15:741–9.
12. Li G, DeFrate LE, Sun H, Gill TJ. In vivo elongation of the anterior cruciate ligament and
posterior cruciate ligament during knee flexion. Am J Sports Med. 2004;32:1415–20.
13. Otsubo H, Shino K, Suzuki D, Kamiya T, Suzuki T, Watanabe K, Fujimiya M, Iwahashi T,
Yamashita T. The arrangement and the attachment areas of three ACL bundles. Knee Surg
Sports Traumatol Arthrosc. 2012;20:127–34.
9 Biomechanics of the Knee 213
14. Kanamori A, Zeminski J, Rudy TW, Li G, Fu FH, Woo SLY. The effect of axial tibial torque
on the function of the anterior cruciate ligament. Arthroscopy. 2002;18:394–8.
15. Fukuda Y, Woo SLY, Loh JC, Tsuda E, Tang P, Mcmahon PJ, Debski RE. A quantitative anal-
ysis of valgus torque on the ACL: a human cadaveric study. J Orthop Res. 2010;21:1107–12.
16. Woo SLY, Debski RE, Zeminski J, Abramowitch SD, Chan Saw SS, Fenwick JA. Injury and
repair of ligaments and tendons. Annu Rev Biomed Eng. 2000;2:83–118.
17. Li G, Rudy TW, Allen C, Sakane M, Woo SLY. Effect of combined axial compressive and
anterior tibial loads on in situ forces in the anterior cruciate ligament: a porcine study. J
Orthop Res. 1998;16:122.
18. Voos JE, Mauro CS, Wente T, Warren RF, Wickiewicz TL. Posterior cruciate ligament: anat-
omy, biomechanics, and outcomes. Am J Sports Med. 2012;40:222–31.
19. Gill TJ, DeFrate LE, Wang C, Carey CT, Zayontz S, Zarins B, Li G. The biomechanical effect
of posterior cruciate ligament reconstruction on knee joint function: kinematic response to
simulated muscle loads. Am J Sports Med. 2003;31:530–6.
20. Inoue M, Mcgurk-Burleson E, Hollis JM, Woo SLY. Treatment of medial collateral ligament
injury: I. The importance of anterior cruciate ligament on the varus-valgus knee laxity. Am J
Sports Med. 1987;15:15–21.
21. Javier RF, Nicolas P. Recruitment order of quadriceps motor units: femoral nerve vs. direct
quadriceps stimulation. Eur J Appl Physiol. 2013;113:3069–77.
22. Bell D, Trigsted S, Post E, Walden C. Hip strength in patients with quadriceps strength defi-
cits after ACL reconstruction. Med Sci Sports Exerc. 2016;48:1886–92.
23. Segal NA, Glass NA. Is quadriceps muscle weakness a risk factor for incident or progressive
knee osteoarthritis? Phys Sportsmed. 2011;39:44–50.
24. Ko HI, Jeon SY, Kim SH, Nam PK. Comparison of hip extensor muscle activity including
the adductor magnus during three prone hip extension exercises. Physiother Theory Pract.
2019;35:451–7.
25. Cleather DJ. An important role of the biarticular hamstrings is to exert internal/external rota-
tion moments on the tibia during vertical jumping. J Theor Biol. 2018;455:101–8.
26. Wang ZW, Liu YL, Lin KJ, Qu TB, Dong X, Cheng CK, Hai Y. The effects of implantation
of tibio-femoral components in hyperextension on kinematics of TKA. Knee Surg Sports
Traumatol Arthrosc. 2012;20:2032–8.
27. Giffin JR, Vogrin TM, Zantop T, Woo SLY, Harner CD. Effects of increasing tibial slope on
the biomechanics of the knee. Am J Sports Med. 2004;32:376–82.
28. Arendt E, Dick R. Knee injury patterns among men and women in collegiate basketball and
soccer: NCAA data and review of literature. Am J Sports Med. 1995;23:694–701.
29. Hefzy MS, Kelly BP, Cooke TD. Kinematics of the knee joint in deep flexion: a radiographic
assessment. Med Eng Phys. 1998;20:302–7.
30. Hung CH, Wei HW, Chang ZW, Cheng CK. Anatomy and biomechanics of knee. In: Chan
KM, editor. Practical arthroscopic surgery. Beijing: People’s Medical Publishing House;
2008. p. 78–90.
31. Chung JY, Song HK, Jung MK, Oh HT, Kim JH, Yoon JS, Min BH. Larger medial femoral
to tibial condylar dimension may trigger posterior root tear of medial meniscus. Knee Surg
Sports Traumatol Arthrosc. 2016;24:1448–54.
32. Lafortune M, Cavanagh P, Sommer HJ, Kalenak A. Three-dimensional kinematics of the
human knee during walking. J Biomech. 1992;25:347–57.
33. Zhang Y, Yao Z, Wang S, Huang W, Ma L, Huang H, Xia H. Motion analysis of Chinese
normal knees during gait based on a novel portable system. Gait Posture. 2015;41:763–8.
34. Hollis JM, Takai S, Adams DJ, Horibe S, Woo SLY. The effects of knee motion and external
loading on the length of the anterior cruciate ligament (ACL): a kinematic study. J Biomech
Eng. 1991;113:208.
35. ASTM. Standard guide for total knee replacement loading profiles. West Conshohocken, PA:
ASTM International; 2017. p. 1–33.
36. Fox AJ, Wanivenhaus F, Rodeo SA. The basic science of the patella: structure, composition,
and function. J Knee Surg. 2012;25:127–42.
214 H. Wang et al.
37. Akbarshahi M, Fernandez JW, Schache AG, Pandy MG. Subject-specific evalua-
tion of patellofemoral joint biomechanics during functional activity. Med Eng Phys.
2014;36:1122–33.
38. Hsu RWW, Cheng CK, Yu SY, Chen TH. The laxity of knee joint in normal Chinese adults.
Biomed Eng Appl Basis Commun. 1994;6:186–92.
39. Shultz SJ, Nguyen AD, Schmitz RJ. Differences in lower extremity anatomical and postural
characteristics in males and females between maturation groups. J Orthop Sports Phys Ther.
2008;38:137–49.
40. Byl T, Cole JA, Livingston LA. What determines the magnitude of the Q angle? A prelimi-
nary study of selected skeletal and muscular measures. J Sport Rehabil. 2000;9:26–34.
41. Griffin LY, Albohm MJ, Arendt EA, Bahr R, Beynnon BD, DeMaio M, Dick RW, Engebretsen
L, Garrett WE, Hannafin JA. Understanding and preventing noncontact anterior cruciate lig-
ament injuries: a review of the Hunt Valley II meeting, January 2005. Am J Sports Med.
2006;34:1512–32.
42. Ali N, Rouhi G. Barriers to predicting the mechanisms and risk factors of non-contact ante-
rior cruciate ligament injury. Open Biomed Eng J. 2010;4:178–89.
43. Shimokochi Y, Shultz SJ. Mechanisms of noncontact anterior cruciate ligament injury. J Athl
Train. 2008;43:396–408.
44. Finsterbush A, Frankl U, Matan Y, Mann G. Secondary damage to the knee after isolated
injury of the anterior cruciate ligament. Am J Sports Med. 1990;18:475–9.
45. Lecoq FA, Parienti JJ, Murison J, Ruiz N, Bouacida K, Besse J, Morin V, Padiolleau G,
Cucurulo T, Graveleau N. Graft choice and the incidence of osteoarthritis after anterior cru-
ciate ligament reconstruction: a causal analysis from a cohort of 541 patients. Am J Sports
Med. 2018;46:2842–50.
46. Smith BA, Livesay GA, Woo SL. Biology and biomechanics of the anterior cruciate liga-
ment. Clin Sports Med. 1993;12:637–70.
47. Gföller P, Abermann E, Runer A, Hoser C, Pflüglmayer M, Wierer G, Fink C. Non-operative
treatment of ACL injury is associated with opposing subjective and objective outcomes over
20 years of follow-up. Knee Surg Sports Traumatol Arthrosc. 2019;27:2665–71.
48. Fink C, Hoser C, Hackl W, Navarro R, Benedetto K. Long-term outcome of operative or
nonoperative treatment of anterior cruciate ligament rupture-is sports activity a determining
variable? Int J Sports Med. 2001;22:304–9.
49. Noyes F, Mooar L, Moorman C, McGinniss G. Partial tears of the anterior cruciate ligament.
Progression to complete ligament deficiency. J Bone Joint Surg Br Vol. 1989;71:825–33.
50. Feagin JA, Curl W. Isolated tear of the anterior cruciate ligament: 5-year follow-up study. Am
J Sports Med. 1976;4:95–100.
51. Sandberg R, Balkfors B, Nilsson B, Westlin N. Operative versus non-operative treatment of
recent injuries to the ligaments of the knee. A prospective randomized study. J Bone Joint
Surg Am Vol. 1987;69:1120–6.
52. Feagin JA, Pierce CM, Geyer MR. ACL primary repair: what we did, the results, and how it
helps today to tailor treatments to the patient and the pathology. In: The ACL-deficient knee.
London: Springer; 2012. p. 97–104.
53. Sipe JD. Tissue engineering and reparative medicine. Ann N Y Acad Sci. 2002;961:1–9.
54. Badylak SF, Freytes DO, Gilbert TW. Extracellular matrix as a biological scaffold material:
structure and function. Acta Biomater. 2009;5:1–13.
55. Fisher MB, Liang R, Jung HJ, Kim KE, Zamarra G, Almarza AJ, McMahon PJ, Woo
SLY. Potential of healing a transected anterior cruciate ligament with genetically modified
extracellular matrix bioscaffolds in a goat model. Knee Surg Sports Traumatol Arthrosc.
2012;20:1357–65.
56. Fleming BC, Carey JL, Spindler KP, Murray MM. Can suture repair of ACL transection restore
normal anteroposterior laxity of the knee? An ex vivo study. J Orthop Res. 2008;26:1500–5.
57. Gisselfält K, Edberg B, Flodin P. Synthesis and properties of degradable poly (urethane urea)
s to be used for ligament reconstructions. Biomacromolecules. 2002;3:951–8.
9 Biomechanics of the Knee 215
58. Farraro KF, Sasaki N, Woo SLY, Kim KE, Tei MM, Speziali A, McMahon PJ. Magnesium
ring device to restore function of a transected anterior cruciate ligament in the goat stifle joint.
J Orthop Res. 2016;34:2001–8.
59. Chambat P, Guier C, Sonnery-Cottet B, Fayard J-M, Thaunat M. The evolution of ACL
reconstruction over the last fifty years. Int Orthop. 2013;37:181–6.
60. Parchi PD, Ciapini G, Paglialunga C, Giuntoli M, Picece C, Chiellini F, Lisanti M, Scaglione
M. Anterior cruciate ligament reconstruction with LARS artificial ligament-clinical results
after a long-term follow-up. Joints. 2018;6:75–9.
61. Barenius B, Nordlander M, Ponzer S, Tidermark J, Eriksson K. Quality of life and clinical
outcome after anterior cruciate ligament reconstruction using patellar tendon graft or qua-
drupled semitendinosus graft: an 8-year follow-up of a randomized controlled trial. Am J
Sports Med. 2010;38:1533–41.
62. Aune AK, Holm I, Risberg MA, Jensen HK, Steen H. Four-strand hamstring tendon autograft
compared with patellar tendon-bone autograft for anterior cruciate ligament reconstruction: a
randomized study with two-year follow-up. Am J Sports Med. 2001;29:722–8.
63. Laboute E, James-Belin E, Puig P, Trouve P, Verhaeghe E. Graft failure is more frequent
after hamstring than patellar tendon autograft. Knee Surg Sports Traumatol Arthrosc.
2018;26:3537–46.
64. Tashman S, Collon D, Anderson K, Kolowich P, Anderst W. Abnormal rotational knee
motion during running after anterior cruciate ligament reconstruction. Am J Sports Med.
2004;32:975–83.
65. Salmon LJ, Russell VJ, Refshauge K, Kader D, Connolly C, Linklater J, Pinczewski
LA. Long-term outcome of endoscopic anterior cruciate ligament reconstruction with patel-
lar tendon autograft: minimum 13-year review. Am J Sports Med. 2006;34:721–32.
66. Nebelung W, Becker R, Merkel M, Röpke M. Bone tunnel enlargement after anterior cruciate
ligament reconstruction with semitendinosus tendon using endobutton fixation on the femo-
ral side. Arthroscopy. 1998;14:810–5.
67. Lai CC, Ardern CL, Feller JA, Webster KE. Eighty-three per cent of elite athletes return to
preinjury sport after anterior cruciate ligament reconstruction: a systematic review with meta-
analysis of return to sport rates, graft rupture rates and performance outcomes. Br J Sports
Med. 2018;52:128–38.
68. Chen CH, Chang CH, Su CI, Wang KC, Liu HT, Yu CM, Wong CB, Wang IC. Arthroscopic
single-bundle anterior cruciate ligament reconstruction with periosteum-enveloping ham-
string tendon graft: clinical outcome at 2 to 7 years. Arthroscopy. 2010;26:907–17.
69. Nagelli CV, Cook JL, Kuroki K, Bozynski C, Ma R, Hewett TE. Does anterior cruciate liga-
ment innervation matter for joint function and development of osteoarthritis? J Knee Surg.
2017;30:364–71.
70. Guidoin MF, Marois Y, Bejui J, Poddevin N, King MW, Guidoin R. Analysis of retrieved
polymer fiber based replacements for the ACL. Biomaterials. 2000;21:2461–74.
71. Englund M, Guermazi A, Gale D, Hunter DJ, Aliabadi P, Clancy M, Felson DT. Incidental
meniscal findings on knee MRI in middle-aged and elderly persons. N Engl J Med.
2008;359:1108–15.
72. Zhang F, Kumm J, Svensson F, Turkiewicz A, Frobell R, Englund M. Risk factors for menis-
cal body extrusion on MRI in subjects free of radiographic knee osteoarthritis: longitudinal
data from the osteoarthritis initiative. Osteoarthr Cartil. 2016;24:801–6.
73. Kalliakmanis A, Zourntos S, Bousgas D, Nikolaou P. Comparison of arthroscopic meniscal
repair results using 3 different meniscal repair devices in anterior cruciate ligament recon-
struction patients. Arthroscopy. 2008;24:810–6.
74. Arnoczky SP, Warren RF. The microvasculature of the meniscus and its response to injury: an
experimental study in the dog. Am J Sports Med. 1983;11:131–41.
75. Allaire R, Muriuki M, Gilbertson L, Harner CD. Biomechanical consequences of a tear of
the posterior root of the medial meniscus: similar to total meniscectomy. J Bone Joint Surg.
2008;90:1922–31.
216 H. Wang et al.
76. Jarraya M, Roemer FW, Englund M, Crema MD, Gale HI, Hayashi D, Katz JN, Guermazi
A. Meniscus morphology: does tear type matter? A narrative review with focus on relevance
for osteoarthritis research. Semin Arthritis Rheum. 2017;46:552–61.
77. McDermott I. Meniscal tears, repairs and replacement: their relevance to osteoarthritis of the
knee. Br J Sports Med. 2011;45:292–7.
78. Roos H, Laurén M, Adalberth T, Roos EM, Jonsson K, Lohmander LS. Knee osteoarthritis
after meniscectomy: prevalence of radiographic changes after twenty-one years, compared
with matched controls. Arthritis Rheum. 1998;41:687–93.
79. Ihn J, Kim S, Park I. In vitro study of contact area and pressure distribution in the human knee
after partial and total meniscectomy. Int Orthop. 1993;17:214–8.
80. Papageorgiou CD, Gil JE, Kanamori A, Fenwick JA, Woo SL, Fu FH. The biomechanical
interdependence between the anterior cruciate ligament replacement graft and the medial
meniscus. Am J Sports Med. 2001;29:226–31.
81. Jorgensen U, Sonne-Holm S, Lauridsen F, Rosenklint A. Long-term follow-up of meniscec-
tomy in athletes. A prospective longitudinal study. J Bone Joint Surg Br Vol. 1987;69:80–3.
82. McNicholas M, Rowley D, McGurty D, Adalberth T, Abdon P, Lindstrand A, Lohmander
L. Total meniscectomy in adolescence: a thirty-year follow-up. J Bone Joint Surg Br Vol.
2000;82:217–21.
83. Milachowski K, Weismeier K, Wirth C. Homologous meniscus transplantation. Int Orthop.
1989;13:1–11.
84. van der Wal RJP, Thomassen BJW, van Arkel ERA. Long-term clinical outcome of open
meniscal allograft transplantation. Am J Sports Med. 2009;37:2134–9.
85. Stone K, Adelson W, Pelsis J, Walgenbach A, Turek T. Long-term survival of concurrent
meniscus allograft transplantation and repair of the articular cartilage: a prospective two-to
12-year follow-up report. J Bone Joint Surg Br Vol. 2010;92:941–8.
86. Verdonk PC, Verstraete KL, Almqvist KF, De Cuyper K, Veys EM, Verbruggen G, Verdonk
R. Meniscal allograft transplantation: long-term clinical results with radiological and magnetic
resonance imaging correlations. Knee Surg Sports Traumatol Arthrosc. 2006;14:694–706.
87. Rodkey WG, DeHaven KE, Montgomery WH, Baker CL, Beck CL, Hormel SE, Steadman
R, Cole BJ, Briggs KK. Comparison of the collagen meniscus implant with partial meniscec-
tomy: a prospective randomized trial. J Bone Joint Surg. 2008;90:1413–26.
88. Krishnasamy P, Hall M, Robbins S. The role of skeletal muscle in the pathophysiology and
management of knee osteoarthritis. Rheumatology. 2018;57:iv22–33.
89. Miller RH. Joint loading in runners does not initiate knee osteoarthritis. Exerc Sport Sci Rev.
2017;45:87–95.
90. McAlindon T, Formica M, Schmid C, Fletcher J. Changes in barometric pressure and ambient
temperature influence osteoarthritis pain. Am J Med. 2007;120:429–34.
91. Hofstede S, Vlieland TPV, Ende CH, Mheen PJM, Nelissen R, Bodegom-Vos L. Designing
a strategy to implement optimal conservative treatments in patients with knee or hip osteo-
arthritis in orthopedic practice: a study protocol of the BART-OP study. Implement Sci.
2014;9:22–8.
92. Wise BL, Niu J, Mei Y, Lane NE, Harvey W, Felson DT, Hietpas J, Nevitt M, Sharma L, Torner
J. Patterns of compartment involvement in tibiofemoral osteoarthritis in men and women and
in Caucasians and African Americans: the multicenter osteoarthritis study. Arthritis Care Res.
2012;64:847–52.
93. Willis-Owen CA, Brust K, Alsop H, Miraldo M, Cobb JP. Unicondylar knee arthroplasty in
the UK National Health Service: an analysis of candidacy, outcome and cost efficacy. Knee.
2009;16:473–8.
94. Cao Z, Mai X, Wang J, Feng E, Huang Y. Unicompartmental knee arthroplasty vs high tibial
osteotomy for knee osteoarthritis: a systematic review and meta-analysis. J Arthroplasty.
2017;33:952–9.
95. Lambert KL. The weight-bearing function of the fibula. A strain gauge study. J Bone Joint
Surg Am Vol. 1971;53:507–13.
9 Biomechanics of the Knee 217
96. Takebe K, Nakagawa A, Minami H, Kanazawa H, Hirohata K. Role of the fibula in weight-
bearing. Clin Orthop Relat Res. 1984;184:289–92.
97. Pho RW, Levack B, Satku K, Patradul A. Free vascularised fibular graft in the treatment of
congenital pseudarthrosis of the tibia. J Bone Joint Surg Br Vol. 1985;67:64–70.
98. Pho RW. Malignant giant-cell tumor of the distal end of the radius treated by a free vascular-
ized fibular transplant. J Bone Joint Surg Am Vol. 1981;63:877–84.
99. Solonen KA. Free vascularized bone graft in the treatment of pseudarthrosis. Int Orthop.
1982;6:9–13.
100. Hall FM. Treatment of avascular necrosis of the femoral head. Hip. 1983;165:876–7.
101. Wang J, Wang K, Chen W, Wu T, Zhang F, Zhang Y. The influence of partial fibula osteotomy
on hip knee and ankle joint. J Hei Bei Med Univ. 2015;36:84–5.
102. Yang ZY, Chen W, Li CX, Wang J, Shao DC, Hou ZY, Gao SJ, Wang F, Li JD, Hao JD. Medial
compartment decompression by fibular osteotomy to treat medial compartment knee osteoar-
thritis: a pilot study. Orthopedics. 2015;38:e1110–e4.
103. Zou GP, Lan WB, Zeng YY, Xie JH, Chen SL, YR Q. Early clinical effect of proximal fibular
osteotomy on knee osteoarthritis. Biomed Res. 2017;28:9291–4.
104. Nie Y, Ma J, Huang ZY, Xu B, Tang S, Shen B, Kraus VB, FX P. Upper partial fibulectomy
improves knee biomechanics and function and decreases knee pain of osteoarthritis: a pilot
and biomechanical study. J Biomech. 2018;71:22–9.
105. Yazdi H, Mallakzadeh M, Mohtajeb M, Farshidfar SS, Baghery A, Givehchian B. The effect
of partial fibulectomy on contact pressure of the knee: a cadaveric study. Eur J Orthop Surg
Traumatol. 2014;24:1285–9.
106. Sandoval EAP, Mesa AC, Valera DH, Pérez JAQ, Quiñones AD, Villanueva FS. Complications
of proximal fibular ostectomy in patients with painful genu varum. Revista Cubana de
Medicina Militar. 2018;47:1–9.
107. Zhang Y-Z. Innovations in orthopedics and traumatology in China. Chin Med J. 2015;128:2841.
108. Qi XZ, Chang CM, Tan CM, Zhang YZ, Cheng CK. Evaluation of unicondylar knee
arthroplasty and fibulectomy from a biomechanical viewpoint. J Med Biomech.
2015;30:479–87.
109. Laskin RS. Unicompartmental tibiofemoral resurfacing arthroplasty. JBJS. 1978;60:182–5.
110. Insall J, Aglietti P. A five to seven-year follow-up of unicondylar arthroplasty.
JBJS. 1980;62:1329–37.
111. Kulshrestha V, Datta B, Kumar S, Mittal G. Outcome of unicondylar knee arthroplasty vs total
knee arthroplasty for early medial compartment arthritis: a randomized study. J Arthroplast.
2017;32:1460–9.
112. Smith JR, Robinson JR, Porteous AJ, Murray JR, Hassaballa MA, Artz N, Newman JH. Fixed
bearing lateral unicompartmental knee arthroplasty—short to midterm survivorship and knee
scores for 101 prostheses. Knee. 2014;21:843–7.
113. Howell RE, Lombardi AV Jr, Crilly R, Opolot S, Berend KR. Unicompartmental knee arthro-
plasty: does a selection bias exist? J Arthroplast. 2015;30:1740–2.
114. Koskinen E, Paavolainen P, Eskelinen A, Pulkkinen P, Remes V. Unicondylar knee replace-
ment for primary osteoarthritis: a prospective follow-up study of 1,819 patients from the
Finnish Arthroplasty Register. Acta Orthop. 2007;78:128–35.
115. Liddle A, Pandit H, Judge A, Murray D. Optimal usage of unicompartmental knee arthro-
plasty: a study of 41 986 cases from the National Joint Registry for England and Wales. Bone
Joint J. 2015;97:1506–11.
116. Liddle AD, Pandit H, Judge A, Murray DW. Effect of surgical caseload on revision rate fol-
lowing total and unicompartmental knee replacement. JBJS. 2016;98:1–8.
117. Yoshida K, Tada M, Yoshida H, Takei S, Fukuoka S, Nakamura H. Oxford phase 3 unicom-
partmental knee arthroplasty in Japan—clinical results in greater than one thousand cases
over ten years. J Arthroplast. 2013;28:168–71.
118. Mckeever DC. The choice of prosthetic materials and evaluation of results. Clin Orthop Relat
Res. 1955;6:17–21.
218 H. Wang et al.
119. Gunston FH. Prosthetic simulation of normal knee movement. J Bone Joint Surg Br Vol.
1971;53:272–7.
120. Marmor L. The modular knee. Clin Orthop Rel Res. 1973;94:242–8.
121. Goodfellow JW, O’Connor J. The mechanics of the knee and prosthesis design. J Bone Joint
Surg Br Vol. 1978;60:358–69.
122. American Joint Replacement Registry. Annual report. 2018. http://www.ajrr.net/images/
annual_reports/AAOS-AJRR-2018-Annual-Report-final.pdf.
123. Australian Orthopaedic Association National Joint Replacement Registry. Hip, knee & shoul-
der annual report. 2018. https://aoanjrr.sahmri.com/documents/10180/576950/Hip%2C%20
Knee%20%26%20Shoulder%20Arthroplasty.
124. Swedish Knee Arthroplasty Register. Annual report. 2017. http://www.myknee.se/pdf/
SVK_2017_Eng_1.0.pdf.
125. National Joint Registry. 15th Annual Report National Joint Registry for England. Wales,
Northern Ireland and the Isle of Man. https://www.hqip.org.uk/wp-content/uploads/2018/11/
NJR-15th-Annual-Report-2018.pdf.
126. Vasso M, Corona K, D’Apolito R, Mazzitelli G, Panni AS. Unicompartmental knee arthro-
plasty: modes of failure and conversion to total knee arthroplasty. Joints. 2017;5:44–50.
127. Riff AJ, Sah AP, Della Valle CJ. Outcomes and complications of unicondylar arthroplasty.
Clin Sports Med. 2014;33:149–60.
128. Wang X-H, Song D-Y, Dong X, Suguro T, Cheng C-K. Motion type and knee articular con-
formity influenced mid-flexion stability of a single radius knee prosthesis. Knee Surg Sports
Traumatol Arthrosc. 2019;27:1595–603.
129. Liu YL, Lin KJ, Huang CH, Chen WC, Chen CH, Chang TW, Lai YS, Cheng CK. Anatomic-
like polyethylene insert could improve knee kinematics after total knee arthroplasty—a com-
putational assessment. Clin Biomech. 2011;26:612–9.
130. Moonot P, Shang M, Railton GT, Field RE, Banks SA. In vivo weight-bearing kinematics
with medial rotation knee arthroplasty. Knee. 2010;17:33–7.
131. Fang C-H, Chang C-M, Lai Y-S, Chen W-C, Song D-Y, McClean CJ, Kao H-Y, Qu T-B, Cheng
C-K. Is the posterior cruciate ligament necessary for medial pivot knee prostheses with regard
to postoperative kinematics? Knee Surg Sports Traumatol Arthrosc. 2015;23:3375–82.
132. Cadambi A, Engh GA, Dwyer KA, Vinh TN. Osteolysis of the distal femur after total knee
arthroplasty. J Arthroplasty. 1994;9:579–94.
133. Huang CH, Hsu LI, Chang TK, Chuang TY, Shih SL, Lu YC, Chen CS, Huang CH. Stress
distribution of the patellofemoral joint in the anatomic V-shape and curved dome-shape fem-
oral component: a comparison of resurfaced and unresurfaced patellae. Knee Surg Sports
Traumatol Arthrosc. 2017;25:263–71.
134. Chang MA, Rand JA, Trousdale RT. Patellectomy after total knee arthroplasty. Clin Orthop
Rel Res (1976–2007). 2005;440:175–7.
135. Jan N, Fontaine C, Migaud H, Pasquier G, Valluy J, Saffarini M, Putman S. Patellofemoral
design enhancements reduce long-term complications of postero-stabilized total knee arthro-
plasty. Knee Surg Sports Traumatol Arthrosc. 2019;27:1241–50.
136. Huang CH, Liau JJ, Ho FY, Lin CY, Young TH, Cheng CK. Polyethylene failure of the
patellar component in New Jersey low-contact stress total knee arthroplasties. J Arthroplast.
2005;20:202–8.
137. Huang CH, Hsu LI, Lin KJ, Chang TK, Cheng CK, Lu YC, Chen CS, Huang CH. Patellofemoral
kinematics during deep knee flexion after total knee replacement: a computational simula-
tion. Knee Surg Sports Traumatol Arthrosc. 2014;22:3047–53.
Chapter 10
Biomechanics of Foot and Ankle
Duo Wai-Chi Wong, Ming Ni, Yan Wang, and Ming Zhang
Both surgical and conservative methods have been adopted to treat foot problems
and diseases. Most of the interventions targeted the treatment on the problem site
but the effect of the foot problems could extend beyond the site to adjacent regions
or the entire foot. Assessment and evaluation of the biomechanical environment and
the variations before and after the intervention could help intervention design, plan-
ning and thus minimizes complications.
Experimental methodologies, such as motion capture analysis, pedobarogra-
phy, and cadaveric experiments have been developed to quantify foot biomechan-
ics. However, the internal biomechanics of the foot, such as the stress distributions
within bones and soft tissues is not easy to measure by experiments. Computational
methods or platforms such as the finite element (FE) analysis are useful tools to
investigate biomechanics of the musculoskeletal structures. This evaluation
method has been used to study injury mechanism, improve prosthetic and orthotic
designs, and predict surgery outcome, in addition to promoting fundamental
understanding of foot biomechanics. In this chapter, four common foot problems
and interventions of the foot and ankle were analyzed by FE models including
hallux valgus, ankle arthroplasty, tarsometatarsal joint fusion, and calcaneal
fractures.
Summary
Hallux valgus (or hallux abducto valgus) has been one of the most common foot
problems and associated with functional disability. Clinical studies were conducted
that featured the pathoanatomy and pathomechanics of hallux valgus. However, the
effectiveness of current treatments remains unsatisfactory, while complications and
recurrence rates are undeniable. It is pragmatically demanding to further understand
the biomechanics of hallux valgus to improve intervention. This study constructed
a three-dimensional foot model of the subject and simulated with hypermobility.
Besides, partial models consisted of isolated first ray and were constructed and
compared among the normal participant and hallux valgus scenario. The model par-
ticipant was invited to participate in a gait analysis session to acquire the boundary
and loading condition for the simulation of walking stance. The finite element (FE)
analysis outcome suggested that the hypermobility case generated higher joint force
at the metatarsocuneiform and metatarsophalangeal joint. In the medial-lateral
direction, the metatarsocuneiform joint force switched sharply that could indicate a
potential risk of metatarsus primus varus.
10 Biomechanics of Foot and Ankle 221
1.1 Background
Hallux valgus (or hallux abducto valgus) is represented by a lateral and medial
deviation deformity of the hallux and the first metatarsal, respectively. The enlarged
and swollen medial projected eminence is also a characteristics of hallux valgus and
thus it is also known as the bunion. Hallux valgus imposes significant public health
burden as supported by the incident rate of related foot surgery. There could also be
secondary problems such as poor gait patterns and impaired coordination and stabil-
ity that may lead to falling risks to the elderly population [1–5].
The prevalence of hallux valgus was found strongly associated with family his-
tory, while gender is also suggested to be a contributing factor [6]. The shape of the
foot bones, arch height, and the level of hypermobility may be affected by heredity
[7]. In fact, the incidence of hallux valgus on women is more than two times higher
than that of men [8]. Similarly, the different sex presents different bone morphology
and alignment, in addition to the level of hypermobility or laxity [9–11]. Other
influencing factors included footwear, body weight, habit, and occupation [12].
In fact, hallux valgus ranks high in the list of common foot complaint. More than
one-quarter of adults in the United Kingdom suffered from the problem [8], while
nearly 15% of students reported hallux valgus as reported by Owoeye et al. [13].
The same study reported that 9% of the students with hallux valgus showed pain
and 14% demonstrated difficulty in prolong walking. Survey on the elderly popula-
tion reported even higher prevalence [14] which results in economic burden on
hospitalization and society. More than 50 k surgeries were taken to treat hallux
valgus which accounted for more than one-quarter of all forefoot surgeries [15].
More than 2 absent days are incurred by relevant surgery, resulting in more than
AUS$3800 productivity loss and AUS$3700 hospitalization cost in Australia in
2008 [16–19].
Despite that surgical correction for hallux valgus are not few, the outcome
remains unsatisfactory with failures, complications, and recurrence. The complica-
tion rates and recurrence rates can be up to 55% and 16%, respectively [20, 21],
contributed by over-correction, avascular necrosis, etc. [22]. Surgery using soft tis-
sue correction procedure produces even higher failure rates [22].
Biomechanical studies on hallux valgus and its intervention have been conducted to
better understand the pathogenesis of the disease and improve treatment outcomes.
The hypermobility of the first ray was first introduced by Morton and Lapidus which
attracted debates on the appropriate interventions [23, 24]. Arthrodesis procedure
was proposed aiming to address the problem of stability. The evaluation of stability
was also included in other studies on the outcome of osteotomy of hallux valgus
222 D. W.-C. Wong et al.
[25]. Currently, mobility and stability are assessed by manual dorsal excursion [26],
load-bearing radiographs [12], or custom-made mechanical devices [27]. Yet the
quantification of hypermobility has been believed to be subjective and confined to
static measurement [23].
Foot types and deformities are often classified using plantar pressure measure-
ment [28]. Hallux valgus was found to impair the loading at the hallux region. The
peak pressure of the forefoot was found shifting laterally [29], as evidenced by the
heightened peak pressure and pressure time-integral at the central forefoot [30].
Conversely, some study discovered that the plantar pressure pattern moved medially
[31, 32]. There could be other factors, including secondary deformities, that may
influence the pressure pattern on the medial metatarsal region [33]. Hallux valgus
patients shall have a persistent reduction in hallux loading compensated by the cen-
tral metatarsal area [33].
Pedobarographic assessment has been used as a biomechanical tool to evaluate
surgical interventions of hallux valgus. By expanding the contact area under hal-
lux, Mittal et al. [34] suggested that McBride procedure could restore some of the
hallux functions. Another operation carried out by Saro et al. [35] suggested that
the operation did not produce positive biomechanical effect as indicated by the
significant reduction in peak pressure under the hallux and heel region. Some stud-
ies compared different osteotomy procedures (Mitchell and Scarf) using plantar
pressure distribution [36].
Since consensus was not reached on the interpretation of plantar pressure distribu-
tion towards clinical outcome of the surgeries, researchers endeavored to investigate
how hallux valgus attenuates the biomechanical environment of the foot by means of
manual examination and further plantar pressure studies. This approach has been
achieved by the finite element (FE) simulations that could investigate the internal
stress or strain pattern as well as the load transfer characteristics noninvasively [37].
A major extrinsic factor of hallux valgus, the wearing of high-heeled shoes, was
considered in the study of Yu et al. [38] using FE model. In addition, a simplified first
ray model was used to understand the linkage between first ray hypermobility and
hallux valgus [39]. Meanwhile, we have constructed a FE model to imitate hallux
valgus and simulated the walking stance. Five instants were extracted from the simu-
lation findings and the outcome on stress, strain, and joint loading were used to
evaluate the changes of kinematics and stability induced by hallux valgus.
The model geometry of the normal foot and hallux valgus foot (asymptomatic) was
reconstructed from magnetic resonance (MR) images of two female subjects. They
were both aged 28, 165 cm tall, while the body weights were 54 kg and 56 kg,
respectively, for the normal and hallux valgus foot participants. Both participants
10 Biomechanics of Foot and Ankle 223
1.3.2 Material Properties
The elastic modulus of the bone was 7300 MPa with the Poisson’s ratio of 0.3
[40], and was assumed isotropic and homogeneous. The material of the encapsu-
lated soft tissue was hyperelastic with data experimented by Lemmon et al. [41].
The supporting ground was bilayers. The bottom layer was rigid, while the upper
layer was deformable (40 GPa), representing concrete ground. The elastic modu-
lus of all ligaments was 264.8 MPa, which was the reported average elasticity of
rearfoot ligaments [42]. The thickness of ligaments was 1.5 mm as reported by
Milz et al. [43].
Ground reaction force (GRF) and tibial inclination from the gait experiment of the
model subject was input as the loading and boundary conditions to simulate stance
phase in five consecutive static steps named: heel strike (0% stance), GRF first peak
(27% stance), GRF valley, (45% stance), initial push-off (60%), and GF second
peak (75%). The magnitude of muscle forces was adopted from literature [44–46]
based on the level of muscle activation and maximum capacity.
Frictionless contact was assigned at all joint facets. The coefficient of friction
between the ground and tissue contact was 0.6 [47]. The constructed geometrical
of the bony components were illustrated in Fig. 10.1, demonstrating a predicted
metatarsophalangeal (MTP) dorsiflexion angle of 27.3° at 75% stance. The axial
direction was defined as the longitudinal axis along the tibia and fibula segment.
Both the direction of the metatarsocuneiform (MC) joint force and the MTP joint
force were defined as acting from the proximal side to the distal side.
1.4 Study Design
The study was divided into two parts. The first part involved with the reconstruction
of the entire foot and ankle model for the normal subject while comparing to that of
the hypermobile foot. By weakening the deep transverse metatarsal ligament
(DTML) between the first and second metatarsals, the hypermobility of the first ray
224 D. W.-C. Wong et al.
Axial Direction
AP Direction
Resultant
MC Joint Force
Resultant
MTP Joint Force
Fig. 10.1 Illustration of the joint forces and defined axes directions. (Reprinted from Medical
Engineering & Physics, 36, Wong et al., Biomechanics of first ray hypermobility: An investigation
on the joint force during walking using finite element analysis, 1388–1393, Copyright (2014), with
permission from Elsevier)
was resembled on the normal foot [48]. The MC and MTP joint forces were output
from the simulation. The second part of the study involved with the building of
partial foot model (the first ray) for the normal and hallux valgus participant. The
bone alignment and displacement were extracted for analysis.
1.5.1 Model Validation
Model validation was performed by comparing the measured and predicted plantar
pressure. The participant was asked to walk at their self-comfortable speed. An in-
sole plantar pressure measurement system (F-Scan, Tekscan, USA) was utilized to
assess the plantar pressure distribution. Figure 10.2 shows the comparison of pre-
diction and measurement at the instant of GRF first peak and second peak. During
GRF first peak, the predicted maximum peak pressure was 0.49 MPa, locating at the
heel while the measurement value was 0.46 MPa. On the other hand, the plantar
pressure was concentrated at the hallux, toes, and metatarsals at GRF second peak.
The predicted and measured maximum peak pressures at the hallux region were
0.52 MPa and 0.46 MPa, respectively, which was generally agreeable.
10 Biomechanics of Foot and Ankle 225
Heelstrike GRF First Peak GRF Valley Initial Push-off GRF Second
% Stance
Peak
0.00 0.04 0.07 0.11 0.14 0.18 0.22 0.25 0.29 0.33 0.36 0.40 0.43 MPa
Fig. 10.2 Comparison of FE predicted and experimental result on plantar pressure distribution.
(Reprinted from Medical Engineering & Physics, 36, Wong et al., Biomechanics of first ray hyper-
mobility: An investigation on the joint force during walking using finite element analysis,
1388–1393, Copyright (2014), with permission from Elsevier)
The MC joint forces of the normal and the hypermobile foot in AP, axial, and medio-
lateral (ML) directions during stance are shown in Fig. 10.3. Before the instant of
GRF first peak, joint forces in all directions were quite small, but increased
apparently after the instant of GRF valley. On the other hand, the force in both axial
and AP directions increased consistently until terminal stance, while that in medial-
lateral direction switched laterally during late stance.
The deviation of MC joint force became prominent during late stance. There
were about one-tenth leverage in the medial direction and one-fifth leverage in the
axial and AP direction joint forces upon hypermobility during initial push-off. The
increase was more than 25% at GRF second-peak instant in axial and AP directions,
and skyrocketed to 200% in the medial direction.
226 D. W.-C. Wong et al.
–15 –50
% Stance % Stance
0 10
0 20 40 60 80
Normal
–50 Normal 5
Hypermobile
Hypermobile 0
–100 0 20 40 60 80
–5
–150
–10
–200 –15
–250 –20
% Stance % Stance
500 250
Normal Normal
400 Hypermobile 200
Hypermobile
300 150
200 100
100 50
0 0
0 20 40 60 80 0 20 40 60 80
–50
Fig. 10.3 Metatarsocuneiform (MC) and metatarsophalangeal (MTP) joint force comparing nor-
mal foot and foot with hypermobile first ray. ML direction (+ Medial), axial direction (+ Inferior),
AP direction (+ Anterior). The dotted lines were interpolated from the results of the five instants
for illustration. (Reprinted from Medical Engineering & Physics, 36, Wong et al., Biomechanics of
first ray hypermobility: An investigation on the joint force during walking using finite element
analysis, 1388–1393, Copyright (2014), with permission from Elsevier)
The MTP joint force demonstrated comparable trend as that of the MC joint
force. Joint forces along the ML and AP directions advanced with time (percentage
stance phase) and the deviation became obvious after the GRF valley instant.
Notwithstanding, the joint force in the axial direction turned from superior to infe-
rior at 75% stance. In fact, the MTP joint force differences among the normal and
10 Biomechanics of Foot and Ankle 227
hypermobile foot were generally less than that of the MC joint force, while the larg-
est difference between the normal and hypermobile foot occurred in the axial
direction.
MTP and MC joint force increase with hypermobility which could be due to the
reduction in shock attenuation capacity of the arch. To maintain the stability and
integrity of the foot, the load on the first ray may be increased. However, joint prob-
lems could be occurred with the burden of heavy joint force. The sharp change of
the MC joint force direction could result in joint instability and may consequently
lead to the malalignment of the first metatarsal. Since the attenuation of joint force
primarily happened at the MC joint rather than the MTP joint, it was suggested that
hallux abducto valgus deformity could be secondary to the metatarsus primus varus.
The intermetatarsal angle (IMA) and the hallux valgus angle (HVA) were used to
quantify bone alignment. The former parameter represents the angulation between
the first and second metatarsal, while the latter was calculated by the intersection of
the first metatarsal and phalanx axes. To estimate the alignment angles, we esti-
mated the longitudinal axis of the bone shaft by means of regression algorithm and
assumed that the bones were cylindrical. For normal condition, HVA and IMA were
9.1° and 15.0°, respectively. However, imposing a hallux valgus condition elevated
these parameters to 10.4° and 25.7°, respectively. The angles advanced with increas-
ing forefoot loading. Hallux valgus patients demonstrated higher deterioration of
IMA, which was 20% higher than that of normal foot. The finding also supported
the fact that hallux abducto valgus deformity could be secondary to the metatarsus
primus varus.
1.6 Conclusion
The outcome on MC and MTP joint force provides insights on the development of
hallux valgus upon the laxity effect constituted by first ray hypermobility. The
increased joint loading may contribute to the risk of joint problems, whereas the
abrupt change on the loading direction at the MC joint may indicate potential devel-
opment of metatarsus primus varus.
Summary
Ankle arthrodesis is considered as the standard treatment for some ankle problems,
including degenerative deformity, traumatic arthritis. However, many complications
228 D. W.-C. Wong et al.
2.1 Background
Ankle arthritis is a common foot and ankle problem, while osteoarthritis (OA) sec-
ondary to fracture and trauma was highly prevalent. Survey indicated that about
15% of the people was affected by the disabling conditions and pain ascribed to OA
and approximately 6% have OA of the ankle [49]. Another survey in the United
Kingdom estimated that the number of ankle OA patients requiring hospitalization
or clinical care was 47.7 per 100,000 [50].
Ankle arthritis is a prevalent disabling condition that impair one’s physical func-
tion and quality of living. Glazebrook et al. [51] reported that patients with end-
stage ankle arthritis scored significantly lower in the evaluation of function and
quality of living. Further, the consequence with respect to mental and physical dis-
ability could be as severe as that of the end-stage hip arthrosis [52]. Ankle arthrod-
esis, firstly performed in 1879, was considered as the salvage procedure for end-stage
ankle arthritis. Despite significant relief of pain and retaining plantigrade foot func-
tion, ankle arthrodesis is known to cause gait alterations as well as arthritic changes
in the subtalar and other distal foot joints [53–56]. Existing retrospective clinical
studies revealed that there was a high incidence rate of ipsilateral hind- and mid-foot
arthritis associated with the deterioration [56, 57].
2.3 Computational Analysis
2.3.1 Geometry Construction
A normal adult (height 164 cm and body weight 54 kg) was recruited in the study.
The MR images of her right foot were extracted under a setting of 0.625 mm pixel
resolution and 2 mm slice interval. The three-dimensional FE model was recon-
structed featuring the foot-and-ankle complex with 28 bony components, encapsu-
lated soft tissue, 103 ligaments, plantar fascia, and major muscles [63, 64]
(Fig. 10.4). The reconstruction process was carried out in the segmentation soft-
ware, Mimics (Materialise, Leuven, Belgium), and meshed in the finite element
package, Abaqus (Dassault Systèms Simulia Corp., Providence, RI, USA).
We resembled the ligaments and plantar fascia using wires that connected the
insertions points on the bony structures and meshed them with tension-only truss
elements. Nine major muscle groups were assumed by axial connectors. The bilayer
ground plate consisted of an upper layer with high elastic modulus and a rigid lower
layer. The coefficient of friction between the foot plantar surface and the ground
support surface was set to 0.6 [47].
2.3.2 Material Property
Most of the components were assumed to be isotropic, linearly elastic, and homo-
geneous, including the osseous structure and soft tissue, and the Young’s modulus
and Poisson’s ratio of these components were adopted from existing literature
[42, 65–67]. The bulk soft tissue and skin were assigned as hyperelastic material
with nonlinear properties. Most of the three-dimensional components were
meshed as linear tetrahedral elements, while that of the plantar fascia and liga-
ments were meshed as truss. The mesh and material properties are shown in
Table 10.1.
230 D. W.-C. Wong et al.
Fixed surfaces
Achilles
Tendon
a
Muscle Forces
x
y
Fascia
Plantar z
f=0.6
ctio n
d Rea
Groun rces
Fo
a
Fig. 10.4 The three-dimensional finite element model of the foot and ankle and application of
boundary and loading conditions. (Reprinted from PlosOne, Wang et al., Effects of Ankle
Arthrodesis on Biomechanical Performance of the Entire Foot, e0134340 (2015), under CC-BY)
Table 10.1 Material property and mesh element type for the foot model components
Element Young’s modulus E Poisson’s Cross-section area
Component type (MPa) ratio v (mm2)
Bone C3D4 7300 0.3 –
Encapsulated soft C3D4 and Hyperelastic – –
tissue S3
Cartilage C3D4 1 0.4
Ligaments T3D2 260 – 18.4
Plantar fascia T3D2 350 – 58.6
Ground C3D8R 17,000 0.1 –
The model participant was invited to conduct a gait analysis using the motion cap-
ture system (Vicon, Oxford Metrics, Oxford, UK) to obtain the boundary and load-
ing conditions for the input of the FE analysis. We targeted on seven body segments
with 16 retroreflective markers attached on the lower extremity of the participant.
The participant stood on the force platform (AMTI, Watertown, MA, USA) to con-
duct the static calibration for the motion capture system. The orientation of the
shank was defined by the three-dimensional angulation between the shank segment
and the global system, denoted as α in Fig. 10.4 with the time series in Fig. 10.5.
The force plate would detect the ground reaction force in the vertical, anterior-
posterior, and medial-lateral directions. In Fig. 10.5, we extracted the first-peak,
midstance, and second-peak instants of the ground reaction force curve in the verti-
cal direction for further analysis.
10 Biomechanics of Foot and Ankle 231
The proximal side of the FE model was fixed, including the superior surfaces
of the tibial, fibula, and the encapsulated soft tissue (Fig. 10.4). The ground reac-
tion force was applied on the rigid ground plate to mimic the setting of the force
platform. The foot-shank angle was applied through the rotational degree of free-
dom of the ground plate whereas the translational degree of freedom was not fixed.
The muscle forces were approximated by the cross-sectional area, the normalized
electromyography, and the muscle gain of the muscles during walking stance [46,
68, 69], and later adjusted based on another study [45].
450
Force (N)
350
250
150
50
–50 0 10 20 30 40 50 60 70 80 90 100
–150
80 Ground-Shank Angle
60
Angle (º)
40
20
0
0
–20 10 20 30 40 50 60 70 80 90 100
% Stance Phase
Fig. 10.5 Ground reaction forces and ground-shank angle recorded in the gait analysis and the
three instants, first peak, midstance, and second peak, for simulation. The three instants were
marked in the curves. (Reprinted from PlosOne, Wang et al., Effects of Ankle Arthrodesis on
Biomechanical Performance of the Entire Foot, e0134340 (2015), under CC-BY)
232 D. W.-C. Wong et al.
2.4.1 Model Validation
Plantar pressure during balanced standing and walking stance was assessed and
compared between the experiments and predictions from the FE model. Model
validation was completed by comparing these findings. As shown in Fig. 10.6, the
pressure pattern was generally agreeable with the peak pressure located at the
heel region.
2.4.2 Plantar Pressure
Figure 10.7 shows the plantar pressure distribution of the intact foot and the foot
after arthrodesis at the three stance instants (first peak, midstance, and second peak).
At the first-peak instant, there was a slight difference in peak plantar pressure
between the conditions, which was 0.33 MPa and 0.36 MPa for the intact foot and
arthrodesis foot, respectively. The peak plantar pressures for the intact foot between
the midstance and second-peak instant were both 0.68 MPa. However, that of the
arthrodesis foot was higher and increased chronologically. The peak plantar pres-
sure was 0.78 MPa and 0.93 MPa, respectively, for the midstance and second-peak
condition for the arthrodesis foot. Compared to the intact condition, the center of
pressure after arthrodesis has shifted slightly towards the medial side and appeared
beneath the third metatarsal bone.
Figure 10.8 summarizes the maximum contact pressure of the articular joints
between the intact and arthrodesis condition at the first-peak, midstance, and
second-peak instants. There were observable increases of maximum pressure at the
talonavicular and intertarsal joints after the ankle arthrodesis procedure. In consid-
eration of the average of the maximum contact pressure across the time instants, the
talonavicular joint remained highest and was increased from 0.80 to 1.21 MPa; 1.14
to 1.59 MPa; and 2.00 to 2.14 MPa after surgery at the three instants.
The medial cuneonavicular joints carried higher maximum contact pressure
compared with the other cuneonavicular joints. The maximum contact pressure of
the joint increased from 0.60 to 0.79 MPa; from 0.97 to 1.20 MPa; and from 1.90 to
1.97 MPa after ankle arthrodesis, respectively, for the first-peak, midstance, and
second-peak instants. However, the highest magnitude of the change due to the
surgical procedure was evident at the lateral cuneonavicular joint. The percentage
changes were 80.3%, 64.7%, and 11.6%, respectively, at the first-peak, midstance,
and second-peak instants.
Distally, considering the tarsometatarsal joints, the second tarsometatarsal
joint sustained the highest maximum contact pressure compared with the other
10 Biomechanics of Foot and Ankle 233
a
CPRESS (MPa)
(KPa)
0.191
0.150 > = 150
0.138
0.125 138
0.113 225mm2 232mm2 125
0.100 0.051MPa
0.088 0.058MPa 113
0.075 100
0.063 88
0.050
0.038 75
0.025 63
0.013
0.000 50
38
25
13
>=0
150mm2 155mm2
0.168MPa 0.157MPa
b CPRESS (MPa)
(KPa)
0.332
0.300 > = 300
0.275
0.250 275
0.225
0.200 250
0.175 225
0.150
0.125 200
0.100 175
0.075
0.050 150
0.025 125
0.000
100
75
50
25
>=0
150mm2 155mm2
0.300MPa 0.307MPa
Fig. 10.6 Comparison of the plantar pressure between computational prediction and experimental
measurement in (a) balanced standing position, (b) the first-peak instant, and (c) the second-peak
instant for validation. (Reprinted from PlosOne, Wang et al., Effects of Ankle Arthrodesis on
Biomechanical Performance of the Entire Foot, e0134340 (2015), under CC-BY)
234 D. W.-C. Wong et al.
0.125 0.150
0.100 0.125
0.075 15 mm 0.100
0.050 0.075
0.025 0.050
0.000 0.025
0.000
Max: 0.332
CPRESS (MPa)
CPRESS (MPa)
0.683 0.779
0.300 Max: 0.683 0.300
0.275 0.275
0.250 16 mm 0.250
0.225 0.225
0.200 0.200
0.175 0.175
0.150
Mid-stance
0.125 0.150
0.100 0.125
0.075 0.100
Max: 0.779 0.075
0.050
0.025 0.050
0.000 0.025
0.000
0.150 0.150
0.125 0.930 0.125
0.100
0.100
0.075 Max 0.075
0.050
0.025 0.683 0.050
0.000 0.025
0.000
Fig. 10.7 Comparison of the plantar pressure distribution between normal foot model and ankle
arthrodesis foot model at the three instants. (Reprinted from PlosOne, Wang et al., Effects of Ankle
Arthrodesis on Biomechanical Performance of the Entire Foot, e0134340 (2015), under CC-BY)
10
1.5
1.0
Biomechanics of Foot and Ankle
0.5
First Peak
First Peak
First Peak
First Peak
First Peak
First Peak
First Peak
First Peak
First Peak
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Fig. 10.8 Comparison of the contact pressure at nine joints in the hind- and mid-foot between the normal foot model and the ankle arthrodesis foot model at
the first-peak, midstance, and second-peak instants. (Reprinted from PlosOne, Wang et al., Effects of Ankle Arthrodesis on Biomechanical Performance of the
Entire Foot, e0134340 (2015), under CC-BY)
235
236 D. W.-C. Wong et al.
tarsometatarsal joints. The maximum contact pressure was increased from 0.60 to
0.90 MPa; from 0.91 to 1.19 MPa; and from 1.72 to 1.88 MPa after the ankle
arthrodesis procedure, respectively, for the three stance instants. In fact, the surgi-
cal procedure imposed the largest influence of the third tarsometatarsal joint
accounting for the percentage changes of 62.8%, 51.4%, and 2.0% at the three
stance instants. The fourth and fifth tarsometatarsal joints did not demonstrate
consistent trend throughout the stance instants. The former showed a maximum
contact pressure reduction after surgery at the first peak and midstance, whereas
the latter presented a reduction at the midstance and second-peak instant.
Ankle arthrodesis reduced the maximum contact pressure of the subtalar and cal-
caneocuboid joints. The maximum contact pressure of the calcaneocuboid joint was
decreased from 0.32 to 0.24 MPa; from 0.48 to 0.39 MPa; and from 0.51 to 0.49 MPa
after surgery at the three stance instants, respectively. Furthermore, the maximum
contact pressure of the subtalar joint was decreased from 0.39 to 0.37 MPa; from
0.54 to 0.51 MPa; and from 0.78 to 0.70 MPa after surgery, respectively, at the three
instants. The reduction of maximum contact pressure of the hindfoot joints may sug-
gest that subtalar arthritis may not be a consequence of ankle arthrodesis. We specu-
lated that the progression could be attributed to the pre-existing degenerative changes.
Figure 10.9 compares the joint contact forces among the intact and surgery condi-
tions at the selected instants of walking stance. The contact force elevated chrono-
logically (first-peak, midstance, and then second-peak instants) for the talonavicular,
cuneonavicular, and the first and second tarsometatarsal joints. Apparent change
appeared at the first-peak instance whereas most of the joint presented their highest
value at the second-peak instant.
The talonavicular joint acted as the highest load-bearing bone in the foot and
ankle complex. The joint forces were 181 N, 259 N, and 514 N, respectively, at the
first-peak, midstance, and second-peak instants. The forces were increased more
than half for the first peak and midstance and reached to 578 N at the second peak
which was slightly higher than the applied vertical ground reaction force (540 N).
Conversely, the load transfer towards the calcaneocuboid declined after arthrodesis
by 31%, 17%, and 8%, and reached to 33 N, 55 N, and 74 N, respectively, for the
three instants of walking stance.
In general, the first ray sustained a higher contact force compared to that of the
second and third ray. They experienced greater contact force at the second-peak
instants. The medial cuneonavicular and first tarsometatarsal joints of the first ray
increased by 31% and 75%, respectively, at the first-peak instant, while the interme-
diate cuneonavicular and second tarsometatarsal joints of the second ray increased
by 52% and 79%, in addition to the 74% and 71% increase at the lateral cuneona-
vicular and third tarsometatarsal joints of the third ray.
Figure 10.10 demonstrates the differences in load transfer between the intact and
arthrodesis model at the first-peak instant, which was more prominent compared to
that of the other two instants. Approximately one-third of the body weight was
10
700
600 Normal
Comparison of Joint Contact Force
500 Arthrodesis
400
300
200
Biomechanics of Foot and Ankle
100
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Midstance
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Second Peak
Talonavicular Calcaneoc- Medial Intermediate Lateral First Second Third Forth Fifth
uboid Cuneonavi- Cuneonavi- Cuneonavi- Tarsometat- Tarsometat- Tarsometat- Tarsometat- Tarsometat-
cular cular cular arsal arsal arsal arsal arsal
Fig. 10.9 Comparison of the contact forces at ten joints in the hind- and mid-foot between the normal foot model and ankle arthrodesis model at the first-peak,
midstance, and second-peak instants. (Reprinted from PlosOne, Wang et al., Effects of Ankle Arthrodesis on Biomechanical Performance of the Entire Foot,
e0134340 (2015), under CC-BY)
237
238 D. W.-C. Wong et al.
0.07 0.06
0.11
0.0
9
0.34
0.58
0.23 0.33
Fig. 10.10 Load transfer (times of body weight) in the normal and ankle arthrodesis foot model at
the first-peak instant. (Reprinted from PlosOne, Wang et al., Effects of Ankle Arthrodesis on
Biomechanical Performance of the Entire Foot, e0134340 (2015), under CC-BY)
transferred through the talonavicular joint to the medial joints while less than 10%
of the body weight was transmitted through the calcaneocuboid joint to the fourth
and fifth rays. From mid-foot to forefoot, 23% of the body weight was conveyed
medially and 11% laterally. The ankle arthrodesis imposed additional burden on
load transferred which was increased to 58% at the talonavicular joint and reduced
that of the calcaneocuboid joint. Similarly, the midfoot-to-forefoot transfer increased
to 34% in the medial three rays and decreased to 7% in the lateral two rays. The
change in load transfer pattern may induce risk of foot pain the medial columns
which may constitute to foot deformity in long term. Orthotic intervention such as
wedge insole may help revise the load transfer pattern after surgery.
2.4.5 Bone Stress
As shown in Fig. 10.11, ankle arthrodesis increased the von Mises stress of the bone
in general. At the first-peak instant, the von Mises stress increased apparently by
19% and 52%, respectively, in the first and third metatarsal bone, while the lateral
metatarsal only processed small slight changes. However, all the metatarsal showed
an obvious elevation in stress at the midstance and second-peak instants. The sec-
ond metatarsal represented the part with the highest stress which was 42 MPa and
52 MPa, respectively, for the normal and arthrodesis condition. The third metatarsal
comes after the second metatarsal. The stress was 20 MPa at midstance and increased
to 34 MPa at the second-peak instant.
As shown in Fig. 10.12, the predicted results showed different joint position and foot
deformation between the intact and surgery conditions, particularly at the second-
peak instant. The foot-shank angle for both conditions were 30°. However, the ori-
entation angle was 28° for the intact foot and 44° for the arthrodesis foot, respectively,
between the horizontal plane and the first ray axis at the second-peak instant.
10 Biomechanics of Foot and Ankle 239
15.010 16.675
13.346 15.012
11.683 13.350
10.019 11.687
8.356 10.025
6.692 8.362
5.029 6.700
3.365 5.037
1.702 3.375
0.039 1.712
0.050
S, Mises S, Mises
(Avg: 75%) (Avg: 75%)
25.505 31.439
20.000 20.000
18.340 18.340
16.679 16.679
Mid-stance
15.019 15.019
13.358 13.359
11.698 11.698
10.037 10.038
8.377 8.377
6.716 6.717
5.056 5.057
3.395 3.396
1.735 1.736
0.074 0.076
S, Mises S, Mises
(Avg: 75%) (Avg: 75%)
42.133 51.911
20.000 20.000
18.341 18.344
Second Peak
16.682 16.689
15.023 15.033
13.364 13.378
11.706 11.722
10.047 10.067
8.388 8.411
6.729 6.756
5.070 5.100
3.411 3.445
1.752 1.789
0.093 0.134
Fig. 10.11 Comparison of von Mises stress in five metatarsal bones in normal foot model and ankle
arthrodesis foot model at three instants. (Reprinted from PlosOne, Wang et al., Effects of Ankle
Arthrodesis on Biomechanical Performance of the Entire Foot, e0134340 (2015), under CC-BY)
2.4.7 Limitations
The FE model in this study inherited some simplifications and assumptions. The
ankle arthrodesis was realized by constraining the contact at the ankle articulation
rather than reconstructing and installing the models of the screws or pins. It would
be better to reconstruct the screws to examine its behavior and influences. Besides,
the suggested Young’s modulus and Poisson’s ratio was not relatively accurate [40].
It was derived from a weighted average elasticity of the cortical and trabecular
based on their volumetric contribution [66]. Further model refinement shall be made
to advance a more accurate model or material representation.
240 D. W.-C. Wong et al.
30 º
30 º
Y X
X Y
GRFx 28 º 44 º
GRFx
Fig. 10.12 Angular positions of normal foot and ankle arthrodesis foot at a second-peak instant in
the sagittal plane. (Reprinted from PlosOne, Wang et al., Effects of Ankle Arthrodesis on
Biomechanical Performance of the Entire Foot, e0134340 (2015), under CC-BY)
2.5 Conclusions
Ankle arthrodesis is a common surgical procedure that aims at pain relief and func-
tion improvement for patients with ankle degeneration. However, the fusion and
constraints on the ankle joint, which is the major foot joint, corrupt the normal
biomechanics of both the ankle joint and the entire foot, leading to potential postop-
erative complications.
The biomechanical consequences of ankle arthrodesis are indicated by the
differences in biomechanical parameters, including plantar pressure, contact
10 Biomechanics of Foot and Ankle 241
force, stresses, and deformation, which could explain the mechanism of some
clinical findings. The high load on the medial rays may result in pain and dete-
riorate the arthritic condition, while the large stress on the second and third
metatarsals could impose risk of bone fractures. There was a decrease in subtalar
joint loading after ankle arthrodesis such that we believe the postoperative
arthritis of the subtalar joint may not be a consequence of the ankle arthrodesis
procedure.
The model used in this study was regarded as a representation of the normal
physique and may be regarded as a baseline reference from a biomechanical point
perspective. Foot in valgus position may lateralize the loading. In addition,
orthotic intervention may help to relieve the negative effects of ankle arthrodesis.
Further evaluations are required to decide the effectiveness of these
interventions.
Summary
The Lisfranc joint complex is also named as the tarsometatarsal joint (TMT) and
is recognized as a crucial connection that facilitates stability of the midfoot and
forefoot. The joint comprises sophisticated osseous and capsuloligamentous
structure such that the injury of the TMT joint may lead to disabling osteoarthritis
(OA) requiring surgery. Acknowledging the effectiveness of surgical intervention
and minimizing risk of complication are imperative in surgical design. Determining
the internal biomechanical environment, such as plantar pressure distribution,
stress of the bone, and deformation of soft tissue, attenuated by surgical interven-
tion facilitates evaluation. In this study, we aimed to evaluate the influence of
TMT joint fusion biomechanically using a finite element model of the foot and
ankle complex, which consisted of 28 bony structures, 72 ligaments, plantar pres-
sure, and an encapsulated bulk tissue. Our finite element prediction suggested that
TMT joint fusion elevated the plantar pressure by 0.42, 19, and 37% in three
featured time instants compared to the intact foot. In particular, the load transfer
across the cuneonavicular and the fifth cuboideometatarsal joints were increased
by 27% and 40%, respectively, at the second-peak instant that may impose risk of
joint arthritis. On the other hand, the maximum stress of the second metatarsal
bone was increased by more than 20% that may lead to vulnerability to stress
fracture.
3.1 Background
The tarsometatarsal (TMT) joint, or the Lisfranc joint, is named after a French sur-
geon, Jacques Lisfranc (1790–1847), who served in the Napoleon’s [70]. The TMT
joint plays an important role in bridging the midfoot and the forefoot. In addition,
242 D. W.-C. Wong et al.
the arrangement of the five metatarsals on the distal side and the four midfoot bones
(three cuneiforms and cuboid) facilitates structural support for the transverse arch
along with the attaching ligamentous structures. The TMT joint is often divided into
three columns along the anterior-posterior direction by the articulation of the sec-
ond and third TMT joints. The TMT joint can have little sagittal plane motion with
a range of 0.6 mm, 3.5 mm, and 13 mm, respectively, for the central, medial, and
lateral columns [70, 71].
In the United States, fractures of the TMT joint accounted for 0.2% of all frac-
tures, in which there was one fracture in every 55,000 persons each year [72]. The
treatment to TMT joint injuries, however, remains no consensus. Nonoperative
intervention could be indicated for nondisplaced fracture. Surgical methods using
either closed or open reduction would be used to treat displaced fracture assisted
with different implants, such as Kirschner wires, screws, plates, or suture-button
devices. Correct reduction and alignment with sufficient stability is the key to satis-
factory clinical outcome [73].
While joint fusion procedure aims to restore some normal foot functions, it
may produce some undesirable biomechanical effects. The constraints on the
joint may induce additional stress onto the bony structures leading to irritation,
loosening of implants, and breakage. A mid-term follow-up on TMT joint fixa-
tion patients reported that more than 10% of them developed posttraumatic
arthritis and nearly 3% developed subluxation [74]. In addition, approximately
7% of them developed flatfoot or severe symptoms that required a secondary
surgery [74].
that offer additional insights to the problem. In this study, we simulated a TMT
joint fusion procedure on a comprehensive 3D FE foot and ankle model to evalu-
ate the biomechanical performance of the intervention.
The FE model was reconstructed from the right foot of a female (54 kg, 1.64 m).
The model consisted of a volume of encapsulated soft tissue embracing 28 body
segments and other soft tissues [63, 64]. The first and second tarsometatarsal joint
fusion procedure was realized in the simulation platform [76]. Clinically, the
affected joint is linked by screws whereas the process was simplified in the simula-
tion by fusing the articular contact interface among the medial and intermediate
cuneiform, and the first and second metatarsal bones (Fig. 10.13).
a b
Fig. 10.13 Surgery of first and second tarsometatarsal joint fusion (a), and four tied bones in the
model for simulation (b). Articular surfaces among the first and second metatarsal bones and
medial and intermediate cuneiforms were tied together to simulate the fixed joints. (Reprinted from
Medical Engineering & Physics, 36, Wang et al., Biomechanical study of tarsometatarsal joint
fusion using finite element analysis, 1394–1400, Copyright (2014), with permission from Elsevier)
244 D. W.-C. Wong et al.
The model subject was recruited to perform locomotion analysis using the motion
capture system, as well as the force platform and electromyography. Reflective mark-
ers were attached to the interested body segments whereas the kinematic and kinetic
information during gait were extracted. As shown in Fig. 10.14, the GRFs with time
were obtained featuring the first-peak, midstance, and the second-peak instants.
The first peak of the vertical GRF happened at 25% stance phase while that of the
second peak occurred at 70%. The midstance was defined as the valley of the curve
and occurred between the first- and second-peak instants. Extrinsic muscle forces
were estimated and applied according to the linear EMG-force assumption, mea-
sured EMG and suggested cross-sectional area of the muscles [68, 69]. The input of
the boundary and loading conditions in the four simulated time instants are sum-
marized in Table 10.2.
The muscles in the simulation were modelled as connectors jointing the relevant
attachment points. Tendon Achilles were assumed by five axial connectors and
forces were applied onto these connectors equally. Figure 10.15 illustrates the other
Table 10.2 Boundary conditions of the four simulated instants: balanced standing, first peak,
midstance, and the second peak
Loadings Balanced standing First peak Midstance Second peak
GRF (N) 270 578.6 519.3 600
Ankle-shank angle (rad) 0 0.113 0.216 0.485
Tibialis anterior (N) – 0 0 0
Tibialis posterior (N) – 34 42.5 0
Achilles tendon (N) 135 500 900 1100
Extensor digitorum longus (N) 0 0 0 0
Flexor digitorum longus (N) 0 40 20 96
Flexor hallucis longus (N) 0 30 0 284
Peroneus longus (N) 0 0 41.25 0
Peroneus brevis (N) 0 20 22 91.8
10 Biomechanics of Foot and Ankle 245
Fixed Surfaces
Achilles Tendon
Force
Muscle Forces
Z
GRF
Y
Fig. 10.15 Boundary and loading conditions for the simulation of gait instants. The superior sur-
faces of soft tissue, tibia and talus bones were fixed. Ground reaction forces of anteroposterior and
vertical directions were applied. Muscle forces were applied to muscle representatives. (Reprinted
from Medical Engineering & Physics, 36, Wang et al., Biomechanical study of tarsometatarsal joint
fusion using finite element analysis, 1394–1400, Copyright (2014), with permission from Elsevier)
muscle construction represented by the red dotted lines. The proximal end of the
encapsulated soft tissue, tibia, and fibula were fixed along with GRFs applied beneath
the rigid ground plate. The ground plate was also rotated according to the foot-shank
orientation profile sourced from the locomotion analysis. Besides, balanced walking
was also mimicked with half body weight (270 N). It was also assumed that only
Achilles tendon was activated and other muscle forces were negligible.
Plantar pressure distribution, contact pressure of the articular joint at the midfoot and
hindfoot, as well as the maximum von Mises stress of the metatarsal bones would be
compared between the intact and the surgical conditions. In addition, model validation
was carried out by comparing the plantar pressure and joint contact pressure between
FE prediction and physical experiment which demonstrated a reasonable agreement.
Figure 10.16 demonstrated that the plantar pressure distribution between the intact
and surgical conditions were similar but the surgical model showed a larger peak
pressure value. The peak plantar pressure was increased by 0.42%, 19%, and 37%
after surgery, respectively, at the first-peak, midstance, and second-peak instants,
which was more prominent during later stance. The corresponding peak pressure
values from intact to surgical conditions were: from 0.50 to 0.51 MPa; from 0.60 to
0.72 MPa; and from 0.64 to 0.88 MPa.
246 D. W.-C. Wong et al.
Fig. 10.16 Plantar pressure distributions in the normal and fused tarsometatarsal joint models in
three instants. (Reprinted from Medical Engineering & Physics, 36, Wang et al., Biomechanical
study of tarsometatarsal joint fusion using finite element analysis, 1394–1400, Copyright (2014),
with permission from Elsevier)
Arch height stiffness and contact area are also determinants to contact pressure.
The arch height stiffness was reflected by its flexibility which was quantified by the
change in arch height during balanced standing (measured from the dorsal peak of
intermediate cuneiform to the tuberosity of the calcaneus bone). Our study showed
that the surgery reduced about a quarter flexibility compared to that of the intact foot.
The joint fusion surgery elevated the maximum contact pressure of the ankle, talo-
navicular, intermediate cuneonavicular, cuboideonavicular, and the fifth cuboideo-
metatarsal joints. Figure 10.17 illustrates the maximum joint contact pressure
normalized to that of the ankle joint during the first-peak instant.
10 Biomechanics of Foot and Ankle 247
3.5
Ratio of Contact Pressure (100%)
2.5
1.5
0.5
0
1st Peak
Midstance
2nd Peak
1st Peak
Midstance
2nd Peak
1st Peak
Midstance
2nd Peak
1st Peak
Midstance
2nd Peak
1st Peak
Midstance
2nd Peak
Ankle Talonavicular NavicoICune Navicocuboid Meta5Cuboid
Normal Fusion
Fig. 10.17 Comparison of normalized contact pressure at five joints between normal foot and foot
with two tarsometatarsal joint fusion. These five joints showed increased contact pressure after the
joints fusion. All contact pressures were divided by that of the ankle joint during the first-peak
instant. (Reprinted from Medical Engineering & Physics, 36, Wang et al., Biomechanical study of
tarsometatarsal joint fusion using finite element analysis, 1394–1400, Copyright (2014), with per-
mission from Elsevier)
Figure 10.17 shows that the maximum contact pressure progressed chronologi-
cally along the stance phase, particularly dominated by the ankle joint. The fusion
surgery did not produce considerable change at the ankle joint with a percentage
increase of 12%, 14%, and 0.58%, respectively, for the three stance instants. The
joint between the navicular and cuboid bone presented the largest percentage change
during midstance despite relatively small magnitude.
Figure 10.18 illustrates the von Mises stress plot during midstance. While metatar-
sal bones were relatively more susceptible to fracture due to its long and thin shape
with large load transfer, von Mises stress was believed to be one of the key predic-
tors for stress fracture [77].
The maximum stress was located at the second metatarsal bone at midstance.
The magnitude increased from 26 MPa under normal condition to 31 MPa under the
fusion condition. Similarly, the stress was also elevated by 16% and 14%, respec-
tively, during first peak and second peak. Besides, the maximum von Mises stress of
the fifth metatarsal bone was increased by 5.1% and 9.5% after fusion, respectively,
248 D. W.-C. Wong et al.
during the first-peak and midstance instants. On the other hand, there were no sub-
stantial change of the maximum stress for the first and fourth metatarsal bone com-
paring the normal and the fusion conditions.
Stress fractures are more prevalent at the second and third metatarsals which was
reported as one of the major complications after foot surgeries [78, 79]. With approxi-
mately one-fifth of stress increase, our prediction supported the fact that TMT joint
fusion between the first and second rays induce risk of second metatarsal bone fracture.
3.6 Conclusions
TMT joint fusion could be a salvage procedure to repair severe flatfoot. The stiffen-
ing created by the fusion attenuated the internal stress and strain of bones and soft
tissue, as well as the plantar pressure distribution. The peak plantar pressure increased
in all simulated instants whereas the largest increase happened during push-off.
10 Biomechanics of Foot and Ankle 249
The surgical procedure produced relatively pronounced raise in the peak contact
pressure between the navicular and cuboid joint, whereas increases in contact pres-
sure were also predicted in other joints, including the ankle, talonavicular joint, the
fifth cuboideometatarsal, and intermediate cuneonavicular joints. Based on the find-
ings, we believed that the cuboideonavicular and the ankle joints were more suscep-
tible to arthritis due to the large contact pressure. Likewise, the large von Mises
stress on the second metatarsal induced by the procedure could probably increase
the risk of stress fracture which in line with the higher complication rates of second
metatarsal stress fracture in clinical findings.
Effective interventions are expected to remedy the pain, impaired functions, and
quality of life because of the disease or problems, even though surgery may inter-
fere natural anatomy and physiology resulting in complications and other negative
outcomes. Biomechanical information provides additional information for physi-
cians to understand the limitations or potential consequences of an intervention and
support their decision-making process.
Summary
Calcaneus fracture comprises 1–2% of all fractures and about 60% of those affect-
ing the foot, with approximately 75% as intra-articular fractures. Fractures of the
calcaneus are typically produced by axial compressive force yet relevant research
using cadavers demonstrated variations in experimental design and outcome.
Biomechanical tolerance of calcaneus towards axial collision impact also remains
unclear. Finite element (FE) analysis provides a versatile platform to estimate the
internal features of the calcaneus in a controlled environment. In this study, a com-
prehensive FE analysis was adopted to evaluate the influence of axial impact on the
risk of calcaneal fractures. An impact at 5.0 m/s induced a maximum stress (von
Mises) of 3.21 MPa and 2.41 MPa, respectively, for the calcaneus and talus trabecu-
lar, in addition to maximum shear stress (Tresca) of 3.46 and 2.55 MPa.
Approximately one-fifth of the volume surpassed the yielding point of compressive
stress and more than 80% exceeded that of the shear stress. These volumes were
centered at the talocalcaneal articulation and the inferior calcaneal tuberosity that
resembled common sites of fracture. This study can provide insights into injury
prevention and fracture management for high energy trauma.
4.1 Background
Calcaneus plays an important role in the foot by its unique morphology and its vari-
ous connections to other soft tissue attachment. It sustains the pulling force of the
plantar fascia, tendon, and ligaments during weight-bearing and forming a tenseg-
rity structure to facilitate the stability of the longitudinal arch and lateral column of
250 D. W.-C. Wong et al.
the foot [80]. Besides, calcaneus transmits majority of the body weight to the ground
and acts as a strong lever for the greatest plantarflexor of the foot during propulsion.
Calcaneus is the most common fracture site of the hindfoot, and the consequence
could be devastating. Calcaneal fractures occur during the high-energy events, such
as a car crash or a fall from height. The annual incidence was 11.5 in every 100,000
population and males had a 2.4 times higher occurrence than females [81]. In males,
the annual incidence was 16.5 in every 100,000 population, with a peak incidence
of 21.6 in the age group 20–29. In females, the overall incidence was 6.26, with a
more evenly distributed rate across the age cohorts and showing a gradual increase
towards the post-menopause.
Calcaneal fracture is difficult to treat and requires a lengthy recovery period.
Treatment involves both conservative and surgical interventions to reconstruct the
normal anatomy of the heel and restore mobility so that patients can return to nor-
mal activity [82]. But even with appropriate treatment, some fractures may result in
long-term complications, such as pain, swelling, loss of motion, and arthritis. Many
complications can be prevented and treated with the considerations of biomechanics
during surgical planning.
The ankle joint that encompassed the calcaneus and talus stands close to the load
line. Therefore, the calcaneus is more vulnerable to high axial compressive force
that leads to calcaneal fracture [83, 84]. From the biomechanical point of perspec-
tive, it is worth knowing the detailed mechanism of the fracture, while relevant
studies examined the influence of collision impact during an accident and the toler-
ance of the foot and ankle [83]. The fracture was induced to cadaveric specimens
with a prescribed impact velocity or energy [85], while some mimicked a pedal
impact using different pre-dorsiflexed ankle position, as well as different impact
forces and velocities [86–89].
Among hindfoot fracture, calcaneus was one of the most frequent sites of injury,
followed by talus and ankle fracture [84]. Despite, cadaveric studies in support to
this fact reported mixed results probably due to the variation in research protocols.
One study envisaged an extremely high impact velocity at 12 m/s to simulate a
pedal impact upon a landmine explosion on the vehicle [87]. Different loading
profiles were also considered in quasi-static scenarios [88]. Even though within a
single study, the reports of fracturing load on different specimens can range from
3.7 to 8.3 kN [84].
Computational simulation provides an alternative to evaluate the potential of cal-
caneal fracture upon impact otherwise difficult to be conducted ethically using
physical experiments [90]. The advantage of the finite element (FE) method included
the assessment of internal biomechanical environment of complex structures in
well-controlled conditions which support different designs and clinical applications
[91–94]. Previous studies had also utilized dynamic analyses in applications such as
10 Biomechanics of Foot and Ankle 251
car crash, landing, and running [2, 3, 95]. A pedal impact on the forefoot was also
simulated using a foot-shank model complex by Shin et al. and his colleagues [96].
The aim of this research was to study how different impact velocity attenuate the
stress of the heel and thus the risk of fracture by means of FE analysis. Prior to the
dynamic simulation, we constructed an anatomically detailed FE model of the foot
and ankle complex and performed a validation. The outcome measure of the simula-
tion included the reaction forces and the volume fraction of the yielding bone upon
different impact velocities.
4.3.1 Model Reconstruction
We recruited a healthy female (age 28; height 165 cm; body mass 54 kg) to partici-
pate in the study. She did not reported any musculoskeletal pain, disorder or previ-
ous foot surgeries. The coronal magnetic resonance (MR) images of the right foot
was scanned using A 3.0-T scanner (TrioTim, Siemens Medical Solutions, Erlangen,
Germany) at neutral position and under no weight-bearing. The clinical images
were processed in an image segmentation software (Mimics 10, Materialise, Leuven,
Belgium) where 30 bones and the encapsulated tissue were segmented and further
optimized in another software (Rapidfrom XOR2, INUS technology Ltd., Seoul,
Korea). The calcaneus and talus bones were further segmented into the cortical
layer and the trabecular core according to the settings of Sabry et al. [97]. Since the
cartilage was difficult to identify in the clinical images, we simplified the layer
geometry into a nonlinear contact stiffness [65] along with a frictionless behavior
[98]. The ligaments, tendons, and fascia were constructed using trusses, surfaces,
and connectors jointing the insertion points on the constructed osseous geometry
which was further confirmed by anatomy expert. The model was an extension to our
previous work in which the validation process using both plantar pressure measure-
ment and cadavers, and the mesh convergence test were conducted [99]. The recon-
structed finite element model of the foot and ankle complex is shown in Fig. 10.19.
4.3.2 Material Properties
The material properties of the model parts were all sourced from literature as
shown. The calcaneus and talus were segmented into trabecular and cortical core
and assumed linearly elastic with a Young’s modulus of 0.4 MPa and 17 GPa, and
the Poisson’s ratio of 0.3 [100] while that of the other bones without segmentation
were assigned 7.3 GPa [40]. Hyperelastic material property was assigned to the
encapsulated soft tissue and skin using the constitutive equation of the second
order polynomial strain energy potential and first-order Ogden model, respectively
[41, 101]. The suggested average elasticity of rearfoot ligament from literature
252 D. W.-C. Wong et al.
Side view
Trabecular Core
Cortical Layer Fixed end
Achilles tendon
force
Plantar Fascia
Retinaculum
Encapsulated
soft tissue
Foam plate
Rigid plate
Impactor
Top view
Fig. 10.19 Finite element model of the foot and ankle complex. Finite element model of the foot
and ankle complex showing the top and side view of the parts geometry and demonstrating the
boundary and load conditions used in the simulation. (Reprinted from PlosOne, Wong et al., Finite
Element Analysis of Foot and Ankle Impact Injury: Risk Evaluation of Calcaneus and Talus
Fracture, e0154435 (2016), under CC-BY)
was assigned to our model with a value of 264.8 MPa [42, 102]. The forefoot liga-
ments were assumed as truss and assigned with a cross-sectional area of 18.4 mm2
[63], while the other ligaments modelled with shell surface have a thickness of
1.5 mm [43, 98]. The footplate was regarded as a 19-mm thick foam padding with
a higher elastic modulus (15 MPa) sticking on top of another plate with rigid defi-
nition [84].
The simulation was carried out using commercially available finite element package
(Abaqus 6.11, Dassault Systèmes, RI, USA). The coefficient of friction bounded by
the encapsulated soft tissue and the ground plate was set to 0.6 [47] and the bones
were tied to the encapsulated soft tissue.
Firstly, two sets of simulations were conducted for model validation which
attempted to resemble the two sets of conditions in a cadaveric study [84].
According to the study protocol. The encapsulated bulk tissue and the proximal end
of tibial and fibular were encastré. As shown in Fig. 10.22, a foot pedal was driven
by an impact plate striking at 5.0 m/s such that the pedal would eventually meet the
10 Biomechanics of Foot and Ankle 253
plantar foot surface [84]. The mass of the impactor was assigned 7 kg while that of
the foot pedal was 4.5 kg and comprised one deformable and one rigid layer [86].
On the other hand, a vertical ground reaction force resembling half body weight of
a 54 kg person was applied on the pedal in superior direction once the foot came
into contact. The second load case for validation presented the same settings as the
first load case, except that the Achilles tendon would be loaded 1940 N. Immediately
after the simulation for validation, a parametric analysis with different impact
velocities from 2.0 to 7.0 m/s would be conducted based on the aforementioned
boundary and loading conditions for validation.
4.3.4 Data Analysis
Ground reaction force (GRF; as computed by the contact between the plantar foot
and pedal) and tibial reaction force (TRF) would be evaluated on the two scenarios
(i.e., with and without Achilles tendon load). TRF is the predicted reaction force to
maintain the fixture of the proximal tibial end.
By varying the impact velocity at 1.0 m/s interval from 2.0 to 7.0 m/s, we inves-
tigated the influence on GRF, TRF, as well as the maximum von Mises stress and
shear (Tresca) stress of the calcaneal and talar trabecular core. The volume fraction
of bone exceeding yield in compression and shear would be regarded as the risk
indicator for bone fracture. The compressive and shear yield was 1.8 MPa and
0.79 MPa, respectively [103, 104].
Figure 10.20 illustrates the GRF and TRF under impact with and without Achilles
tendon load and compared the outcome between our FE predictions and existing
study [84]. Regarding GRF, the differences between prediction and experiment find-
ings were approximately 10% under both the conditions with and without Achilles
tendon load. FE analysis found that the GRFs were 7400 and 6000 N with and without
Achilles tendon load conditions. Besides, the differences of TRF between simulation
and experiment were relatively prominent (27.5%) under the condition with Achilles
tendon, while the difference (600 N) was less without the Achilles tendon load.
10
8
Force (kN)
2
n=6 n=6 n=7 n=7
0
GRF TRF GRF TRF
Impact with
Pure Impact Achilles Tendon Loaded
Fig. 10.20 Validation of finite element model by comparing the prediction results to existing lit-
erature. Comparison of finite element prediction and cadaveric experiment results from Funk et al.
[84] on the ground reaction force (GRF) and tibial reaction force (TRF) under pure impact and
impact with Achilles tendon loading. (Reprinted from PlosOne, Wong et al., Finite Element
Analysis of Foot and Ankle Impact Injury: Risk Evaluation of Calcaneus and Talus Fracture,
e0154435 (2016), under CC-BY)
14
12
10
Force (kN)
0
2.0 3.0 4.0 5.0 6.0 7.0
Impact Velocity (m/s)
GRF TRF
Fig. 10.21 Ground reaction force (GRF) and tibial reaction force (TRF) under different impact
velocity (2.0–7.0 m/s). (Reprinted from PlosOne, Wong et al., Finite Element Analysis of Foot and
Ankle Impact Injury: Risk Evaluation of Calcaneus and Talus Fracture, e0154435 (2016),
under CC-BY)
10 Biomechanics of Foot and Ankle 255
a b
Cortical layer
Fig. 10.22 Von Mises stress of calcaneus and talus. (a) Cross section view of von Mises stress of
the calcaneus and talus at 5.0 m/s impact velocity. Orange arrows indicate compressive stress.
Cyan arrows indicate tensile stress. (b) X-ray of a typical patient with a compressive fracture of the
calcaneus. Arrows indicate regions of fractures. (Reprinted from PlosOne, Wong et al., Finite
Element Analysis of Foot and Ankle Impact Injury: Risk Evaluation of Calcaneus and Talus
Fracture, e0154435 (2016), under CC-BY)
respectively, at the highest impact velocity. Their differences ranged from 33% to
47% in the study range of impact velocities.
Figure 10.22a displays the von Mises stress of the calcaneus and talus. In addition,
as shown in Figure 10.22b, the fracture sites of one typical patient presented similar
pattern to that of the location of the peak stress. Figures 10.24 and 10.25 suggested
that the increase of impact velocity resulted in the increase of both the maximum
von Mises stress and Tresca stress. The Tresca stress was 7% higher than the maxi-
mum von Mises stress at 2.0 m/s impact and the difference was enlarged with
increasing impact velocity. At 7.0 m/s impact velocity, the maximum von Mises
stress was 5.06 MPa compared to 5.47 MPa of Tresca stress.
Figures 10.23 and 10.24 illustrates the volume fraction of the trabecular bone of
calcaneus that was higher than the yielding point. Interestingly, the yielding was dom-
inant by shear stress. Nearly 80% of the bone experienced yielding at 4.0 m/s and
nearly total fraction exceeded shear yield at 7.0 m/s. The trabecular bone experienced
less yield and only two-third of the volume exceeded compressive yield at 7.0 m/s.
The volume of trabecular calcaneus that exceeded yield is presented in Fig. 10.24.
The trabecular calcaneus underwent shear yielding predominantly compared to
compressive yield. The volume of bone with shear yielding increased considerably
256 D. W.-C. Wong et al.
(MPa)
0.00 0.25 0.50 0.75 1.00 1.25 1.50 1.75 2.00 2.25 2.50 2.75 3.00
b Talus Trabecular
Von Mises stress
Tresca stress
(MPa)
0.00 0.25 0.50 0.75 1.00 1.25 1.50 1.75 2.00 2.25 2.50 2.75 3.00
Fig. 10.23 Von Mises and Tresca stresses of the (a) calcaneus trabecular and (b) talus trabecular
at different impact velocities. (Reprinted from PlosOne, Wong et al., Finite Element Analysis of
Foot and Ankle Impact Injury: Risk Evaluation of Calcaneus and Talus Fracture, e0154435 (2016),
under CC-BY)
from 36.2% at 3.0 m/s impact to 79.7% at 4.0 m/s. Nearly all trabecular bone of the
calcaneus exceeded the yielding point of shear at 7.0 m/s impact. The trabecular
bone that exceeded the compressive strength was relatively mild. About one-fifth
and two-thirds of the volume reached the compressive yield at 5.0 m/s and 7.0 m/s
impact, respectively (Fig. 10.24).
The maximum stress and Tresca of trabecular talus are demonstrated in
Figs. 10.23 and 10.25. The line of shear stress was consistently larger than that of
the von Mises stress, while both parameters increased with increasing impact veloc-
ity. The stresses were 0.48 and 0.55 MPa for the maximum von Mises and Tresca at
2.0 m/s impact and were increased to 3.68 and 3.90 MPa, respectively, at 7.0 m/s.
Nearly one-fifth of the bone volume experienced a compressive yield which was
then increased to half from 5.0 to 7.0 m/s.
Figure 10.23 shows that both the peak of von Mises stress and shear (Tresca)
were located at the inferior calcaneal tuberosity and talus articulation, which was
generally agreeable with the findings of cadaveric study and common fracture sites
[84, 87]. The stress concentration at the posterior talocalcaneal articulation, as
well as the superior side of the talar trochlea suggested that talus was also suscep-
tible to fracture during compressive impact [87]. Lateral malleolar articulation and
10 Biomechanics of Foot and Ankle 257
6 100.0%
90.0%
60.0%
3 50.0%
40.0%
2
30.0%
20.0%
1
10.0%
0 0.0%
2 3 4 5 6 7
Impact velocity (m/s)
Fig. 10.24 Maximum von Mises and Tresca stress with yielding volume of trabecular calcaneus
against impact velocity. (Reprinted from PlosOne, Wong et al., Finite Element Analysis of Foot and
Ankle Impact Injury: Risk Evaluation of Calcaneus and Talus Fracture, e0154435 (2016), under CC-BY)
6 100.0%
90.0%
% Volume of trabecular bone
5
80.0%
70.0%
Stress (MPa)
4
60.0%
3 50.0%
40.0%
2
30.0%
20.0%
1
10.0%
0 0.0%
2 3 4 5 6 7
Impact velocity (m/s)
Fig. 10.25 Maximum von Mises and Tresca stress with yielding volume of trabecular talus against
impact velocity. (Reprinted from PlosOne, Wong et al., Finite Element Analysis of Foot and Ankle
Impact Injury: Risk Evaluation of Calcaneus and Talus Fracture, e0154435 (2016), under CC-BY)
258 D. W.-C. Wong et al.
4.5 Conclusion
An axial compressive impact (5.0 m/s) induced yielding to the trabecular bone of
the heel which predisposes potential fracture. Our prediction showed that the frac-
ture could be dominant by shear failure and compounded by compression. Future
study shall consider the sensitivity of different loading patterns on fracture which
could provide additional insights to design measures for injury prevention and inter-
vention for fracture management.
In the future, this computational platform can investigate the relationship between
different loading modes and fracture pattern/mechanism that could support design
for the prevention of injury, as well as management of the fracture.
Acknowledgments The work of this chapter was supported by the Key R&D Program granted by
the Ministry of Science and Technology of China (2018YFB1107000), NSFC granted by the
National Natural Science Foundation of China (11732015), and General Research Fund granted by
the Hong Kong Research Grant Council (PolyU152065/17E).
References
1. Abhishek A, Roddy E, Zhang W, Doherty M. Are hallux valgus and big toe pain
associated with impaired quality of life? A cross-sectional study. Osteoarthr Cartil.
2010;18(7):923–6.
2. Cho JR, Park SB, Ryu SH, Kim SH, Lee SB. Landing impact analysis of sports shoes using
3-d coupled foot-shoe finite element model. J Mech Sci Technol. 2009b;23(10):2583–91.
3. Cho NH, Kim S, Kwon DJ, Kim HA. The prevalence of hallux valgus and its association with
foot pain and function in a rural korean community. J Bone Joint Surg Br. 2009a;91b(4):494–8.
4. Menz HB, Roddy E, Thomas E, Croft PR. Impact of hallux valgus severity on general and
foot-specific health-related quality of life. Arthritis Care Res Hoboken. 2011;63(3):396–404.
5. Meyr AJ, Adams ML, Sheridan MJ, Ahalt RG. Epidemiological aspects of the surgical cor-
rection of structural forefoot pathology. J Foot Ankle Surg. 2009;48(5):543–51.
6. Wu D, Louie L. Does wearing high-heeled shoe cause hallux valgus? A survey of 1,056 chi-
nese females. Foot Ankle Online J. 2010;3(5):3.
7. Bonney G, McNab L. Hallux valgus and hallux rigidus—a critical survey. J Bone Joint Surg
Br. 1952;34-B(3):366–85.
8. Roddy E, Zhang W, Doherty M. Prevalence and associations of hallux valgus in a primary-
care population. Ann Rheum Dis. 2006;65:403–4.
10 Biomechanics of Foot and Ankle 259
9. Coughlin MJ, Shurnas PS. Hallux valgus in men. Part II: first ray mobility after bunionec-
tomy and factors associated with hallux valgus deformity. Foot Ankle Int. 2003a;24(1):73–8.
10. Ferrari J, Hopkinson DA, Linney AD. Size and shape differences between male and female
foot bones: is the female foot predisposed to hallux abducto valgus deformity? J Am Podiatr
Med Assoc. 2004;94(5):434–52.
11. Wilkerson RD, Mason MA. Differences in men's and women's mean ankle ligamentous lax-
ity. Iowa Orthop J. 2000;20:46.
12. Coughlin MJ, Jones CP. Hallux valgus and first ray mobility. A prospective study. J Bone
Joint Surg Am. 2008;90(5):1166–7.
13. Owoeye BA, Akinbo SR, Aiyegbusi AL, Ogunsola MO. Prevalence of hallux valgus among
youth population in Lagos, Nigeria. Nigerian Postgrad Med J. 2011;18(1):51–5.
14. Menz HB, Lord SR. The contribution of foot problems to mobility impairment and falls in
community-dwelling older people. J Am Geriatr Soc. 2001;49(12):1651–6.
15. Thompson FM, Coughlin MJ. The high price of high-fashion footwear. J Bone Joint Surg
Am. 1994;76a(10):1586–93.
16. Access Economics. The economic impact of podiatric surgery. In: Report for the Australasian
College of Podiatric Surgeons. Sydney: Access Economics Pty Ltd; 2008.
17. Australian Bureau of Statistics. Australian National Accounts: national income, expenditure
and product. Canberra: Australian Bureau of Statistics; 2008.
18. Courtney TK, Matz S, Webster BS. Disabling occupational injury in the us construction
industry, 1996. J Occup Environ Med. 2002;44(12):1161–8.
19. Mathers CD, Vos ET, Stevenson CE, Begg SJ. The burden of disease and injury in Australia.
Bull World Health Organ. 2001;79(11):1076–84.
20. Caminear DS, Addis-Thomas E, Brynizcka AW, Saxena A. Revision hallux valgus surgery.
In: Saxena A, editor. International advances in foot and ankle surgery. London: Springer-
Verlag; 2012. p. 71–82.
21. Lehman DE. Salvage of complications of hallux valgus surgery. Foot Ankle Clin.
2003;8(1):15–35.
22. Coughlin MJ, Mann RA. Hallux valgus. In: Coughlin MJ, Saltzman CL, Mann RA, editors.
Surgery of the foot and ankle. 8th ed. Pennsylvania: Mosby Elsevier; 2012.
23. Faber FWM, Kleinrensink GJ, Mulder PGH, Verhaar JAN. Mobility of the first tarsometatar-
sal joint in hallux valgus patients: a radiographic analysis. Foot Ankle Int. 2001;22(12):965–9.
24. Glasoe WM, Allen MK, Saltzman CL, Ludewig PM, Sublett SH. Comparison of two meth-
ods used to assess first-ray mobility. Foot Ankle Int. 2002;23(3):248–52.
25. Kim JY, Park JS, Hwang SK, Young KW, Sung IH. Mobility changes of the first ray after
hallux valgus surgery: clinical results after proximal metatarsal chevron osteotomy and distal
soft tissue procedure. Foot Ankle Int. 2008;29(5):468–72.
26. Smith BW, Coughlin MJ. The first metatarsocuneiform joint, hypermobility, and hallux val-
gus: what does it all mean? Foot Ankle Surg. 2008;14(3):138–41.
27. Klaue K, Hansen ST, Masquelet AC. Clinical, quantitative assessment of first tarsometatar-
sal mobility in the sagittal plane and its relation to hallux valgus deformity. Foot Ankle Int.
1994;15(1):9–13.
28. Hillstrom HJ, Song JS, Kraszewski AP, et al. Foot type biomechanics part 1: structure and
function of the asymptomatic foot. Gait Posture. 2013;37(3):445–51.
29. Stokes IAF, Hutton WC, Stott JRR, Lowe LW. Forces under the hallux valgus foot before and
after surgery. Clin Orthop Relat Res. 1979;142:64–72.
30. Kernozek TW, Elfessi A, Sterriker S. Clinical and biomechanical risk factors of patients diag-
nosed with hallux valgus. J Am Podiatr Med Assoc. 2003;93(2):97–103.
31. Martínez-Nova A, Cuevas-García JC, Sánchez-Rodríguez R, Pascual-Huerta J, Sánchez-
Barrado E. Study of plantar pressure patterns by means of instrumented insoles in subjects
with hallux valgus. Rev Esp Cir Ortop Traumatol (Engl Ed). 2008;52(2):94–8.
32. Mickle KJ, Munro BJ, Lord SR, Menz HB, Steele JR. Gait, balance and plantar pressures in
older people with toe deformities. Gait Posture. 2011;34(3):347–51.
260 D. W.-C. Wong et al.
33. Wen JM, Ding QC, Yu ZY, Sun WD, Wang QN, Wei KL. Adaptive changes of foot pressure
in hallux valgus patients. Gait Posture. 2012;36(3):344–9.
34. Mittal D, Raja S, Geary NPJ. The modified mcbride procedure: clinical, radiological, and
pedobarographic evaluations. J Foot Ankle Surg. 2006;45(4):235–9.
35. Saro C, Andrén B, Felländer-Tsai L, Lindgren U, Arndt A. Plantar pressure distribution
and pain after distal osteotomy for hallux valgus: a prospective study of 22 patients with
12-month follow-up. Foot. 2007;17(2):84–93.
36. Dhukaram V, Hullin MG, Senthil Kumar C. The Mitchell and scarf osteotomies for hallux
valgus correction: a retrospective, comparative analysis using plantar pressures. J Foot Ankle
Surg. 2006;45(6):400–9.
37. Cheung JT-M, Nigg BM. Clinical applications of computational simulation of foot and ankle.
Sports Orthop Traumatol. 2008;23(4):264–71.
38. Yu J, Cheung JTM, Fan YB, Zhang Y, Leung AKL, Zhang M. Development of a finite ele-
ment model of female foot for high-heeled shoe design. Clin Biomech. 2008;23:S31–8.
39. Kai T, Wang C-T, Wang D-M, Wang X. Primary analysis of the first ray using a 3-dimension
finite element foot model. Paper read at Engineering in Medicine and Biology Society, 2005.
In: 27th Annual International Conference of the IEEE-EMBS 2005. 2006.
40. Nakamura S, Crowninshield RD, Cooper RR. An analysis of soft tissue loading in the foot—
a preliminary report. Bull Prosthet Res. 1981;10:27–34.
41. Lemmon D, Shiang TY, Hashmi A, Ulbrecht JS, Cavanagh PR. The effect of insoles in thera-
peutic footwear—a finite element approach. J Biomech. 1997;30(6):615–20.
42. Siegler S, Block J, Schneck CD. The mechanical characteristics of the collateral ligaments of
the human ankle joint. Foot Ankle. 1988a;8(5):234–42.
43. Milz P, Milz S, Steinborn M, Mittlmeier T, Putz R, Reiser M. Lateral ankle ligaments and
tibiofibular syndesmosis—13-MHz high-frequency sonography and mri compared in 20
patients. Acta Orthop Scand. 1998;69(1):51–5.
44. Arnold EM, Ward SR, Lieber RL, Delp SL. A model of the lower limb for analysis of human
movement. Ann Biomed Eng. 2010;38(2):269–79.
45. Froberg A, Komi P, Ishikawa M, Movin T, Arndt A. Force in the achilles tendon during walk-
ing with ankle foot orthosis. Am J Sports Med. 2009;37(6):1200–7.
46. Perry J, Davids JR. Gait analysis: normal and pathological function. J Pediatr Orthop.
1992;12(6):815.
47. Zhang M, Mak AFT. In vivo friction properties of human skin. Prosthetics Orthot Int.
1999;23(2):135–41.
48. Coughlin MJ. Instructional course lectures, the american academy of orthopaedic surgeons-
hallux valgus. J Bone Joint Surg. 1996;78(6):932–66.
49. Valderrabano V, Horisberger M, Russell I, Dougall H, Hintermann B. Etiology of ankle
osteoarthritis. Clin Orthop Relat Res. 2009;467(7):1800–6.
50. Goldberg AJ, MacGregor A, Dawson J, et al. The demand incidence of symptomatic
ankle osteoarthritis presenting to foot & ankle surgeons in the United Kingdom. Foot.
2012;22(3):163–6.
51. Glazebrook M, Daniels T, Younger A, et al. Comparison of health-related quality of life between
patients with end-stage ankle and hip arthrosis. J Bone Joint Surg. 2008;90(3):499–505.
52. Slobogean GP, Younger A, Apostle KL, et al. Preference-based quality of life of end-stage
ankle arthritis treated with arthroplasty or arthrodesis. Foot Ankle Int. 2010;31(7):563–6.
53. Coester LM, Saltzman CL, Leupold J, Pontarelli W. Long-term results following ankle
arthrodesis for post-traumatic arthritis. J Bone Joint Surg Am. 2001;83a(2):219–28.
54. Fuchs S, Sandmann C, Skwara A, Chylarecki C. Quality of life 20 years after arthrodesis of
the ankle—a study of adjacent joints. J Bone Joint Surg Br. 2003;85b(7):994–8.
55. Mazur JM, Schwartz E, Simon SR. Ankle arthrodesis - long-term follow-up with gait analy-
sis. J Bone Joint Surg Am. 1979;61(7):964–75.
56. Thomas R, Daniels TR, Parker K. Gait analysis and functional outcomes following ankle
arthrodesis for isolated ankle arthritis. J Bone Joint Surg Am. 2006;88a(3):526–35.
10 Biomechanics of Foot and Ankle 261
57. Muir DC, Amendola A, Saltzman CL. Long-term outcome of ankle arthrodesis. Foot Ankle
Clin. 2002;7(4):703–8.
58. Beyaert C, Sirveaux F, Paysant J, Mole D, Andre JM. The effect of tibio-talar arthrodesis
on foot kinematics and ground reaction force progression during walking. Gait Posture.
2004;20(1):84–91.
59. Flavin R, Coleman SC, Tenenbaum S, Brodsky JW. Comparison of gait after total ankle
arthroplasty and ankle arthrodesis. Foot Ankle Int. 2013;34(10):1340–8.
60. Piriou P, Culpan P, Mullins M, Cardon JN, Pozzi D, Judet T. Ankle replacement versus
arthrodesis: a comparative gait analysis study. Foot Ankle Int. 2008;29(1):3–9.
61. Ramlee MH, Kadir MRA, Murali MR, Kamarul T. Finite element analysis of three com-
monly used external fixation devices for treating type iii pilon fractures. Med Eng Phys.
2014;36(10):1322–30.
62. Vazquez AA, Lauge-Pedersen H, Lidgren L, Taylor M. Finite element analysis of the initial
stability of ankle arthrodesis with internal fixation: flat cut versus intact joint contours. Clin
Biomech. 2003;18(3):244–53.
63. Cheung JTM, Zhang M, Leung AKL, Fan YB. Three-dimensional finite element analysis of
the foot during standing - a material sensitivity study. J Biomech. 2005;38(5):1045–54.
64. Cheung JTM, Zhang M. A 3-dimensional finite element model of the human foot and ankle
for insole design. Arch Phys Med Rehabil. 2005;86(2):353–8.
65. Athanasiou KA, Liu GT, Lavery LA, Lanctot DR, Schenck RC. Biomechanical topography
of human articular cartilage in the first metatarsophalangeal joint. Clin Orthop Relat Res.
1998;348:269–81.
66. Gefen A, Megido-Ravid M, Itzchak Y, Arcan M. Biomechanical analysis of the three-
dimensional foot structure during gait: a basic tool for clinical applications. J Biomech Eng
Trans ASME. 2000;122(6):630–9.
67. Wright DG, Rennels DC. A study of the elastic properties of plantar fascia. J Bone Joint Surg.
1964;46(3):482–92.
68. Dul J. Development of a minimum-fatigue optimization technique for predicting individual
muscle forces during human posture and movement with application to the ankle musculature
during standing and walking. Ph.D. thesis, Vanderbilt University. 1983.
69. Kim K-J, Kitaoka HB, Luo Z-P, et al. In vitro simulation of the stance phase in human gait. J
Musculoskelet Res. 2001;5(02):113–21.
70. Eleftheriou KI, Rosenfeld PF, Calder JDF. Lisfranc injuries: an update. Knee Surg Sports
Traumatol Arthrosc. 2013;21(6):1434–46.
71. Arastu MH, Buckley RE. Tarsometatarsal joint complex and midtarsal injuries. Acta Chir
Orthop Traumatol Cechoslov. 2012;79(1):21–30.
72. Desmond EA, Chou LB. Current concepts review: Lisfranc injuries. Foot Ankle Int.
2006;27(8):653–60.
73. Watson TS, Shurnas PS, Denker J. Treatment of lisfranc joint injury: current concepts. J Am
Acad Orthop Surg. 2010;18(12):718–28.
74. Ghate SD, Sistla VM, Nemade V, Vibhute D, Shahane SM, Samant AD. Screw and wire fixa-
tion for lisfranc fracture dislocations. J Orthop Surg. 2012;20(2):170–5.
75. Lakin RC, DeGnore LT, Pienkowski D. Contact mechanics of normal tarsometatarsal joints.
J Bone Joint Surg Am. 2001;83a(4):520–8.
76. Wang Y, Li Z, Zhang M. Biomechanical study of tarsometatarsal joint fusion using finite ele-
ment analysis. Med Eng Phys. 2014;36(11):1394–400.
77. Keyak JH, Rossi SA. Prediction of femoral fracture load using finite element models: an
examination of stress-and strain-based failure theories. J Biomech. 2000;33(2):209–14.
78. Weatherall JM, Chapman CB, Shapiro SL. Postoperative second metatarsal fractures associ-
ated with suture-button implant in hallux valgus surgery. Foot Ankle Int. 2013a;34(1):104–10.
79. Weatherall JM, Chapman C, Shapiro S. Comment on “postoperative second metatarsal frac-
tures associated with suture-button implant in hallux valgus surgery” response. Foot Ankle
Int. 2013b;34(6):919–20.
262 D. W.-C. Wong et al.
80. Epstein N, Chandran S, Chou L. Current concepts review: intra-articular fractures of the
calcaneus. Foot Ankle Int. 2012;33(1):79–86.
81. Mitchell MJ, McKinley JC, Robinson CM. The epidemiology of calcaneal fractures. Foot.
2009;19(4):197–200.
82. Lim EVA, Leung JPF. Complications of intraarticular calcaneal fractures. Clin Orthop Relat
Res. 2001;391:7–16.
83. Forman JL, Lopez-Valdes FJ, Duprey S, et al. The tolerance of the human body to automobile
collision impact–a systematic review of injury biomechanics research, 1990–2009. Accid
Anal Prev. 2015;80:7–17.
84. Funk JR, Crandall JR, Tourret LJ, et al. The axial injury tolerance of the human foot/ankle
complex and the effect of achilles tension. J Biomech Eng Trans ASME. 2002;124(6):750–7.
85. Gallenberger K, Yoganandan N, Pintar F. Biomechanics of foot/ankle trauma with variable
energy impacts. Ann Adv Automot Med. 2013;57:123.
86. Crandall JR, Portier L, Petit P, et al. Biomechanical response and physical properties of the
leg, foot, and ankle. SAE Technical Paper. 1996.
87. McKay BJ, Bir CA. Lower extremity injury criteria for evaluating military vehicle occupant
injury in underbelly blast events. Stapp Car Crash J. 2009;53:229.
88. Parenteau CS, Viano DC, Petit PY. Biomechanical properties of human cadaveric ankle-
subtalar joints in quasi-static loading. J Biomech Eng Trans ASME. 1998;120(1):105–11.
89. Yoganandan N, Pintar FA, Kumaresan S, Boynton M. Axial impact biomechanics of the
human foot-ankle complex. J Biomech Eng. 1997;119:433–7.
90. Chang CY, Rupp JD, Kikuchi N, Schneider LW. Development of a finite element model to
study the effects of muscle forces on knee-thigh-hip injuries in frontal crashes. Stapp Car
Crash J. 2008;52(52):475–504.
91. Cheung JT-M, Yu J, Wong DW-C, Zhang M. Current methods in computer-aided engineering
for footwear design. Footwear Sci. 2009;1(1):31–46.
92. Telfer S, Erdemir A, Woodburn J, Cavanagh PR. What has finite element analysis taught
us about diabetic foot disease and its management. A systematic review. PLoS One.
2014;9(10):e109994.
93. Wang Y, Li ZY, Wong DWC, Zhang M. Effects of ankle arthrodesis on biomechanical perfor-
mance of the entire foot. PLoS One. 2015;10(7):e0134340.
94. Wang Y, Wong DWC, Zhang M. Computational models of the foot and ankle for pathome-
chanics and clinical applications: a review. Ann Biomed Eng. 2016;44(1):213–21.
95. Farhang B, Araghi FR, Bahmani A, Moztarzadeh F, Shafieian M. Landing impact analysis of
sport surfaces using three-dimensional finite element model. Proc Inst Mech Engrs P J Sports
Eng Technol. 2016;230(3):180–5.
96. Shin J, Yue N, Untaroiu CD. A finite element model of the foot and ankle for automotive
impact applications. Ann Biomed Eng. 2012;40(12):2519–31.
97. Sabry FF, Ebraheim NA, Mehalik JN, Rezcallah AT. Internal architecture of the calcaneus:
implications for calcaneus fractures. Foot Ankle Int. 2000;21(2):114–8.
98. Wong DWC, Zhang M, Yu J, Leung AKL. Biomechanics of first ray hypermobility: an
investigation on joint force during walking using finite element analysis. Med Eng Phys.
2014;36(11):1388–93.
99. Wong DWC, Wang Y, Zhang M, Leung AKL. Functional restoration and risk of non-union
of the first metatarsocuneiform arthrodesis for hallux valgus: a finite element approach. J
Biomech. 2015;48(12):3142–8.
100. Morales-Orcajo E, Bayod J, de Las Casas EB. Computational foot modeling: scope and
applications. Arch Comput Methods Eng. 2016;23(3):389–416.
101. Gu YD, Li JS, Ren XJ, Lake MJ, Zeng YJ. Heel skin stiffness effect on the hind foot biome-
chanics during heel strike. Skin Res Technol. 2010;16(3):291–6.
102. Siegler S, Chen J, Schneck CD. The effect of damage to the lateral collateral ligaments on the
mechanical characteristics of the human ankle joint. In: Proceedings of the annual interna-
10 Biomechanics of Foot and Ankle 263
tional conference of the IEEE Engineering in Medicine and Biology Society, Pts 1–4, 1988b.
p. 622–3.
103. Mittra E, Rubin C, Gruber B, Qin YX. Evaluation of trabecular mechanical and microstruc-
tural properties in human calcaneal bone of advanced age using mechanical testing, mu ct,
and dxa. J Biomech. 2008;41(2):368–75.
104. Sanyal A, Gupta A, Bayraktar HH, Kwon RY, Keaveny TM. Shear strength behavior of
human trabecular bone. J Biomech. 2012;45(15):2513–9.
105. Kelly N, McGarry JP. Experimental and numerical characterisation of the elasto-plastic prop-
erties of bovine trabecular bone and a trabecular bone analogue. J Mech Behav Biomed
Mater. 2012;9:184–97.
Chapter 11
Biomechanics of Human Motion
Keywords Hip joint · Knee joint · Spine joint · Kinematic · Common pathologies
Joint arthroplasty
Studies on the hip biomechanics about hip movement or structure can be valuable
to underpin the understanding of joint function, etiology, and treatment of hip-
related diseases. Although ongoing endeavors to improve arthroplasty materials are
of value in increasing longevity of implants, component positioning and dynamic
function have gained attention as significant factors affecting joint mobility, stabil-
ity, and survivorship after total hip arthroplasty. Thus, it is critical to understand and
appreciate hip kinematics on the normal hip, the diseased hip, and the replaced hip
for the improvement of hip disease treatment in orthopedics. The following sections
will discuss the structural factors contributing to the balance between hip stability
and mobility throughout the healthy status, common pathologies, as well as hip
arthroplasty follow-ups.
The hip joint connects the femur and acetabulum of the pelvis. The hip joint plays
an important role to dynamically sustain and transmit the rotational moment, the
gravitational force of the body, and the ground reaction force of the lower extremi-
ties throughout an extensive range of motion. The hip joint has to maintain balance
and stability of the body in an erect position during standing. While moving, a com-
bination of both static and dynamic stabilizers helps to prevent joint dislocation and
to maintain mechanical efficiency.
1.1.1 Stability
The acetabulum results from the triradiate cartilage growth center of the pelvic
innominate bone at the union of the ilium, ischium, and pubic bones. Though the
acetabular cup appears to have a hemispherical shape, it is spherical only in the
upper 1/3rd or in the dome, allowing the required maximal distribution of force in
areas for weight-bearing during upright stance. As mentioned above, the inclined
acetabulum gives an acetabular index (slope of the acetabulum on an anterior-pos-
terior (AP) radiograph) an average of 38° in males and 40° in females allowing for
increased abduction but decreased adduction (Fig. 11.1a) [1]. The osseous structure
on the femoral side of the joint also contributes to joint stability during static or
dynamic loading. The femoral head is semispherical in shape, ranging from hemi-
spherical to 2/3rds of a full sphere. The head quickly tapers into the femoral neck
that connects to the body of the femur. The distance between the center of the femo-
ral head and the anatomic axis of the femur represents the “head-neck offset” and is
directly associated with the moment arm and efficiency of the hip abductors. Similar
to the anteversion of the acetabulum, the femoral head/neck is anteverted relative to
the anatomic axis by approximately 10–20° in adults (Fig. 11.1b).
Though the spherical acetabular design imparts a high degree of stability and
additional ligamentous support to the hip joint, acting as checkrein to all 6 degrees
of freedom during hip motion, the extra-articular ligaments consist of band-like
thickenings of the capsule that allow for hip motion within a certain range of arc.
Cadaveric studies have demonstrated some variability and controversy regarding
the exact nature and discrete nomenclature of these bands [1]. However, it is gener-
11 Biomechanics of Human Motion 267
a b
Femoral anteversion
angle
Anterior/posterior axis
Inclination
Planar anteversion
True anteversion
Fig. 11.1 Pelvic and femoral version. (a) True acetabular anteversion is the angle between the
anterior/posterior axis and the interception line of transverse plane and the cup opening plane.
Planar anteversion is the complement to the angle between the normal axis of the cup opening
plane and the anterior/posterior axis. Inclination is the angle of the cup rotating along the anterior/
posterior axis with respect to the medial/lateral axis. The anterior/posterior axis is perpendicular to
the frontal pelvic plane formed by two ASIS and pubic symphysis. (b) Femoral anteversion is the
inclination of the femoral neck axis with reference to the retrocondylar line. With the long axis
determined by the best-fit cylinder of the femoral shaft, the neck axis is defined as the centerline of
the femoral head and neck model. The retrocondylar line is determined from the postfemoral con-
dyles (Reprinted with permission from Wolters Kluwer Health)
ally described that the capsule has four extra-capsular ligaments consisting of lon-
gitudinal fibers of the iliofemoral, the ischiofemoral, the pubofemoral ligaments as
well as the annular fibers of the zona orbicularis. These ligaments play a crucial role
in different joint positions during hip motion, which is supported by the change in
motion after removal of the ligaments.
Dynamic stabilizers of the hip joint include flexors, extensors, internal and exter-
nal rotators, abductors, and adductors. Similar to the rotator cuff stabilizing shoul-
der, antagonist function from dynamic stabilizers help to maintain the femoral head
within the acetabulum. However, the interactions among muscle groups in the hip
are much less well understood compared to that among the muscles of the rotator
cup. Conceptually, it is assumed that antagonistic muscle groups help to counter
each other’s motion to lead to a hip rotation with minimal translation, as mentioned
above. For example, with gluteus minimus and medius function during hip abduc-
tion, the adductor complex eccentrically contracts to help translate the femoral head
back into the center of the acetabulum, thereby maintaining concentric and primar-
ily rotatory motion. However, since the insertion points on hip abductors and
the abductor complex are not directly located opposite on the femur, the resulting
268 R. Cheng et al.
difference in lever arms and moments results in a more complex intermuscular rela-
tionship than, say, that of the subscapularis and infraspinatus. Moreover, additional
dynamic stabilizers also participate in direct stabilization of the hip joint, acting as
physical blocks to femoral head escape. In particular, muscles that pass directly over
or even attach to the hip capsule may physically confine the translation of the femo-
ral head with applied loads.
1.1.2 Mobility
compared to the osseous range of motion particularly in extension and flexion with
adduction (Fig. 11.2). Conversely, Turley et al. [5] found that osseous impingement
occurred in vivo at the extremes of adduction in an upright stance as well as abduc-
tion at 90 degrees of flexion corresponding to impingement of the lesser trochanter
100
50
90 50 50 90
Abduction (degrees) Abduction (degrees)
50
Extension (degrees)
Fig. 11.2 The difference in the hip range of motion is presented between the predicted and real-
ized motions. Flexion/extension and abduction/adduction motion differences between predicted
models of motion based on osseous factors versus motion observed from the in vivo measure-
ments. The yellow area corresponds to a range of motion observed in vivo from patients during a
series of motions involved in activities of daily living. Purple areas correspond to the predicted
range of hip motion based on osseous factors alone. Red areas correspond to in vivo hip motions
where osseous impingement limits further motion (Reprinted with permission from Springer Nature)
270 R. Cheng et al.
on the pubis and the greater trochanter on the lateral acetabular rim. This is particu-
larly salient with regard to total hip replacement because an implant or osseous
impingement versus soft tissue impingement presumably puts the patient at higher
risk of dislocation and earlier wear/failure.
The human gait cycle has long been studied using high-speed cameras. Observational
studies on gait analysis were conducted, particularly in injured or weakened patients.
We will discuss the hip functions in the gait cycle. The human gait comprises two
main phases, stance phase corresponding to 60% of gait and swing phase, making
up the rest 40%. The simplification of the stride pattern observed in humans serves
as a blueprint for analyzing the position of the hip and the points at which static and
dynamic factors are involved throughout the gait cycle.
The main role of the hip is to support the weight of the body, transferring this
weight force through the pelvis and into the lower extremities throughout the gait
cycle. However, the hip must transfer force through a dynamic range, depending on
the combination of static and dynamic stabilizers not only to maintain concentric
alignment of the joint but also to facilitate the transition in the gait cycle and locomo-
tion. At the beginning of the gait cycle, the hip is flexed, slightly externally rotated,
and in neutral adduction centering the femoral head in the acetabulum, which allows
for a large degree of the femoral head coverage. After heel strike, the hip extends
when the gait progresses towards toe-off. Just before toe-off, the hip begins to flex
and abduct to allow for the leg to swing through during the swing phase and clear the
ground. This meets with internal rotation through the acceleration phase followed
again by external rotation at the end of swing phase to maintain a centered femoral
head as the leg passes from the behind the body to the front of the body.
At any given point of the gait cycle, the forces on the hip are the sum of both
moment forces produced by hip musculature and loading forces distributed from the
head, arms, and thorax. The net sum of these forces is the total amount of force
across the joint, also known as joint reactive force. The direction and magnitude of
the hip joint reactive force vector change through the gait cycle, ranging from 3.5 to
5 times the subject’s total body weight [6]. It is the interplay between concentrically
contracting muscles and dynamic and static stabilizers that are dramatically altered
in some pathologic conditions of the hip and following the surgical solutions.
Imaging modalities and diagnostic criteria of hip pathology nowadays are frequently
based on static findings (i.e., radiographs, lab values). Still, it is important to note
that all pathologies of the hip involve dynamic scenarios and that disease develop-
ment cannot occur without eventual effects on motion. Moreover, past and current
innovations for THA have only focused on improving fixation methods and bearing
11 Biomechanics of Human Motion 271
surfaces, so that the current challenges with the contemporary THA involve in vivo
dynamic phenomena such as edge loading, impingement, and dislocation. Thus, a
kinematic understanding of the hip is critical for evaluating the etiology of hip
pathology as well as improving the restoration of hip function through arthroplasty.
Over the past decade, the development and implementation of 2D/3D registration
techniques have been employed to address the growing need for higher accuracy in
human kinematic and implant wear measurements. These techniques evaluate a
single fluoroscopic image of the hip using a stored 3D model of the same prosthetic
implant to match the orientation of components to the silhouette on fluoroscopy.
The position of the components can be reconstructed by doing so. Through evaluat-
ing a series of sequential images, the implant motions and relationship between
implants and bones can be evaluated. These methods have provided initial advance-
ments to our current understanding of THA motion in vivo, particularly with regard
to femoral head/cup separation during gait and abduction activities, intraoperative
stability assessment, and position during extremes of motion [9–12]. However, the
accuracy of such methods has been significantly inferior to other methods of mea-
surement, namely RSA, thus limiting the use for further kinematic evaluation, par-
ticularly during motion [12].
Most recently, methods that employ the combination of orthogonal fluoroscopic
images, as well as 2D/3D registration techniques, have been developed, known as
dual fluoroscopic imaging system (DFIS) [13]. This technique involves an initial
CT image of the patient’s hip that is subsequently used to generate a 3D adaptive
global surface model of the femur, pelvis, and any implanted components [14, 15].
As a result, a patient-specific model of osseous geometry is generated and allows
for more accurate measurements than previous techniques that relied on general-
ized implant models (Fig. 11.4). Patients are then imaged at a rate of 30 Hz through-
11 Biomechanics of Human Motion 273
a b
Fig. 11.4 Dual fluoroscopic imaging system imaging registration. (a) Experimental setup of mea-
surement of a subject walking on a treadmill using DFIS. (b) An example of a total hip arthroplasty
registered to images in the computer-simulated dual fluoroscopic imaging system (DFIS).
(Redrawn from Tsai, T.-Y., J.-S. Li, et al. A novel dual fluoroscopic imaging method for determina-
tion of THA kinematics: in-vitro and in-vivo study. Journal of biomechanics, 2013. 46(7):
1300–1304)
out the activity of interest, for example, gait, with two fluoroscopic images
positioned orthogonally to one another (Fig. 11.4a). The fluoroscopes obtain
images synchronously, producing a tandem view of the hip from orthogonal posi-
tions and reducing the artifacts from parallax and beam scatter otherwise experi-
enced by single beam methods [13]. These images are then processed in conjunction
with the 3D CT images, which virtually position the 3D models to match the exact
positions of the patient’s pelvis, femur, and implants observed in the orthogonal
X-ray images (Fig. 11.4b). All images in the series from fluoroscopy are analyzed
in the way mentioned above to generate a series of 3D images that depict the exact
motion of the patient’s pelvis, femur, and implants during gait. This provides solid
information about the hip motion in all 6 degrees of freedom as well as allows for
predictions of angular momentum, force, and even wear characteristics. DFIS has
provided the exciting potential for more accurately measuring small perturbations
in components separation and also in vivo motion trajectory without the need for
invasive markers as in RSA [16]. The accuracy of DFIS is comparable to RSA and
able to measure translational and rotational changes with a measurement error of
0.2 ± 0.3 mm and 0.2 ± 0.8°, respectively [17]. However, although DFIS is a non-
invasive tool, it does inherently increase radiation exposure beyond prior 2D/3D
registration or RSA techniques but remains on the order of 5–6 mSv with current
methods [16].
274 R. Cheng et al.
The hip geometry and its kinematics represent the summation of complex interac-
tions between the static and dynamic stabilizers. The most common changes to this
function can be attributed to alterations in anatomy originating with the acetabular
shape and position (i.e., pincer lesion, DDH) or changes in the femoral head and
neck shapes (i.e., cam lesion, Perthes, SCFE, altered femoral version). Frequently
these changes are reciprocal, and both femoral and acetabular effects must be evalu-
ated, respectively. Though many of these changes originate in infancy or childhood,
the effects of these changes can often go unrecognized or even undiagnosed well
into adulthood. Consequently, the adult hip can represent the response to a lifetime’s
worth of altered hip kinematics that must be recognized and accounted for by the
arthroplasty surgeon.
The most common anatomic changes affecting native hip kinematics are cam and
pincer lesions. Cam lesions represent additional bony contours to the femoral head,
typically over the superior lateral base of the femoral head. The resulting loss in
overall head sphericity results in a functional decrease in the head-neck offset of the
hip [18]. As previously mentioned, this offset is critical for the hip range of motion
and results in a contracted arc of abduction. At the extremes of motion (i.e., flexion
and abduction), the flattened head-neck junction impinges on the acetabular labrum
and eventually osseous rim [19]. Known as femoroacetabular impingement (FAI),
the constant cycling of the labrum between the misshapen femoral head and acetab-
ulum results in accelerated wear and deterioration of the labrum. This cyclical dam-
age also results in consequent changes, including cysts, sclerosis, and microfracture,
in the underlying bone. Similarly, the pincer lesion represents an overgrowth of the
acetabular rim, particularly in the anterosuperior quadrant. The resulting functional
overcovering of the femoral head again can lead to a contracted arc of motion due
to impingement.
Regardless of the etiology of labral pathology, the loss of labral integrity results
in a breakdown of the articular seal and significantly increases the joint reactive
forces. Among other actions, the labrum acts as a gasket to seal the hip joint through-
out the motion. Upon loading, articular cartilage extrudes synovial fluid at a rate
determined by the adhesive and cohesive properties of the fluid and cartilage, the
pressure in the system, and the rate of loading [20]. The intact labrum allows the
pressure in the articular system to increase through a hydraulic effect providing a
transfer and dispersion of the applied force to the entire joint and joint fluid.
Consequently, loss of the labral seal results in a leak of the synovial fluid leading to
a significant decrease in the fluid pressure and causing increase in cartilage loading
rates, creep rates (i.e., extrusion of synovial fluid), and overall joint reactive forces
[21, 22]. Biomechanical modelings predict that the loss of labral integrity increases
11 Biomechanics of Human Motion 275
the contact stresses on the weight-bearing articular cartilage of the hip by up to 90%
[21]. Moreover, the loss of the hydraulic effect with labral pathology shifts the peak
pressures of the system laterally, resulting in an increase in overall joint reactive
force primarily over the lateral acetabular edge [21].
As we continue to elicit more details about the kinematics of the native hip, how
arthroplasty or resurfacing alters the mechanics of the hip remains largely unknown
and controversial. Current arthroplasty techniques require not only substitution of
the natural hip mechanics for those interjected by artificial implants but also altera-
tions in much other static and even dynamic stabilizers of the hip. Dramatic changes
in both dynamic and static stabilizers result from the surgical approach for
THA. However, many of the same factors that can affect native hip function also
plague the total arthroplasty function. Factors such as femoral neck offset to prevent
impingement, over- and under-coverage of the femoral head by the acetabular com-
ponent to prevent escape and dislocation, and also soft tissue balancing to prevent
translation and edge loading are all even more critical factors regarding to total hip
arthroplasty.
The hip is critical for some parameters of the gait cycle and is important in deter-
mining stride length, leg progression, and single-leg stance stability. Previous inves-
tigations utilizing external skin markers to evaluate primarily gait efficiency and
cadence demonstrated significantly reduced gait velocities as well as stride lengths
following THA. Kyriazis and Rigas [23] demonstrated that among 20 females with
severe hip OA, gait velocity and stride lengths were significantly lower than healthy
age-matched controls both before and after THA using external markers and force
plate analysis. However, they not only found a significant improvement in these
metrics in patients post THA compared to pre THA, but there was continued
improvement such that at 8–10 years follow-up, gait velocity and stride length
approached but did not reach that of healthy controls. In a similar vein, Wykman
et al. [24] reported a significant improvement of overall gait velocity, stride length,
and duration of single-leg stance of the affected hip following THA compared to
preoperation. They made the further distinction that this improvement was most
pronounced in patients with bilateral hip involvement though this benefit was most
notably observed after the second hip was replaced. Together, these studies suggest
that after THA, the affected hip has a significantly increased range of motion lead-
ing to the overall increase in the duration of the gait cycle and overall mechanical
efficiency. However, after THA, the significant improvement providing more effi-
cient hip mechanics and lower energy expenditures perhaps never quite reaches that
of the age-matched native hip [25, 26].
Though these studies provide intriguing information about the gait alterations
following THA, they are limited in part by the methods of obtained data; namely
surface mounted markers. More recently, Tsai et al. [16] describe THA kinematics
with the use of DFIS, directly evaluating implant position throughout the gait cycle.
276 R. Cheng et al.
1 10
Non-operated side
0
Operated side 0.5
0
-20 0
-10
-0.5
←ER/IR→ (º)
←M/L→ (mm)
←A/P→ Tilt (º)
-40 a d g
-1 -20
0 20 40 60 80 100 0 20 40 60 80 100 0 20 40 60 80 100
1 10
20
0.5 5
11 Biomechanics of Human Motion
10 0 0
-0.5 -5
←ER/IR→ (º)
←AB/AD→ (º)
0 b e h
-1 -10
40 0.5 5
0 0
20
-0.5 -5
←P/A→ (mm)
←EXT/FLEX→ (º)
←Lift/Drop→ (º)
0 c f i
-1 -10
0 20 40 60 80 100 0 20 40 60 80 100 0 20 40 60 80 100
Fig. 11.5 Average and standard deviation of hip kinematics for the operated and nonoperated hips in unilateral THA patients during gait. Significant differ-
ences in hip rotations, hip translations, and pelvic A/P tilt were observed. (Redrawn from Tsai, T.-Y., J.-S. Li, et al. Does component alignment affect gait
symmetry in unilateral total hip arthroplasty patients? Clinical Biomechanics, 2015. 30(8): 802–807)
277
278 R. Cheng et al.
situation of the maximal range of motion with maximal stability. Tsai et al. [32]
demonstrated that an average 5.1° increase in the internal rotation was observed in
the implanted hip than the contralateral non-implanted hip and internal rotation was
significantly correlated with a linear combination of the increase of cup anteversion,
medial cup translation, and leg-lengthening.
The knee joint comprises two distinctly separate joints/articulations, the tibiofemo-
ral (TF) joint and the patellofemoral (PF) joint. The main functions of the knee joint
are (1) to allow locomotion with (a) minimum energy requirements from the mus-
cles and (b) stability, accommodating for different terrains, and (2) to transmit,
absorb, and redistribute forces caused during the activities of daily life [33].
The tibiofemoral joint motion is movement with 6 degrees of freedom (DOF), three
in rotation, and three in translation. The three degrees of freedom in rotation are
flexion–extension (F–E) in the sagittal plane (primary), varus–valgus (V–V) (or
adduction–abduction) in the frontal plane and internal–external (I–E) rotation in the
horizontal plane. The three degrees of freedom in translation are anterior–posterior
(A–P) movement, mediolateral (M–L) translation, and compression. The 6 DOF
movement could be described in a clinical joint coordinate system (Fig. 11.6) [34].
Full extension (i.e., zero-degree flexion) is usually defined when the long axis of the
tibia and femur are aligned in the sagittal plane [33].
Flexion–Extension
The F–E axis has a different definition from many researchers. Morrison assumed
that a fixed axis of rotation was coincident with the axis of rotation of the knee joint
in the position of full extension [35]. The line through the center of medial and lat-
eral femoral condyles are defined as the F–E axis. Some surgeons used the transepi-
condylar axis (TEA) as the F–E rotation axis in total knee arthroplasty (TKA).
Berger defined surgical TEA as a line connecting the sulcus of the medial epicon-
dyle and the lateral epicondylar prominence [36]. The angle between the surgical
TEA and the posterior condylar line is defined as the posterior condylar angle.
Another TEA definition is called clinical TEA. It is defined as a line connecting the
medial and lateral epicondylar prominence by Yoshioka [37].
11 Biomechanics of Human Motion 279
Joint
distraction
F-E
A-P
V-V
M-L
I-E
Fig. 11.6 The six DOF of TF joint motion expressed in a clinical joint coordinate system. M–L
translation and F–E occur along and about an epicondylar femoral axis. Joint distraction and (I–E)
rotation occur along and about a tibial long axis. A–P translation and V–V rotation occur along and
about a floating axis, which is perpendicular to both femoral epicondylar and tibial long axes
(Reprinted with permission from Elsevier: Masouros, S., A. Bull, and A. Amis, (i) Biomechanics
of the knee joint. Orthopaedics Trauma, 2010. 24(2):84–91)
Internal–External Rotation
Many studies have shown that the I–E rotation axis is located near the center of the
medial tibial plateau. Wang et al. used human knee joint specimens to study the
effect of flexion and rotation on the length patterns of the ligaments of the knee.
Their results showed that the I–E rotation axis was located slightly posterior to the
center of the medial plateau for mid-flexion angles (30°, 60°, 90°), but at 0 and 120°
flexion, this axis passed through the mid-point of the medial spine of tibial emi-
nence [38]. These results were in line with Shaw and Murray’s research experimen-
tally showing that the I-E rotation axis passes through the medial spine of the
intercondylar eminence [39].
280 R. Cheng et al.
Varus–Valgus
Patella is the largest sesamoid bone in the human body. It is embedded in quadriceps
tendon and its function is to increase the mechanical leverage of the quadriceps. At
full knee extension, the patella contacts femur at the distal end of the patella
(Fig. 11.7a). As the flexion angle increases, the patella engages into the femoral
trochlear groove and the contact area spreads across the width of the patella and
moves proximally (Fig. 11.7b) [34]. The increase of PF contact area with knee flex-
ion is a clever mechanism that controls the magnitude of stresses by spreading the
increasing PF joint load with knee flexion over a larger area.
a PF b
Q PF
PT
Q
PT
Q
Q
PF
PF PT
PT
Fig. 11.7 PF joint force at (a) extension and (b) 90° flexion, showing geometrically the increase
of PF joint force with flexion. (PT patellar tendon, Q quadriceps muscles, PF patellofemoral, TF
tibiofemoral) (Reprinted with permission from Elsevier: Masouros, S., A. Bull, and A. Amis, (i)
Biomechanics of the knee joint. Orthopaedics Trauma, 2010. 24(2): 84–91)
11 Biomechanics of Human Motion 281
The soft tissues in the knee joint provide dynamic stability. The muscles stabilize
the joint according to the movement and provide stability of the joint. Ligaments
and meniscus provide passive stability. The ligament elongates as a response to
external force, and thus maintain the joint stability.
Two cruciate ligaments, anterior cruciate ligament (ACL) and posterior cruciate
ligament (PCL) are arranged in a crossed formation in the knee joint. The primary
function of ACL is to prevent excessive tibial anterior translation. The primal func-
tion of PCL is to restrain posterior tibial translation. Two cruciate ligaments act
together to control the A-P rolling and sliding of TF joint during F–E [42].
In the M–L direction, the two collateral ligaments stabilize the knee joint by
restraining the V–V and I–E rotation [34]. The medial collateral ligament (MCL) is
the primary restraint to valgus and internal tibial rotation [43]. The lateral collateral
ligament (LCL) is the primary restraint to varus angulation but slackens with knee
joint flexion, which reduces its restraining capability [44].
The main ligament in PFJ is medial patellofemoral ligament (MPFL). It acts as
the primary passive restraint to lateral patellar displacements and assists in control-
ling patellar motion, especially guiding the patella into the trochlear groove in early
knee flexion.
Gait analysis is based on an optical motion tracking system and is widely used in
investigating in vivo joint kinematics and kinetics. Gait cycle could be divided into
two phases, stance phase and swing phase. The stance phase is defined as the period
between heel strike and toe off. Swing phase is defined as the period between toe off
and next heel strike. In gait analysis, healthy knee joint motion features a large sag-
ittal plane range of motion (ROM), small rotation in the horizontal plane and frontal
plane. The knee joint kinematics of healthy adults is shown in Fig. 11.8 [45].
For F–E angles, there are two peaks in one gait cycle. The first peak is smaller
and in the mid-stance phase, which has a value of 20° flexion. The second peak is
larger and in the early stage of the stance phase, which has a value of 60° flexion.
In the frontal plane, knee joint movements involve both abduction and adduction.
During the stance phase, the knee joint undergoes abduction. After the start of the
swing phase, the knee joint transform to adduction. At the end of the swing phase,
the knee joint turns back to the abduction. Overall, the abduction/adduction angle is
small during the gait cycle.
In the horizontal plane, at the beginning of the stance phase, tibia has a slight
external rotation according to the femur. After heel strike, both femur and tibia
undergo internal rotation. The tibia has a faster speed of internal rotation. As a
result, the knee joint has an internal rotation in the stance phase. After the middle-
stance phase, both femur and tibia undergo external rotation and keep external rotat-
ing until the middle-swing phase.
282 R. Cheng et al.
60
40
flexion/extension. deg.
f6
f5
20
f4
f3
f2
0
f1
–20
10
5
q1 q2
rotation.deg. abd/add. deg.
–5
10
Y2
0
Y1
–5
0 20 40 60 80 100
% of gait cycle
stance swing
Fig. 11.8 Chao et al. evaluated 148 subjects’ lower limb gait. All the subjects were allowed to
walk at their preferred speed and wore shoes [45] (Reprinted with permission from Elsevier: Chao,
E., R. Laughman, et al. Normative data of knee joint motion and ground reaction forces in adult
level walking. 1983. 16(3): 219–233)
11 Biomechanics of Human Motion 283
Knee Osteoarthritis
Osteoarthritis (OA) is one of the most common sources of locomotor disability and
musculoskeletal pain worldwide, and the prevalence is as high as over 40% in older
adults [46]. Factors that may increase the risk of osteoarthritis include older age,
sex, obesity, joint injuries, and deformities. Accepted by most surgeons, knee OA
patients are classified into two categories: (1) idiopathic: those with no presently
known prior event or disease related to the OA; and (2) secondary: those with known
events or disease associated with OA. The knee OA patients can be further divided
into five stages: (1) stage 0: no radiographical signs or pain; (2) stage 1: minor bone
spur growth in X-rays of knee joints; (3) stage 2: greater bone spur growth and
healthy size of cartilage; (4) stage 3: damaged cartilage and minor narrow space
between bones; and (5) stage 4: the cartilage is completely gone and the joint space
between bones is dramatically reduced. Osteoarthritis symptoms often develop
slowly and worsen over time. Knee OA patients always have signs of tenderness,
loss of flexibility, grating sensation, stiffness, and pain. The biomechanical behavior
of knee OA joints is correspondingly changed compared to normal knees, especially
in end-stage knee OA patients. Kinematics and kinetics parameters are crucial for
the evaluation of osteoarthritis severity and treatment outcomes.
gression of osteoarthritis. The decreases of hip internal rotation and ankle dor-
siflexion angle from late stance to early swing were also found in the progression
of knee OA [52].
The change of joint loading has a crucial effect on articular cartilage. Native joint stress
should be homogeneously distributed under proper lubrication. The increase of joint
loading and repeated impact of joint are considered to induce joint wear and decide the
severity of knee OA. Uneven stress distribution may cause the damage and loss of
articular cartilage and the secondary degeneration of knee osteoarthritis. External knee
adduction moment, especially the mid-stance knee adduction moment, is a key param-
eter, which is closely associated with joint loading. Two knee adduction moment peaks
showed in the early and late stance phase. The first knee adduction moment peak is of
great clinical significance because it is correlated with cartilage thickness and joint
loading [53]. In the past decade, the mid-stance knee adduction moment attracted more
and more attention, which is not dependent on the stride velocity and is taken as an
ideal indication for knee OA diagnosis. Expect for the operative management; gait
retraining is an effective treatment for correct knee adduction moment. So far, several
gait patterns were reported to reduce the adduction moment, such as toe-out gait, slow
velocity, stride length reduction, side swing, and so on [54–56].
Through the progression of knee OA, conservative treatment would have few effects
on the patients with tremendous pain, deformities, and disabilities. Total knee
arthroplasty (TKA) is the most effective surgical interventions for pain relief and
functional recovery in patients with advanced degenerative arthritis or rheumatoid
arthritis [57]. Aging of the society has led to increase in the prevalence of arthritis
and the incidence of TKA for end-stage arthritis. With the development of TKA
techniques and design, the demand for TKA increased in the past 20 years, and over
1300,000 patients took TKA procedures annually [58]. TKA has three components:
femoral components, tibial components, and patellar components. Following differ-
ent criteria, TKA prosthesis can be classified as (1) cruciate retaining, and posterior
stabilized prosthesis; (2) mobile-bearing and fixed bearing prosthesis; (3) cemented
and uncemented prosthesis. The knee prosthesis can be further divided into medial
pivot, single radius, and multi-radius based on geometric design.
Although total knee arthroplasty has been developed for many years, complica-
tion rates are still high after primary TKA procedure [59]. John Insall proposed that
total knee arthroplasty should meet the following requirements: (1) correct align-
ment axes; (2) equivalent joint gap during flexion and extension; (3) balanced col-
lateral ligament tension; (4) –3° to 130° of knee flexion; (5) 3 mm changes of joint
line; and (6) balanced patellar trajectories. These biomechanical factors should be
taken into consideration to avoid secondary complication like polyethylene wear,
11 Biomechanics of Human Motion 285
With the retaining of the PCL, CR prosthesis is considered to restore normal knee
biomechanical function compared to PS prosthesis. PCL retention in total knee
replacement has the advantages of enhanced inherent stability, a greater range of
motion, improved proprioception, and increased rollback associated with the native
PCL retention and joint line. Tsai et al. investigated the articular cartilage contact
kinematics of the knee before and after a cruciate retaining arthroplasty. In their
study, the distances between medial and lateral contact locations in TKA knees
were significantly larger than those of the OA knees (Fig. 11.9) [60]. Further, the
50%
Native
40% Knee
30% TKA
Anterior
0%
-10%
OA
Posterior
-20%
TKA
-30%
-40%
-50%
-50% -40% -30% -20% -10% 0% 10% 20% 30% 40% 50%
Medial Lateral
Fig. 11.9 Average medial and lateral articular contact locations and the center points between
medial and lateral contacts of the knees before and after TKA operations were depicted. The solid
and dotted lines represent the distances between medial and lateral contact locations at full exten-
sion and maximal flexion, respectively (Reprinted with permission from John Wiley and Sons: Li,
C., A. Hosseini, T.Y. Tsai, et al. Articular contact kinematics of the knee before and after a cruciate
retaining total knee arthroplasty. J Orthop Res, 2015. 33(3):349–358)
286 R. Cheng et al.
contact points on the medial and lateral compartments were more laterally located
than those of the OA knees. The centers of medial and lateral contact locations have
shifted from the medial side of the tibial plateau of the OA knees to the lateral side
of the TKA knees. The bearing surface design of polyethylene liner has been recog-
nized as a major factor that could affect the postoperative kinematics and contact
patterns of the knee. Failure of polyethylene liner has been reported to account for
approximately 50% of CR-TKA revision procedures. The small contact locations in
the TKA knees found in postoperative patients imply that the contact forces induced
by repeated sliding and rolling during knee motion were localized in these small
medial and lateral contact areas, which may induce unfavorable surface fatigue wear.
In the normal knees, the medial and lateral femoral condyles roll backward to
assist further knee flexion as the degree of flexion increases. However, CR prosthe-
sis was reported to show evidence of the paradoxical anterior femoral movement
during mid-flexion [61]. Improved CR prosthesis with femoral component oversiz-
ing and reducing femoral radius can control the paradoxical anterior femoral
movement efficiently [62]. The survivorship of improved CR prosthesis was up to
94% after 10 years follow-ups [63].
For the patients with injured/contracted/degenerated PCL, over 20° of flexion con-
tracture. Without the cruciate ligament retaining, the PS total knee arthroplasty is
more applicable in advanced stage patients. Due to the reduction in the difficulty of
surgery, the PS prosthesis is widely used in the clinical field. The PCL substituted
with a post-cam device, which constrains the posterior femoral offset at 70° of knee
flexion and helps the knee joint to roll back during flexion. The PS prosthesis is
easier to realize gap balance and allow minor changes of joint line, for which the
prosthesis wear can be reduced. Tsai et al. compared the in vivo kinematics of the
knee after a posterior cruciate-substituting total knee arthroplasty between Caucasian
and South Korean patients. Paradoxical anterior translation of contact points was
observed from 45° to 90° of flexion in the Caucasian patients, whereas contact
points of the South Korean patients moved posteriorly from full extension to 60° of
flexion and then remained relatively constant until 90° of flexion. South Korean
TKA patients had more anterior contact points on the medial compartment, more
lateral contact points on the lateral compartment, and bicondylar femoral posterior
translation rather than medial pivot pattern compared to the Caucasian TKA patients.
Different tibiofemoral articular contact kinematics should be considered for the
design of bearing articular surface in Asian patients (Fig. 11.10).
Four important ligaments stabilize the knee joint, including the anterior cruciate
ligament (ACL), posterior cruciate ligament (PCL), medial collateral ligament
(MCL), and lateral collateral ligament (LCL). Each separate ligament prevents
11 Biomechanics of Human Motion 287
30
Fig. 11.10 Contact kinematics of Caucasian and South Korean total knee arthroplasty (TKA)
knees in a mediolateral translation. (a) Normalized femoral condylar motions. (b) Normalized
tibiofemoral contact kinematics. Positive values mean medial translation. ∗p < 0.05 (Reprinted
with permission from The Knee Surgery and Related Research: Bae, J. H., et al. In vivo Kinematics
of the Knee after a Posterior Cruciate-Substituting Total Knee Arthroplasty: A Comparison
between Caucasian and South Korean Patients. Knee surgery & related research, 2016. 28(2):
110–117)
excessive motion of the knee. When some of the ligament was damaged, the knee
would become unstable in the direction that the injured ligament stabilized. Different
types of physical tests in clinical practice (anterior drawer test, posterior drawer test,
etc.) are based on the abnormal behavior of knee after ligament injuries. The most
common injured ligaments in the clinical field are ACL and PCL. This section will
focus on the kinematics and kinetics characteristics after ACL/PCL injury and
reconstruction.
ACL is one of the major knee stabilizers, which constrains not only the anterior
tibial translation but also the internal-external rotation stability. Zhao et al. investi-
gated the kinematics of ACL-deficient patients and healthy subject in the Chinese
population. In their study, individuals with ACL deficiency exhibit greater knee
internal rotation during higher demand activities, such as ascending and descending
steps or jogging. Compared to Caucasian population, Asian patients showed smaller
tibial varus [64]. An anomalous tibial rotation will change the loading distribution
on the meniscus and cause secondary meniscal damage. Zhang et al. reported that
ACL deficiency with meniscal injury would alter the kinematics when compared to
isolated ACL injury [65].
Previous studies showed that after ACL tear more than 50% of individuals
showed radiographic evidence of osteoarthritis 10–12 years post-injury [66, 67]. An
ACL reconstruction is one of the standard treatments for ACL injury. Operative
treatment showed superior outcomes for knee symptoms and function, and in knee-
specific and health-related quality of life, compared to patients who chose nonsurgi-
288 R. Cheng et al.
-5.0 -15.0
0.0 20.0 40.0 60.0 80.0 100.0 0.0 20.0 40.0 60.0 80.0 100.0
% of step-up % of step-up
c d
-15.0 20.0
Medial (mm) Lateral
-20.0 15.0
-25.0 10.0
0.0 20.0 40.0 60.0 80.0 100.0 0.0 20.0 40.0 60.0 80.0 100.0
% of step-up % of step-up
Intact Pre-op 6M Post-op 36m Post-op
Fig. 11.11 Cartilage contact locations after ACL reconstruction during the step-up motion.
Anterior-posterior translation of cartilage contact locations on the (a) medial and (b) lateral tibial
plateau; Medial-lateral translation of the cartilage contact locations on (c) medial and (d) lateral
tibial plateau (Reprinted with permission from Elsevier: Lin, L., J.-S. Li, W.A. Kernkamp,
A. Hosseini, C. Kim, P. Yin, L. Wang, T.-Y. Tsai, et al. Postoperative time-dependent tibiofemoral
articular cartilage contact kinematics during step-up after ACL reconstruction. Journal of biome-
chanics, 2016. 49(14): 3509–3515)
cal treatment [68]. Tsai et al. measured the articular cartilage contact kinematics
after ACL reconstruction (Fig. 11.11) [69]. This study showed that the tibiofemoral
cartilage contact locations of the knee changes with time after ACL reconstruction.
Li also measured the tibiofemoral cartilage contact biomechanics in patients after
ACL reconstruction. These research revealed that the abnormal posterior and lateral
shift of cartilage contact location to smaller regions of thinner cartilage seen in
ACL-deficient knees persisted in the ACL-reconstructed knees around 0° and 15°,
resulting in a persistent increase of cartilage contact deformation at those flexion
angles [70]. Kernkamp WA et al. predicted the isometry and strain for ACL recon-
struction, and an area of least anisometry was found in the proximal-distal direction
just posterior to the intercondylar notch (Fig. 11.12). Moving the femoral socket
positions in the anterior-posterior direction affected the theoretical ACL strains
significantly. Posterior femoral attachments resulted in decreased lengths with
increasing flexion angles, whereas anterior-distal grafts increased in length with
increasing flexion angles.
11 Biomechanics of Human Motion 289
a >18
16
14
12
Strain (%)
10
8
6
4
2
0
Anteromedial Central Posterolateral
b >18
16
14
12
Strain (%)
10
8
6
4
2
Fig. 11.12 Medial view of a 3D femur model in 90° of flexion. The “heat map” illustrates the
isometry distribution (mean maximum strain-minimum strain) over the medial aspect of the lateral
femoral condyle for single point-to-point curves when connected to the anteromedial, central, or
posterolateral tibial attachment during the dynamic step-up (a) and sit-to-stand motion (b). The
darkest blue area on the femur represents the most isometric attachment area, whereas the red areas
highlight those with a high degree of anisometry. Specifically, the circle represents the most iso-
metric attachment. The black cross (×) on the femur shows the “over the top” position as would be
achieved by transtibial drilling; the black dot shows the center of the ACL footprint as described
by Parkar et al. (Reprinted with permission from Elsevier: Kernkamp, W.A., A.J.T. Jens,
N.H. Varady, E.R.A. van Arkel, et al. Anatomic is better than isometric posterior cruciate ligament
tunnel placement based upon in vivo simulation. Knee Surg Sports Traumatol Arthrosc, 2018)
PCL injuries occupy up to 44% of all acute knee injuries, but the prevalence of
isolated PCL injuries (PCL injuries in which no other ligamentous injuries
require repair and reconstruction) was reported as little as 3.5% [71, 72]. In vivo
and in vitro drawer tests reported that PCL is the primary restraint to the poste-
rior translation of the knee [73, 74]. Li performed an in vivo kinematics study on
PCL-deficient knees compared with contralateral knees, data of which demon-
strated that PCL injuries caused an increase of about 5 mm posterior tibial trans-
lation through all the flexion angle and an increase of about 1 mm lateral tibial
translation at 90° of knee flexion [75]. Anatomic studies showed that PCL orien-
tated anteromedially on femur through the whole range of knee flexion. This
structure implies that PCL injury is not only a loss of posterior stabilizer but also
290 R. Cheng et al.
a medial-lateral stabilizer. The abnormal contact pattern along with the ACL-
deficient kinematics characteristics will yield secondary joint degeneration when
treated nonoperatively.
Isolated PCL injuries have been treated nonoperatively and often have good
functional results, but the 5-year incidence of osteoarthritis in nonsurgically treated
PCL-deficient knees has been reported to be up to 80% and 50% on the medial
femoral condyle and patella, respectively [76–78]. Tunnel selection is a crucial
parameter for PCL reconstruction to restore native PCL function. Kernkamp et al.
simulated the PCL elongation with in vivo kinematics and found that too proximal
(i.e., nonanatomical) femoral attachments are unable to replicate anatomical graft
length changes [79]. However, recent studies reported that isolated PCL reconstruc-
tion could not restore either anteroposterior or rotational stability compared to the
healthy knee. A supplemental surgical technique is required to overcome residual
external rotation instabilities, varus laxity [80].
The spine is made up of the vertebrae and the intervertebral discs, facet joints, and
ligaments, forming the central axis of the human body, and transmitting the load as
well as protecting the spinal cord. The spine provides motion functions for trunk
such as three-dimensional physiological activities.
3.1.1 Vertebral Body
The vertebral body is mainly composed of cancellous bone, and the surface is a
thin layer of cortical bone [81, 82]. It presents as a short cylinder with a slightly
thin central portion and a large expansion at both ends. The upper and lower sides
are rough and can be divided into two areas: the central part is concave and porous,
filled by the cartilage onto the edge; the edge part bulges and is cortical bone,
firmly attached to the intervertebral disc. The vertebral body is a complicated struc-
ture composed of cartilage endplates, cancellous bones, and cortical bones. The
function of these different ingredients contributes to their unique biomechanical
properties. Vertebrae rely on their natural structure to resist various stresses.
Mainly, the vertebrae are almost composed of cancellous bone (net structure)
entirely, but a hard shell covers the outer part. Since the axial stress of the vertebra
presents the largest, the trabecular bone along the vertical direction exhibits the
hardest. The trabecular bones are arranged along the direction of the normal stress
(axial force) to exert their biological function as much as possible. The horizontal
trabecular bone acts as a lateral bracket to prevent vertical deformation of the tra-
becular bone.
11 Biomechanics of Human Motion 291
The vertebral body primarily sustains compressive forces. As the weight of the
vertebral body increases from top to bottom, the vertebral body also becomes larger
from top to bottom. For example, the shape of the lumbar vertebral body is thicker
and broader than that of the thoracic and the cervical vertebrae and is sustained by
a larger load. The mechanical properties of the vertebral body are related to the
anatomical features and bone masses.
3.1.2 Intervertebral Disc
The intervertebral disc is located between adjacent vertebral bodies, and the adjacent
vertebral bodies are firmly connected aiming to maintain the arrangement of the
vertebral canals [81, 83]. The thickness of the intervertebral disc is about one-fourth
of the total length of the spine above the sacrum. Intervertebral disc is a kind of solid
viscoelastic material, which has the characteristics of creep, relaxation, and hyster-
esis, and can absorb the energy of vibration. Under a slight load, the deformation
disappears after unloading. If the load is too excessive, the irreversible deformation
will occur. The intervertebral disc consists of three parts: nucleus pulposus, fibrous
annulus, and cartilage endplate. The nucleus pulposus is a liquid mass located in the
center of the intervertebral disc. The lower lumbar vertebra is more backward. It
contains a large amount of hydrophilic glucosamine and present as gelatinous tissue.
Its water content varies greatly with age and load. When young, the water content is
up to 90%, but with the aging of people, the water content decreases gradually.
Under compression load, water in nucleus pulposus seeps through the endplate, and
the volume of nucleus pulposus decreases. When the load decreases, water returns
and the volume of nucleus pulposus recovers. A fibrous annulus is composed of
fibrocartilage tissue. There are many intersecting collagen bundles in fibrocartilage.
The unique alignment of annular fibers helps the intervertebral disc maintain a cer-
tain degree of torsion resistance. The posterior part of the fibrous ring is woven by
the posterior longitudinal ligament (PLL). The cartilage endplate between the verte-
bral body and the intervertebral disc is composed of hyaline cartilage.
It is an essential biomechanical function of an intervertebral disc that it can bear
and distribute the loads and restrict excessive activity. Compression load acts on
nucleus pulposus and annulus fibrosus through the end plate. Hydraulic pressure
generated inside the nucleus pulposus causes annulus fibrosus to expand outward.
The outer annulus fibrosus bears the maximum tensile stress. The inner annulus
fibrosus bears less tensile stress than the outer one, but it bears a part of compres-
sive stress.
The type of the spinal segment motion depends on the direction of the articular
surface of the intervertebral joint, and the direction of the articular surface varies
among the whole spinal vertebrae [81, 83]. The joint plane of the lower cervical
292 R. Cheng et al.
spine is parallel to the coronal plane and 45° to the horizontal plane. It allows the
cervical spine to flex forward, extend backward, bend laterally, rotate, and extend to
a certain extent. The articular surface of the lumbar spine is perpendicular to the
horizontal plane and 45° to the coronal plane, allowing flexion, extension, and lat-
eral bending, but limiting rotation.
In addition to guiding segmental motion, the facet joints also bear different kinds
of loads, such as compressive force, tensile force, shear force, and torsion force. The
amount of loads they bear vary with the different movements of the spine. The load
on the facets of the joints is the greatest at extension, accounting for 30% of the total
load (another 70% of the load from the intervertebral disc). The load on the facet
joint is also higher when flexion and rotation occur. The tensile loading of facet joints
mainly occurs as the lumbar forward bending. When the lumbar forward bending
reaches the maximum limitation, 39% of the tension load is borne by facet joints.
3.1.4 Ligament
The main components of the ligament are collagen fibers and elastic fibers [81]. The
collagen fibers give the ligaments a certain strength and rigidity, and the elastic fibers
give the ligaments the ability to extend under the loading. Most of the fibers of the liga-
ment are nearly parallel [83, 84], so their functions are more specific and often only
bear the loading along a single direction. The function of the spinal ligaments is to
provide a proper physiological activity for adjacent spines, as well as maintain the
stability of the spine. The specimen of the spine in vitro still maintains a certain inter-
vertebral disc pressure under the traction load. The prestress is derived from the tension
of the ligament and is most prominent in the ligament flavum. All ligaments possess the
ability to resist tension, but they are fatigued quickly under compressive forces. The
ligament strength is closely related to the cross-sectional area of the ligament.
The ligaments of the spine bear most of the distraction loads of the spine. They
act like rubber bands. When the load direction is consistent with the fiber direction,
the ligaments have the strongest bearing capacity. When the spinal motion segments
are applied by different forces and moments, the corresponding ligaments are
stretched and stabilized for the motion segments.
It is generally believed that the anterior longitudinal ligament (ALL) is the tough-
est, and together with the PLL, can prevent the spine from excessive flexion and
extension, but the effect of limiting axial rotation and lateral flexion is not obvious.
The facet joint capsule ligament plays a key role in resisting torsion and lateral flex-
ion. The main function of the interspinous ligament is to stabilize the spine by limit-
ing the ability to flex.
3.1.5 Muscle
The dynamic stability of the spine is correlated with the function of the complete
paraspinal muscle during activities. The muscles associated with the spinal activity
can be divided into the front and back groups according to their locations [85]. The
11 Biomechanics of Human Motion 293
muscles located behind the lumbar vertebrae can be further categorized into three
groups: the deep layer, the middle layer, and the superficial layer [81, 83, 84]. The
deep muscles include the interspinous muscles that end at the adjacent spinous pro-
cesses, the intertransverse muscles that start at the adjacent transverse processes,
and the rotator muscles that start and end at the transverse processes and spinous
processes. The middle layer muscles mainly refer to the multifidus muscles of the
transverse process and the spinous process of the upper vertebral body. The superfi-
cial muscles can be divided into three groups: iliocostalis, longissimus, and spinalis.
a b
Fig. 11.13 (a) Experimental set-up of dynamic in vivo imaging using the DFIS. (b) Virtual DFIS
used to recreate intervertebral kinematics throughout the dynamic motion (Reprinted with permis-
sion from Elsevier: Zhong, W., S.J. Driscoll, T.Y. Tsai, et al. In vivo dynamic changes of dimen-
sions in the lumbar intervertebral foramen. Spine J, 2015. 15(7): 1653–9)
than during the LRL rotation at each intervertebral level. During LRL rotation, the
later COR corresponds to a smaller ROM. The data obtained from this study can
provide insight into the improvement of motor-preserving prosthesis design and
surgical implantation techniques designed to restore normal neck function and pre-
vent adjacent segmental degenerative cervical surgery.
In vivo studies of the lumbar spine showed that during weightlifting, L4-5 and
L5-S1 of lower lumbar segments showed larger anterior-posterior (AP) and
proximal-distal (PD) translations than higher vertebral segments, respectively [89,
90]. Body and lumbar foramen size showed segmental characteristics during
dynamic weightlifting. Also, the movement behavior of the upper lumbar facet
joints (L2-L3 and L3-L4) was similar but different from that of the lower lumbar
joints (L4-L5).
Spinal degenerative diseases are most common among all spinal diseases, and disc
herniation caused by degenerative disc disease is the most common cause of pain in
the lower back and neck [91, 92].
Degenerative disc disease’s symptom is usually lower back or neck pain caused
by aging and wear of the disc. When the herniated disc tissue compresses the spinal
nerves or the spinal cord, it can also cause weakness in the arms or legs, numbness
and burning shot pain (radicular pain) [93, 94]. Cervical and lumbar vertebrae
undergo maximum exercise and pressure and are most susceptible to disc degenera-
tion, so there is a high probability of painful disc degeneration. Disc degeneration is
not a disease and usually does not cause long-term disability, and most cases can be
11 Biomechanics of Human Motion 295
treated with nonsurgical approach. Degenerative disc disease varies in nature and
severity. Degenerative discs do not always cause pain or other symptoms. Because
the intervertebral disc itself is short of innervation, it causes or accelerates the onset
of other spinal disorders when the degenerated disc affects other structures in the
spine, such as muscles, joints, or nerve roots.
Discogenic pain usually results from two main factors: One is inflammation.
When the disc is degraded, inflammatory factors from the interior of the disc may
leak, causing swelling of the surrounding spine. Therefore, inflammation causes
muscle tension, muscle spasm, and tenderness in the back or neck. Pain and numb-
ness may radiate to the arms and shoulders (the symptom of cervical radiculopathy),
or into the buttocks or legs (the symptom of lumbar disc herniation), due to inflamed
nerve roots.
The other is unstable micro-motion. When the outer layer (fibrous ring) of the
disc degrade, the cushioning and support of the intervertebral disc usually reduce,
resulting in small, unnatural movements between the vertebrae. Unstable spinal seg-
ments can induce micro-motions and cause tension and irritation in the surrounding
muscles, joints, and nerve roots, leading to recurrent severe pain episodes. In most
cases, however, the disc herniation itself is not painful, but the tissue leaking from
the intervertebral disc, inflaming or stimulating nearby nerves, causes nerve root
pain. Leg pain from sensitive nerves is often referred to as sciatica.
Spondylolisthesis is a forward translation of a vertebral segment below which a
vertebral segment is located. Typical symptoms are an activity-related history of
low back pain and painful spinal activity and hamstring tension without radiculopa-
thy sign and imaging. Plain films, computed tomography, and single photon emis-
sion computed tomography are useful in determining the diagnosis. The best way to
treat the symptomatic stress response of the spinal joint or adjacent vertebral struc-
tures is to immobilize the spine and activity restriction.
3.3.2 Spinal Fusion
The spinal surgery usually applies a rigid internal fixation device to secure the spine,
aiming to correct deformities, relieve pain, stabilize the spine, and protect the nerves
[95, 96]. Spinal fusion surgery includes a variety of surgical procedures: for example,
lumbar fusion, cervical fusion, and OLF [91]. Although each surgery is different, they
are all designed to help relieve pain or neurological symptoms caused by the interver-
tebral joints, depending on whether the patient is undergoing surgery for degenerative
disc disease, lumbar spondylolisthesis, or other diseases. During spinal fusion sur-
gery, two adjacent vertebrae are combined to eliminate the underlying mechanism of
pain. Fusion of joints eliminate instability of the spinal segments and reduce pain
caused by micro-motion, muscle tension, and inflammation. Moreover, the most
important thing is that fusion can solve the nerve compression problem completely.
Fusion surgery establishes a mechanism of bone growth that occurs months after
surgery. For this reason, the complete recovery process for fusion surgery can last up
to a year, although most patients return to normal activity within 6 weeks.
296 R. Cheng et al.
It is recommended to use a back or neck brace to keep the spine stable and to
minimize painful movements that may disrupt the healing process after surgery.
Also, rehabilitation (physical therapy for example) is often recommended to modu-
late the muscles to support the spine better and to provide painkillers to control
postoperative pain when necessary [97].
References
1. Fuss FK, Bacher A. New aspects of the morphology and function of the human hip joint liga-
ments. Am J Anat. 1991;192(1):1–13.
2. Levangie PK, Norkin CC. Joint structure and function: a comprehensive analysis. 3rd ed.
Philadelphia: F.A. Davis Company; 2000.
3. Heller MO, Bergmann G, Deuretzbacher G, Durselen L, Pohl M, Claes L, Haas NP and Duda
GN. Musculo-skeletal loading conditions at the hip during walking and stair climbing. J
Biomech. 2001;34:883–93.
4. Heller MO, Bergmann G, Deuretzbacher G, Claes L, Haas NP, Duda GN. Influence of femoral
anteversion on proximal femoral loading: measurement and simulation in four patients. Clin
Biomech. 2001;16(8):644–9.
5. Turley GA, Williams MA, Wellings RM, Griffin DR. Evaluation of range of motion restriction
within the hip joint. Med Biol Eng Comput. 2013;51(4):467–77.
6. Crowninshield RD, Johnston RC, Brand RA, Pedersen DR. Pathologic ligamentous constraint
of the hip. Clin Orthop Relat Res. 1983;(181):291–7.
7. Wu G, Siegler S, Allard P, Kirtley C, Leardini A, Rosenbaum D, Whittle M, D’Lima DD,
Cristofolini L, Witte HJJob. ISB recommendation on definitions of joint coordinate system
of various joints for the reporting of human joint motion—Part I: ankle, hip, and spine. J
Biomech. 2002;35(4):543–8.
8. Dandachli W, Islam SU, Richards R, Hall-Craggs M, Witt JJHI. The influence of pelvic tilt on
acetabular orientation and cover: a three-dimensional computerised tomography analysis. Hip
Int. 2013;23(1):87–92.
9. Ferguson S, Bryant JT, Ganz R, Ito K. An in vitro investigation of the acetabular labral seal in
hip joint mechanics. J Biomech. 2003;36(2):171–8.
10. Lombardi AV Jr, Mallory TH, Dennis DA, Komistek RD, Fada RA, Northcut EJ. An
in vivo determination of total hip arthroplasty pistoning during activity. J Arthroplast.
2000;15(6):702–9.
11. Tanino H, Ito H, Harman MK, Matsuno T, Hodge WA, Banks SA. An in vivo model for intra-
operative assessment of impingement and dislocation in total hip arthroplasty. J Arthroplast.
2008;23(5):714–20.
12. Koyanagi J, Sakai T, Yamazaki T, Watanabe T, Akiyama K, Sugano N, Yoshikawa H,
Sugamoto K. In vivo kinematic analysis of squatting after total hip arthroplasty. Clin Biomech.
2011;26(5):477–83.
13. Li G, Wuerz TH, DeFrate LE. Feasibility of using orthogonal fluoroscopic images to measure
in vivo joint kinematics. J Biomech Eng. 2004;126(2):313–8.
14. Gronenschild E. The accuracy and reproducibility of a global method to correct for geometric
image distortion in the X-ray imaging chain. Med Phys. 1997;24(12):1875–88.
15. Gronenschild E. Correction for geometric image distortion in the X-ray imaging chain: local
technique versus global technique. Med Phys. 1999;26(12):2602–16.
16. Tsai T-Y, Li J-S, Wang S, Lin H, Malchau H, Li G, Rubash H, Kwon Y-M. A novel dual fluo-
roscopic imaging method for determination of THA kinematics: in-vitro and in-vivo study. J
Biomech. 2013;46(7):1300–4.
11 Biomechanics of Human Motion 297
17. Lin H, Wang S, Tsai T-Y, Li G, Kwon Y-M. In-vitro validation of a non-invasive dual flu-
oroscopic imaging technique for measurement of the hip kinematics. Med Eng Phys.
2013;35(3):411–6.
18. Ito H, Song Y, Lindsey DP, Safran MR, Giori NJ. The proximal hip joint capsule and the zona
orbicularis contribute to hip joint stability in distraction. J Orthop Res. 2009;27(8):989–95.
19. Bedi A, Lynch EB, Sibilsky Enselman ER, Davis ME, DeWolf PD, Makki TA, Kelly BT,
Larson CM, Henning PT, Mendias CL. Elevation in circulating biomarkers of cartilage dam-
age and inflammation in athletes with femoroacetabular impingement. J Bone Joint Surg.
2013;41(11):2585–90.
20. Soltz MA, Ateshian GA. Experimental verification and theoretical prediction of cartilage
interstitial fluid pressurization at an impermeable contact interface in confined compression. J
Biomech. 1998;31(10):927–34.
21. Ferguson S, Bryant J, Ganz R, Ito K. The acetabular labrum seal: a poroelastic finite element
model. Clin Biomech. 2000;15(6):463–8.
22. Ferguson S, Bryant J, Ganz R, Ito K. The influence of the acetabular labrum on hip joint carti-
lage consolidation: a poroelastic finite element model. J Biomech. 2000;33(8):953–60.
23. Kyriazis V, Rigas C. Temporal gait analysis of hip osteoarthritic patients operated with cement-
less hip replacement. Clin Biomech. 2002;17(4):318–21.
24. Wykman A, Olsson E. Walking ability after total hip replacement. A comparison of gait analy-
sis in unilateral and bilateral cases. J Bone Joint Surg. 1992;74(1):53–6.
25. Arborelius MM, Carlsson A, Nilsson B. Oxygen intake and walking speed before and after
total hip replacement. Clin Orthop Relat Res. 1976;121:113–5.
26. Bennett D, Humphreys L, O’brien S, Kelly C, Orr J, Beverland D. Gait kinematics of age-
stratified hip replacement patients—a large scale, long-term follow-up study. Gait Posture.
2008;28(2):194–200.
27. Tsai T-Y, Dimitriou D, Li J-S, Nam KW, Li G, Kwon Y-M. Asymmetric hip kinematics during
gait in patients with unilateral total hip arthroplasty: in vivo 3-dimensional motion analysis. J
Biomech. 2015;48(4):555–9.
28. Blumenfeld TJ, Glaser DA, Bargar WL, Langston GD, Mahfouz MR, Komistek RDJO. In vivo
assessment of total hip femoral head separation from the acetabular cup during 4 common
daily activities. Orthopedics. 2011;34(6):e127–32.
29. Kung PL, Ries MD. Effect of femoral head size and abductors on dislocation after revision
THA. Clin Orthop Relat Res. 2007;465:170–4.
30. Nevelos J, Johnson A, Heffernan C, Macintyre J, Markel DC, Mont MA. What factors affect
posterior dislocation distance in THA? Clin Orthop Relat Res. 2013;471(2):519–26.
31. D’lima DD, Urquhart AG, Buehler KO, Walker RH, Colwell CW. The effect of the orientation
of the acetabular and femoral components on the range of motion of the hip at different head-
neck ratios. J Bone Joint Surg. 2000;82(3):315–21.
32. Tsai T-Y, Li J-S, Dimitriou D, Kwon Y-M. Does component alignment affect gait symmetry in
unilateral total hip arthroplasty patients? Clin Biomech. 2015;30(8):802–7.
33. Masouros S, Bull A, Amis A. Biomechanics of the knee joint. Orthopaed Trauma.
2010;24(2):84–91.
34. Wu G, Cavanagh PR. ISB recommendations for standardization in the reporting of kinematic
data. J Biomech. 1995;28(10):1257–61.
35. Morrison J. The mechanics of the knee joint in relation to normal walking. J Biomech.
1970;3(1):51–61.
36. Berger RA, Rubash HE, Seel MJ, Thompson WH, Crossett LS. Determining the rotational
alignment of the femoral component in total knee arthroplasty using the epicondylar axis. J
Clin Orthopaed Relat Res. 1993;(286):40–7.
37. Yoshioka Y, Siu D, Cooke T. The anatomy and functional axes of the femur. J Bone Joint Surg
Am. 1987;69(6):873–80.
38. Wang C-J, Walker PS, Wolf B. The effects of flexion and rotation on the length patterns of the
ligaments of the knee. J Biomech. 1973;6(6):587–96.
298 R. Cheng et al.
39. Shaw JA, Murray DG. The longitudinal axis of the knee and the role of the cruciate ligaments
in controlling transverse rotation. J Bone Joint Surg Am. 1974;56(8):1603–9.
40. Crowninshield R, Pope M, Johnson R. An analytical model of the knee. J Biomech.
1976;9(6):397–405.
41. Pennock G, Clark K. An anatomy-based coordinate system for the description of the kinematic
displacements in the human knee. J Biomech. 1990;23(12):1209–18.
42. Butler D, Noyes F, Grood E. Ligamentous restraints to anterior-posterior drawer in the human
knee. J Bone Joint Surg Am. 1980;62(2):259–70.
43. Robinson JR, Bull AM, Thomas RR dW, Amis AA. The role of the medial collat-
eral ligament and posteromedial capsule in controlling knee laxity. Am J Sports Med.
2006;34(11):1815–23.
44. Davies H, Unwin A, Aichroth P. The posterolateral corner of the knee: anatomy, biomechanics
and management of injuries. Injury. 2004;35(1):68–75.
45. Chao E, Laughman R, Schneider E, Stauffer R. Normative data of knee joint motion and
ground reaction forces in adult level walking. J Biomech. 1983;16(3):219–33.
46. Ma VY, Chan L, Carruthers KJ. Incidence, prevalence, costs, and impact on disability of com-
mon conditions requiring rehabilitation in the United States: stroke, spinal cord injury, trau-
matic brain injury, multiple sclerosis, osteoarthritis, rheumatoid arthritis, limb loss, and back
pain. Arch Phys Med Rehabil. 2014;95(5):986–995.e1.
47. Boyer KA, Hafer JF. Gait mechanics contribute to exercise induced pain flares in knee osteo-
arthritis. BMC Musculoskelet Disord. 2019;20(1):107.
48. Kaufman KR, Hughes C, Morrey BF, Morrey M, An KN. Gait characteristics of patients with
knee osteoarthritis. J Biomech. 2001;34(7):907–15.
49. Baliunas AJ, Hurwitz DE, Ryals AB, Karrar A, Case JP, Block JA, Andriacchi TP. Increased
knee joint loads during walking are present in subjects with knee osteoarthritis. Osteoarthr
Cartil. 2002;10(7):573–9.
50. Lewek MD, Scholz J, Rudolph KS, Snyder-Mackler L. Stride-to-stride variability of knee
motion in patients with knee osteoarthritis. Gait Posture. 2006;23(4):505–11.
51. Hurwitz DE, Ryals AB, Case JP, Block JA, Andriacchi TP. The knee adduction moment during
gait in subjects with knee osteoarthritis is more closely correlated with static alignment than
radiographic disease severity, toe out angle and pain. J Orthop Res. 2002;20(1):101–7.
52. Astephen JL, Deluzio KJ, Caldwell GE, Dunbar MJ, Hubley-Kozey CL. Gait and neuromus-
cular pattern changes are associated with differences in knee osteoarthritis severity levels. J
Biomech. 2008;41(4):868–76.
53. Chehab EF, Favre J, Erhart-Hledik JC, Andriacchi TP. Baseline knee adduction and flexion
moments during walking are both associated with 5 year cartilage changes in patients with
medial knee osteoarthritis. Osteoarthr Cartil. 2014;22(11):1833–9.
54. Charlton JM, Hatfield GL, Guenette JA, Hunt MA. Toe-in and toe-out walking require dif-
ferent lower limb neuromuscular patterns in people with knee osteoarthritis. J Biomech.
2018;76:112–8.
55. Cheung RTH, Ho KKW, Au IPH, An WW, Zhang JHW, Chan ZYS, Deluzio K, Rainbow
MJ. Immediate and short-term effects of gait retraining on the knee joint moments and symp-
toms in patients with early tibiofemoral joint osteoarthritis: a randomized controlled trial.
Osteoarthr Cartil. 2018;26(11):1479–86.
56. Lin C-J, Lai K-A, Chou Y-L, Ho C-S. The effect of changing the foot progression angle on the
knee adduction moment in normal teenagers. Gait Posture. 2001;14(2):85–91.
57. Jüni P, Reichenbach S, Dieppe P. Osteoarthritis: rational approach to treating the individual.
Best Pract Res Clin Rheumatol. 2006;20(4):721–40.
58. Kurtz SM, Ong KL, Lau E, Widmer M, Maravic M, Gómez-Barrena E, de Fátima de Pina M,
Manno V, Torre M, Walter WL, de Steiger R, Geesink RGT, Peltola M, Röder C. International
survey of primary and revision total knee replacement. Int Orthop. 2011;35(12):1783–9.
59. Baldini A, Castellani L, Traverso F, Balatri A, Balato G, Franceschini V. The difficult primary
total knee arthroplasty: a review. Bone Joint J. 2015;97-b(10 Suppl A):30–9.
60. Li C, Hosseini A, Tsai TY, Kwon YM, Li G. Articular contact kinematics of the knee before
and after a cruciate retaining total knee arthroplasty. J Orthop Res. 2015;33(3):349–58.
11 Biomechanics of Human Motion 299
61. Vince K. Mid-flexion instability after total knee arthroplasty: woolly thinking or a real con-
cern? Bone Joint J. 2016;98-b(1 Suppl A):84–8.
62. Donadio J, Pelissier A, Boyer P, Massin P. Control of paradoxical kinematics in posterior
cruciate-retaining total knee arthroplasty by increasing posterior femoral offset. Knee Surg
Sports Traumatol Arthrosc. 2015;23(6):1631–7.
63. Duffy GP, Berry DJ, Rand JA. Cement versus cementless fixation in total knee arthroplasty.
Clin Orthop Relat Res. 1998;356:66–72.
64. Zhao C, Lin C, Wang W, Zeng C, Fang H, Pan J, Cai D. Kinematics of anterior cruciate
ligament-deficient knees in a Chinese population during stair ascent. J Orthop Surg Res.
2016;11(1):89.
65. Zhang Y, Huang W, Yao Z, Ma L, Lin Z, Wang S, Huang H. Anterior cruciate ligament inju-
ries alter the kinematics of knees with or without meniscal deficiency. Am J Sports Med.
2016;44(12):3132–9.
66. Oiestad BE, Engebretsen L, Storheim K, Risberg MA. Knee osteoarthritis after anterior cruci-
ate ligament injury: a systematic review. Am J Sports Med. 2009;37(7):1434–43.
67. Lohmander LS, Englund PM, Dahl LL, Roos EM. The long-term consequence of anterior cru-
ciate ligament and meniscus injuries: osteoarthritis. Am J Sports Med. 2007;35(10):1756–69.
68. Ardern CL, Sonesson S, Forssblad M, Kvist J. Comparison of patient-reported outcomes
among those who chose ACL reconstruction or non-surgical treatment. Scand J Med Sci
Sports. 2017;27(5):535–44.
69. Lin L, Li J-S, Kernkamp WA, Hosseini A, Kim C, Yin P, Wang L, Tsai T-Y, Asnis P, Li
G. Postoperative time dependent tibiofemoral articular cartilage contact kinematics during
step-up after ACL reconstruction. J Biomech. 2016;49(14):3509–15.
70. Hosseini A, Van de Velde S, Gill TJ, Li G. Tibiofemoral cartilage contact biomechan-
ics in patients after reconstruction of a ruptured anterior cruciate ligament. J Orthop Res.
2012;30(11):1781–8.
71. Fanelli GC, Giannotti BF, Edson CJ. The posterior cruciate ligament arthroscopic evaluation
and treatment. Arthroscopy. 1994;10(6):673–88.
72. Shelbourne KD, Davis TJ, Patel DV. The natural history of acute, isolated, nonopera-
tively treated posterior cruciate ligament injuries. A prospective study. Am J Sports Med.
1999;27(3):276–83.
73. Gupte CM, Bull AM, Thomas RD, Amis AA. The meniscofemoral ligaments: secondary
restraints to the posterior drawer. Analysis of anteroposterior and rotary laxity in the intact and
posterior-cruciate-deficient knee. J Bone Joint Surg Br. 2003;85(5):765–73.
74. Noyes FR, Stowers SF, Grood ES, Cummings J, VanGinkel LA. Posterior subluxations of the
medial and lateral tibiofemoral compartments. An in vitro ligament sectioning study in cadav-
eric knees. Am J Sports Med. 1993;21(3):407–14.
75. G L, R P, M L, J B, KW N, D A, T G. Effect of posterior cruciate ligament deficiency on
in vivo translation and rotation of the knee during weightbearing flexion. Am J Sports Med.
2008;36(3):474–9.
76. Patel DV, Allen AA, Warren RF, Wickiewicz TL, Simonian PT. The nonoperative treat-
ment of acute, isolated (partial or complete) posterior cruciate ligament-deficient knees: an
intermediate-term follow-up study. HSS J. 2007;3(2):137–46.
77. Strobel MJ, Weiler A, Schulz MS, Russe K, Eichhorn HJ. Arthroscopic evaluation of
articular cartilage lesions in posterior cruciate ligament—deficient knees. Arthroscopy.
2003;19(3):262–8.
78. Parolie JM, Bergfeld JA. Long-term results of nonoperative treatment of isolated posterior
cruciate ligament injuries in the athlete. Am J Sports Med. 1986;14(1):35–8.
79. Kernkamp WA, Jens AJT, Varady NH, van Arkel ERA, Nelissen R, Asnis PD, LaPrade RF,
Van de Velde SK, Li G. Anatomic is better than isometric posterior cruciate ligament tunnel
placement based upon in vivo simulation. Knee Surg Sports Traumatol Arthrosc. 2019;27(8):
2440–9.
80. Lee D-Y, Park Y-J, Kim D-H, Kim H-J, Nam D-C, Park J-S, Hwang S-C. The role of isolated
posterior cruciate ligament reconstruction in knees with combined posterior cruciate ligament
and posterolateral complex injury. Knee Surg Sports Traumatol Arthrosc. 2018;26(9):2669–78.
300 R. Cheng et al.
81. Jastrow H, Vollrath L. Anatomy online: presentation of a detailed WWW atlas of human gross
anatomy--reference for medical education. Clin Anat. 2002;15(6):402–8.
82. Bland JH, Boushey DR. Anatomy and physiology of the cervical spine. Semin Arthritis
Rheum. 1990;20(1):1–20.
83. Turinsky AL, Fanea E, Trinh Q, Dong X, Stromer JN, Shu X, Wat S, Hallgrimsson B, Hill JW,
Edwards C, Grosenick B, Yajima M, Sensen CW. Integration of genomic and medical data into
a 3D atlas of human anatomy. Stud Health Technol Inform. 2008;132:526–31.
84. Pait TG, Elias AJ, Tribell R. Thoracic, lumbar, and sacral spine anatomy for endoscopic sur-
gery. Neurosurgery. 2002;51(5 Suppl):S67–78.
85. Baaj AA, Papadimitriou K, Amin AG, Kretzer RM, Wolinsky JP, Gokaslan ZL. Surgical anat-
omy of the diaphragm in the anterolateral approach to the spine: a cadaveric study. J Spinal
Disord Tech. 2014;27(4):220–3.
86. Aronson AS, Hoist L, Selvik G. An instrument for insertion of radiopaque bone markers.
Radiology. 1974;113(3):733–4.
87. Zhu Z, Li G. An automatic 2D-3D image matching method for reproducing spatial knee joint
positions using single or dual fluoroscopic images. Comput Methods Biomech Biomed Engin.
2012;15(11):1245–56.
88. Yu Y, Mao H, Li JS, Tsai TY, Cheng L, Wood KB, Li G, Cha TD. Ranges of cervical interver-
tebral disc deformation during an in vivo dynamic flexion-extension of the neck. J Biomech
Eng. 2017;139(6):0645011–7.
89. Zhong W, Driscoll SJ, Tsai TY, Wang S, Mao H, Cha TD, Wood KB, Li G. In vivo dynamic
changes of dimensions in the lumbar intervertebral foramen. Spine J. 2015;15(7):1653–9.
90. Wu M, Wang S, Driscoll SJ, Cha TD, Wood KB, Li G. Dynamic motion characteristics of
the lower lumbar spine: implication to lumbar pathology and surgical treatment. Eur Spine
J. 2014;23(11):2350–8.
91. Heider FC, Mayer HM. Surgical treatment of lumbar disc herniation. Oper Orthop Traumatol.
2017;29(1):59–85.
92. Truumees E. A history of lumbar disc herniation from Hippocrates to the 1990s. Clin Orthop
Relat Res. 2015;473(6):1885–95.
93. Karademir M, Eser O, Karavelioglu E. Adolescent lumbar disc herniation: impact, diagnosis,
and treatment. J Back Musculoskelet Rehabil. 2017;30(2):347–52.
94. Corniola MV, Tessitore E, Schaller K, Gautschi OP. Lumbar disc herniation—diagnosis and
treatment. Rev Med Suisse. 2014;10(454):2376–82.
95. Sonntag VK, Marciano FF. Role of spinal instrumentation in fusion for degenerative disease of
the lumbosacral spine. West J Med. 1995;162(3):262–3.
96. Frymoyer JW. Low back pain. The role of spine fusion. Neurosurg Clin N Am. 1991;2(4):933–54.
97. Greenwood J, McGregor A, Jones F, Mullane J, Hurley M. Rehabilitation follow-
ing lumbar fusion surgery: a systematic review and meta-analysis. Spine (Phila Pa 1976).
2016;41(1):E28–36.
Chapter 12
Biomechanics of the Fracture Fixation
Yingze Zhang, Hongde Wang, Tianrui Wang, Wei Chen, and Yanbin Zhu
Y. Zhang (*)
Department of Orthopaedic Surgery, The Third Hospital, Hebei Medical University,
Shijiazhuang, Hebei, People’s Republic of China
Key Laboratory of Biomechanics of Hebei Province,
Shijiazhuang, Hebei, People’s Republic of China
Chinese Academy of Engineering, Beijing, People’s Republic of China
H. Wang · T. Wang · W. Chen · Y. Zhu
Department of Orthopaedic Surgery, The Third Hospital, Hebei Medical University,
Shijiazhuang, Hebei, People’s Republic of China
Key Laboratory of Biomechanics of Hebei Province,
Shijiazhuang, Hebei, People’s Republic of China
1 Introduction
Biologic and mechanical problems are the two underlying problems, when fracture
implants fail prior to fracture union [1–3]. Patient’s systemic biology as biologic
factors including chronic diseases, smoking, medications, and many other causes
may cause delayed union and fixation failure [4–6]. Many biologic etiologies of
fixation failure can be treated by the physician, but there are still many biologic
etiologies beyond the surgeon’s control [7, 8]. Therefore, surgeons should make
many efforts to preserve soft tissue and vascularity, and respect the zone of injury
[9]. Meticulous surgical technique, wound closure, and appropriate perioperative
antibiotic therapy can all reduce the risk of infection and treatment failure.
Mechanical problems are usually the main cause, when failure occurs acutely or
prior to the expected time that fracture healing [10–13]. To determine the appropri-
ate investigation and intervention, surgeons should understand biomechanical prin-
ciples underlying the stable fixation and fixation failure.
There are some biomechanical differences among pins, rods, and nails used for
fracture fixation. Pins only resist alignment changes, rods resist deviations in align-
ment and translation, and nails resist changes in alignment, translation, and rotation.
Kirschner wires and Steinmann pins are often used for both provisional and defini-
tive fracture fixation. Due to its poor bending resistance, it should be used in con-
junction with bracing or casting. They are usually inserted as final fixation with
limited open reduction or percutaneously. They should be inserted slowly and stop
drilling frequently. To prevent thermal damage to bone and soft tissues. To make
their removal easier after fracture healing, we recommend using smooth wires.
Threaded wires can better fix the fractures compared with temporary fixation,
but the fracture fragments must be fixed together when insertion wire to avoid dis-
traction. If the cortical bone is hard, there is a risk of pin breakage. For small frag-
ments in metaphyseal and epiphyseal regions, especially in the fracture of distal
foot, forearm, and hand, such as Colles fracture, and in displaced metacarpal and
phalangeal fracture after closed reduction, pin or wire fixation is usually sufficient
[14, 15]. In most cases, pins are inserted under the control of an image intensifier.
This can protect the soft tissues from further damage, theoretically allowing for
maximum bone regeneration; however, it must be noted that nerves and tendons are
not wound around the pin during insertion. Wire fixation is used alone or in combi-
nation with other implants for definitive fixation of some metaphyseal fractures,
such as in the cervical spine, proximal humerus, and patella [16]. The wire should
not be notched as it may reduce the fatigue life of the implant. It is rare touse wire
alone to provide adequate stability for functional rehabilitation of the extremity [17].
For some intra-articular fractures, simple rigid fixation using screws or plates for
a long time might result in some complications such as articular degeneration,
12 Biomechanics of the Fracture Fixation 303
a b c
Fig. 12.1 (a) Components of the elastic bionic fixation; (b) Assembly of the fixation device; (c)
Post-operative X ray film of the fixation device
arthritis, and the limited articular mobility. Given that, our research team used the
biomechanical principle to invent an elastic fixation device for treatment of ankle
fracture combined with distal tibiofibular syndesmosis (DTS). As presented, the
elastic bionic fixation device included elastic cable, a bolt, and a button (Fig. 12.1),
which could provide both rigid and elastic fixation, and therefore reduce the possi-
ble intra-articular issues, compared to the traditional rigid fixation. With this device,
we treated 17 cases of ankle fracture combined with DTS, and at the final follow-up
all of them obtained excellent and good outcomes according to the AOFAS score.
3 Screw Fixation
Screws are made up of four parts: head, shaft, thread, and tip. The head of a screw
acts as an attachment for the screwdriver and can be hexagonal, cruciate, slotted, or
Phillips. The head also acts as a reaction, and the pressure generated by the screw
acts on the bone. The shaft or shank is the smooth portion of the screw between the
304 Y. Zhang et al.
head and the threaded portion. The thread is defined by its root (or core) diameter,
its thread (or outside) diameter, its pitch (or distance between adjacent threads), and
its lead (or distance it advances into the bone with each complete turn). The root
area determines the resistance of the screw to pullout forces and relates to the area
of the bone at the thread interface and the root area of the tapped thread. The cross-
sectional design usually is a buttress (ASIF screws) or V-thread (usually used in
machine screws). The tip of the screw is either round (require spretapping) or self-
tapping (fluted or trocar). Clinically, if it is necessary to consider pulling out from
the screw because of soft bone, it is more inclined to use a larger thread diameter,
and if the bone is stronger and more fatigue is needed, the screw with a wider root
diameter has a higher anti-fatigue failure ability. Screws also usually are divided
into machine-type screws and ASIF screw.
The use of screws to convert torque forces into compressive forces through a
fracture is a valuable technique. Its success requires the application of a screw in
such a way that the proximal portion of the screw is allowed to slide in the near bone
and the thread is formed in the opposite cortex so that the head of the screw can
exert load and forces the fracture to heal. Careful selection of the angle of the screw
corresponding to the fracture is necessary to prevent the fracture fragments from
slipping when compressed.
Screw Breakage by Shearing During Insertion
A screw is a mechanical device that is used to convert rotary load (torque) into com-
pression between a plate and a bone or between bone fragments. The basic compo-
nents of a screw are shown in Fig. 12.2. As shown in Fig. 12.3, when the thread of
the screw is released from the shaft, it is actually a slope or a slope, and the lower
bone is pulled toward the fixing plate, causing compression between the bone and
the bone. In order to achieve this effect, the screw head and shaft should rotate
freely within the steel plate; otherwise, the compressive force generated may be
limited. The locking screw passes through the plate hole; although this fixed inter-
face is beneficial in some clinical situations, it prevents compression between the
plate and the bone.
In the cortical bone, a tap is necessary so that the torque applied by the surgeon
translates into compression rather than cutting the thread, thereby overcoming the
friction between the screw thread and the bone (Fig. 12.3) that it is being driven into
(Fig. 12.4). In some cases such as inserting screw into a dense bone or inserting a
smaller diameter screw, using a separate tap and then inserting the screw, the screw
can be pushed into the bone. Most modem screw designs have self-tapping screw
tips that cuts the path of the thread when the screw is inserted. Screws with multiple
cutting flutes at the up of the screw appear to be the easiest to insert and have a
greater grip. Tapping in cancellous bone is less advantageous because tapping
reduces the pullout strength of the screw. In some cases, it may beneficial to tap the
cancellous bone. A clinical example is when treating a femoral neck fracture with a
physiologically older patient versus a younger patient; you may need to use a tap to
make a thread in the denser bone of a younger patient. The reason for using tap in
the dense bone is to prevent the frictional force from causing femoral head to rotate
12 Biomechanics of the Fracture Fixation 305
Major diameter
Pitch diameter
Root
diameter Single depth
Helix angle
Pitch
Root
Fig. 12.2 Nomenclature of screws. The root diameter is the inner diameter of the screw and the
pitch defines the distance between threads
during the insertion of screw resulting malreduction. In particularly hard bones the
frictional forces become so great that it becomes difficult to advance the screw [18].
One problem with the placement of the screw is that the shear failure of the
screw, the head twisting off usually, making it difficult to remove the shaft from the
bone. This can occur especially if the tap is not used before insertion, or when a
smaller (less than 4-mm diameter) screw is inserted into the dense bone. The stiff-
ness and strength of the screw are related to the fourth power of its radius (the effect
of moment of inertia for screws of the same material). The 6-mm diameter screw is
306 Y. Zhang et al.
Ft
Fn
Fz
about five times stiffer than a 3-mm diameter screw and 16 times more resistant to
shear damage than a 3 mm diameter screw. The junction of the screw head and
threaded portion of the screw is the transition point of shape and size. Therefore, it
acts as a stress concentrator, usually where the screw breaks.
12 Biomechanics of the Fracture Fixation 307
Screw Pullout
Especially, in cancellous bone, the maximum force that a screw can withstand along
its axis. The pullout force depends on the size of the screw and the density of the
bone placed. As shown in Fig. 12.5, when the force acting on the screw exceeds its
pullout strength, the screw will be pulled out or “stripped” out of the hole, and the
sheared bone will be placed in thread, greatly reducing the nail holding force and
fixing force. The larger the diameter of the screw, the larger the number of threads
per unit length, and the longer the insertion length of the screw shaft, the greater the
pulling force. And a greater density of the bone it is placed into. The diameter and
length of the embedded screw can be considered to define the outer surface of a
cylinder along that screw shears. Given a maximum stress that bone of a particular
308 Y. Zhang et al.
density can withstand, increasing the surface area of the screw cylinder increases
the pullout force (because force = stress multiplied by the area over which it acts).
In order to enhance screw purchase, it is conceivable to insert the screw of largest
diameter into the bone of the highest density with the length of the implant as long
as possible. However, placing screws of as large a diameter as possible has disad-
vantages. Larger screws occupy a larger volume in small fracture fragments, limit-
ing the number of possible fixation sites and propagating adjacent fracture lines.
In cancellous bone, the extraction of the screw becomes a more important prob-
lem because the porosity of cancellous bone reduces its density and thus the shear
strength. Drilling preparation, especially drilling, rather than tapping, can increase
the pullout strength of cancellous bone (such as pedicle screws in the vertebral
body). The reason why the cancellous bone is knocked down is that the tap is
removed from the hole or placed in the hole, which can effectively increase the
diameter of the hole and reduce the amount of bone material that interacts with the
thread. When the bone density is reduced, the tapping is more harmful, and the
pullout strength can be reduced from 8 to 27%. The pullout strength can also be
related to the time after insertion. As the bone heals, it can remodel around the
screw, possibly doubling its initial pullout strength [19].
Recent research has focused on whether pullout strength is an appropriate mea-
sure of screw performance in cancellous bone. In nonlocking steel and screw con-
structions, the stability of most structures comes from the friction created by
compression between the plate and the bone. When the screw is inserted into the
bone, if it is able to generate high values of insertional torque, the compression of
the steel plate to the bone is increased, and the stability is increased. As the maxi-
mum insertional torque is reached and then exceeded, the screw will then “strip out”
and lose its supporting force in the bone. Although there is a relationship between
maximum insertional torque, screw pitch, and compression forces, it has been found
that the pullout strength has no correlation with either the maximum insertional
torque or screw pitch. Therefore, this may be a better way to measure screw perfor-
mance and optimize screw characteristics.
Screw Breakage by Cyclic Loading
Once screw is successfully inserted and the construction is completed, the screw
will be subjected to cyclic bending forces as the patient begins to mobilize
(Fig. 12.6). Ideally, a nonlocking screw initially tightens the plate to achieve the
maximal torque, which is translated into the maximum compression between the
plate and the bone (Fig. 12.4). The screw on the bone portion is in frictional contact,
depending on the friction generated between the plate and the bone. The frictional
force is directly dependent on the compressive force generated by the screws. If a
slip occurs between the plate and the bone, the bending load will be transferred from
the screw head into the plate, at which point the screw comes into contact with the
plate. The bending load perpendicular to the axis of the screw, coupled with possible
stress corrosion and fretting corrosion, may cause the screw to fail rapidly in fatigue.
Zand et al.’s research shows that when the screw is fastened to the steel plate, it is
subjected to a maximum load of less than 1000 loading cycles due to bending
12 Biomechanics of the Fracture Fixation 309
fatigue, compared to a fully fastened screw capable of withstanding more than 2.5
million loading cycles. The load is less than 10–15% of the maximum load. This
emphasizes the clinical importance of ensuring screw tightness during the fixation
of the plate.
Locking the screws on the board can reduce the problem, since the problem is
less subjective when the screw head is fully fastened to the plate hole. Small-
fragment screws (3.5- to 4-mm outer diameter) can fatigue because of their small
core diameters. Although the use of locking screws with a larger core diameter and
shallower thread can reduce the possibility of fatigue failure, a smaller core d iameter
and deeper thread can increase purchase strength in the bone. Screws with smaller
core diameters are more likely to fatigue than larger ones. The fatigue strength of
the screw must be weighed against the purchasing power of the screw and the size
of the screw associated with the size of the bone fragment. The surgeon sometimes
must make a decision between a screw with a large core diameter with shallower
thread, which maximizes fatigue strength, and a smaller core diameter screw with
deeper threads, which maximizes purchase power.
Cannulated screws are used for fixation when the insertion of a guide wire is
helpful to guide the future path of the screw. However, as the bone density increases
and the diameter of the guide wire increases, the drilling accuracy of the guide wire
decreases. Cannulated screws follow the same mechanical principles as solid
screws, but material must be removed from the center of the screw to accommodate
the passage of the guidewire. Manufacturers commonly increase the diameter of the
screw at the base of the thread to fit the loss of this central material. The same size
310 Y. Zhang et al.
of cannulated screws usually has less thread depth than solid screws. The result
depends on the size of screw, rather than pullout strength. For screws with a diam-
eter of 4 mm, the tensile strength of the cannulated screws of the same outer diam-
eter is about 16% lower. Alternatively, to maintain the same thread depth, the outer
diameter of the screw may be increased. Another consideration is that the cannu-
lated screw is much more expensive than the solid screw [20].
Fully Threaded Lag Screws
The lag screw is a very effective device which can produce amount of pressure
across fracture fragments and the fracture site. The head of the screw and upper por-
tion of the shaft must be allowed to glide in the near broken pieces to pull the far
broken fragments toward it, thereby creating compression on the fracture surface.
As shown in Fig. 12.7, a fully threaded lag screw can prevent the gliding action
between the two fracture fragments. The fully and partly threaded lag screws were
used to compare the compressive forces across the fracture site. The results showed
that the average compressive force at the opposite cortex (i.e., the force in the screw
itself) was about 50% greater when a partly threaded screw was used.
3.1 Machine Screws
The machine screws are threaded throughout the length and can be self-tapping or
threaded before insertion. Most are self-tapping; there is a cutting flute that cuts the
screw threads as the screw is inserted. Machine screws are mainly used to fix the hip
compression screw on the femoral shaft. The size of the machine screw holes is
critical. A hole that is too large can cause the thread to be unsafe, and a hole that is
too small can cause the screw to be inserted or fractured during insertion. The drill
bit selected should be slightly smaller than the screw after the screw minus the
12 Biomechanics of the Fracture Fixation 311
thread. For a self-tapping screw, the drill point is used to drill holes in soft bone. The
size of the screw and drill tip should be checked before surgery.
3.2 Asif Screws
Screws developed by the Swiss ASIF Group for bone fixation techniques and prin-
ciples are widely used. The threads are more horizontal than the machine screws,
and these screws rarely seldom self-tapping; the drill must be tapped with a cutting
tapper before inserting the screws. ASIF screws are available in cortical, cancellous,
and malleolar designs. Mini screws for fixation of small fragments and small bones,
and the standard cancellous and cortical screws, come in multiple lengths and diam-
eters. Standard cancellous and cortical screw heads have a hexagonal recess for a
special screwdriver, while the smaller screws have a Phillips head.
3.2.1 Cortical Screws
The full length thread of the cortical ASIF screw is available in a variety of diame-
ters (4.5, 3.5, 2.7, 2.0, and 1.5 mm). If the hole in the near cortex is drilled too deep,
the cortical screws can be used as a positional or lag screws for inter-fragmentary
compression.
3.2.2 Cancellous Screws
These screws have larger threads that provide more support for soft cancellous
bone, making them more suitable for use on the metaphyseal areas. The cancellous
screws are available in 6.5- and 4.0-mm diameters with thread lengths of 16 and
32 mm, respectively. Regardless of the lengths of the screw, both lengths are
threaded. The malleolar 4.5-mm screw is also included in this group, but it is unique
in that it has a self-tapping thread. Choosing the right drill size and tapping the hole
is the key to a safe purchase. Plastic and metal washers are commonly used with
these types of screws to reattach ligament tears or to increase compression between
the fragments by providing a larger cortical surface area for screw head.
The self-tapping screws are the same sizes as cortical screws. A small portion of the
ends of these screws are used to remove bone fragments. Self-tapping screws have
a lower pullout strength due to their structure. These screws are preferably used for
external fixation pins [21].
312 Y. Zhang et al.
3.2.4 Locking Screws
The locking screw is a self-tapping screw with a locking screw at the head. These
screws require accurate predrilling so that the locking plate is tightly attached to the
locking plate and requires a special screw drivers for implantation.
Pauwels et al. firstly defined and applied the tension band principle to fix fractures
and nonunion. This engineering principle applies to the transformation of the tensile
force into a compressive force on the convex side of an eccentrically loaded bone.
This is accomplished by placing a tension band (bone plate) across the fracture on
the tension (or convex) side of the bone. The tension is counteracted by the tension
band at this position and converted into a compressive force. If the plate is applied
to the compression (or concave) side of the bone, it is likely to bend, fatigue, and fail
[22]. Therefore, a basic principle of tension band plating is that it must be applied to
the tension, and the bone itself will get a compressive force, so the tension band
device does not require a heavy and rigid tension band principle and is also used for
some olecranon and patellar fractures. Tension bands and axial compression are
often combined when using plates and screws [23–25].
We have found that almost all plate hole breaks occur at the plate hole near frac-
ture area [26]. Therefore, the hole area seems to be the weakest part of the board,
and naturally it is a place for improvement. We only widened the locking compres-
sion plate (LCP) in the hole area to make it a gourd-shaped LCP to increase the
strength of the plate and reduce the plate breakage (Fig. 12.8) [27, 28]. After a series
of axial loading single cycle to failure test, torsion single cycle to failure test, four-
point bending single cycle to failure test, and dynamic four-point bending test, it is
concluded that the gourd-shaped LCP structure has greater stiffness, strength, and
longer fatigue life than LCP. This may be a more reasonable LCP that can reduce
the rate of clinical breaks [29].
Our team used a new anatomical plate and compression bolt fixation technique,
combined with a small incision of the posterior foot, to treat intra-articular calcaneal
fractures and achieved good or excellent clinical results, and had fewer soft-tissue
problems (Fig. 12.9) [30]. Compared to the conventional plate and cancellous
screws technique, our fixation technique requires higher loads to cause structural
failure, which may be related to the design of the implant. According to the mea-
surement of the calcaneus specimen and the data of the three-dimensional CT image
of the calcaneus, the anatomical steel plate and the compression bolt were designed.
The use of conventional anatomical plates and cancellous screws to fix a calcaneal
fracture is characterized by compression of the plate to the lateral wall of the calca-
neus. The actual stability lies in the friction between the plate and the bone, which
b
314 Y. Zhang et al.
can easily be achieved by our anatomic plate and compression bolts. Putting the
compression bolts together can be seen as another piece of plate that is compressed
onto the inner side wall of the bone specimen to provide higher friction and restore
the width of the calcaneus.
Another example was the application of compression bolt in the treatment of
Schatzker type II–VI tibial plateau fractures (Fig. 12.10), in which joint surface
widening and collapse are commonly accompanied [31, 32]. The traditional metal
plates and screw fixation of fractures of such types are commonly associated with a
high rate of postoperative reduction loss, which likely result in the development of
traumatic arthritis. Our preliminary reports of using this compression bolt presented
the favorable results, both in biomechanics and clinical effectiveness.
12 Biomechanics of the Fracture Fixation 315
compression bolt
4.1 Locking Plates
Locking plates are a combination of steel plate technology and percutaneous bridge
plating technology using locking screws as fixed angle devices [33]. Locking steel
plates provide a stronger, longer lasting fix than nonlocking steel plates [34]. They
have been proven to allow for greater loads bearing than regular plates. The Less
Invasive Stabilization System (LISS) (Synthes, Inc., West Chester, PA) uses unicor-
tical locking screws that allow for greater elastic deformation than conventional
plating systems. The locking plate can also be used in combination with locking and
unlocking screws, mechanically similar to a pure locking structure. The locking
plate works best on osteoporotic bones, where pulling out the steel plate is problem-
atic. They also provide sufficient load-bearing strength to avoid the distal femur,
proximal humerus, and medial and lateral plates of the tibial plateau. However, the
locking plate structure also has an inherent that inhibits the movement of the frac-
ture site, making it insufficient to stimulate callus formation. Therefore, the locking
plate of the distal femoral fracture can result in insufficient and asymmetrical ankle
formation, with minimal deposits in the proximal cortex. A recent study of locking
plates for distal femoral fractures confirmed this and showed a nonhealing rate of
10–23% and a reoperation rate of open fractures of 31%.
Satisfactory stable intramedullary fixation of the fracture is possible under the fol-
lowing conditions:
316 Y. Zhang et al.
Just as plate, intramedullary nail has an anatomic and functional name. The central
body nail is inserted into the bone in a straight line with the medullary canal. It
interferes longitudinally with the bone through multiple points of contact [44–49].
They rely on restoring bone contact and stability to avoid axial deformation of the
fracture during rotation [50, 51]. The classic Küntscher cloverleaf and Sampson
nails are examples of centromedullary nails. The condylocephalic nails enter the
bone of the condyles of the metaphysis and usually enter the opposite metaphyseal-
epiphyseal area. They are usually inserted into groups to increase rotational stabil-
ity. Ender and Hackenthall pins are examples of condylocephalic nails.
Cephalomedullary nails have a centromedullary portion but it is also allowed to be
fixed to the femoral head. The Küntscher Y-nail and Zickel subtrochanteric nail are
examples of this type [52, 53].
Interlocking techniques further improves these classics by adding interlocking
centromedullary and interlocking cephalomedullary nails [54–56]. Interlocking
nails allow longer working length of the interlocking nail screw axial and rotational
deformation resistance of fracture. Modney first designed the first interlocking nail.
Küntscher also designed an interlocking nail (the detensor nail), which was modi-
fied by Klemm and Scheilman and later modified by Kempf et al. These pioneers
318 Y. Zhang et al.
developed techniques and implants that form the basis of some designs and
techniques used today. Cephalomedullary interlocking nails, designed to treat com-
plex fractures and the proximal femur, were axially and rotationally unstable, such
as complex subtrochanteric fractures, pathologic fractures, and ipsilateral hip and
shaft fractures. These nails can be secured with bolts, nails, and special lag screws
such as Russell-Taylor reconstruction nails, Williams y nails, and Uniflex nails [57].
The current intramedullary nails for femoral fixation design reflect regional internal
fixation nails. Antegrade femoral nails can be performed through the piriformis or
trochanteric inlet. The retrograde femoral nail passes through the entrance between
the femoral condyles [58, 59].
Interlocking fixation is defined as a dynamic, static, and double lock. Dynamic
fixation controls bending and rotational deformation but allows axial load transfer
of the bone. Axial stable fractures and partial nonunion can be fixed by power. Static
fixation controls the rotation, bending, and axial load, so that the implant has more
bearing potential and reduce the fatigue life of the equipment. It is particularly use-
ful in crushing, nonisthmal fractures of the femur and tibia. The double-locked
mode controls bending, rotational forces, and some axial deformation, but some
shortening occurs due to the ability of the screw to translate axially within the nail.
This type of fixation is often used for humerus fracture with delayed union and not
healing.
The dynamics of the interlocking nails were originally designed to avoid fracture
healing, [60] as it is theoretically believed that static interlocking will stop the repair
of the fracture. This technique involves conversion of the static mode to a dynamic
mode by removing the screws from the longest fragment. Dynamization increases the
fatigue life of the nail by reducing the load-carrying capacity of the nail while increas-
ing the compressive force at the break point; however, if there is insufficient cortical
stability or bone regeneration before exercise, shrinkage may occur [45, 61, 62].
For patients with multiple fractured long bone fractures, the need for reaming for
intramedullary nailing has been controversial [63]. Physicians who support non-
reamed nails emphasize the lack of physiological effects of reaming, such as fat
embolism in the lungs [64, 65]. Experimental evidence suggests that reaming has an
adverse effect on lung function. This adverse effect does not appear to be apparent
in most clinical patients; however, some authors believe that the development of
pulmonary complications may be related to the severity of the associated chest
injury, rather than to the reaming of the medullary cavity. Studies supporting the
reaming nail showed no statistically significant difference in the incidence of pul-
monary complications in patients with and without reaming [66]. Due to various
factors leading to the development of adult lung failure syndrome, it is difficult to
determine which patients’ lung expansion may be harmful. Whether the long bone
12 Biomechanics of the Fracture Fixation 319
6 External Fixation
Total force on
hip
Force component
which creates
binding of screw in
barrel
Force
component
which causes
sliding
135 degress
150 degress
Fig. 12.11 The joint reaction force in the femoral head can be divided into two major components.
The one parallel to the axis of the femoral neck produces sliding and impaction of the fracture
components and the other, transverse to the femoral neck, causes the screw component of the
femoral hip screw to bind, resisting sliding. The higher-angle hip screw has a screw axis more
closely aligned with the joint reaction force so the force component that produces sliding is larger
whereas the transverse force component resisting sliding is smaller
322 Y. Zhang et al.
mechanical interlocking for improved stability. Therefore, the goal of the femoral
neck fixation system is to use the component of the joint force parallel to the femo-
ral neck to encourage the fracture surfaces to slide together. This is the basic prin-
ciple for selecting high angle hip screw when possible.
The following points regarding the sliding hip screw device also apply to the
nail/tension screw device. When the screw slips, since the structure is staggered by
the fracture, the screw is supported by the barrel to prevent the femoral head from
bending down. Adhering to two basic mechanical principles will increase the ability
of the screw to slide within the side plates or nail holes. As mentioned above, higher
angle hip screws are more effective in adjusting slip [97–101]. In addition, the screw
should engage as deep as possible within the barrel. For the force acting on the
femoral end of the screw, if the internal force of the screw in contact with the barrel
is small, the remaining amount of the screw shaft in the barrel is small, and the
internal force in the barrel is increased. This is because the moment (bending load)
generated by the force acting transversely on the screw axis at the femoral head
(Fig. 12.12) acts on the longer force arm or the vertical distance L (the force x is
perpendicular to the edge of the barrel, i.e. the fulcrum)). The balance arm Lb is
shorter because there are fewer screws left in the barrel. Since Fh acts on a longer
arm and F acts on a shorter arm, Fb increases. When the screw is in contact with the
barrel, its internal force Fb produces greater frictional resistance, which requires
more friction to overcome the friction and allow slippage. Sliding hip screws with
two- or four-hole side plates seems to provide an equivalent anti-physiological com-
pression load. There are several factors that affect the fixation strength of the femo-
ral neck using multiple screws, but the number of screws used (3 or 4) is not a
significant factor.
Factors that increase this type of fixed strength include more long-axis screws
with transverse fracture lines, larger femoral skull mass density in the position of
the screw, and less comminuted fractures, shorter arm loads on the arm (shorter The
distance from the center of the femoral head fracture line). However, the most
important factor is the quality of the reduction because of the importance of cortical
support in reducing fracture displacement. Under physiological load, several mech-
anisms of fixed failure were observed (Fig. 12.13). In some cases, the screw bends
downwards, especially when it is unable to support a fracture surface below the
screw due to fracture comminution. If a washer is not used to distribute the screw
load to the bone, when the cortex is thin, the screw head will pass through the cortex
near the greater trochanter. Finally, if the screws do not support well down through
the fracture, they may rotate downwards, causing the femoral head to invert.
Supporting the hypodermis with at least one screw is a mature clinical technique
that may help prevent this from happening.
Both supracondylar fractures of the femur and tibial plateau fractures are challeng-
ingly stable because they usually involve the fixation of multiple small cancellous
bones [102–104]. Mechanically comparable alternative methods for supraorbital
12 Biomechanics of the Fracture Fixation 323
Hip force Fh
Force of
Le barrel on
screw
Lb
Fb
Fulcrum
Le
Fh
L’b
F ’b
Fh * Le = Fb * Lb = F ’b * L’b
Lb decreases,
Fb increases
If Fb increases,
resistance to sliding
increasese
Fig. 12.12 The greater the length of the sliding screw within the barrel, the lower its resistance to
sliding. In this diagram Fh is the component of the joint reaction force perpendicular to the axis of
the screw. The inferior edge of the proximal end of the barrel is the location of the fulcrum in bend-
ing. An internal force, Fb from the surface of the barrel acts against the screw to counteract Fh. For
equilibrium, the moments produced by Fh(Fh × Le) and Fb(Fb × Lb) must be equal. If Lb, the distance
from the point of application of internal force Fb to the fulcrum, decreases, Fb must increase to
therefore the resistance to screw sliding will increase (Le is the length of the screw beyond
the barrel)
324 Y. Zhang et al.
a b
Moment arm of load
component which
creates bending
Bone density at
screw purchase
sites
Degree of inferior
comminution
Angle of
fracture line
Fig. 12.13 (a) Some factors that decrease the strength of femoral neck fracture fixation include
decreased bone density, a more vertical fracture surface (which reduces buttressing against bend-
ing), and a longer moment arm or distance of the center of the femoral head to the fracture line. (b)
Observed mechanisms of failure of femoral neck fixation using screws include bending of the pins,
displacement of the screw heads through the thin cortex of the greater trochanter, especially if
washers are not used, and rotation of the screws inferiorly through the low-density cancellous bone
of the Ward triangle area until they settle against the inferior cortex
fixation include condylar plates, plates and plates that use lag screws at the fracture
site. All equipment tests seem to provide similar structural stiffness [105–108]. The
most important factor in determining plate fixation is to maintain contact at the
cortex opposite the fixture. A fixed structure without cortical contact is only 20%
harder than a fixed structure with cortical support. It has been found that the use of
a retrograde IM supracondylar nail results in a 14% reduction in axial compression
strength and a 17% reduction in torsional strength compared to fixed-angle side
panels. However, longer nails (36 cm) enhance fixation stability compared to shorter
nails (20 cm). Several new fixation systems have been described as stable for supra-
condylar fractures of the femur. The Minimally Invasive Stabilization System
(LISS) uses a low profile plate with a single cortical screw distal end, which is also
locked to the plate. Compared to a conical screw or a support plate, the LISS plate
produces a structure with greater elastic deformation and less sedimentation.
The tibial plateau fracture is difficult to stabilize [109–111]. Given the patient’s
prognosis, risk factors for reduced reduction have been shown to include patients
older than 60 years, premature weight bearing, fracture comminution, and severe
12 Biomechanics of the Fracture Fixation 325
Screw in
cancellous bone
a Applied
b
load
Tension in screw
with loading
Bending
of plate
Screw in
cortical
bone
Fig. 12.14 Two alternative methods of fixation of tibial plateau fractures: (a) transverse screws
combined with a buttress plate and (b) transverse screws alone. The buttress plate provides addi-
tional support in bending as the tibial fracture component is loaded in an inferior direction and
allows the screws to engage the thicker, more distal cortical bone
Proximal humerus fractures fixed with a locking plate provided greater stability
against torsional loading, but is similar to blade plate structure when bent, as both
fixation devices are loaded as tension bands in bending [127]. When comparing dif-
ferent types of blade plate structures, the hardest structure uses an eight-hole, low-
contact dynamic compression plate that is shaped into the shape of the blade and
secured with a diagonal screw that is triangular to the end of the blade. This arrange-
ment is quite harder than other blade plates or T-plate and screw structures. A poten-
tial problem is the screw penetration of the subchondral bone in patients with
osteoporosis. Due to the stiffness of the locking plate-screw structure, if there is any
“settling” in the fracture site, the locking screw may penetrate the joint. The inci-
dence of intra-articular screw penetration in the proximal humeral locking plate was
significantly higher than that of conventional implants [14].
6.1.4 Fixation of Spine
For the treatment of spinal fractures, the goals are to reduce the fracture, protect the
neurological function, and accelerate functional recovery [128]. The theory of
3-column model is the basis of the treatment rationale in spinal fractures [129].
Injuries that represent 3-column instability require operative stabilization even if
there is no n eurological deficit. The attachments of spinal fixation system consist of
hooks, wires and screws, which produce different types of holding force [130, 131].
Wires could resist tension, hooks could resist driving force against the bone, while
screws could resist forces from all directions except rotation. Therefore, screws are
widely used for spinal fixation because of the superiority.
Posterior internal fixation system with pedicle screws has become popular for the
treatment of spinal fracture. When applying lumbar spinal fixation, some principles
can be considered. Screws are vulnerable to toggling when they are placed into
pedicles. The screw tends to toggle about the base of the pedicle because of the
cortical bone. In order to reduce toggling, the screw head should be locked to the
rod of plate (Fig. 12.15).
Longer fixation could reduce forces acting on the screws because of the effect of
the greater lever arm of a longer rod along with more vertebrae. Whereas it is not
beneficial for a clinical perspective because the reduced spinal motion. It is also
important to add a fusion cage to reduce forces in the fixation. Coupler bars could
connect the fixation rods to form an H configuration, and prevent the rods from
rotating medially or laterally, as shown in (Fig. 12.16). The coupler bars could sig-
nificantly enhance the torsional and lateral bending stability of the implant.
68.3% of pelvic fractures are unstable fractures, which are serious injuries, and the
mortality rate is up to 19%. The stability of the pelvis is mainly related to the integ-
rity of posterior pelvic ring. There are many methods available, including iliosacral
12 Biomechanics of the Fracture Fixation 327
Vertical loading
(IS) screws, sacral bars, tension band plate (TBP), triangular osteosynthesis, and so
on. IS screw fixation is a well-recognized technique for treating the posterior pelvic
ring disruption. It is implanted in the supine or prone position and has such merits
as short operative time, slight trauma, and minimal invasion. However, it remains a
technically demanding procedure, and both doctors and patients are exposed to
large amounts of radiation as continuous fluoroscopic or computerized tomography
(CT) guidance for appropriate screw insertion. In addition, higher rates of iatro-
genic injury is one of the disadvantages, seriously affecting the clinical use of this
technology. To avoid these limitations, our team developed a novel minimally inva-
sive adjustable plate (MIAP) (Fig. 12.17). This MIAP is designed according to the
anatomy of the pelvic ring and simulated the sacroiliac complex structure of
“bridge.” It can be better attached the posterior aspect of the sacroiliac joint without
bending and adjusted the length of the connecting rod to pressure or separation of
328 Y. Zhang et al.
Fracture
d
vertebra
Link 4
vertebra
Link 1
Link 1
Fig. 12.16 Without a coupler bar between two longitudinal rods (left), they can rotate when a
lateral moment or axial torsion is applied (right). A coupler connecting the rods to form an H con-
figuration reduces this effect
fracture end. Moreover, during the operation, two small incision were made for
placing the MIAP, which can effectively reduce the blood loss and shortened the
operation time.
occur. In recent years, the suture button as a flexible fixation has been applied. The
suture button allows physiologic motion in the tibiofibular joint and maintains the
reduction of the ankle. However, the suture between buttons can gradually release
under daily motion. To avoid these drawbacks, our team developed a novel technique
called “bionic fixation” (Fig. 12.18). The screw segment may afford an improved
rigidity and stability. The high strength non-absorbable suture located between the
Leading portion
Threaded portion
Break-off groove
Screw nut
High strength
non-absorbable
suture
Fixing button
Fig. 12.18 (1): Schematic diagrams showing the bionic fixation construct (2): Schematic dia-
grams showing the three techniques for fixation of the tibiofibular syndesmosis. (a) A hole was
drilled with a 2.8-mm drill bit from posterolateral fibula to anteromedial tibia. (b, c) A 3.5-mm
cortical screw was then inserted through the hole from the fibular side. (d) After the cortical screw
was removed, the hole was over-drilled with a 4.0-mm drill bit. (e) A 3.5-mm main screw was
passed through the hole from the fibular side and the fixing button of the screw-tail was tightly
attached to the fibula, then the screw nut was installed and adjusted on the tibial side to make the
construct tightened properly. (f) The exposed leading portion of the screw was broken off. (g) The
bionic fixation construct was removed. (h) The non-absorbable suture of the fixing construct was
pulled from the fibular side to the tibial side. (i) The suture was threaded into the tibial button,
looped, traversed through and securely tied over the fibular button. This process was repeated until
there were three independent groups of sutures in the channel
330 Y. Zhang et al.
a b c
d e f
g h i
tibia and fibula may retain the motion of the syndesmosis to the maximum degree.
Comparing with the Endo button fixation, the bionic fixation can provide more sta-
ble fixation force and retaining the motion function of syndesmosis. Besides, this
technique has a low cost and is easy to perform.
Operative reduction and internal fixation is the standard treatment for unstable pos-
terior column acetabular fractures to allow early mobilization and decrease the risk
of posttraumatic arthritis. The conventional methods of fixation involve lag screws
and reconstruction plates, or both in combination. Conventional fixation depends on
the structure of the acetabulum and the surgical technique because of the specific
anatomy of posterior column of acetabulum. The conventional reconstruction plates
need to be bended based on the size of the size of the acetabulum. Using screws and
two reconstruction plates to obtain better fixation is a potentially serious traumatic
complication. Our team designed a W-shaped acetabular angular plate (WAAP) for
posterior columns of the acetabulum fractures (Fig. 12.19). This novel fixation
includes a W-shaped locking plate and the guide apparatus. Comparing with other
12 Biomechanics of the Fracture Fixation 331
reconstruction plates, the WAAP provides some advantages. First, the WAAP is
anatomically pre-contoured and could match the surface of the posterior acetabu-
lum column properly. Second, the extended fixation range spans from the greater
sciatic notch to the rim of the posterior acetabulum. Third, the WAAP has locking
holes which can achieve angular stability.
References
1. Agel J, Rockwood T, Barber R, Marsh JL. Potential predictive ability of the orthopaedic
trauma association open fracture classification. J Orthop Trauma. 2014;28:300–6.
2. Centers for Disease Control and Prevention. Fatalities and injuries from falls among older
adults—United States, 1993–2003 and 2001–2005. MMWR Morb Mortal Wkly Rep.
2006;55:1221–4.
3. Glatt V, Bartnikowski N, Quirk N, Schuetz M, Evans C. Reverse dynamization: influence of
fixator stiffness on the mode and efficiency of large-bone-defect healing at different doses of
rhBMP-2. J Bone Joint Surg Am. 2016;98:677–87.
4. Bonyun M, Nauth A, Egol KA, et al. Hot topics in biomechanically directed fracture fixation.
J Orthop Trauma. 2014;28(Suppl 1):S32–5.
5. Carter DR, Blenman PR, Beaupre GS. Correlations between mechanical stress history and
tissue differentiation in initial fracture healing. J Orthop Res. 1988;6:736–48.
6. Carter DR, Wong M. The role of mechanical loading histories in the development of diarthro-
dial joints. J Orthop Res. 1988;6:804–16.
7. Crichlow RJ, Andres PL, Morrison SM, Haley SM, Vrahas MS. Depression in orthopaedic
trauma patients. Prevalence and severity. J Bone Joint Surg Am. 2006;88:1927–33.
8. Giorgi M, Verbruggen SW, Lacroix D. In silico bone mechanobiology: modeling a multifac-
eted biological system. Wiley Interdiscip Rev Syst Biol Med. 2016;8:485–505.
9. Boerckel JD, Uhrig BA, Willett NJ, Huebsch N, Guldberg RE. Mechanical regulation of vas-
cular growth and tissue regeneration in vivo. Proc Natl Acad Sci U S A. 2011;108:E674–80.
10. Arazi M, Yalcin H, Tarakcioglu N, Dasci Z, Kutlu A. The effects of dynamization and destabi-
lization of the external fixator on fracture healing: a comparative biomechanical study in dogs.
Orthopedics. 2002;25:521–4.
11. Augat P, Burger J, Schorlemmer S, Henke T, Peraus M, Claes L. Shear movement at the frac-
ture site delays healing in a diaphyseal fracture model. J Orthop Res. 2003;21:1011–7.
12. Augat P, Simon U, Liedert A, Claes L. Mechanics and mechano-biology of fracture healing in
normal and osteoporotic bone. Osteoporos Int. 2005;16(Suppl 2):S36–43.
332 Y. Zhang et al.
13. Bizzarri M, Palombo A, Cucina A. Theoretical aspects of systems biology. Prog Biophys Mol
Biol. 2013;112:33–43.
14. Adamczyk MJ, Riley PM. Delayed union and nonunion following closed treatment of diaphy-
seal pediatric forearm fractures. J Pediatr Orthop. 2005;25:51–5.
15. Anderson R. A new method of treating fractures in the distal third of the femur. Can Med
Assoc J. 1935;32:625–9.
16. Bassuener SR, Mullis BH, Harrison RK, Sanders R. Use of bioabsorbable pins in surgical fixa-
tion of comminuted periarticular fractures. J Orthop Trauma. 2012;26:607–10.
17. Flannery W, Balts J, McCarthy JJ, Swick J, Noonan KJ, Olson J. Are terminally threaded guide
pins from cannulated screw systems dangerous? Orthopedics. 2011;34:e374–7.
18. Bottlang M, Doornink J, Fitzpatrick DC, Madey SM. Far cortical locking can reduce stiff-
ness of locked plating constructs while retaining construct strength. J Bone Joint Surg Am.
2009;91:1985–94.
19. Barber FA, Dockery WD. Long-term absorption of poly-l-lactic acid interference screws.
Arthroscopy. 2006;22:820–6.
20. Downey MW, Kosmopoulos V, Carpenter BB. Fully threaded versus partially threaded screws:
determining shear in Cancellous bone fixation. J Foot Ankle Surg. 2015;54:1021–4.
21. De Bastiani G, Aldegheri R, Renzi Brivio L. The treatment of fractures with a dynamic axial
fixator. J Bone Joint Surg Br. 1984;66:538–45.
22. Claes L, Augat P, Suger G, Wilke HJ. Influence of size and stability of the osteotomy gap on
the success of fracture healing. J Orthop Res. 1997;15:577–84.
23. Aalto K, Holmstrom T, Karaharju E, Joukainen J, Paavolainen P, Slatis P. Fracture repair dur-
ing external fixation. Torsion tests of rabbit osteotomies. Acta Orthop Scand. 1987;58:66–70.
24. Chrisovitsinos JP, Xenakis T, Papakostides KG, Skaltsoyannis N, Grestas A, Soucacos
PN. Bridge plating osteosynthesis of 20 comminuted fractures of the femur. Acta Orthop
Scand Suppl. 1997;275:72–6.
25. Harbacheuski R, Fragomen AT, Rozbruch SR. Does lengthening and then plating (LAP)
shorten duration of external fixation? Clin Orthop Relat Res. 2012;470:1771–81.
26. Stoffel K, Dieter U, Stachowiak G, Gachter A, Kuster MS. Biomechanical testing of the LCP—
how can stability in locked internal fixators be controlled? Injury. 2003;34(Suppl 2):B11–9.
27. Xu GH, Liu B, Zhang Q, et al. Biomechanical comparison of gourd-shaped LCP versus
LCP for fixation of comminuted tibial shaft fracture. J Huazhong Univ Sci Technol Med Sci.
2013;33(2):250–7.
28. Xu G, Chen W, Zhang Q, et al. Flexible fixation of syndesmotic diastasis using the assembled
bolt-tightrope system. Scand J Trauma Resusci Emerg Med. 2013;21(1):1–9.
29. Gautier E, Sommer C. Guidelines for the clinical application of the LCP. Injury. 2003;34(Suppl
2):B63–76.
30. Wang H, Yang Z, Wu Z, et al. A biomechanical comparison of conventional versus an anatomic
plate and compression bolts for fixation of intra-articular calcaneal fractures. J Huazhong Univ
Sci Technol Med Sci. 2012;32(4):571–5.
31. Uhthoff HK, Finnegan M. The effects of metal plates on post-traumatic remodelling and bone
mass. J Bone Joint Surg Br. 1983;65:66–71.
32. Uhthoff HK, Poitras P, Backman DS. Internal plate fixation of fractures: short history and
recent developments. J Orthop Sci. 2006;11:118–26.
33. Lujan TJ, Henderson CE, Madey SM, Fitzpatrick DC, Marsh JL, Bottlang M. Locked plat-
ing of distal femur fractures leads to inconsistent and asymmetric callus formation. J Orthop
Trauma. 2010;24:156–62.
34. Eichinger JK, Herzog JP, Arrington ED. Analysis of the mechanical properties of locking
plates with and without screw hole inserts. Orthopedics. 2011;34:19.
35. Nahm NJ, Como JJ, Wilber JH, Vallier HA. Early appropriate care: definitive stabilization of
femoral fractures within 24 hours of injury is safe in most patients with multiple injuries. J
Trauma. 2011;71:175–85.
12 Biomechanics of the Fracture Fixation 333
36. Schreiber VM, Tarkin IS, Hildebrand F, et al. The timing of definitive fixation for major frac-
tures in polytrauma—a matched-pair comparison between a US and European level I centres:
analysis of current fracture management practice in polytrauma. Injury. 2011;42:650–4.
37. Stannard JP, Lopez-Ben RR, Volgas DA, et al. Prophylaxis against deep-vein thrombosis fol-
lowing trauma: a prospective, randomized comparison of mechanical and pharmacologic pro-
phylaxis. J Bone Joint Surg Am. 2006;88:261–6.
38. Kempf I, Grosse A, Beck G. Closed locked intramedullary nailing. Its application to commi-
nuted fractures of the femur. J Bone Joint Surg Am. 1985;67:709–20.
39. Lottes JO. Blind nailing technique for insertion of the triflange medullar nail: report of three
hundred nailing for fractures of the shaft of the tibia. J Am Med Assoc. 1954;155:1039–42.
40. Ly TV, Travison TG, Castillo RC, Bosse MJ, MacKenzie EJ, Group LS. Ability of lower-
extremity injury severity scores to predict functional outcome after limb salvage. J Bone Joint
Surg Am. 2008;90:1738–43.
41. Makridis KG, Tosounidis T, Giannoudis PV. Management of infection after intramedullary nail-
ing of long bone fractures: treatment protocols and outcomes. Open Orthop J. 2013;7:219–26.
42. Sassoon A, Riehl J, Rich A, et al. Muscle viability revisited: are we removing Normal muscle?
A critical evaluation of dogmatic debridement. J Orthop Trauma. 2016;30:17–21.
43. Slobogean GP, O’Brien PJ, Brauer CA. Single-dose versus multiple-dose antibiotic prophy-
laxis for the surgical treatment of closed fractures. Acta Orthop. 2010;81:256–62.
44. Acker JH, Murphy C, D’Ambrosia R. Treatment of fractures of the femur with the Grosse-
Kempf rod. Orthopedics. 1985;8:1393–401.
45. Basumallick MN, Bandopadhyay A. Effect of dynamization in open interlocking nailing of
femoral fractures. A prospective randomized comparative study of 50 cases with a 2-year fol-
low-up. Acta Orthop Belg. 2002;68:42–8.
46. Patton MS, Lyon TD, Ashcroft GP. Levels of systemic metal ions in patients with intramedul-
lary nails. Acta Orthop. 2008;79:820–5.
47. Qin YX, Kaplan T, Saldanha A, Rubin C. Fluid pressure gradients, arising from oscillations in
intramedullary pressure, is correlated with the formation of bone and inhibition of intracortical
porosity. J Biomech. 2003;36:1427–37.
48. Tornetta P 3rd, Collins E. Semiextended position of intramedullary nailing of the proximal
tibia. Clin Orthop Relat Res. 1996;328:185–9.
49. Tran W, Foran J, Wang M, Schwartz A. Postsurgical bleeding following treatment of a chronic
Morel-Lavallee lesion. Orthopedics. 2008;31:814.
50. Rose DM, Smith TO, Nielsen D, Hing CB. Expandable intramedullary nails in lower limb
trauma: a systematic review of clinical and radiological outcomes. Strategies Trauma Limb
Reconstr. 2013;8:1–12.
51. Betz A, Baumgart R, Schweiberer L. [First fully implantable intramedullary system for callus
distraction—intramedullary nail with programmable drive for leg lengthening and segment
displacement principles and initial clinical results]. Chirurg. 1990;61:605–9.
52. Bong MR, Kummer FJ, Koval KJ, Egol KA. Intramedullary nailing of the lower extremity:
biomechanics and biology. J Am Acad Orthop Surg. 2007;15:97–106.
53. Brumback RJ, Uwagie-Ero S, Lakatos RP, Poka A, Bathon GH, Burgess AR. Intramedullary
nailing of femoral shaft fractures. Part II: fracture-healing with static interlocking fixation. J
Bone Joint Surg Am. 1988;70:1453–62.
54. Utvag SE, Korsnes L, Rindal DB, Reikeras O. Influence of flexible nailing in the later phase of
fracture healing: strength and mineralization in rat femora. J Orthop Sci. 2001;6:576–84.
55. Utvag SE, Reikeras O. Effects of nail rigidity on fracture healing. Strength and mineralisation
in rat femoral bone. Arch Orthop Trauma Surg. 1998;118:7–13.
56. Uzer G, Thompson WR, Sen B, et al. Cell mechanosensitivity to extremely low-magnitude
signals is enabled by a LINCed nucleus. Stem Cells. 2015;33:2063–76.
57. Uhthoff HK, Dubuc FL. Bone structure changes in the dog under rigid internal fixation. Clin
Orthop Relat Res. 1971;81:165–70.
334 Y. Zhang et al.
58. Cole JD, Justin D, Kasparis T, DeVlught D, Knobloch C. The intramedullary skeletal kinetic
distractor (ISKD): first clinical results of a new intramedullary nail for lengthening of the
femur and tibia. Injury. 2001;32(Suppl 4):SD129–39.
59. Cullinane DM, Fredrick A, Eisenberg SR, et al. Induction of a neoarthrosis by precisely con-
trolled motion in an experimental mid-femoral defect. J Orthop Res. 2002;20:579–86.
60. Tigani D, Fravisini M, Stagni C, Pascarella R, Boriani S. Interlocking nail for femoral shaft
fractures: is dynamization always necessary? Int Orthop. 2005;29:101–4.
61. Claes L, Blakytny R, Besse J, Bausewein C, Ignatius A, Willie B. Late dynamization by
reduced fixation stiffness enhances fracture healing in a rat femoral osteotomy model. J Orthop
Trauma. 2011;25:169–74.
62. Foxworthy M, Pringle RM. Dynamization timing and its effect on bone healing when using the
Orthofix dynamic axial fixator. Injury. 1995;26:117–9.
63. Court-Brown CM, Gustilo T, Shaw AD. Knee pain after intramedullary tibial nailing: its inci-
dence, etiology, and outcome. J Orthop Trauma. 1997;11:103–5.
64. Dietch ZC, Petroze RT, Thames M, Willis R, Sawyer RG, Williams MD. The “high-risk” deep
venous thrombosis screening protocol for trauma patients: is it practical? J Trauma Acute Care
Surg. 2015;79:970–5; discussion 975
65. Study to Prospectively Evaluate Reamed Intramedullary Nails in Patients with Tibial Fractures
Investigators, Bhandari M, Guyatt G, et al. Randomized trial of reamed and unreamed intra-
medullary nailing of tibial shaft fractures. J Bone Joint Surg Am. 2008;90:2567–78.
66. Keating JF, O’Brien PJ, Blachut PA, Meek RN, Broekhuyse HM. Locking intramedullary nail-
ing with and without reaming for open fractures of the tibial shaft. A prospective, randomized
study. J Bone Joint Surg Am. 1997;79:334–41.
67. Klein P, Opitz M, Schell H, et al. Comparison of unreamed nailing and external fixation of
tibial diastases—mechanical conditions during healing and biological outcome. J Orthop Res.
2004;22:1072–8.
68. Oh JK, Hwang JH, Sahu D, Jun SH. Complication rate and pitfalls of temporary bridging
external fixator in periarticular communited fractures. Clin Orthop Surg. 2011;3:62–8.
69. Eggers GW. Internal contact splint. J Bone Joint Surg Am. 1948;30A:40–52.
70. Gerber C, Mast JW, Ganz R. Biological internal fixation of fractures. Arch Orthop Trauma
Surg. 1990;109:295–303.
71. Lebel E, Blumberg N, Gill A, Merin O, Gelfond R, Bar-On E. External fixator frames as
interim damage control for limb injuries: experience in the 2010 Haiti earthquake. J Trauma.
2011;71:E128–31.
72. Lenarz C, Bledsoe G, Watson JT. Circular external fixation frames with divergent half pins: a
pilot biomechanical study. Clin Orthop Relat Res. 2008;466:2933–9.
73. Lerner A, Fodor L, Soudry M. Is staged external fixation a valuable strategy for war injuries to
the limbs? Clin Orthop Relat Res. 2006;448:217–24.
74. Eijer H, Hauke C, Arens S, Printzen G, Schlegel U, Perren SM. PC-fix and local infection
resistance—influence of implant design on postoperative infection development, clinical and
experimental results. Injury. 2001;32(Suppl 2):B38–43.
75. Paffrath T, Lefering R, Flohe S, TraumaRegister DGU. How to define severely injured
patients?—an injury severity score (ISS) based approach alone is not sufficient. Injury.
2014;45(Suppl 3):S64–9.
76. Pape HC, Tornetta P 3rd, Tarkin I, Tzioupis C, Sabeson V, Olson SA. Timing of fracture fixa-
tion in multitrauma patients: the role of early total care and damage control surgery. J Am Acad
Orthop Surg. 2009;17:541–9.
77. Park SH, O’Connor K, Sung R, McKellop H, Sarmiento A. Comparison of healing process in
open osteotomy model and closed fracture model. J Orthop Trauma. 1999;13:114–20.
78. Gomez-Benito MJ, Garcia-Aznar JM, Kuiper JH, Doblare M. A 3D computational simulation
of fracture callus formation: influence of the stiffness of the external fixator. J Biomech Eng.
2006;128:290–9.
79. Siguier T, Glorion C, Langlais J, Rouvreau P, Pouliquen JC. External fixation in fractures of
the lower limb in children. Rev Chir Orthop Reparatrice Appar Mot. 1995;81:157–62.
12 Biomechanics of the Fracture Fixation 335
80. Marsh JL, Nepola JV, Wuest TK, Osteen D, Cox K, Oppenheim W. Unilateral external fixa-
tion until healing with the dynamic axial fixator for severe open tibial fractures. J Orthop
Trauma. 1991;5:341–8.
81. Paley D. PRECICE intramedullary limb lengthening system. Expert Rev Med Devices.
2015;12:231–49.
82. Gardner TN, Evans M, Kenwright J. The influence of external fixators on fracture motion
during simulated walking. Med Eng Phys. 1996;18:305–13.
83. Huang C, Ogawa R. Mechanotransduction in bone repair and regeneration. FASEB
J. 2010;24:3625–32.
84. Ilizarov GA. The principles of the Ilizarov method. Bull Hosp Jt Dis Orthop Inst.
1988;48:1–11.
85. Ilizarov GA, Frankel VH. The Ilizarov external fixator, a physiologic method of orthopaedic
reconstruction and skeletal correction. A conversation with Prof. G. A. Ilizarov and Victor
H. Frankel. Orthop Rev. 1988;17:1142–54.
86. Gandhi RR, Overton TL, Haut ER, et al. Optimal timing of femur fracture stabilization in
polytrauma patients: a practice management guideline from the eastern Association for the
Surgery of trauma. J Trauma Acute Care Surg. 2014;77:787–95.
87. Gillespie WJ, Walenkamp GH. Antibiotic prophylaxis for surgery for proximal femoral and
other closed long bone fractures. Cochrane Database Syst Rev. 2010;(3):CD000244.
88. Ozaki A, Tsunoda M, Kinoshita S, Saura R. Role of fracture hematoma and periosteum dur-
ing fracture healing in rats: interaction of fracture hematoma and the periosteum in the initial
step of the healing process. J Orthop Sci. 2000;5:64–70.
89. Pacicca DM, Moore DC, Ehrlich MG. Physiologic weight-bearing and consolidation of new
bone in a rat model of distraction osteogenesis. J Pediatr Orthop. 2002;22:652–9.
90. Wu CC. The effect of dynamization on slowing the healing of femur shaft fractures after
interlocking nailing. J Trauma. 1997;43:263–7.
91. Domb BG, Sponseller PD, Ain M, Miller NH. Comparison of dynamic versus static external
fixation for pediatric femur fractures. J Pediatr Orthop. 2002;22:428–30.
92. Farouk O, Krettek C, Miclau T, Schandelmaier P, Tscherne H. Effects of percutaneous and
conventional plating techniques on the blood supply to the femur. Arch Orthop Trauma Surg.
1998;117:438–41.
93. Glatt V, Evans CH, Matthys R. Design, characterisation and in vivo testing of a new, adjustable
stiffness, external fixator for the rat femur. Eur Cell Mater. 2012;23:289–98.. discussion 299
94. Glatt V, Miller M, Ivkovic A, et al. Improved healing of large segmental defects in the rat
femur by reverse dynamization in the presence of bone morphogenetic protein-2. J Bone
Joint Surg Am. 2012;94:2063–73.
95. Glatt V, Tepic S, Evans C. Reverse dynamization: a novel approach to bone healing. J Am
Acad Orthop Surg. 2016;24:e60–1.
96. Nickerson TP, Zielinski MD, Jenkins DH, Schiller HJ. The Mayo Clinic experience with
Morel-Lavallee lesions: establishment of a practice management guideline. J Trauma Acute
Care Surg. 2014;76:493–7.
97. Verkerke GJ, Schraffordt Koops H, Veth RP, et al. Design of a lengthening element for a
modular femur endoprosthetic system. Proc Inst Mech Eng H. 1989;203:97–102.
98. Wang L, Li JY, Zhang XZ, et al. Involvement of p38MAPK/NF-kappaB signaling path-
ways in osteoblasts differentiation in response to mechanical stretch. Ann Biomed Eng.
2012;40:1884–94.
99. Wang Q, Huang C, Xue M, Zhang X. Expression of endogenous BMP-2 in periosteal pro-
genitor cells is essential for bone healing. Bone. 2011;48:524–32.
100. Weyts FA, Bosmans B, Niesing R, van Leeuwen JP, Weinans H. Mechanical control of
human osteoblast apoptosis and proliferation in relation to differentiation. Calcif Tissue Int.
2003;72:505–12.
101. Wolf S, Janousek A, Pfeil J, et al. The effects of external mechanical stimulation on the
healing of diaphyseal osteotomies fixed by flexible external fixation. Clin Biomech (Bristol,
Avon). 1998;13:359–64.
336 Y. Zhang et al.
102. Haas N, Hauke C, Schutz M, Kaab M, Perren SM. Treatment of diaphyseal fractures of the
forearm using the point contact fixator (PC-fix): results of 387 fractures of a prospective
multicentric study (PC-fix II). Injury. 2001;32(Suppl 2):B51–62.
103. Hak DJ, Toker S, Yi C, Toreson J. The influence of fracture fixation biomechanics on fracture
healing. Orthopedics. 2010;33:752–5.
104. Hake ME, Young H, Hak DJ, Stahel PF, Hammerberg EM, Mauffrey C. Local antibiotic
therapy strategies in orthopaedic trauma: practical tips and tricks and review of the literature.
Injury. 2015;46:1447–56.
105. Goodship AE, Kenwright J. The influence of induced micromovement upon the healing of
experimental tibial fractures. J Bone Joint Surg Br. 1985;67:650–5.
106. Klein P, Schell H, Streitparth F, et al. The initial phase of fracture healing is specifically sensi-
tive to mechanical conditions. J Orthop Res. 2003;21:662–9.
107. Klein-Nulend J, Bakker AD, Bacabac RG, Vatsa A, Weinbaum S. Mechanosensation and
transduction in osteocytes. Bone. 2013;54:182–90.
108. Konda SR, Lack WD, Seymour RB, Karunakar MA. Mechanism of injury differentiates risk
factors for mortality in geriatric trauma patients. J Orthop Trauma. 2015;29:331–6.
109. Gorman SC, Kraus KH, Keating JH, et al. In vivo axial dynamization of canine tibial frac-
tures using the Securos external skeletal fixation system. Vet Comp Orthop Traumatol.
2005;18:199–207.
110. Grundnes O, Reikeras O. Effects of instability on bone healing. Femoral osteotomies studied
in rats. Acta Orthop Scand. 1993;64:55–8.
111. Haller JM, Holt D, Rothberg DL, Kubiak EN, Higgins TF. Does early versus delayed span-
ning external fixation impact complication rates for high-energy Tibial plateau and plafond
fractures? Clin Orthop Relat Res. 2016;474:1436–44.
112. Young H, Topliss C. Complications associated with the use of a titanium tibial nail. Injury.
2007;38:223–6.
113. Zucman J, Maurer P. Two-level fractures of the tibia. Results in thirty-six cases treated by
blind nailing. J Bone Joint Surg Br. 1969;51:686–93.
114. Wang L, Zhang Y, Song Z, et al. A novel method of using elastic bionic fixation device for
distal tibiofibular syndesmosis injury. Int Orthop. 2018;42(9):2219–29.
115. Tejwani SG, Cohen SB, Bradley JP. Management of Morel-Lavallee lesion of the knee:
twenty-seven cases in the national football league. Am J Sports Med. 2007;35:1162–7.
116. Thompson WR, Rubin CT, Rubin J. Mechanical regulation of signaling pathways in bone.
Gene. 2012;503:179–93.
117. Marsh JL, Slongo TF, Agel J, et al. Fracture and dislocation classification compen-
dium—2007: orthopaedic trauma association classification, database and outcomes commit-
tee. J Orthop Trauma. 2007;21:S1–133.
118. Marumo K, Sato Y, Suzuki H, Kurosaka D. MRI study of bioabsorbable poly-l-lactic acid
devices used for fixation of fracture and osteotomies. J Orthop Sci. 2006;11:154–8.
119. Morgan EF, Gleason RE, Hayward LN, Leong PL, Palomares KT. Mechanotransduction and
fracture repair. J Bone Joint Surg Am. 2008;90(Suppl 1):25–30.
120. Wang B, Zheng Z, Liu H, et al. The preliminary clinical application of Zhang’ compression
bolt for treatment of tibial plateau fracture. J Hebei Med Univ. 2018;
121. Farouk O, Krettek C, Miclau T, Schandelmaier P, Guy P, Tscherne H. Minimally invasive
plate osteosynthesis: does percutaneous plating disrupt femoral blood supply less than the
traditional technique? J Orthop Trauma. 1999;13:401–6.
122. Jansen JH, Weyts FA, Westbroek I, et al. Stretch-induced phosphorylation of ERK1/2 depends
on differentiation stage of osteoblasts. J Cell Biochem. 2004;93:542–51.
123. Kato M, Namikawa T, Terai H, Hoshino M, Miyamoto S, Takaoka K. Ectopic bone forma-
tion in mice associated with a lactic acid/dioxanone/ethylene glycol copolymer-tricalcium
phosphate composite with added recombinant human bone morphogenetic protein-2.
Biomaterials. 2006;27:3927–33.
12 Biomechanics of the Fracture Fixation 337
124. Kelly DJ, Jacobs CR. The role of mechanical signals in regulating chondrogenesis and osteo-
genesis of mesenchymal stem cells. Birth Defects Res C Embryo Today. 2010;90:75–85.
125. Hente R, Cordey J, Rahn BA, Maghsudi M, von Gumppenberg S, Perren SM. Fracture heal-
ing of the sheep tibia treated using a unilateral external fixator. Comparison of static and
dynamic fixation. Injury. 1999;30(Suppl 1):A44–51.
126. Huang Z, Wang B, Chen F, et al. Fast pinless external fixation for open tibial fractures: pre-
liminary report of a prospective study. Int J Clin Exp Med. 2015;8:20805–12.
127. Gardner MJ, Griffith MH, Demetrakopoulos D, et al. Hybrid locked plating of osteoporotic
fractures of the humerus. J Bone Joint Surg Am. 2006;88:1962–7.
128. Garwe T, Cowan LD, Neas B, Cathey T, Danford BC, Greenawalt P. Survival benefit of
transfer to tertiary trauma centers for major trauma patients initially presenting to nontertiary
trauma centers. Acad Emerg Med. 2010;17:1223–32.
129. Tanno M, Furukawa KI, Ueyama K, Harata S, Motomura S. Uniaxial cyclic stretch induces
osteogenic differentiation and synthesis of bone morphogenetic proteins of spinal ligament
cells derived from patients with ossification of the posterior longitudinal ligaments. Bone.
2003;33:475–84.
130. Yacoub AR, Joaquim AF, Ghizoni E, Tedeschi H, Patel AA. Evaluation of the safety and
reliability of the newly-proposed AO spine injury classification system. J Spinal Cord Med.
2017;40:70–5.
131. Yanagisawa M, Suzuki N, Mitsui N, Koyama Y, Otsuka K, Shimizu N. Effects of compressive
force on the differentiation of pluripotent mesenchymal cells. Life Sci. 2007;81:405–12.
Chapter 13
Biomechanical Principles in Designing
Custom-Made Hip Prosthesis
Jia Hua
Abstract Total hip replacement has increasingly become popular in treating osteo-
arthritis, relieving pain, restoring joint function and correcting the deformities.
However, due to the wide variations of the anatomy in femoral bone between indi-
viduals, the commonly used off-the-shelf standard hip stems cannot consistently
achieve the good fit, thus the clinical outcome is compromised. On the other hand,
an increasing demand for restoring the normal life quality after total hip replace-
ment including play sports and other activities, especially with the coming era of the
precision medicine and digital orthopaedics, and the availability of the advanced
surgical navigation systems and 3-D printing technologies, the patient-specific hip
prostheses have become ever favourable.
The rationale for using the custom stem is not only for the complex cases, but also
for the routine cases in order to achieve better clinical outcomes and longer survivor-
ship. This chapter will focus on how to design a custom hip stem for an individual
patient based on the bone geometry, deformities and pathological conditions. The
advantages and disadvantages of each design options are considered and analysed.
Keywords Hip joint arthroplasty · Total hip replacement · Design of hip stem
Custom hip stem · Osteoarthritis
1 Introduction
Constantly on the rise in quantities and qualities, total hip arthroplasty (THA) has
become ever popular in the treatment of hip diseases, and in relieving pain, restoring
function and correcting deformities [1]. THA is now one of the most successful
surgical intervention in the century for improving life quality of the patients.
J. Hua (*)
Department of Natural Science, Faculty of Science and Technology, Middlesex University,
London, UK
e-mail: j.hua@mdx.ac.uk
According to the 14th Annual Report of the National Joint Registry, over a hundred
thousand hip surgeries were performed in 2016 across the UK except for Scotland,
a 3.5% increase from the previous year, 90% of which were for the treatment of
osteoarthritis [2]. The joint registry data from the USA showed that 443,219 hip
surgeries have been reported between 2012 and 2017; among them revision surger-
ies accounted for 10.7% [3]. One of the concerns is that some of the revision surger-
ies were performed at very early stages after the first procedure, meaning that the
revision patients are no longer old and inactive; some of them now fall into the
physically active group, while the clinical results for revision THR are still inferior
to that for primary THR [4]. In the past, most cases are performed using standard
off-the-shelf hip stems; only a few of the very difficult cases have used the custom
designed prosthesis to correct deformities and provide best stem fixation and better
hip functions.
However, due to wide variations of anatomy in femoral bone and increasing
demand for the normal function of the hip including sports activity, especially with
the availability of the 3D printing technology, the patient-specific custom designed
hip prostheses have become more favourable. This chapter will focus on how to
design a custom hip stem for an individual patient based on their unique bone geom-
etry, deformities and pathological conditions. For those readers who are not design-
ing hip stems such as surgeons and researchers, may probably find the information
in this chapter can also be very useful.
In order to design a stem that can exactly fit with the femoral cavity, accurately
determining the boundary of cortical bone from the images, either from normal
X-rays or CT scan, is crucially important. Some of the factors can affect the deter-
mination of the boundary.
For normal plain X-ray images, it should normally include the pelvis and down
to the knee joint on AP and lateral views, with the scaled ruler placed on the side
of the femur at the level of the bone. The opposite side is ideally also included
for comparison. This allows the designer to assess the leg length discrepancy,
orientation of the knee and fixed deformities of the lumbar spine and the knee
joint, as these may affect reconstruction of the new centre of the hip.
The images should be clear with adequate contrast, brightness and resolution in
order for designers to identify the boundary, which involves proper adjustment of
kV and exposure time. However, for digital X-rays that have now been exclu-
sively used, the contrast and brightness can be manipulated on the screen; thus,
13 Biomechanical Principles in Designing Custom-Made Hip Prosthesis 341
the line of boundary showed on the images could be altered as a result, which
may not reflect the true boundary of the cortex. For using of CT scan, the deter-
mination of cortical boundary become critical.
Some of the other factors that could also affect the determination of the boundary
are the nature of the diseases and the density of the bone. For example, young
and active patients with osteoarthritis usually have hard bone, especially for a
band of trabecular bone adjacent to the cortex, which are sometimes too hard to
remove. On the other hand, for rheumatoid arthritis, the bone is quite softer,
especially for those under treatment of steroids. Therefore, the cavity can easily
be overreamed and the stem should be designed slightly tighter than normal. All
these should be considered when assessing the boundary of cortex for designing
custom hip stems.
To design a hip stem that achieves the best fit with the cavity is always considered
to be of critical importance in order to obtain a long-term success of the surgery.
This requires the geometry of the stem to match that of the femoral cavity. However,
for a stem to reach 100% fill of the cavity is unpractical in which the stem will not
be able to be inserted. Therefore, the designer should not only focus on which part
of the bone the stem should fit with but also assess whether the stem can be inserted
or not. In principle, the proximal medial curve of the femur in calcar region is
regarded as the most priority region which requires an exact fit in order to achieve
immediate stability of the stem and physiological stress transfer to the bone
(Fig. 13.1). However, this design mostly depends on the condition of the bone being
strong enough to carry the load.
The cortical bone on the lateral side of the femur is equally thick and strong as that
on the medial side due to the tensile stress across this region when the bone is loaded.
The lateral curve can provide stem with additional axial stability due to its wedge effect
and avoid stem from valgus tilt over time. However, one of the considerations is how
far should the stem follow the curve. This really depends on the position of the greater
trochanter and how wide is the opening at the neck resection. If the greater trochanter
is too medialized or the opening of the neck resection is too narrow, the lateral flare of
the stem needs to be reduced in order to facilitate the insertion of the stem.
For the anterior part of the femur, the stem geometry should not completely fol-
low the curve of the femur; otherwise the stem will be too thick to be inserted.
Nevertheless, the anterior curve of the stem is very useful in enhancing the torsional
stability when comparing with the thin and symmetrical design.
The design of the posterior part of the stem is simple; basically it is straight.
342 J. Hua
From biomechanical and clinical points of view, the stem length should be as short
as possible in order for the femoral cavity being least disturbed. The longer the
stem, the more damage to the bone can occur, which leaves revision surgery more
difficult. However, when the stem being too short it can cause instability and high
stress concentration on the lateral bone at the tip of the stem. In most cases, if the
stem geometry can match the normal proximal medial curve of the bone, a total
length of 90–100 mm (around 2 cm below the lesser trochanter) should be adequate
to provide stem stability as well as the physiological distribution of the stresses.
This is because when the proximal stem is properly supported by the medial calcar
bone, the stem tip will not be tilted under load and distribute lower contact stresses
13 Biomechanical Principles in Designing Custom-Made Hip Prosthesis 343
to the bone. In this regard, the design of the short stem is adequate. However, if the
stem fit in the calcar region is not appropriate or uncertain, then the length of the
stem should be increased to engage more cavity in diaphyseal region to avoid stress
concentration on the bone at the tip of the stem.
In most of the cases, to determine the diameter of the stem should be straightfor-
ward, which simply follows the diameter of the femoral canal. However, sometimes
the diameter needs to be adjusted in certain circumstances. The reason for this is
that the shape of the femoral cavity in the diaphyseal region is more likely oval, but
the distal stem is usually in round shape, so that cross-section of the distal stem will
not completely fit with the canal. Therefore, the diameter of the stem can be custom-
ized depending on how much bone can be overreamed on the narrow side.
In some unusual situations where the canal diameter is too small such as less than
6 mm while the patient has normal body weight, the stem diameter designed accord-
ingly could cause a fracture. In this case, a deliberately overream of the bone may
become necessary in order to increase the diameter of the stem, should the thickness
of the cortical wall and quality of the bone be allowed. If in certain cases that the
increase of the stem diameter being impossible which could cause weakness of the
bone, reducing offset of the neck or changing the material of the implant such as
using Cobalt Chrome instead become an alternative option.
On the other hand, if the diameter of the canal being too large such as over
16 mm, then the distal stem designed for may be too stiff that could lead to stress
shielding of the bone. In order to reduce the stiffness of the stem, a few methods can
be applied. One of the common methods is to make a slot in the middle of the stem,
or to remove part of the metal on the medial side towards to distal so that the shape
of the distal stem looks like a half-moon. Both designs can equally fulfil the pur-
pose, but offer different biomechanical benefits, so which method should be applied
depend on the actual cases.
The design of the stems outside the medullary canal is basically to determine the
centre of the femoral head that mainly involves the height, offset and angle of the
femoral stem neck. Different stem neck angle can influence the range of motions of
the hip joint and impingement between the neck of the stem and acetabular cup or
pelvic bone. The neck angle for the standard implants are usually around 45°, or
135° in neck-shaft angle. However, the neck angle for a custom implant can vary,
which depends on the anatomy of the particular patient and surgical requirement, so
344 J. Hua
that the height and offset of the femoral neck are adjusted in each case to suit for the
patients. Nowadays, the modular femoral heads have been exclusively used; there-
fore if the angle of the neck is more vertical, then any plus heads will increase the
height more than increasing the offset, and vice versa.
One of the challenges in designing custom hip stem is to determine the height of the
femoral head. It is always attempted to either maintain or equalize the leg length. In
most of the cases, the contralateral side of the femoral head can be referenced for the
affected side in order to make both leg lengths equal. However, if the leg length dis-
crepancy is too large, then only partial correction may be possible in order to prevent
overstretches of the nerves and vessels. For some of the cases, the soft tissues and joint
capsule may need to be extensively released during surgery to allow for pulling down
the leg and reducing the hip joint. In revision total hip replacements, the leg can nor-
mally be pulled down by 2.5 cm roughly; but in the situation such as developmental
dysplasia of the hip (DDH), only limited leg length can be corrected due to long-
lasting dislocation of the hip and shortening of the nerves, vessels and soft tissues. In
some situations, if the cup needs to be placed into the true anatomical acetabulum to
restore anatomical and biomechanical normality, conducting a sub-trochanteric oste-
otomy and removing a segment of the bone may become necessary.
Often less attention has been paid to adjust and optimize the offset of the femoral
neck when designing a hip stem. However, it can be of paramount importance in
affecting the hip joint biomechanically and functionally. The change of the neck
offset directly affects the forces across the hip joint. If the offset is increased, the
forces across the hip joint will decrease in a single leg stance, because the forces
from the abductor muscles are reduced. This can be calculated based on the balance
of the moments from two sides—the centre of the body weight and the force from
the abduct muscles. However, increasing the offset will increase the bending force
across the hip stem. If the stem diameter is too small, the stem fracture will cause
for concern; then the patients’ body weights need to be taken into consideration.
In the situation that the diameter of the stem cannot be increased due to small
cavity of the femur and overream of the bone is not attempted, in order to avoid stem
fracture, reducing offset of the femoral neck can be one of the choices to reduce
bending force across the hip stem. To do so, the distance of the lever arm of the
abductor muscles is decreased, in which more forces will be generated from the
muscle contraction in order to balance the moment of the hip joint when the patient
is in single leg stance. Therefore, the forces across the hip joint will be increased,
which subsequently increase the wear particles from the bearing surface of the joint.
13 Biomechanical Principles in Designing Custom-Made Hip Prosthesis 345
Stress shielding of the bone following total hip replacements has caused major con-
cerns over the past, and many research projects have been carried out to study the
stem geometries that can distribute the normal stresses over the bone [5]. According
to Wolff’s law, if the bone is under-stresses, bone resorption will occur [6]. The
clinical follow-up study showed that 20% of reduction in bone density at the calcar
region has been observed at 1 year after total hip replacements [7]. This may further
lead to bone fracture or loosening of the implants.
A number of factors can contribute to the stress shielding of the bone following
THR. One of the main reasons is the mismatch of the material modulus between the
implant and the bone so that most of the loads are carried by the implant rather than
the femoral bone. The higher Young’s modulus of elasticity for the implant materi-
als (110 GPa for Titanium alloy; and 230 GPa for Cobalt Chrome) comparing with
the bone (17 GPa for cortical bone at mid-shaft of the femur under compression
load) seem making this occurrence of stress shielding inevitable. In the past, some
of the hip stems have been designed with lower Young’s modulus such as Identifit
implant, but the clinical results were very disappointing, with 17% of revision rates
over the 2 years [8]. The reason for this is that reducing the stiffness of the implant
will increase interface micromotion between the stem and bone, which will inhibit
the bone ingrowth into the stem. Some of the other factors such as stem fails to
closely contact with the bone in the calcar region resulting in less stress transfer to
the bone also play a critical role. Therefore, the match of the stem geometry with the
bone in order to achieve a line-to-line fit and maintain long-term bone qualities is of
the most importance.
However, some researches have shown a wide variation in the geometry of the
femur between individuals, in which three types of femoral shape have been sum-
marized. The three shapes include normal, stovepipe and champagne bottle, which
represent different ratios in width between proximal and distal canals [9]. For this
reason, the off-the-shelf standard hip stems cannot consistently provide an optimal
fit for each patient.
In the occurrence of loading carried by the distal stem, where the distal fit is too
tight but the proximal bone contact is not achieved, the proximal stem will transfer
less stresses onto the bone. Over the time, the proximal part of the bone will undergo
stress shielding and bone resorption while the distal part will show the formation of
the para-prosthetic pedestal bone [10].
On the contrary, the custom designed hip stem can reliably provide the best fit
and transfer close to normal load onto the bone. A clinical follow-up of custom hip
stems showed that the bone densities measured by DEXA scan at 2 years
postoperatively were maintained at 97% when compared with that measured imme-
diately after surgery [11].
Therefore, in most of the cases for uncemented fixation, the distal stems should
only be designed for sliding fit to avoid distal loading, in which the function of the
distal stems is only served to prevent the stems from tilting.
346 J. Hua
Among all other factors that may have detrimental effects to the survivorship of the
hip implants, failing to obtain immediate fixation of the stem is the most critical. In
situations where the proximal bone is too poor to load, distal fixation should always
be regarded as an alternative. Primary factors relating to the stability of the stems
include relative motion between stem and bone, and migration of the stem over time.
Relative motion is defined as a recoverable relative position of a component
relative to its host bone after a cyclic loading (Fig. 13.2). This is due to elastic or
plastic deformation of the materials or geometric mismatch between bone and com-
ponent. The amount of relative motion for uncemented stems can vary but should be
around 10–150 μm. It can occur either in localized areas or over the entire interface.
If the motion is over 150 μm, osseointegration and biological fixation of the stem
will unlikely take place [12]. This phenomenon is due to higher tolerance of fibrous
tissue to strain energy in the interface than that of bone tissue. Furthermore, the rela-
tive motion can induce more shear stresses and strains in the bone, which is not
favourable to bone tissue ingrowth.
Therefore, in order to achieve long-term stability and biological fixation of the
stem through osteointegration, immediate mechanical fixation of the stem with the
bone is of critical importance. This to large degrees relies on the geometry of the
stem designed to match the medullary canal. To fit stem in the proximal medial
region of the canal has always been regarded by many designers as the priority for
achieving immediate fixation of the stems.
Migration is defined as a permanent change of position or subsidence of a com-
ponent relative to its host bone after many cyclic loadings over a period of time
(Fig. 13.2), in which the distance between the final position and initial position of
the stem is measured. This can similarly be due to elastic or plastic deformation, but
the geometric mismatch between bone and component plays large roles. In situa-
tions that stems have been in situ for many years, progressive bone resorption and
RELATIVE MOTION BETWEEN
TOTAL MIGRATION
STEM AND FEMUR
MICROMOTION
MOST MIGRATION AT 2500 CYCLES
OCCURS IN EARLY
CYCLES
Fig. 13.2 Diagram shows axial migration and micromotion of the hip stem in relation to the
femoral bone
13 Biomechanical Principles in Designing Custom-Made Hip Prosthesis 347
extensive osteolysis can also lead to considerable migration of the stem, eventually
causing aseptic loosening and failure of the stems.
Previously a number of studies have shown that early migration of the stem cor-
relates with the long-term clinical success. If the stems were initially stable and only
migrated less than 2 mm within 2 years, they were likely to be successful in long
term; otherwise if the stems were not stable at beginning and migrated over 2 mm
in 2 years, these stems were likely to fail [13–15]. In general, uncemented stems
need more time to stabilize which could take up to 1–3 months after surgery.
Therefore the uncemented stems may be migrated more at early stage which mostly
depends on the geometry of the stem, and also how tightly the stem has been
impacted during surgery. Once the stem has settled, the uncemented stem should be
as stable as the cemented stem for the initial stage. With porous and hydroxyapatite
coating for most of the uncemented stems, bone ingrowth and long-term biological
fixation will take place. Progressive migration of the stems for both cemented and
uncemented fixation indicates that a failure of the stem will soon occur.
The custom-made hip stems have achieved superior results for both relative
motion and migration when compared with the standard stems. A laboratory testing
showed that relative axial motion for custom-made stems was 11 μm in average
after 2500 cyclic loading, while for standard stems was 19 μm [16]. In a clinical
follow-up study, the migration for custom-made primary and revision stems were
less than 2 mm in 2 years, which were more stable than the standard stems [17].
Both relative motion and migration of the stem are three-dimensional referred to
the axis systems in the medullary canal. Relative motion can basically only be mea-
sured in vitro using LVTDs mounted onto the bone and attached to the stem. The
relative motion of the stem along the long axis of the femur, the horizontal motion
at the tip of the stem on both medial-lateral and anterior-posterior direction and the
rotational motion at the neck of the stem are usually measured. The differences in
reading before and after loading in a single cycle are calculated as the magnitude of
the relative motion. A number of cyclic loadings to pre-set the stem into the position
are necessary in order to achieve a stable and consistent reading and to reflect the
true value of relative motion. To measure relative motion after the stem is inserted
into patients is normally impossible, unless two X-rays are taken in a sequence. The
first X-ray is taken when the patient is standing on the effected leg (in loading situ-
ation) and the second X-ray is taken when the patient is standing on the alternative
leg (in off-loading situation). The distance between the stem and bone before and
after the leg is loaded can be measured and compared on the images, and the amount
of difference can be calculated as the value for relative motion.
Migration of the stem can be measured both in vitro using LVTDs and in vivo
using medical images. There are basically three types of methods that can be applied
to measure the migration.
Using the pen to mark the landmarks on the stem and the bone and measure them
by the ruler on plain X-rays. This method is simple and no additional equipment
is required. However, the accuracy is low, which can only be used in clinical for
a gross comparison and general assessment.
348 J. Hua
Applying a digitize technique on the plain X-rays either on the digitize table or
on the computer screen with a specially developed software. This method is
more accurate than using the ruler and pen. A previous study showed that the
accuracy of the method can be up to 0.25 mm if the landmarks on the stem and
bone can be clearly defined [15]. The most benefits of using this method are not
only that it can be more accurate but also that it can be applied for a retrospective
study. This means that all clinical plain X-rays taken in the past years can be used
for the study. However, this method can only measure the axial migration of the
stem, other axes such as rotation and varus-valgus tilt of the stem will not be able
to measure due to the two-dimensional views of the images.
Application of Radiostereometric analysis (RSA) is currently the most accurate
technique to measure the migration of the stems. The study showed that the accu-
racy of the method can reach to 0.15 mm. Moreover, it can also measure the
migration of the stem in three-dimensional planes using plain X-rays [18], so
that it can more truly reflect the status of the stem in situ. However, this method
requires special equipment, technical expertise and analysing software. Therefore
it can only be applied in special centres. The RSA techniques involve injecting a
number of tantalum beads into stem and bone during surgery as specially defined
reference marks for the measurements, and the radiograph has to be taken using
the special method of dual X-ray tubes placed at diagonal planes [19]. In this
way, any studies measuring the migration of the stems using RSA techniques has
to be pre-planned and set up prospectively, so that the study will take long times
to complete.
A most recently developed method of measuring migration of the stem using CT
scan has been studied [20]. The method also relies on the injection of the tanta-
lum beads as reference marks for the measurements; thus, it can only be used for
a prospective study. Instead of using dual X-ray tubes, it measures the reference
marks from the CT scan images. From the study, the femoral head was used as
the reference marks for the stem and the different lengths of the femoral heads
(−3.0, 0.0, 2.5, 5.0, 7.5 and 10 mm) were regarded as the migration of the stem
related to the bone. The accuracy of the method was claimed to be 0.11 mm [20],
even better than the RSA method. However, this method has only been validated
on the cadaveric bone, its real value for clinical application needs further
evaluation.
The 28 mm diameter of the femoral head has been mostly used in the past. However,
larger sizes of the femoral heads such as 32 and 36 mm have become more favour-
able for some of the surgeons. According to the report from American Joint
Replacement Registry, 36 mm femoral heads had become the most popular size,
followed by 32 mm [1]. This is because large femoral heads have the advantages of
providing stable joints, being less likely to dislocate, and potentially offering more
13 Biomechanical Principles in Designing Custom-Made Hip Prosthesis 349
range of motion of the hip. It can also increase the bearing surfaces and lead to
decreased contact stresses and thus reduced linear wear; but on the other hand, large
femoral heads cause more volumetric wear and number of wear particles.
Small femoral heads such as 22 mm diameter are usually only used due to the
restriction of the small size of the acetabular cups. However, small femoral heads do
provide biomechanical advantages such as producing lower friction between the
femoral head and the acetabular cup, offering shorter travel distance for the same
range of motion, and thus generating fewer wear particles.
Restoring range of motion of the hip should always be part of the design objectives
for the hip replacement. Although how the acetabular cup being placed can affect
the range of motion, the diameter of the femoral heads in relation to the sizes of the
stem neck can be one of the determining factors. In general, the bigger the size of
the femoral head, the more the range of the motion it can achieve, but this is not
always true. Strictly speaking, the range of motion of the hip is only associated
with the head/neck ratio. While for most of the cases where the size of the neck is
constant, the bigger size of the head will obtain more range of the motion. However,
for a small head such as 22.2 mm, it can achieve the same range of flexion and
extension as that for 28 mm head as long as a mini spigot on the anterior and pos-
terior side of the neck is produced, which make head/neck ratio the same as for
28 mm head.
Among other complex cases, developmental dislocation of the hip (DDH) is one of
the most challenge cases to design a hip stem for, due to its unusual bone geometry,
with excessive anteversion of the femoral neck, and often accompanied by a mal-
developed acetabular socket. The disease usually started when the baby is born,
gradually further developed and deteriorated over time. If the proper treatment is
not undertaken in time, the hip joint becomes so deformed that severely affect the
hip functions, and finally requires a total hip replacement. One of the difficulties in
designing a DDH stem is that the proximal medial part of the stem needs to be
gradually anteverted to follow the deformity of the femoral canal, while the stem
neck needs to be retroverted in order for the femoral head to match the normal ori-
entation of the acetabular cup (Fig. 13.3). With this design, the hip can be reduced
and the further dislocation of the hip can be avoided; but the full function of the hip
has still not been restored, because the greater trochanter and abductor muscle are
still at the posterior side of the hip.
350 J. Hua
Fig. 13.3 Hip stem designed for DDH case. The stem follows the geometry of the femoral cavity
and the femoral neck retroverted to the normal position of the acetabular cup
Another concern is how to secure the stem with the cavity below the osteotomy
site. The proximal and the distal part of the bones need to be held together tightly by
the stem, so bone rotation around the stem will occur after the stem undergoes tor-
sional loading. A stem with distal cutting flutes is usually designed to provide the
torsional stability for the distal stem, while the proximal stability is normally
achieved by the geometry fit with the bone. Sometimes, the transverse screws
through the distal bone and stem can be used to provide the torsional stability, pro-
vided the diameter of the stem is large enough for the hole. However, this option has
been less favourable because it may cause destruction and weakness for both bone
and stem. If the diameter of the femoral canal in the diaphyseal region is too small,
both designs may not be suitable. In this situation, two-stage surgeries, with de-
rotational and bone shortening osteotomy performed first followed by total hip
replacement, may be the choice.
In overall, the design for revision stems are more complicated than that for primary
stems because there are large variations on the quality of the remaining bone, as
well as the different locations of the bone defects. However, the principles for
designing a revision stem should be kept the same as that for a primary stem, in
which to achieve the initial stability of the stem is the priority. In revision situations,
the design rationales for each case can be different, which are mostly determined by
the condition of the remaining bone. From previous experience of designing custom-
made hip prostheses, the design rationales for each revision stem have been sum-
marized into four types [12], which were correspondent to Paprosky’s classification
of cavitary defects in revision situation [22].
• Type 1: The quality of the bone is uniformly preserved along the entire femoral
canal. The density of the bone especially in the calcar region is normal which can
be used for load bearing. In this case, the revision stem can be designed short just
like a usual primary stem, regardless of the length of the original stem. The fixa-
tion can be relied on the calcar and metaphyseal bone.
• Type 2A and B: The proximal bone is reasonably maintained. The implant can
be fully or partially supported by the metaphysis. The diaphyseal bone is mostly
intact but there are localized osteolytic lesions, particularly around the previous
distal stem. For this situation, proximal fixation is still obtainable, but the stem
should be designed longer in order to bypass the defects. The optimal length of
stem to bypass the defect should be 2.5–3.0 times the canal diameter in order to
avoid the distally concentrated stresses further weakening the defect and causing
bone fracture. The stress concentration caused by the tip of the stem is mostly
around the lateral side. Therefore, the stem should pass more if the defect is on
the lateral side of the bone.
352 J. Hua
Comparing with the clinical results of the standard hip implants, the information for
custom hip implants are much less published. Furthermore, the evaluation of the
clinical results for custom hip stems is more difficult because none of the implants
is designed the same, especially if the stems are designed from the different compa-
nies. In overall, most of the clinical results for custom hip stems are excellent.
A clinical follow-up study of the primary custom hip stems designed by the
author and produced by Stanmore Implants Worldwide (SIW) showed that among
the 114 consecutive CAD-CAM custom-made primary hip stems, inserted by a
single surgeon from one hospital, the survivorship was 100% with the average
13.2 years of follow-up (ranged from 10 to 17 years) and mean age of 46.2 years old
(range from 24 to 62 years old), although after 12 years, some cases have changed
the acetabular liner and femoral head but none of the femoral stems need replace-
ment. A radiographic study showed no aseptic loosening around the stem [23, 24].
Also, a study of the 158 consecutive revision custom hip stems with minimal
10 years follow-up showed that all femoral components were well fixed radiologi-
cally, with 97% survivorship at 10 years. Among the revised cases, some were not
related to the femoral components [23, 24]. One of the observation from the radio-
13 Biomechanical Principles in Designing Custom-Made Hip Prosthesis 353
graphic study showed that there were very little osteolysis occurring, even after
some of the femoral heads and acetabular liners being revised. This suggests that the
close geometric fit of the stem with the cavity may have additional benefits of
achieving layers of osteointegration thus seal the stem-bone interface. If this can
happen, the wear particles generated from the bearing surface will be prevented
from travelling into the femoral cavity, furthermore prevent the macrophages and
osteoclasts from reacting to the wear particles. As a result, osteolysis can be avoided.
Apart from many benefits of using custom stems, some of the disadvantages should
also be considered, which could be the main reasons to prevent the custom stems
from being widely used. However, with the advance of the new technologies, some
of the issues may be able to solve. From surgical point of view, one of the major
disadvantages is that no sequence of the tools can be used to prepare the femoral
cavity and to facilitate insertion of individual implant. Usually only the final shape-
matched custom reamer and rasp can be provided for each case. Therefore, some of
the surgeons may find difficulty to insert a custom hip stem, because all the custom
stems are designed to have line-to-line fit with the cavity, any misalignment at the
initial insertion point will cause the implant jam inside the cavity. With the advance
and wide use of the surgical navigation system, the surgical procedures can be more
precise, accurate and easier. Another disadvantage of using custom stem could be
that the stem may not fit with the bone, in which the stem may be unable to be
inserted. A number of factors can cause this happen. One of the common factors is
the quality of the images makes the boundary of the cortical bone not well defined
or misinterpreted. Other factors could also be possible such as the time between the
images taken and surgery is too long so that the bone geometry has changed since.
Other disadvantages related to the custom stems such as costing and delivery timing
should not be the restricting factors now because the increasingly popular 3D print-
ing technology can make manufacture process of custom stems more quickly, effi-
ciently and economically.
11 Summary
In total hip replacements, the standard stems have been playing a major role for a
long time. However, since individual patients have significantly different require-
ments, such as bone conditions, anatomy, pathologies, clinical problems, and expec-
tations for hip functions/activities/quality of life, custom stems are becoming ever
more popular and demanding in performance. This is true not only for the complex
cases, but also for the routine cases to achieve better clinical outcomes and longer
survivorship. While we are expecting the coming era of precision medicine and
354 J. Hua
digital orthopaedics, advanced surgical navigation systems and 3-D printing tech-
nologies are paving the way for using patient specific custom stems to solve patient
specific clinical problems. Although the advanced and rapid development of future
science and technology will change people’s view and expectation on total hip
replacements, the basic principles of implant design will remain the same. I would
be very pleased if this chapter can be of some contribution and useful to those dedi-
cated to the design of hip stems.
References
Berry D. In: Fourth American Joint Replacement Registry (AJRR) 2017 annual report on
hip and knee arthroplasty data, Rosemont, IL. ISSN 2375-9100 (print) and ISSN 2375-9119
(online). 2017.
Green M, Howard P, Porter MWilkinson M and Wishart N. In: 14th annual report National
Joint Registry for England, Wales, Northern Ireland and the Isle of Man, Surgical data to 31
December 2016; ISSN 2054-1821 (print). 2017.
Bozic K. In: Fifth American Joint Replacement Registry (AJRR) 2018 annual report on hip
and knee arthroplasty data, Rosemont, IL. ISSN 2375-9100 (print) and ISSN 2375-9119
(online). 2018.
Postler AE, Beyer F, Wegner T, Lützner J, Hartmann A, Ojodu I, Günther KP. Patient-
reported outcomes after revision surgery compared to primary total hip arthroplasty. Hip Int.
2017;27(2):180–6.
Behrens BA, Wirth CJ, Windhagen H, et al. Numerical investigations of stress shielding in
total hip prostheses. Proc Inst Mech Eng H. 2008;222(5):593–600.
Chamay A, Tschantz P. Mechanical influences in bone remodeling. Experimental research on
Wolff’s law. J Biomech. 1972;5(2):173–80.
Tapaninen T, Kröger H, Venesmaa P. Periprosthetic BMD after cemented and uncemented total
hip arthroplasty: a 10-year follow-up study. J Orthop Sci. 2015;20(4):657–62.
Robinson RP, Clark JE. Uncemented press-fit total hip arthroplasty using the identifit custom-
molding technique. J Arthroplast. 1996;11(3):247–54.
Dorr LD, Faugere MC, Mackel AM, et al. Structural and cellular assessment of bone quality of
proximal femur. Bone. 1993;14(3):231–42.
Agathangelidis F, Boutsiadis A, Petsatodis G. Pedestal sign in cementless total hip replace-
ment. Hippokratia. 2014;18(4):378.
Hua J, Walker PS, Muirhead-Allwood SK, Engelhardt F, Bentley G. Custom uncemented revi-
sion stems based on a femoral classification. Hip Int. 2010;20:18–25.
Soballe K, Brockstedt-Rasmussen H, Hansen ES, Bunger C. Hydroxyapatite coating modifies
implant membrane formation. Controlled micromotion studied in dogs. Acta Orthop Scand.
1992;63:128–40.
Freeman MAR, Plante-Bordeneuve P. Early migration and late aseptic failure of proximal
femoral prostheses. J Bone Joint Surg Br. 1994;76-B:432–8.
Walker PS, Mai SF, Cobb AG, Bentley G, Hua J. Prediction of clinical outcome of THR from
migration measurements on standard radiographs: a study of cemented Charnley and Stanmore
femoral stems. J Bone Joint Surg Br. 1995;77-B:705–14.
Hua J, Walker PS. Relative motion of hip stems under load: an in vitro study of symmetrical,
asymmetrical and custom asymmetrical designs. J Bone Joint Surg. 1994;76-A(1):95–103.
Walker PS, Culligan S, Hua J, Muirhead-Allwood S, Bentley G. Stability and bone preserva-
tion in custom designed revision hip stems. Clin Orthop. 2000;373:164–73.
Alfaro-Adrian J, Gill HS, Murray DW. Cement migration after THR: a comparison of Charnley
Elite and Exeter femoral stems using RSA. J Bone Joint Surg Br. 1999;81(1):130–4.
13 Biomechanical Principles in Designing Custom-Made Hip Prosthesis 355
Chung VWJ. Leveraging the use of existing C-arms for roentgen stereophotogrammetric anal-
ysis. University of British Columbia; 2015.
Boettner F, Sculco P, Lipman J, Renner L, Faschingbauer M. A novel method to measure
femoral component migration by computed tomography: a cadaver study. Arch Orthop Trauma
Surg. 2016;136(6):857–63.
Zadeh HG, Hua J, Walker PS, Muirhead-Allwood SK. Uncemented total hip replacement with
subtrochanteric derotational osteotomy in hip joint osteoarthritis with concomitant severe
femoral anteversion. J Arthroplast. 1999;14(6):682–8.
Paprosky WG, Lawrence J, Cameron H. Femoral defect classification: clinical application.
Orthop Rev. 1990;19(Suppl):9–15.
Muirhead-Allwood S, Sandiford N, Skinner JA, Hua J, Kabir C, Walker PS. Uncemented
custom computer-assisted design and manufacture of hydroxyapatite-coated femoral compo-
nents—survival at 10 to 17 years. J Bone Joint Surg Br. 2010a;92-B(8):1079–84.
Muirhead-Allwood S, Sandiford NA, Skinner JA, Hua J, Muirhead W, Kabir C, Walker
PS. Uncemented computer-assisted design-computer-assisted manufacture femoral compo-
nents in revision total hip replacement: a minimum follow-up of ten years. J Bone Joint Surg
Br. 2010b;92(10):1370–5.
Hua J, Walker PS. Closeness of fit of uncemented stems improves the strain distribution in the
femur. J Orthop Res. 1995;13(3):339–46.
Chapter 14
Biomechanics of Orthopedic Rehabilitation
Abstract This chapter discusses four main areas that clinicians should understand to
improve the effectiveness of orthopedic rehabilitation. The first part is about the biome-
chanical factors that affect orthopedic rehabilitation process. The second part is about
the types of exercises during orthopedic rehabilitation and their underlying biome-
chanical principles. The third part is about how to apply these biomechanical principles
to manage common orthopedic disorders. The fourth part is about the biomechanical
guidelines for the use of assistive devices during orthopedic rehabilitation.
A. A. Mohamed
Rehabilitation Engineering Lab, Department of Kinesiology and Community Health,
University of Illinois at Urbana-Champaign, Champaign, IL, USA
Department of Basic Science and Biomechanics, Faculty of Physical Therapy,
Beni-Suef University, Beni Suef, Egypt
Y.-K. Jan (*) · I. M. Rice
Rehabilitation Engineering Lab, Department of Kinesiology and Community Health,
University of Illinois at Urbana-Champaign, Champaign, IL, USA
e-mail: yjan@illinois.edu
F. Pu
Advanced Innovation Center for Biomedical Engineering, School of Biological Science
and Medical Engineering, Beihang University, Beijing, China
C.-K. Cheng
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
e-mail: ckcheng2020@sjtu.edu.cn
© Springer Nature Singapore Pte Ltd. 2020 357
C.-K. Cheng, S. L-Y. Woo (eds.), Frontiers in Orthopaedic Biomechanics,
https://doi.org/10.1007/978-981-15-3159-0_14
358 A. A. Mohamed et al.
Learning Objectives
After reading this chapter, the readers will be able to:
1. Understand the biomechanical factors of the musculoskeletal tissues (muscles,
tendons, and ligaments) in the human body that can help to improve orthopedic
rehabilitation
2. Understand the biomechanical principles of common exercise types in orthope-
dic rehabilitation
3. Recognize the advantages of applying biomechanical principles to improve
effectiveness of orthopedic rehabilitation
4. Know the common orthopedic disorders of the upper and lower joints and how
biomechanical principles can improve their interventions during orthopedic
rehabilitation
5. Use the biomechanical principles to prescribe appropriate assistive devices
(splints, walkers, crutches, and canes) to improve orthopedic rehabilitation.
There are several biomechanical factors that should be well understood when per-
forming orthopedic rehabilitation to the human musculoskeletal tissues. This part
discusses the factors that clinicians should understand when applying different
rehabilitation therapies. We provide examples of key factors to clarify the role of
these factors during orthopedic rehabilitation.
1.1 The Viscoelasticity
The creep phenomenon is the first common viscoelastic property that the musculo-
skeletal tissues follow when they are subjected to stress. The creep phenomenon is
defined as the gradual lengthening (shortening) of a viscoelastic material with time
when a constant tensile (compressive) stress is placed on it. When a constant tension
is placed on a muscle for a period of time, this tension is distributed on all of the
sarcomeres in an equal manner. An increase (decrease) in one sarcomere length due
to this tension produces a change in its isometric tension. This causes a lengthening
(shortening) of this sarcomere at a fixed speed that matches the amount of applied
tension (speed of sarcomere change is proportionate to the magnitude of applied
tension). For example, use of splints for stretching shortened musculoskeletal tis-
sues (lengthening effect) functions primarily through the creep phenomena. These
tissues change their shape after a period of time in response to the constant tension
of the applied splint.
The hysteresis is the second common viscoelastic property that the musculoskel-
etal tissues follow when they are subjected to stress. The hysteresis can be defined
as the energy loss in the form of heat. It is considered a recovery process after the
removal of a certain load. The amount of energy loss as heat is based on the rate
of applied strain to deform the body tissues. The continual loading and unloading
can explain the increase in the temperature of the viscoelastic material during
loading and unloading (Fig. 14.1). For example, when a shortened ligament is
repetitively stretched with a constant stretching force (stretch then relax then
stretch), hysteresis is developed and the ligament length limit increases with each
cycle. Thus, an extra ligament length occurs in the same session by performing a
repetitive stretching manner.
Strain
360 A. A. Mohamed et al.
The stress–relaxation relationship is the third common viscoelastic property that the
musculoskeletal tissues follow when they are subjected to stress. The stress–relax-
ation relationship can be defined as the reduction in the stress due to the same degree
of strain produced in the viscoelastic tissues. To explain this, when musculoskeletal
tissues (viscoelastic materials) are held under tensile stress for a prolonged time,
there is an increase in this stress over time. Thus, the stress should be increased to
produce the same required strain (Fig. 14.2). For example, in static stretching, when
a constant stretch (strain) is applied on a muscle for a prolonged time, the magnitude
of the stretching force (stress) is less than the firstly applied stretching force.
The force–velocity relationship is the fourth common viscoelastic property that the
musculoskeletal tissues exhibit when they are subjected to stress. The force–veloc-
ity relationship can be defined as the amount of muscle force in relation to the veloc-
ity of motion or vice versa. Based on this relationship, the maximum possible
tension occurs with the eccentric contraction (Fig. 14.3). This clarifies why it is
easier to lower a load and stop it than lifting it and why it is easier to descend from
a certain height and land than to jump up to the same height.
Viscous material
Relaxation beginning line
Viscoelastic material
Elastic material
Fig. 14.2 The stress relaxation curve. There are differences in the relaxation behaviors for the vis-
cous, elastic, and viscoelastic materials in the stress–relaxation curve (σ = stress, T = time). The
elastic material returns immediately to its original state after relaxation. The viscoelastic material
exhibits some changes in the return way, loses some energy in the form of heat and an alteration in
its original state occurs. The viscous material continues in the state developed by the stress and does
not relax
14 Biomechanics of Orthopedic Rehabilitation 361
Force
concentric contraction, as
the load increases, there is
an increase in the velocity
of contraction Concentric
Elongation Shortening
Speed
The Hill’s muscle model is another biomechanical factor that can help us to
understand which muscle tissues are responsible for developing each type of the
muscle tension. The Hill’s muscle model demonstrates that there are two sources
participated in the development of the muscle tension (active and passive). The
active tension is developed by the interaction of actin and myosin filaments in the
muscle, while the passive tension is developed by the connective tissue compo-
nents (fascia and tendon) in the muscle when the muscle length exceeds its resting
length. The Hill’s muscle model demonstrates that the active and passive tensions
are depending on each other and each of them cannot be considered as a separate
structural element of the muscle because the connective tissue matrix of the mus-
cle is relatively complex and connected to each other (within and between the
muscles) [11] (Fig. 14.4).
362 A. A. Mohamed et al.
The most common exercise types during orthopedic rehabilitation are stretching,
strengthening, ROM, and mobilization exercise.
2.1 Stretching Exercise
The late effects of stretching exercise are much better than the immediate effects.
The first effect is due to the prolonged stretch that causes a significant improvement
(8–12° or 5–31%) in the ROM after 12 weeks of training. This effect is attributed to
14 Biomechanics of Orthopedic Rehabilitation 363
There are three types of muscle stretching exercise commonly performed in ortho-
pedic rehabilitation (static, dynamic, and pre-contraction). The first type, static
stretching exercise, is most common and regularly used. In static stretching, the
stretching force is constant and includes two subtypes, passive and self-stretching.
Passive stretching exercise occurs when the stretch force is applied by the therapist
or another person. In contrast, self-stretching exercise requires the patient to apply
the stretch force independently.
The second type is the dynamic stretching exercise that occurs when the stretch-
ing force is not constant and includes two subtypes, active and ballistic stretching.
The active stretching exercise is usually applied by moving the limb repeatedly until
the end of the available ROM. The ballistic stretching exercise is conducted by
quick, alternating stretching motions or “bouncing” performed at the end of the
available ROM. This technique is not recommended as there is an increased inci-
dence of the injury to the stretched tissues.
2.2 Strengthening Exercise
muscle strength of the patient. This section discusses two biomechanical principles
of strengthening exercise [18] and lever systems [19].
There are three types of the strengthening exercise (isometric, isotonic, and iso-
kinetic types). The isometric strengthening exercise is a static type of the strength-
ening exercise. During isometric strengthening, muscles contract without any
noticeable alteration in either its length or the joint ROM. Although there is no
movement, the amount of force and tension developed can be measured. The sec-
ond type is the isotonic strengthening exercise. Isotonic strengthening is a dynamic
type of the strengthening exercise in which muscle contraction occurs with joint
movement and excursion. It includes two subtypes, the concentric type, in which
the muscle force is produced through contraction (muscle shortening), and the
eccentric type, in which the force is produced during muscle lengthening. The third
type, isokinetic strengthening exercise, is a dynamic type of strengthening exercise
in which muscle contraction occurs about a joint moving at a fixed velocity.
The maximum force generated with eccentric strengthening contraction is more
than during isometric or concentric contraction due to two causes [20]. First, when
a muscle is stretched before contraction, more cross-bridges are included, and thus
more tension is developed than when muscle contracts from a shortened position.
Second, the cross-bridges cycling interaction can occur with less adenosine triphos-
phate (ATP) hydrolysis than in concentric contractions. However, the eccentric con-
tractions are accompanied by a lesser energy cost and high force output and have a
higher tension production that leads to a damage to the muscle-tendon system.
Thus, using the eccentric contraction during orthopedic rehabilitation should be
performed with caution because of a high incidence of injury.
The lever can be defined as a semirigid or rigid segment that moves on a stationary
point (axis) when a force is applied to this segment (not passing through its axis
point) [19]. The lever has a capability to assist or resist the muscle contraction. This
capability is based mainly on its mechanical advantage. The mechanical advantage
of the lever equals the proportion of muscular force to the resistance force. Levers
are classified into three categories by the relative locations of the axis, muscular
force, and resistance. They include three classes.
For the first class, the axis is located between the force and the resistance.
Thus, the force arm can be greater, equal, or less than the resistance arm. For an
example, in the extension of the neck. The axis (the atlantooccipital joint) is posi-
tioned between the resistance (head weight) and the force (deep cervical exten-
sors). The mechanical advantage may be higher than, less than, or equivalent to 1
14 Biomechanics of Orthopedic Rehabilitation 365
depending on the distance of the head to the joint. Thus, the assistance or resis-
tance to the muscle contraction depends on which the lever arm is larger
(Fig. 14.5a).
For the second class, the resistance is located between the force and the axis.
Thus, the force arm is always bigger than the resistance arm. For an example, the
resistance (body weight) is positioned between the axis (the metatarsophalangeal
joints) and the force (calf muscle) during standing on the toes. The mechanical
advantage of the second class is constantly larger than the first class because the
resistance arm is less than the force arm. Thus, it causes an increase in the force
production. It is also called the force multiplier lever (Fig. 14.5b).
For the third class, the force is located between the resistance and the axis. Thus,
the resistance arm is always bigger than the force arm. For an example, the force
(biceps brachii muscle) is positioned between the axis (the elbow joint) and the
resistance (forearm weight) during elbow flexion. The mechanical advantage is con-
stantly less than the first class because the force arm is lesser than the resistance
arm. Thus, it causes an increase in the speed of movement. It is also called the speed
multiplier lever (Fig. 14.5c).
Using lever systems to graduate the strengthening exercise is considered a safe
and effective way during orthopedic rehabilitation. For an example, during strength-
ening exercise for weak shoulder flexors, the strengthening exercise with elbow
flexion is easier than with elbow extension while the weight of the upper limb is
fixed. This is because when the elbow is flexed the resistance arm is only to the
elbow joint; while, when the elbow is extended the resistance arm is to the fingers.
Increasing the lever arm of the resistance increases the demand on the muscle to
counteract this increase.
R = Head weight
a
F R
b
F= Calf muscle
R = Body weight
F R
A = Metatarsophalangeal joints
c
F = Elbow flexors muscles
Bic
R F
ep
s
A = Elbow joint
R = Forearm weight
Fig. 14.5 Types of lever systems. (a) First class lever. As in the action of the deep cervical exten-
sors muscles. The force is performed by cervical deep extensors muscles to overcome the resis-
tance which is applied by the weight of the head, and the axis is the atlantooccipital joint. (b)
Second class lever. As in the action of the calf muscle when standing on the toes. The force is
performed by the calf muscle to overcome the resistance which is applied by the weight of the
body, and the axis is the metatarsophalangeal joints. (c) Third class lever. As in the action of the
biceps muscle when it flexes the elbow joint. The force is performed by the biceps muscle to over-
come the resistance which is applied by the weight of the forearm, and the axis is the elbow joint.
F muscular force, R resistance, A axis
14 Biomechanics of Orthopedic Rehabilitation 367
Two primary types of movements occur within joints and there are specific exer-
cises for each movement. The first joint movement is the physiological movement,
which is defined as the movement of the two joint bone segments outside the joint
through the available ROM. The second joint movement is the accessory move-
ment; it is defined as the movement of the two joint segments inside the joint cap-
sule through the available ROM “joint play” motion. In the ROM exercises the
focus is on the physiological movement, while the mobilization exercise focuses on
accessory movements.
There are three fundamental planes and axes in which physiological movements
occur and orthopedic rehabilitation team should be aware of which physiologi-
cal movement plane is limited such that a ROM exercise plan can be formu-
lated. The first plane is the sagittal plane; it intersects the body into anterior and
posterior halves. The axis perpendicular to the sagittal plan in which move-
ments occur is known as the frontal axis. The primary movements of the sagittal
plane are flexion and extension. The second plane is the frontal plane which
intersects the body into medial and lateral halves and its movement axis is the
sagittal axis. Primary movements occurring in the frontal plane are adduction
and abduction. The third type is the transverse plane, which intersects the body
into upper and lower halves and its movement axis is the vertical axis. The
movements occurring in the transverse plane are rotation, elevation, and
depression.
There are three main types of ROM exercise commonly used during orthopedic
rehabilitation. The first type is passive ROM exercise, in which no voluntary con-
traction occurs and an external force is used to move the segment through the avail-
able ROM. The external force can be the other limb, gravity, a machine, or a
therapist. The second type is active ROM exercise, in which no external assistance
is used and the segment is moved through the available ROM with active voluntary
contraction created by the muscles crossing that joint. The third type is the active-
assistive ROM; it is a combination of both passive and active ROM. In which the
first part of the ROM is actively performed and the second part is passively p erformed
because the agonist muscles cannot complete the whole ROM alone due to its
weakness.
368 A. A. Mohamed et al.
2.4 Mobilization Exercise
Mobilization exercise can be defined as a force applied to move the articular sur-
faces inside a joint. It is performed to improve the joints mobility, ROM or to reduce
accompanying pain associated with joint structures via passive or manual accessory
joint movement [6]. In order to perform a successful mobilization exercise, the
rehabilitation team should know three main aspects. The first aspect is understand-
ing how the accessory movements work to form the mobilization exercise. The sec-
ond aspect is the biomechanics of the articular surfaces of the treated joint including
the convex–concave rule and the loose- and close-packed positions of this joint and
the grades for this mobilization exercise. The third aspect is the grades of the mobi-
lization exercise and the indications for each. This section discusses types of joint
play movements [24], the convex–concave rule [25], close- and loose-packed posi-
tions [26], and grades of the mobilization exercise [27].
a b c d e
Spinning
Compression
Sliding
llin
Traction
Ro
Femur
Tibia
Fig. 14.6 Types of accessory movements. (a) Gliding movement, in which the same point on one
of the two joint bony surfaces contacts new points on the other bone surface. (b) Rolling move-
ment, in which the new point on one of the two joint bony surfaces contacts new points on the other
bone surface. (c) Spinning movement, in which one joint bony surface rotates around a stationary
axis in clockwise or counterclockwise direction. (d) Traction movement, in which there is an
increase in the vertical linear separation between both joint surfaces away from the treatment
plane. (e) Compression movement, in which there is a decrease in the vertical linear separation
between the joint surfaces away from the treatment plane
joint supination, the radius (concave) moves on the ulna (convex). To perform a
successful mobilization exercise to increase distal radioulnar joint supination, the
mobilizing force should be in a posterior direction (same direction) (Fig. 14.7b).
There are two common positions for any joint. The first position is the close-packed
position; it is a joint position in which the capsule ligaments are taut and there is a
maximum contact between the two contacting bones. It is the most congruent posi-
tion of the joint. The second position is the loose-packed position; it is a joint posi-
tion in which the capsule ligaments are loose and there is a minimum contact
between the two contacting bones. It the loosest position of the joint. The mobiliza-
tion exercise should be performed in the loose-packed position, not in the close-
packed position, because a greater movement can be achieved in this position.
lide
a b rior g
Inferior glide
Ante
1
1
Humerus,
convex,
Moving. Radius, Ulna,
Scapula,
Concave, Convex,
Concave,
Moving. Fixed.
Fixed
Abduction
ation
Pron
2
Fig. 14.7 The convex–concave rule. (a) Mobilizing force. (b) Movement direction. (a) The mobi-
lizing force for increasing shoulder abduction is performed in an opposite direction to the desired
movement (inferior glide) because the convex humeral head moves on the concave glenoid cavity
of the scapula (convex on concave rule). (b) The mobilizing force for increasing the pronation in
the distal radioulnar joint is performed in a same direction of the desired movement (the anterior
glide) because the concave ulnar notch on the radius moves on the convex ulnar head (concave on
convex rule). 1: mobilization force direction and 2: direction of desired movement
oscillations and small amplitude movements are performed at only the beginning of
the available ROM. It is mainly used to decrease pain. Grade II is a low speed, small
oscillations and large amplitude movements are performed near the middle of the
available ROM. It is mainly used to decrease pain. Grade III is a low speed, small
oscillations and large amplitude movements are performed near the end of the avail-
able ROM. It is mainly used to increase the ROM. Grade IV is a low speed, small
oscillations, and small amplitude movements are performed at the end of the avail-
able ROM. It is mainly used to increase the ROM. Grade V is a high speed, large
oscillations and small amplitude movements are performed beyond the end of the
available ROM. It is called manipulation.
The upper limbs consist of several joints, including the shoulder, elbow, wrist, and
hand joints. Upper limb functions typically consist of activities that require a high
degree of mobility.
The shoulder girdle is responsible for complex movements which are important for
functional activities. To perform optimally, the shoulder girdle must work together
in a synchronized and coordinated manner in order to perform overhead activities,
such as hair brushing and reaching to a high shelf [28]. This section discusses the
common biomechanical principles utilized in orthopedic rehabilitation for manage-
ment of shoulder joint stiffness [29, 30], shoulder joint dislocations, and sublux-
ations [31, 32].
∆M = M 1 + M 2 + M 3 (14.1)
M = F × ma (14.2)
372 A. A. Mohamed et al.
rot ward
n
nt ae
Up tion
atio
wn
rot
l
tra
Up zius
mo scapu
wa
a
Do
pe
per
rd
me
r
ato
Rh ome
lev
sm
om nt
bo
s
tu
ra ior
id
r
Se ter
an
Pe m
cto om
ra en
s
trap er
lis t
eziu
Low
m
ino
r
Center of
rotation
Fig. 14.8 The muscular forces included in upward and downward rotations of the scapula. (a)
Upward rotation of the scapula is performed by action of three moments rotate around a fixed axis
of rotation (center of the scapula). These moments include upper trapezius, lower trapezius and the
serratus anterior moments. (b) Downward rotation of the scapula also is performed by action of
three moments rotate around a fixed axis of rotation (center of the scapula). These moments include
levator scapula, rhomboids, and pectoralis minor moments
where M: moment, F: force, and ma: moment arm from the center of scapula (center
of rotation).
According to the previous equations, to increase the upward rotation of the scap-
ula during orthopedic rehabilitation, the rehabilitation team should increase the
force of all three muscles responsible for upward rotation of the scapula. Because
theses muscles act together from different directions to induce rotation, any decrease
in the force production due to weakness or pathology can affect scapular rotation.
Tendons
Fig. 14.9 The Deltoid muscle. It is a multipennate muscle that contain several muscle fiber orien-
tations. PCSA is more accurate than the ACSA in the presentation of all its muscle fibers. The
pennation angle (ϴ) is the angle between the tendon and the fiber orientation. Its force production
equals the summation of all forces generated in it, thus its force production is high
374 A. A. Mohamed et al.
The deltoid muscle is also composed of three types of fibers, including anterior,
middle, and posterior. Thus, deltoid muscle force production is greater than the
unipennate muscles. This increases its force production as well. In the case of a
dislocated or subluxated shoulder joint, therapists should prescribe deltoid strength-
ening to increase the compression force inside the glenohumeral joint. Ultimately,
compression force inside the glenohumeral joint increases and glenohumeral joint
stability improves.
The elbow joint consists of three bones, including humerus, ulna, and radius bones.
The articulations between these bones form the elbow complex. The elbow com-
plex consists of four joints, the humeroulnar (main elbow joint), humeroradial,
superior radioulnar, and inferior radioulnar joints. The orthopedic rehabilitation
protocol for the elbow ligaments injuries should focus on offering the injured liga-
ment to be healed maximally while minimizing the hazards of joint stiffness, resid-
ual instability, and posttraumatic arthritis [33]. This section discusses common
biomechanical principles applied to orthopedic rehabilitation specific to the elbow
joint collateral ligaments, the medial (ulnar) [34, 35] and lateral (radial) [36, 37].
MCL exercise should be performed with the forearm in pronation. Elbow flexion
from a pronated position increases the force production of the biceps muscle and
increases the axial compressive force inside the elbow joint. Biceps brachii muscle
inserts into the radial tuberosity. With forearm pronation, the radial tuberosity
moves away and this increases the length of the biceps muscle. This increases the
compression force between the trochlear notch of the ulna and the trochlea of the
humerus. Also, during elbow flexion exercise, this lengthening in the biceps brachii
before its contraction causes an increase in its force production that increases the
level of the compression force inside the elbow joint during exercises.
Passive mobilization of the deficient MCL should be performed when the fore-
arm is in supination because the forearm supination generates an external rotation
moment on the ulna that closes the medial side of the elbow effectively and this
increases the stability of the elbow joint. The movements of the radius and ulna are
related to each other and it takes place around an axis that passes from the center
of the head of the radius to the base of the styloid process of the ulna. External
rotation of the ulna takes place with forearm supination and internal rotation of the
ulna takes place with forearm pronation. In a forearm supination, the external rota-
tion moment pulls the ulna medially inside the elbow joint; this increases the pull
on the ulna inside the joint and gives a more elbow stability (Fig. 14.10).
14 Biomechanics of Orthopedic Rehabilitation 375
a b
Internal rotation
Internal rotation
moment
moment
Axis of rotation
Pronation
Supination
Fig. 14.10 Types of moments placed on the ulna during elbow supination and pronation. The
movements of the radius and ulna are related to each other and they take place around an axis that
passes from the center of the head of the radius to the base of the styloid process of the ulna.
External rotation of the ulna takes place with forearm supination and internal rotation of the ulna
takes place with forearm pronation. In a forearm supination, the external rotation moment pulls the
ulna medially inside the elbow joint; this increases the pull on the ulna inside the joint and gives a
more elbow stability
The LCL is a key stabilizer which resists both the varus stresses and the rotational
instabilities which prevents elbow joint laxity. Forearm pronation helps to stabilize
the LCL lacking while the arm is passively flexed in a vertical orientation because
the internal rotational torque placed on the wrist to keep the forearm in a pronation
causes the ulna to be hinged over the intact soft tissues on the medial side of the
elbow. This overlapping closes the valgus stress on the lateral side of the elbow.
Thus, patients with acute posterolateral rotatory instability can be actively and pas-
sively mobilized early by keeping the elbow in a full pronation.
Patients with LCL injuries should exercise with the arm positioned in a gravity-
loaded overhead position, because in this position, the gravity and contraction of
the biceps brachii, brachialis, and triceps brachii increase axial compression, con-
gruency, and stability of the elbow joint. When elevating the arm above head, the
gravity and these previous muscles that connect the humerus to the ulna or radius
apply downward forces. These forces help to approximate the joint surface and
increase the compression force inside the elbow joint and consequently improve its
stability.
376 A. A. Mohamed et al.
Wrist and hand are complicated structures that contain high numbers of joints to
allow performance of several fine motor activities such as writing, painting, and
grasping. All of these joints are inter-related and must work together in harmony to
achieve optimal functioning at the hand and wrist. Good understanding of the bio-
mechanics of the hand and wrist joints is crucial to treat these injuries. This section
discusses common biomechanical concepts applied to wrist injuries [38] and in the
carpal tunnel syndrome [39, 40].
F × Fa = R × Ra (14.7)
Thus,
F = ( R × Ra ) / Fa (14.8)
Fa / Ra (14.9)
where F: Force applied by the splint, Fa: length of the forearm component, R:
weight of the hand, and Ra: the length of the palmar component (Fig. 14.11).
According to the previous equations, increase in forearm component length is
recommended to decrease the stress on the healing tissues and to increase the
patient’s comfort level. Increasing the forearm component length decreases the
force required to counteract the weight of the hand to support the splint, and
consequently the resultant stress applied to the healing tissues is less. Reduced
stress on the healing tissues has been associated with increased comfort and
decreased pain.
14 Biomechanics of Orthopedic Rehabilitation 377
RA
FA
Fig. 14.11 The wrist splint. Wrist splint is considered as the first class lever. The forearm piece
should be large to increase the lever arm of the force. This decreases the force needed to overcome
the hand weight of the hand. F splint force, FA force arm, R resistance (hand weight), RA resis-
tance arm
Carpal tunnel syndrome is one of the most common injuries to the wrist joint in
which the median nerve is narrowed across the carpal tunnel between the radial and
ulnar bursa and the synovial membranes of the extrinsic finger flexor tendons.
Extension or flexion of the wrist increases the contact stress and motion friction
between the tendons on the palmar and dorsal surfaces of the carpal tunnel. The
dorsal surface includes the distal head of the radius and the carpal bones. While the
volar surface includes the median nerve and the flexor retinaculum.
In carpal tunnel syndrome, the rehabilitation team should focus on relieving the
stress on the median nerve. The median nerve is more compressed during wrist
flexion than extension because it is located more on the volar side of the carpal tun-
nel. The resultant compression force on the median nerve during flexion can be
calculated by assuming the tendons and nearby structures as a low friction pulley-
belt mechanism (Fig. 14.12). The load distribution for the contacting surfaces, FL, is
related to tendon load, Ft, and the anatomical pulley radius, r:
FL = Ft / r
Thus, the resultant force, FR, on the pulley construction is connected to the ten-
don load and angle of contact between the pulley and the tendon [41].
FR = 2 Ft sin (θ / 2 ) (14.10)
FR
FL
θ
Ft r
Fig. 14.12 Calculations of the forces applied on the median nerve during wrist flexion. If the
tendons and nearby structures are considered as a low friction pulley-belt mechanism, the resultant
force FR is calculated by measuring the load distribution for the contacting surfaces (FL), the
related tendon load (Ft), the anatomical pulley radius (r) and the angle of inclination (θ)
The main function of the lower limbs is the locomotion and body weight support.
Thus, orthopedic rehabilitation should focus on providing more strength to support
the body weight. The lower limb consists of several joints the hip, knee joints, and
ankle and foot joints.
The hip joint has special biomechanical characteristics; it is formed by the articu-
lation between the acetabulum and the femoral head. The acetabulum is deep in
order to apply more stability to the hip joint to enable it to carry the weight of the
body. Osteoarthritis of the hip joint is commonly due to weight-bearing [42].
Total hip arthroplasty can be a treatment to overcome the severe pain and degen-
eration with severe hip osteoarthritis. This section discusses common biomechan-
ical concepts that should be applied during orthopedic rehabilitation after total
hip arthroplasty [43–45].
The main aims of orthopedic rehabilitation after total hip arthroplasty are to min-
imize the pain, return function, and decrease possible deformities in the hip joint.
The hip abductor peak moment reduction is one of the problems occurring with the
total hip arthroplasty that can affect stride length and gait pattern. There are three
elements which can cause a decrease in the hip abduction peak moment at the hip.
14 Biomechanics of Orthopedic Rehabilitation 379
The first one is the muscle strength related to soft tissue damage. The second one is
the surgical approach, as it is supposed that the posterior approach leads to fewer
damage to the hip abductor muscles and consequently improved postoperative gait.
The third one is the vertical distance between the point of action of the abduction
force and the axis of hip rotation.
In total hip arthroplasty, the rehabilitation team must increase the strength of the
hip abductors because during walking it counteracts three times the weight of the
body during normal standing. To maintain the body in equilibrium during any phase
of waking, the muscular moment should counteract the weight of the body moment,
according to the equilibrium equation of moments.
In normal standing,
( M R × M ma ) + ( M L × M ma ) = W × Wma (14.11)
( M × M ma ) = W × Wma (14.12)
where MR: the muscular force of the right hip abductors, ML: the muscular force of
the left hip abductors, Mma: muscle moment arm, W: body weight force, and Wma: is
the weight moment arm. The moment arm is defined as the line between the force
and the center of the affected hip joint.
Both hip abductors contract to counteract the body weight during the double leg
support. In single limb support, the hip abductors of the supporting limb should
increase its force to compensate for the other hip abductor force and movement of
the weight line of action away from the supporting limb. Thus, the hip abductor of
the supporting limb counteracts approximately three times greater body weight than
the body weight that placed on it during normal standing. The other element is the
distance of the vertical distance between the hip abductors and the center of the hip
joint. According to Eq. (14.11), decreased femoral offset decreases the hip abductor
moment arm, and consequently increases the hip abductors force to counteract the
weight of the body. Thus, in orthopedic rehabilitation after total hip arthroplasty, if
the femoral offset is smaller than the normal femoral neck, the rehabilitation team
should increase the strength of the affected hip abductors muscles to counteract the
decrease in the femoral offset (Fig. 14.13).
The knee joint consists of two joints, the tibiofemoral and the patellofemoral joints.
The rehabilitation exercise for the knee joint is mainly conducted in two forms, an
open kinetic chain (OKC) and a closed kinetic chain (CKC) exercises. The OKC
exercise is described as the movement of the knee joint in which the distal segment
(tibia and fibula) is freely moving. Non-weight-bearing exercise is considered a
380 A. A. Mohamed et al.
a b
Hip abductors moment
Fig. 14.13 The difference in femoral offset effect on the hip abductors. A decreased femoral offset
decreases the hip abductor moment arm. This increases the work required from the hip abductor
muscles. (a) Decreased the femoral offset. (b) Normal femoral offset
regular OKC exercises, such as knee extension from seating on a leg extension
machine. The CKC exercise is described as the movement of the knee joint in which
the proximal segment (femur) is moving or the movement in which the distal seg-
ment of the joint encounters significant resistance. The weight-bearing exercise is
considered a typical CKC exercise such as a squat or step-up. This section discusses
the effect of these two exercises on the stability of the tibiofemoral and patellofemo-
ral joints [46].
The tibiofemoral joint is formed by the articulation of the femur and the tibia. This
section discusses the effect of these two types of exercises on the anterior cruciate
ligament (ACL) [46–48] and posterior cruciate ligament (PCL) injuries and recon-
structions [45, 49].
14 Biomechanics of Orthopedic Rehabilitation 381
a b
Fe
mu
r
Tibia
Fig. 14.14 The anterior tibial translation during 45° and 75°. (a) At 45° of knee flexion, there is a
large anterior transition tension which is not recommended during early ACL rehabilitation. (b) At
75° of knee flexion there is a decrease in the anterior transition force because the patellar tendon
exerts approximately a vertical torque (0°) on the tibia in an upward direction
382 A. A. Mohamed et al.
functional activities such as gait, run, stair climb, and jump and cannot be regener-
ated by isolated OKC exercises. Thus, CKC exercises are safely related to anterior
shearing forces placed on the tibia and it can be performed safely in the early phase
of orthopedic rehabilitation following PCL surgery.
The patellofemoral joint is composed of the articulation between the femur (proxi-
mal) and the patella (distal). The stability of this joint is based mainly on the active
and passive restraints within the knee. The main passive restraints include the lateral
patellar and the medial patellofemoral ligaments. It accounts for about 60% of the
whole restrictive force. The lateral retinaculum accounts for 10% and the medial
patellomeniscal ligament accounts for 13% of the resistive components to the lateral
displacement of the patella. While for medial patellar translation, the main passive
resistive components to it are the structures that make the profound and superficial
lateral retinaculum. The deep retinaculum is made by the lateral patellofemoral liga-
ment, the lateral patellotibial ligament, and the deep fibers of the iliotibial band. The
superficial retinaculum is made by fibers from the iliotibial band and vastus lateralis.
In knee flexion training, during CKC flexion training, with the increase in the
flexion, the knee flexion moment arm increases. This requires a high quadriceps and
patellar tendon tension to offset the increase of the flexion moment arm. This leads
to a higher patellofemoral joint reaction force (PFJRF) with knee flexion. During
gait, the PFJRF is equal to the half of the body weight. In ascending and descending
stairs, the PFJRF is about 3–4 times the body weight. In squatting, the PFJRF is
about 7–8 times the body weight. This data can illustrate the increase in patello-
femoral pain with activities that include progressive knee flexion with
weight-bearing.
During OKC extension, there is an increase in the knee flexion moment arm with
a decrease in the patellar extension moment arm. This increases the request for the
quadriceps force to make the knee extension, particularly at full range. This illus-
trates the extensor lag that occurs with quadriceps weakness as in poliomyelitis.
In OKC knee extension, the peak PFJRF at 36° of flexion is 4 times of the weight
of the body. It is decreased to half of the weight of body with full extension. Thus,
the straight leg raise and short arc quadriceps exercises are recommended to per-
form from 20° to 0° during the initial phase of orthopedic rehabilitation. The PFJRF
and patellofemoral contact stress are higher with OKC leg extension exercise in
comparison with the leg press from 0° to 45° knee flexion. While, from 50° to 90°
knee flexion, the PFJRF and contact stress were higher for the leg press in compari-
son with the OKC leg extension exercise.
In knee extension training, with OKC exercises, the forces across the patella
are minimum at 90° knee flexion. With knee extension from 90° of flexion, the
PFJRF rises, and there is a decrease in the patellofemoral contact area. This
causes more contact stress with knee extension until about 20° because the
patella is non-contacting the trochlea. With CKC exercise, there are minimum
384 A. A. Mohamed et al.
a b
Quadriceps moment arm
Femur
Patella
Fig. 14.15 The patellofemoral resultant compression force. The compression force on the patella
increases with the increase in knee flexion (a). Thus, in the painful proximal lesion of the patella,
exercises should be avoided from 60° to 90° of flexion (b). (c) Resultant compression force
forces across the patella at 0° of extension. With knee flexion, PFJRF increases
with an increase in the patellofemoral contact area. This initial increase in the
PFJRF causes a reduction in contact stress, then, the contact stress increases
with flexion progression subordinate to the increase in joint reaction force
(Fig. 14.15).
Both CKC and OKC exercises are performed to treat the patients with patello-
femoral pain if conducted in the pain-free range. CKC exercises for the patello-
femoral joint are better tolerated during 0–45° of knee flexion. During this range,
CKC exercise includes leg presses, step-ups, and mini-squats. In contrast to CKC
exercises, OKC training for the patellofemoral joint is better tolerated in the ranges
during 20–0° and 90–50° of knee flexion. During these ranges, exercises may
include straight leg raises, quadriceps sets, multiple angle isometrics, and short arc
isotonic. The aim of the exercising in these previous ranges is to load the quadriceps
muscles with lower degrees of the stress exerted on the patella.
Ankle and foot articulations are considered as a closed chain system, in which
each bone segments are interdependent and interrelated on each other, and thus
their functions are dependent on each other and cannot move autonomously of the
others. Hypomobility at any joint affects the whole movement of all joints through
a compound sequence of compensations that disturb all other joints.
14 Biomechanics of Orthopedic Rehabilitation 385
a b
Line of gravity
Fig. 14.16 The line of gravity course with normal and supinated foot (posterior view). During
supination, the line of gravity moves medially, this increases the stress applied to the lateral side of
the ankle and causes a lateral ankle sprain. (a) Supinated foot. (b) Normal foot
Lateral collateral ligament sprain is a common ankle injury [50]. It results from
an increase in the supination moment across the subtalar joint, which results from
increasing the magnitude of the upright ground reaction time during initial foot
contact. At this position, if the center of plantar pressure deviates more medially to
the axis of the subtalar joint, a higher moment arm occurs across the subtalar joint
axis. Thus, more supination moment is occurred due to sudden aggressive ankle
supination. When the foot is plantarflexed, the initial touch is performed with the
forefoot. This increases the moment arm between the resultant joint torque and the
subtalar joint axis. Consequently, the sudden aggressive twisting movement results;
this causes ankle sprain injury. Thus, during the rehabilitation, ankle taping or brac-
ing for correcting the ankle joint position at stance phase provides more ankle
mechanical support [51] (Fig. 14.16). Ankle taping or bracing should apply a varus
stress to reduce the stress on the healing tissues and increase the patient comfort.
This part discusses assistive devices that are mainly used for ambulation in patients
with lower limbs orthopedic disorders. Early ambulation is an essential component
to orthopedic rehabilitation because it decreases postoperative complications and
386 A. A. Mohamed et al.
length of stay. Thus, the rehabilitation team should have a complete understanding
of assistive device prescription and the biomechanical advantages associated with
each. The rehabilitation team should prescribe an appropriate assistive device for
each patient in accordance with his/ her abilities and needs and at the correct time
during orthopedic rehabilitation. Assistive devices can also be used to decrease
pain, decrease inflammation, and compensate for muscle weakness in the lower
limbs. The assistive devices include the wheelchairs, walkers, crutches and canes.
4.1 Wheelchairs
The main goal of any orthopedic rehabilitation program is to make patients as inde-
pendent as possible. Wheelchairs are the main way of ambulation for patients who
are unable to ambulate because of both lower limbs amputations. There are several
patterns of wheelchair propulsion that patients can use. This section discusses
guidelines for wheelchair prescription [52] and biomechanical concepts associated
with wheelchair use [53–55].
P= F /a (14.13)
should be supported to avoid overwork of the dorsiflexors which works to keep the
feet in a horizontal position and to prevent planterflexor contractures.
Wheelchair propulsion includes two phases, including the push (stroke, drive) and
the recovery phases. The push phase (the force production phase) occurs when the
hands are in contact with the hand rims. The second phase is the recovery phase (the
non-propulsive phase), in which the hands are repositioned to start the push
phase again.
The wheelchair propulsion pattern is mainly determined via the recovery phase
during the push phase because the upper limbs are moving in a closed kinematic
chain. Movement patterns are mainly two patterns, namely “semicircular” or
“pumping” patterns. The semicircular push pattern occurs when the hand passes
below the push rim through the recovery phase, producing a semicircular move-
ment. The pumping push pattern occurs when the hands only pass slightly below
push rim during the recovery phase. There are two additional propulsion patterns
identified, including single loop over (SL) and double loop over (DL). The SL pat-
tern occurs when the hands rise above push rim through the recovery phase. The DL
pattern occurs when the hands rise above push rim and then cross over and drop
under the push rim through the recovery phase (Fig. 14.17).
Understanding propulsion pattern is important because it can help maximize an
individual’s quality of life through repetitive strain injury minimization and
performances optimization. Importantly, the semicircular propulsion pattern is
a b c d
Pushing Phase
Recovery phase
Fig. 14.17 Wheelchair propulsion patterns. (a) The semicircular push pattern occurs when the
hands pass below the push rim through the recovery phase. (b) The pumping push pattern occurs
when the hands only pass slightly below push rim during the recovery phase. (c) The single loop
over (SL) pattern occurs when the hands rise above push rim through the recovery phase. (d) The
double loop over pattern occurs when the hands rise above push rim and then cross over and drop
under the push rim through the recovery phase
388 A. A. Mohamed et al.
4.2 Walkers
Walkers are the most stable assistive devices used for gait training and usually
orthopedic rehabilitation begins with it. Walkers have several advantages over
crutches and canes, including increasing the patient base of support, anterior sta-
bility, and lateral stability and decreasing the weight-bearing and joint reaction
forces. This section discusses the main guidelines for walker description [56] and
the main biomechanical concepts in weaker-assisted gait [54, 56].
Walkers’ height should be adjusted by the alignment of the top of the frame with the
ulnar styloid process. This technique has been shown to provide between 20° and
30° of elbow flexion. This slight elbow flexion permits the patients for a powerful
downward push on the walker. This downward push permits patients to bear some
of the body weight through the upper limbs. Extended elbow (no flexion angle)
causes a decrease in the downward push force. Excessive elbow flexion angle causes
the patient to excessively bend at trunk anteriorly. Anterior bending of the trunk can
prevent hip extension during the stance phase of gait by moving the center of gravity
anterior to the hip joints (Fig. 14.18).
Strengthening of the elbow extensors should be prescribed by clinicians for all patients
that use walker-assisted gait because maximum elbow extension moment far exceeds
those at the shoulder or wrist joints. Shoulder flexors and adductors should be strength-
ened too. The eccentric moment of the shoulder flexors is needed because this repre-
sents the eccentric demand on the shoulder flexors to control the deceleration and
downward movement of the walker frame. While shoulder adductors counteract the
shoulder abduction moment, this abduction moment increases the distal to proximal
moment arm away from joint centers and increasing the influence of distal joint forces.
Walker-assisted gait should be explained by clinicians to patients because going
too much into the walker during the gait moves the center of gravity backward and
increases the tendency to fall backward. The proper technique for walker use
involves the patients moving the walker slightly forward first followed by move-
ment of the affected leg forward and finally movement of the unaffected leg comes
forward to meet the affected leg.
14 Biomechanics of Orthopedic Rehabilitation 389
4.3 Crutches
Crutches are commonly used in couples. Crutches are commonly used to assist
locomotion by decreasing the load on injured tissue, compensating for the loss of
muscular control or alleviating pain. This section discusses the guidelines for
crutches prescription [54] and the common biomechanical concepts in crutch-
assisted gait [57, 58].
The main types of crutches are the axillary, triceps, platform, and forearm crutches.
The first type is the axillary crutch which is the most common type. In axillary
crutches, the patient must avoid complete weight-bearing on the crutch over the
axilla during walking to avoid excess pressure on the blood vessels and superficial
nerves. This excessive pressure increases incidence of injuries. This excessive
390 A. A. Mohamed et al.
pressure can be decreased by two considerations. Firstly, the upper end of the
crutch must be two to three fingers widths under the patient’s axilla when the
patient is standing with a 30° of elbow flexion. Secondly, putting a sponge rubber
to cover the upper end of the crutch to minimize stress against the user’s chest. The
design of the crutch should include a longer upper end than the lower end to
increase the support area on the chest and reduce energy consumption in the initial
few minutes of walking. The lower end of the crutch should be padded with a rub-
ber to prevent patient slippage during walking. The crutch should be adjusted to
that the lower end of the crutch to be 15 cm from the outside of the patient’s feet to
avoid the ankle trauma caused by the contact of the crutch with the patient legs
during gait (Fig. 14.19).
The other types are less common and each one has a particular purpose. The
second type is the triceps crutches. They are prescribed by clinicians for patients
with paralyzed shoulder muscles. The proximal part has lateral and medial supports
merged with a couple of posterior bands. These supports perform the same action of
the triceps muscle to maintain the elbow in extension. The third type is platform
a b c d
90°
30°
30°
Fig. 14.19 Different types of crutches. All types should be well padded and include a sponge
lower tip to prevent the slippage during gait. (a) Axillary crutch is the common ones, in which
elbow angle is 30°. (b) Triceps crutch is used to help the weak triceps muscles, in which elbow
angle is 0°. (c) Forearm crutch is used when a partial weight bearing only is required, in which
elbow angle is 30°. (d) Platform crutch has a flat support for loading the weight on the forearm, in
which elbow angle is 90°
14 Biomechanics of Orthopedic Rehabilitation 391
crutches; they have a flat support for loading the weight on the forearm. These
crutches are prescribed for patients with the inability to bear weight transmission
over the hand, wrist, or forearm. The fourth type is the forearm crutch, they are
prescribed by clinicians to patients who have mild orthopedic problems and need
only a partial weight-bearing gait.
4.4 Canes
Canes are considered the simplest assistive devices used for ambulation. It is com-
monly used by older people or people with lower limb osteoarthritis. This section
discusses the main guidelines for cane prescription [54, 59, 60] and the biomechani-
cal concepts in cane-assisted gait [61, 62].
Canes are available with several kinds of handles. The basic one is the inverted U
shape which allows patients to suspend the cane on a forearm or back of a chair
while it is not in use. Other handle designs include the pistol grip handles which are
designed to increase the contact area, thus decreasing the pressure on the hand [Eq.
(14.13)]. This design helps to increase comfort and decrease pain. The shaft of the
cane can be folded, height adjusted, depending on the subject height. The cane’s
base should be rubbered with broad deep grooves to prevent its slippage. It should
be kept clean to offer ultimate traction. The base can be single or quadruped. The
quadruped base increases the base of support and gives more stability to the patients.
The cane should be grasped as its handle’s height should permit semi-flexed
elbow about 30°. The cane’s tip should be at a distance of 15 cm forward and
5–10 cm lateral to the foot. Prolonged use of the cane causes a backward bend of the
subject or can interfere with a proper weight that should the cane alleviates. Canes
which are too short cause excessive anterior bending in patients. For canes with a
wide base, the clinicians should ensure that the base edge of the cane is laterally
away from the foot enough to avoid hitting the ankle by the cane base through the
swing phase of gait.
People with lower-limb instability usually use the cane on their ipsilateral side,
while people with muscle weakness or pain use the cane on their unaffected side
(contralateral). The contralateral use of the cane decreases the hip contact forces
more efficiently and encourages the normal reciprocal gait pattern. The decrease
in hip contact forces during cane-assisted gait is mainly argued to the increase in
the moment arm of the cane which decreases the demand on the abductor mus-
cles of the affected side (Fig. 14.20). The hip abductor muscle force is required
14 Biomechanics of Orthopedic Rehabilitation 393
Fa
Fc Fa Fc
B B
C C
Affected side
Affected side
Fw
Fw
Fig. 14.20 The side of holding a cane in a person with an affected hip joint. The cane should be
held in the contralateral side to decrease pain of the affected hip and increase the force of the hip
abductor muscle on the affected side. Using the cane on the contralateral side increases the moment
arm of the cane and decreasing the need of the affected hip abductors. Fc force of the cane, A
moment arm of the cane, Fw force of the body weight, C moment arm of the body weight, Fa force
of affected hip abductor muscle and B moment arm of hip abductor muscle
to balance the pelvis during unilateral stance. When the cane is grasped on the
affected side, the force of the hip abductor muscles can be calculated by the fol-
lowing equation.
Fw × C = ( Fc × A ) + ( Fa × B )) (14.14)
where Fa: hip abductor force, Fc: cane force, Fw: body weight, A: moment arm of
cane, and B: moment arm of the hip abductor and C: moment arm of the body
weight. By increasing (A) there is an increase in the (Fa); this leads to a decrease in
the demand on the hip abductors to produce high force.
It is shown that the hip contact forces can be decreased up to 56% and the gluteus
medius moment can be decreased up to 31.1% with using the cane on the contralat-
eral side in comparison with the normal gait. Thus, clinicians should prescribe the
use of the cane on the unaffected side during cane-assisted gait because the use of
394 A. A. Mohamed et al.
the cane on the uninvolved side significantly decreases demands on the hip a bductors
and consequently decreases compression forces inside the hip joint accompanied
with hip adductors contraction.
References
1. Kassab GS. Biomechanics of the cardiovascular system: the aorta as an illustratory example. J
R Soc Interface. 2006;3(11):719–40.
2. Koontz AM, McCrory JL, Cham R, Yang Y, Wilkinson M. Rehabilitation biomechanics. In:
Wiley encyclopedia of biomedical engineering. 2006.
3. Zhang G. Evaluating the viscoelastic properties of biological tissues in a new way. J
Musculoskelet Neuronal Interact. 2005;5(1):85–90.
4. Saldana RP, Smith DA. Four aspects of creep phenomena in striated muscle. J Muscle Res Cell
Motil. 1991;12(6):517–31.
5. Knudson D. Fundamentals of biomechanics. 2nd ed. New York: Springer; 2007.
6. Manske RC. Fundamental orthopedic management for the physical therapist assistant. 4th ed.
Mosby/Elsevier: St. Louis, MI; 2016.
7. Özkaya N, Nordin M, Goldsheyder D, Leger D. Fundamentals of biomechanics. 3rd ed.
New York: Springer; 2012.
8. Magnusson SP, Simonsen EB, Dyhre-Poulsen P, Aagaard P, Mohr T, Kjaer M. Viscoelastic
stress relaxation during static stretch in human skeletal muscle in the absence of EMG activity.
Scand J Med Sci Sports. 1996;6(6):323–8.
9. Knudson D. Mechanics of the musculoskeletal system. In: Fundamentals of biomechanics.
2nd ed. New York: Springer; 2007. p. 343.
10. Lin DC. Force–velocity relationship of skeletal muscle. In: Encyclopedia of neuroscience.
Berlin: Springer; 2009. p. 1611–5.
11. Knudson D. The biomechanics of stretching. J Exerc Sci Physiother. 2006;2:3–12.
12. Duong B, Low M, Moseley AM, Lee RYW, Herbert RD. Time course of stress relaxation and
recovery in human ankles. Clin Biomech. 2001;16(7):601–7.
13. Minozzo FC, Baroni BM, Correa JA, et al. Force produced after stretch in sarcomeres and
half-sarcomeres isolated from skeletal muscles. Sci Rep. 2013;3:77–86.
14. Decoster LC, Cleland J, Altieri C, Russell P. The effects of hamstring stretching on range of
motion: a systematic literature review. J Orthop Sports Phys Ther. 2005;35(6):377–87.
15. Shrier I. Does stretching improve performance? A systematic and critical review of the litera-
ture. Clin J Sport Med. 2004;14(5):267–73.
16. Page P. Current concepts in muscle stretching for exercise and rehabilitation. Int J Sports Phys
Ther. 2012;7(1):109–19.
17. Haskell WL, Lee I-M, Pate RR, et al. Physical activity and public health: updated recom-
mendation for adults from the American College of Sports Medicine and the American Heart
Association. Circulation. 2007;116(9):1081–93.
18. Kisner C and Colby L. Resistance exercise for impaired muscle performance. In: Theraputic
exercise foundations and techniques. 6th ed. Philadelphia, PA: F.A. Davis Company; 2012.
p. 157–240.
19. Davidovits P. Static forces. In: Physics in biology and medicine. 4th ed. Academic Press; 2012.
p. 8–10.
20. Pasquet B, Carpentier A, Duchateau J, Hainaut K. Muscle fatigue during concentric and eccen-
tric contractions. Muscle Nerve. 2000;23(11):1727–35.
21. Clavet H, Hébert PC, Fergusson D, Doucette S, Trudel G. Joint contracture following pro-
longed stay in the intensive care unit. CMAJ. 2008;178(6):691–7.
22. Macdermid J. Evidence-based orthopaedics: the best answers to clinical questions. 1st ed. In:
Wright JG, editor. Philadelphia, PA: Elsevier Saunders; 2009.
14 Biomechanics of Orthopedic Rehabilitation 395
23. Kisner C, Colby L. Range of motion. In: Theraputic exercise founsations and techniques. 6th
ed. Philadelphia, PA: F.A. Davis Company; 2012. p. 52.
24. Weigel JP, Arnold G, Hicks DA, Millis DL. Biomechanics of rehabilitation. Vet Clin North
Am - Small Anim Pract. 2005;35(6):1255–85.
25. Neumann DA. The convex-concave rules of Arthrokinematics: flawed or perhaps just misinter-
preted? J Orthop Sport Phys Ther. 2012;42(2):53–5.
26. Michael Aloysius MacConaill JB. Muscles and movements: a basis for human kinesiology.
Baltimore: Williams & Wilkins; 1969.
27. Kisner C, Colby L. Peripheral joint mobilization/manipulation. In: Therapeutic exercise foun-
dations and techniques. 6th ed. Philadelphia, PA: F.A. Davis Company; 2012. p. 119–156.
28. Müntener M. Functional anatomy of the shoulder girdle. Helv Chir Acta. 1982;48(5):
523–32.
29. Ludewig PM, Cook TM. Alterations in shoulder kinematics and associated muscle activity in
people with symptoms of shoulder impingement. Phys Ther. 2000;80(3):276–91.
30. Ludewig Paula MJPB. Shoulder impingement: biomechanical considerations in Rehabilitation.
Man Ther. 2011;16(1):33–9.
31. Wilk KE, Macrina LC, Reinold MM. Non-operative rehabilitation for traumatic and atrau-
matic glenohumeral instability. N Am J Sports Phys Ther. 2006;1(1):16–31.
32. Rosso C, Mueller AM, McKenzie B, et al. Bulk effect of the deltoid muscle on the glenohu-
meral joint. J Exp Orthop. 2014;1(1):14.
33. Alolabi B, Gray A, Ferreira LM, Johnson JA, Athwal GS, King GJW. Rehabilitation of the
medial- and lateral collateral ligament-deficient elbow: an in vitro biomechanical study. J
Hand Ther. 2012;25(4):363–73.
34. Morrey BF, AK-N TS. Valgus stability of the elbow: a definition of primary and secondary
constraints. Clin Orthop Relat Res. 1991;26(5):187–195.
35. Armstrong AD, Dunning CE, Faber KJ, Duck TR, Johnson JA, King GJW. Rehabilitation of
the medial collateral ligament-deficient elbow: an in vitro biomechanical study. J Hand Ther.
2000;25(6):1051–7.
36. Patterson SD, Johnson JA, King GJW. Muscle forces and pronation stabilize the lateral liga-
ment deficient elbow. Clin Orthop Relat Res. 2001;388:118–24.
37. Manocha RHK, Kusins JR, Johnson JA, King GJW. Optimizing the rehabilitation of
elbow lateral collateral ligament injuries: a biomechanical study. J Shoulder Elb Surg.
2017;26(4):596–603.
38. Duncan RM. Basic principles of splinting the hand. Phys Ther. 1989;69(12):1104–16.
39. Walker WC, Metzler M, Cifu DX, Swartz Z. Neutral wrist splinting in carpal tunnel syn-
drome: a comparison of night-only versus full-time wear instructions. Arch Phys Med Rehabil.
2000;81(4):424–9.
40. Page MJ, Massy-Westropp N, O’Connor D, Pitt V. Splinting for carpal tunnel syndrome.
Cochrane Database Syst Rev. 2012;7(7):CD010003.
41. Armstrong T. Development of a biomechanical hand model for study of manual activities. In:
Ronald Easterby KHE, Kroemer DBC, editors. Anthropometry and biomechanics: theory and
application. New York: Plenum Press; 1982. p. 188–9.
42. Baker-LePaina JC, Lane NE. Relationship between joint shape and the development of osteo-
arthritis. Curr Opin Rheumatol. 2010;22(5):538–43.
43. Madsen MS, Ritter MA, Morris HH, et al. The effect of total hip arthroplasty surgical approach
on gait. J Orthop Res. 2004;22(1):44–50.
44. Ewen AM, Stewart S, St Clair Gibson A, Kashyap SN, Caplan N. Post-operative gait analysis
in total hip replacement patients-a review of current literature and meta-analysis. Gait Posture.
2012;36(1):1–6.
45. Shiba N, Tagawa Y, Nakashima Y, et al. Biomechanical effect and clinical application of the
hip joint moment reduction brace. Clin Orthop Relat Res. 1998;351:149-157.
46. McGinty G, Irrgang JJ, Pezzullo D. Biomechanical considerations for rehabilitation of the
knee. Clin Biomech (Bristol, Avon). 2000;15(3):160–6.
396 A. A. Mohamed et al.
47. Beynnon BD, Fleming BC, Johnson RJ, Nichols CE, Renstrom PA, Pope MH. Anterior
cruciate ligament strain behavior during rehabilitation exercises in vivo. Am J Sports Med.
1995;23(1):24–34.
48. Fleming BC, Beynnon BD, Renstrom PA, Peura GD, Nichols CE, Johnson RJ. The strain behav-
ior of the anterior cruciate ligament during bicycling. Am J Sports Med. 1998;26(1):109–18.
49. Lutz GE, Palmitier RA, An KN, Chao EY. Comparison of tibiofemoral joint forces during open-
kinetic-chain and closed-kinetic-chain exercises. J Bone Joint Surg Am. 1993;75(5):732–9.
50. Lin C-WC, Hiller CE, de Bie RA. Evidence-based treatment for ankle injuries: a clinical per-
spective. J Man Manip Ther. 2010;18(1):22–8.
51. Fong DT, Chan Y-Y, Mok K-M, Yung PS, Chan K-M. Understanding acute ankle ligamentous
sprain injury in sports. Sports Med Arthrosc Rehabil Ther Technol. 2009;1(1):14.
52. Di Marco A, Russell M, Masters M. Standards for wheelchair prescription. Aust Occup Ther
J. 2003;50(1):30–9.
53. Vanlandewijck Y, Theisen D, Daly D. Wheelchair propulsion biomechanics: implications for
wheelchair sports. Sports Med. 2001;31(5):339–67.
54. Edelstein J. Assistive devices for mobility: canes, crutches, walkers and wheelchairs. In:
Michelle H, Cameron LM, editors. Physical rehabilitation: evidence-based examination, eval-
uation, and intervention. 1st ed: Saunders; 2007. p. 877–96.
55. Koontz AM, Roche BM, Collinger JL, Cooper RA, Boninger ML. Manual wheelchair pro-
pulsion patterns on natural surfaces during start-up propulsion. Arch Phys Med Rehabil.
2009;90(11):1916–23.
56. Bachschmidt RA, Harris GF, Simoneau GG. Walker-assisted gait in rehabilitation: a study of
biomechanics and instrumentation. IEEE Trans Neural Syst Rehabil Eng. 2001;9(1):96–105.
57. Li S, Armstrong CW, Cipriani D. Three-point gait crutch walking: variability in ground reac-
tion force during weight bearing. Arch Phys Med Rehabil. 2001;82(1):86–92.
58. Carpentier C, Font-Llagunes JM, Kövecses J. Dynamics and energetics of impacts in crutch
walking. J Appl Biomech. 2010;26(4):473–83.
59. Dean E, Ross J. Relationships among cane fitting, function, and falls. Phys Ther.
1993;73(8):494–500-4.
60. Kumar R, Roe MC, Scremin OU. Methods for estimating the proper length of a cane. Arch
Phys Med Rehabil. 1995;76(12):1173–5.
61. Ajemian S, Thon D, Clare P, Kaul L, Zernicke RF, Loitz-Ramage B. Cane-assisted gait
biomechanics and electromyography after total hip arthroplasty. Arch Phys Med Rehabil.
2004;85(12):1966–71.
62. Bateni H, Maki BE. Assistive devices for balance and mobility: benefits, demands, and adverse
consequences. Arch Phys Med Rehabil. 2005;86(1):134–45.
Chapter 15
Biomechanics of Osteo-Synthetics
1 Trends in Osteo-Synthetics
2.2 Stress
is based upon the level of vascularity. Earlier, the group also developed an osteo-
genic index and applied the theory on the numerical models to judge the occurrence
of bone, cartilage, and fibrous tissue [10].
Followed by the healing, stress still plays an determinant role in bone remodel-
ing. As noted in the Wolff’s law, the bone structure still undergo an optimization to
fit in the demand of mechanical requirement without redundancy in the intricate
architecture. Guldberg et al. developed an in vivo model of hydraulic bone chamber
in their canine metaphyseal trabecular bone and found convincing evidence of
microstructural adaptation during trabecular bone repair form microcomputed
tomography images [11]. The microstructural adaptation within the bone chamber
was similar to that observed in the spinal cage during bony bridge formation.
Therefore, if the designed internal architecture of osteo-synthetics could provide a
controlled and predictable mechanical stimulation, more reliable construct strength
would be obtained.
Implants can share the load applied to bone, and hence stress on the tissue is
reduced. This phenomenon is known as stress shielding. According to the Wolff’s
law, bone would adapt itself by reducing its mass, either by becoming more porous
(internal remodeling) or by making the structure thinner (external remodeling), to
respond to the decreased load. During spinal fusion, it remains unclear whether the
bone quality of the developing interbody fusion mass will be affected due to the
shield-stress, and if so, what the threshold of the adequate mechanical load would
be required to prevent bone mass loss over the long period. In a more than 8-year
postoperative investigation, Cunningham et al. showed the histological composition
of cervical interbody fusion in thoroughbred horses and they discovered significant
decrease in bone mineral density at the fusion site within the cage compared with
the adjacent vertebral bodies [12]. Similarly, a series of clinical reports showed
resorption in a manner of reduced cortical thickness and increased porosity were
observed in most patients who received noncemented total hip arthroplasty. On the
contrary, van Dijk et al. showed that reduced stiffness of cage contributed to the rate
of lumber interbody fusion [13]. The group used polymeric material poly(l-lactic
acid) (PLLA), which possess an apparently lower elastic modulus (4.2 GPa) com-
pared to titanium (110 GPa), to fabricate a cage with an identical geometry. They
found reduced stiffness significantly enhanced interbody fusion as compared with
titanium cages after 6 months. The results from their large quadrupedal animal mod-
els showed significant improvement in the arthrodesis rate and the quality of tra-
becular bone in the new constructs [13–15]. This result implied that reduced stiffness
of the cage compensates the loss of strain energy to bone tissues.
The relationship between the change of bone density and strain energy was suc-
cessfully quantified in a remodeling algorithm propose by Huiskes et al. [16]. The
model included the idea of a lazy or homeostatic zone where a certain threshold
interval exists. Bone mass increases when the strain energy density is above a cer-
tain level inferring to the modeling from stress fractures. Below a certain threshold,
there is excessive remodeling or absorption of bone; in between these levels is
maintenance of bone structure. Therefore, the reduction of shielded stress would
15 Biomechanics of Osteo-Synthetics 401
ensure bone tissue adjacent to the implant to acquire sufficient energy to maintain
bone mineral density when the rigidity requirement is still placed first in the priority.
2.3 Porosity
It has been shown earlier in series of in vivo studies that implant surface geometry
as a design variable significantly influences long-term implant performance [18–
20]. Early in 1979, Bobyn and colleagues examined the optimal pore size for the
fixation of porous surfaced metal implants by the bone ingrowth [21]. The pore size
was investigated to influence the rate of bone ingrowth and the retained maximum
fixation strength. They concluded that in the shortest period (8 weeks) a pore size
range of approximately 50–400 μm ended up to provide the optimum or maximum
fixation strength of 17 MPa. In an earlier report, the group also showed the surface
configuration played as an additional role affecting the tensile strength of fixation of
implants by bone ingrowth [22]. The results indicated implants with the multiple
particle layer surface configuration develop a greater tensile strength of fixation
than that provided by implants with the single particle layer surface configuration.
Also, they suggested this fixation strength develops more quickly if the cortical
bone is petaled prior to implantation. In a more microscopic investigation, Simmons
et al. reported the differences in osseointegration caused by surface geometry can be
attributed to the alteration of local tissue strains [6]. In their computational model-
ing, local tissue strain was predicted in two different designs of plasma-sprayed and
402 C.-Y. J. Lin et al.
porous-surfaced. The result indicated that porous surface structure provided a larger
secure region that experienced low distortional and volumetric strains, whereas the
plasma-sprayed implant provided little local strain protection to the healing tissue.
Coincident with Pilliar’s study, low distortional and volumetric strains are believed
to favor osteogenesis.
In a more cellular-based study, Carter and Giori suggested that proliferation and
differentiation of the mesenchymal cells responsible for surrounding tissues forma-
tion of implants are regulated by the local mechanical environment [19]. Put
together, the mesenchymal cells tend to be more osteogenic when experiencing low
distortional strain and low compressive hydrostatic stress, provided under an ade-
quate vascularity.
3.2 Porous Material
To regenerate tissues and organs with high vascularity, porous structure has been
considered as a desirable feature of scaffolds because of high void ratio and surface
area. Many biomaterials have been fabricated into porous structures with defined
global and local pore sizes as well as interconnected pore network. In 1972, Hulbert
and the colleagues investigated the tissue reaction with porous ceramics versus non-
porous ones [23]. Tissue around discs of porous ceramics healed faster and pre-
sented a thinner fibrous encapsulation than the impervious implants. Blood vessels
invasion were more rapid in those discs with pore size of 100 to 150 μm, constitut-
ing a richer blood supply.
Currently biodegradable polymers such as poly(lactic-co-glycolic acid) have
been prevalently used in tissue engineering. Peter et al. developed porous, poly-
meric bone flaps with attached vascular pedicle to reconstruct defects and found
blocks of vascularized bone were formed 6 weeks after implantation [7]. Hacking
et al. also showed complete tissue ingrowth throughout a porous tantalum implant.
Tissue ingrowth and sufficient vascularity were noted over time and the attachment
strength was three- to sixfold greater compared to a similar study where the implant
was treated with porous beads [24].
The multiple factors in osteo-synthetics that can be determinant for osseous integra-
tion suggest that new synthetic implants can be optimized to concurrently enhance
stability, biofactor delivery, and mechanical tissue stimulation. Modern structural
optimization can be traced back to 1940s, when the discipline was initially d eveloped
15 Biomechanics of Osteo-Synthetics 403
4.2 Topology Optimization
4.3 Homogenization Theory
5.1 Metals
Metals are perhaps the most common materials used in modern orthopedics. Among
the metals, stainless steel and titanium have been prevalently used for instruments,
particularly when load bearing and mechanical integrity of the new constructs are
required. In spinal applications, anterior interbody cages are often titanium cylin-
ders that are placed in the intervertebral disc space. The cages made of titanium
offer a superior rigidity, and therefore in most single-level fusion cases, additional
instrumentation (e.g., pedicle screws) or postoperative back braces for support are
not needed.
Although biocompatible metals provide desired load bearing, the dense mate-
rial has also created certain obstacles in the real practice . Potential image arti-
facts have been observed in the vicinity of the cage during magnetic resonance
imaging (MRI) and computed tomographic (CT) scanning [31, 32]. Although the
CT images with titanium or titanium, aluminum, and vanadium (Ti-6Al-4V)
alloy present a better result, the amount of MR image artifacts is still comparable
to that with stainless steel [33, 34]. How, lesser field strength and the use of fast
spin–echo techniques have been found to effectively remedy the problem
[35–37].
The second concern with these materials is that solid fusion cannot be easily and
definitely determined from simple radiographic analysis alone. The devices made of
metals are not radiolucent and thus bring the difficulty to determine whether solid
osseous fusion has occurred (osseous trabeculation, evidence of bone formation in
and around the device) on radiographs. Complementary histological examinations
15 Biomechanics of Osteo-Synthetics 405
of the tissue obtained in the hollow spaces from retrieved cages in the studies of
Lange et al. [38] and Carvi et al. [39] confirmed sufficient bone growth in these
areas. However, the reexamination is not feasible in the real practice.
Like the common complication with the use of any metallic device, stress shield-
ing significantly contributes to the relatively high incidence of cage subsidence.
Stress shielding in the segmental fusion achieved by rigid stabilization techniques
with transpedicular screws has been noticed to be associated with disuse osteopenia
in fused vertebra in dog models [40–45]. This osteopenia will likely lead to screw
loosening and instrumentation failures [41]. The mechanically shielded environ-
ment resulted from thick wall or cylindrical thread in conventional cage designs
allows lower intracage pressure propagation [46], which leads to significant bone
mineral density decrease in long term [12]. Consequently, increased incidence of
postoperative complications has been reported such as stress-shielding, the migra-
tion or dislodgement of the cage, pseudarthrodesis, or the combined adverse symp-
toms [14].
5.2 Allograft
5.4 Biodegradable Polymers
[64, 65]. In addition, degradation patterns and half-lives are also different for stereo-
isomers belonging to the same compound [63, 66, 67].
The idea to incorporate bioabsorbable polymers in the surgical implants was first
introduced by Kulkarni et al. [68]. A variety of other applications were then invented,
including sutures, repair of craniofacial defects, appendicular fracture fixation, and
soft-tissue repair [59, 61, 62, 67, 69–73]. A recent attempt also included to fabricate
a PLA interbody fusion cage with the same geometry from the titanium [14, 74, 75].
The use of bioabsorbable materials in skeletal reconstruction has aroused signifi-
cant interests in clinic. The dynamic load transition has been show to lead to higher
bone formation rates, further improving clinical outcomes in specific applications.
Clinical studies are currently underway to evaluate the feasibility to extend their use
in more practice of orthopedic surgery.
6.1 Introduction
The effects of how porous features of a scaffold such as pore size, pore shape, and
interconnectivity affect tissue regenerates have been well studied. These micro-
structural parameters are correlated with mechanical and mass transport properties
of the scaffolds. One delicate way to achieve a viable tissue regenerate is to find
optimal microstructures that achieve prescribed mechanical and mass transport
properties.
To obtain the overarching aim, hierarchical scaffold design appears to be an
adapting optimization scheme to include all these diverse design goals [76]. In the
hierarchical scaffold design scheme, unit microstructures, or unit cells (structural
unit, not biological cells) are selected from unit cell libraries and assembled to form
a scaffold with a global shape that fits into anatomical geometries. The mechanical
and fluidic properties of the scaffold are calculated using the homogenization theory
based on double-scale asymptotic expansion [77, 78]. Furthermore, the pore archi-
tectures can be designed with predefined geometries such as three orthogonal cylin-
drical pores or spherical pores. Hollister et al. optimized pore diameters of scaffolds
with three orthogonal cylindrical pores using the homogenization method and
empirically fitted polynomials that relate pore diameters and the effective stiffness
tensor [79]. The requirements for mass transport were considered by applying a
lower-bound constraint to the porosity.
In more general cases, new microstructures with target properties can be created
using topology optimization [80–82]. Topology optimization distributes material
within a unit microstructure with compose of the final, global architecture that ful-
fills the targeted performances. Lin et al. adapted the topology optimization to find
scaffold microstructures that reach the targeted anisotropic elastic constants [83].
408 C.-Y. J. Lin et al.
Hollister and Lin further extended the method by introducing effective permeability
to the optimization scheme to design scaffolds with the maximized permeability
[84]. However, in that initial attempt, the permeability was not coupled with the
mechanical property in the optimization procedure, so that maximizing permeabil-
ity could affect the mechanically optimized microstructure.
Recently, several multifunctional material design schemes based on the topology
optimization have been reported. Guest and Prevost illustrated a general 3D micro-
structure design scheme using the topology optimization method to achieve maxi-
mized bulk modulus and isotropic permeability [85]. They optimized microstructures
by differentially weighting mechanical and transport terms in the objective, allow-
ing designers to tailor the material properties. de Kruijf et al. found optimal struc-
tures with maximized bulk modulus and thermal conductivity by minimizing both
mechanical and thermal compliance in 2D [86]. The authors explored Pareto opti-
mality by varying weights for mechanical and transport properties. Challis et al.
presented the design of isotropic unit structures with maximized bulk modulus and
isotropic conductivity by a level set method [87]. The authors also explored changes
in design with different combinations of weighting factors.
The design approaches of multifunctional material structures with maximized
properties have gained increasing interests in many engineering fields. Tissue engi-
neering scaffolds, for example, have been tailored to meet a wide range of mechani-
cal and mass transport properties, including cross property relationships that fall
well within the cross-property bounds. For instance, cartilage needs low mass trans-
port and mechanical properties, which lay well within the interior of the mass trans-
port and mechanical cross-property bounds [88].
Thus, in order to design microstructures with ranges of mechanical and mass
transport properties, we adapted a local microstructure topology optimization
scheme based on the SIMP method for target optimization. The target properties
were selected within defined cross-property bounds connecting effective bulk mod-
ulus and isotropic diffusivity. Various microstructures were designed and utilized
within the tissue engineering scaffolds. A porous biodegradable interbody fusion
cage was designed as a biomedical application of multifunctional microstructures
by integrating the layout from the global topology optimization with the local opti-
mized microstructure. The final integrated structures were then built using solid
free-form fabrication techniques.
6.2.1 Material Interpolations
material laws should be defined to relate element densities and local material proper-
ties. In addition, the intermediate density values are penalized to have a final discrete
design. The most common local material law is the Solid Isotropic Microstructure
with Penalization (SIMP) [89]. We utilized the SIMP method for elasticity:
Cijkl = ρ pCijkl
base
, ( p > 1)
where Cijkl is the element stiffness tensor, ρ is the element density, p is a penalization
base
factor, and Cijkl is the stiffness tensor for the base material. For the diffusivity, a
SIMP-like material law can be applied to the interpretation of the intermediate den-
sities with penalization,
Dij = (1 − ρ ) Dijbase ,
p
p >1
where Dij is the element diffusivity tensor, ρ is the element density, p is a penaliza-
tion factor, and Dijbase is the free diffusivity tensor for the fluid phase. With the local
material laws defined for both stiffness and diffusivity, the objective function and
sensitivity derivatives are derived with respect to material density ρ, and the optimi-
zation problem can be solved by updating ρ at each iteration.
For the phase base material, we used unit isotropic diffusivity, D = 1 for the void
phase. For the base material solid phase, we chose Poisson’s ratio equal to 1/3 with
a Young’s modulus of 1, which yields a bulk modulus of 1. In this case, the designed
properties could be easily compared within the cross-property bounds normalized to
base material properties.
In order to tailor the material properties directly, the optimization problem was
defined to minimize the error between the target and the effective bulk moduli and
diffusivities, with constraints on porosity:
2 2
KH DH
minimize f = w1 ∗ − 1 + w2 ∗ − 1 + w3 fcubic
K D
N
1 − ρi
subject to φlb ≤ ∑ ≤ φub ,
i =1 N
0 < ρi ≤ 1
where KH is the homogenized bulk modulus, K∗ is the target bulk modulus, DH is the
homogenized isotropic diffusivity, D∗ is the target isotropic diffusivity, fcubic is the
cubic error function and wi (i = 1, …, 3) are weighting factors, ϕlb and ϕub are the
upper and lower bounds of porosity, ρi is ith element density, and N is the total num-
ber of elements.
410 C.-Y. J. Lin et al.
1 C H + C2222
K H = 1111
H
+ C3333
H
+
H
(
2 C1122 + C2233
H
+ C1133
H
)
3 3 3
In the same way, the effective diffusivities were evaluated as average of the diag-
onal terms in diffusivity tensor.
D11H + D22
H
+ D33H
DH =
3
The cubic error function is defined to minimize the differences among three nor-
mal components, three off-diagonal terms, and three shear terms in the stiffness
tensor components, respectively.
2 2 2
CH C3333
H
C1111
H
fcubic = 2222
H
− 1 +
H − 1 H − 1
+
C1111 C2222 C3333
2 2 2
CH C1133
H
C1122
H
+ 2233
H
− 1 + H
− 1 + H
− 1
C1122 C2233 C1133
2 2 2
CH C1212
H
C2323
H
+ 1313
H
− 1 + H
− 1 + H
− 1
C2323 C1313 C1212
H
where Cijkl are the components of the homogenized stiffness tensor. This multi-
objective formulation can be easily converted to a formulation in which one of the
target properties is optimized while the other is constrained.
6.2.3 Implementation
6.3 Design Results
Our results demonstrate that the properties of the microstructures can be tailored to
meet various scaffold requirements such as stiffness and mass transport using topol-
ogy optimization with SIMP interpolation and sensitivity filtering. Target design
points were chosen close to the cross-property upper bounds. Figure 15.1 illustrates
various microstructural architectures obtained in this study and the achieved proper-
ties are presented in Table 15.1. The mesh resolution for microstructures (a), (c), (e),
(g), and (f) was 60 × 60 × 60, and the mesh resolution for the other microstructures
a b c
d e f
g h
Fig. 15.1 Microstructures obtained by targeting bulk modulus and diffusivity close to the upper
cross-property bounds, for 30% porosity (a, b, and c), 50% porosity (d, e, and f), and 60% porosity
(g, h). (With permission from “Structural and Multidisciplinary Optimization”, Springer Nature)
412 C.-Y. J. Lin et al.
Table 15.1 Properties of microstructures tailored with target bulk moduli and diffusivities (With
permission from “Structural and Multidisciplinary Optimization”, Springer Nature)
Microstructures Porosity Diffusivitya Bulk modulusa Young’s modulusa Poisson’s ratio
A 0.2825 0.1276 0.3734 0.4875 0.2824
B 0.3030 0.1340 0.3565 0.5273 0.2535
C 0.2935 0.1616 0.3317 0.4277 0.2851
D 0.4831 0.3016 0.1512 0.2258 0.2511
E 0.4828 0.3156 0.1624 0.1955 0.2994
F 0.5037 0.3330 0.1522 0.2251 0.2535
G 0.5802 0.3556 0.1246 0.2442 0.1734
H 0.5882 0.4164 0.1114 0.0973 0.3544
The values are normalized to the base material properties
a
0.2
0.1
was 40 × 40 × 40. The designed microstructures were identified within the cross-
property bounds in Fig. 15.2.
Note that the porosities of the designed microstructures satisfied the constraints
despite the lack of an exact match in the corresponding cross-property bounds. This
is because the porosity constraints were set at a small range around the target poros-
ity. For example, the porosity constraints were set less than 52% and greater than
48% for the design of 50% porosity microstructures. Nonetheless, there was excel-
lent agreement between the target and designed bulk moduli and diffusivities
(Fig. 15.3).
Because of the theoretical cross-property bounds for 50~60% porosities, the
maximum normalized diffusivities are 0.4 and 0.5, respectively. Thus, we may con-
sider diffusivity over 0.3 as high diffusivity for 50~60% porosity materials.
15 Biomechanics of Osteo-Synthetics 413
Normalized Diffusivity
0.4
the mirostructures
presented in Fig. 15.1
(a–h, respectively). 0.3
(With permission from
“Structural and 0.2
Multidisciplinary
Optimization”,
0.1
Springer Nature)
0
A B C D E F G H
Microstructures
0.4
0.3
0.2
0.1
0
A B C D E F G H
Microstructures
Target Bulk Mod Achieved Bulk Mod
Microstructures with relatively high diffusivity designed for either 50% or 60%
porosity approached the cross-property upper bound, as depicted in Fig. 15.1d–h.
The properties of the microstructures illustrated in these figures were isotropic. It
should be noted that the designed microstructures have different topologies while
the achieved properties were close to each other. Interestingly, the property pair of
the microstructure in Fig. 15.1f is the closest to the cross-property upper bound,
implying that the structure is optimal. Furthermore those microstructures designed
to have 60% porosity showed lower bulk modulus of approximately 0.1 of that of
the solid phase. When the structures were specified within cross-property bounds,
414 C.-Y. J. Lin et al.
both structures again have near optimal properties because the properties are close
to the upper bounds (Fig. 15.2).
Microstructures targeted a low diffusivity for 30% porosity were also near the cor-
responding cross-property upper bounds (Fig. 15.1a–c). The optimized structures
have thick diagonal in the unit cell domain to obtain a high bulk modulus and small
pore diameter to decrease diffusivity. The normalized diffusivities of these micro-
structures were between 0.12 and 0.17 (Table 15.1). These low diffusivity structures
were also close to the upper cross-property bounds due to high bulk moduli
(Fig. 15.2).
a b
Fig. 15.4 (a) Microstructures with low diffusivity and low bulk modulus, (b) 1/8 of the designed
microstructure, and (c) representative cross-sectional view of the structure. (With permission from
“Structural and Multidisciplinary Optimization”, Springer Nature)
0.1
0.05
0
0 0.05 0.1 0.15 0.2 0.25 0.3 0.35 0.4
Effective Diffusivity, D/D2
Using SFF technique, we fabricated the designed scaffolds with PCL/HA compos-
ites for compression tests. Because the scaffold microstructures were represented
using voxel elements, a three-dimensional version of pixels, the geometry of the
optimized scaffolds was generated by simply repeating the corresponding voxels
using an image-based modeling technique. The dimension of the specimens tested
were 8 mm × 8 mm × 16 mm, with unit microstructures of
2.67 mm × 2.67 mm × 2.67 mm, which represents 3 × 3 × 6 units of microstructures.
The voxel representation of the specimens was then converted to STL files to
accommodate the SFF technique.
A selective laser sintering technique was utilized to fabricate the specimens by
sintering PCL and HA in a powder bed. As shown in Fig. 15.6, the specimens
(a)~(h) corresponds to microstructures (a)~(h) in Fig. 15.1. Compression tests were
conducted with the fabricated specimens and the results were compared to the
designed properties. MTS Alliance RT30 electromechanical test frame (MTS
Systems Corp., MN) was used with a strain rate of 1 mm/min under a preload of
1 lb. TestWorks4 software (MTS Systems Corp., MN) was used to collect load-
displacement responses. Average stress was obtained from the recorded load divided
by the undeformed cross-sectional area of the specimen (~64 mm2). In the similar
manner, average strain was calculated from the displacement divided by the unde-
formed height (~16 mm).
The stress–strain curve is presented in Fig. 15.7. The experimental modulus was
then calculated based on the slope of a line that connects the origin and 1% strain
point. The moduli obtained by the average stress and strain were compared with the
designed Young’s modulus, which was calculated from bulk modulus and Poisson’s
ratio because the optimized microstructures were all cubic symmetric (Table 15.2).
15 Biomechanics of Osteo-Synthetics 417
a b c d
e f g h
Fig. 15.6 Scaffolds with optimal microstructures illustrated in Fig. 15.1 (a–h, respectively) were
designed and fabricated using SFF
6
Stress (MPa)
0
0 0.05 0.1 0.15 0.2
Strain
418 C.-Y. J. Lin et al.
0.4
y = 0.6151x + 0.013
R2 = 0.6845
0.3
0.2
0.1
0.0
0 0.1 0.2 0.3 0.4 0.5 0.6
Theoretical Young’s Modulus
As shown in Fig. 15.8, the theoretical Young’s moduli were correlated to experi-
mental Young’s moduli, although the latter were lower than the former. It should be
noted that small geometric features such as struts and pores in the microstructures
were not accurately fabricated in some cases. For example, disconnected struts were
identified between unit microstructures in Figs. 15.6, 15.7, and 15.8. In the selective
laser sintering process, the laser beam scanning speed and particle size of the pow-
der have been found to affect the energy exposure at a spot, which determines the
minimal curing path size. Geometric features, smaller than the minimum path size,
were skipped based on the preset accuracy of the machine. Considering these manu-
facturing defects, the theoretically designed properties seemed to provide upper
bound for the experimental properties.
15 Biomechanics of Osteo-Synthetics 419
6.4 Discussion
1 N ρi − 0.5
Rconv = ∑
N i =1 0.5
Several studies for microstructure design have shown the composite or porous
structures that are near or on the cross-property upper bounds [85–87]. In these
previous works, two competing properties were maximized simultaneously.
However, one of our main interests in this study was to design microstructures
whose properties are far from the upper bounds.
In particular, our main interest was the design of microstructures with low dif-
fusivity and low bulk modulus. As presented in the result, our design converged to
a minimum. Typically the Rconv index was less than 0.8. If we targeted a design point
far from the upper cross-property bounds, the Rconv index became even smaller. To
evaluate the difficulty of achieving this inner design point, we tested three design
points: (1) K = 0.2 and D = 0.3, (2) K = 0.15 and D = 0.2, and (3) K = 0.1 and
D = 0.15. We used the same problem statement and control parameters for filtering
until convergence at a local minimum was achieved.
The outer point or the point on the upper bounds was easily achieved with an
Rconv index of almost 0.99. For the middle design point, the Rconv index was 0.93,
which means the final design contained a blurry solid-void boundary. However, for
the innermost design point case, the Rconv index was 0.71 and the structure exhibited
a clear gray layer in addition to the black solid structure. This can be noticed in the
histogram as shown in Fig. 15.9 where the number of elements with given densities
were plotted in bins. The case with the inner design point generated a large amount
of elements containing around 0.3–0.4. One possibility was that the presence of the
gray regions represented sub-microstructures were with higher degrees of freedom
in reaching the interior targets than the distinct 0 or 1. This would be more relevant
in the hierarchical structure of biologic tissues, where the feature sizes can range
from the nanometer to centimeter scale.
Another important factor is the practical aspect for manufacturing. Particularly
for the designs with low diffusivities designs, small holes would become the case
that in turn limits the overall diffusivity. Considering the size of unit cells (typically
around 1 mm) in the skeletal tissue scaffolds, the small holes may not be manufac-
turable due to the default resolution of the machine.
2000
1000
0
0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1
Density
15 Biomechanics of Osteo-Synthetics 421
In our study, diffusivity was considered in our design process because the char-
acteristics of diffusion in the scaffold can affect many critical cellular behaviors,
such as migration. The property will also influence mass transportation such as
oxygen and nutrient delivery as well as metabolic waste removal. Mathematical
models of cell migration and tissue regeneration have adapted diffusion in the simu-
lations [101, 102]. In addition, diffusivity and permeability of scaffolds are well
correlated [103]. There are also known cross-property bounds on the effective dif-
fusivity and bulk modulus, suggesting the necessity to include them as design
characteristics.
As a temporary substitute for the extracellular matrix, scaffolds are expected
to provide a favorable milieu to promote new tissue formation. More experi-
mental data are warranted to elucidate what are considered as optimal condi-
tions to augment tissue regeneration as ambiguities have been noticed in
different observations. For example, conflicting findings have been reported
regarding the effect of oxygen diffusion on cartilage regeneration [97]. In this
regard, the method of microstructural topology optimization emerges as a new
platform tool to further this type of studies and help explore the most relevant
scaffold properties.
References
1. Liberman JR, Daluiski A, Einhorn TA. The role of growth factors in the repair of bone.
Biology and clinical application. J Bone Joint Surg. 2002;84-A:1032–44.
2. Sanhu HS. Anterior lumbar interbody fusion with osteoinductive growth factors. Clin Orthop
Relat Res. 2000;371:56–60.
3. Cullinane DM, Fredrick A, Eisenberg SR, Pacicca D, Elman MV, Lee C, et al. Induction of a
neoarthrosis by precisely controlled motion in an experimental mid-femoral defect. J Orthop
Res. 2001;20(2002):579–86.
4. Pilliar RM, Cameron HU, Welsh RP, Binnington AG. Radiographic and morphologic studies of
load-bearing porous-surfaced structured implants. Clin Orthop Relat Res. 1981;156:249–57.
5. Simmons CA, Meguid SA, Pilliar RM. Mechanical regulation of localized and appositional
bone formation around bone-interfacing implants. J Biomed Mater Res. 2001;55:63–71.
6. Simmons CA, Meguid SA, Pilliar RM. Differences in osseointegration rate due to implant sur-
face geometry can be explained by local tissue strains. J Orthop Res. 2001;19(2001):187–94.
7. Peter SJ, Miller MJ, Yasko AW, Yaszemski MJ, Mikos AG. Polymer concepts in tissue engi-
neering. J Biomed Mater Res. 1998;43:442–27.
8. Wolff J. Das gaesetz der transformation der knochen. A. Hirchwild: Berlin; 1982.
9. Carter DR, Blenman PR, Beaupre GS. Correlations between mechanical stress history and
tissue differentiation in initial fracture healing. J Orthop Res. 1988;6:734–48.
10. Carter DR. Mechanical loading history and skeletal biology. J Biomech. 1987;20:
1095–109.
11. Guldberg RE, Caldwell NJ, Guo XE, Goulet RW, Hollister SJ, Goldstein SA. Mechanical
stimulation of tissue repair in the hydraulic bone chamber. J Bone Miner Res.
1997;12(8):1295–302.
12. Cunningham BW, Ng JT, Haggerty CJ. A quantitative densitometric study investigating the
stress-shiedling effects of interbody spinal fusion devices: emphasis on long-term fusions in
thoroughbred racehorses. Trans Orthop Res Soc. 1998;23:250.
422 C.-Y. J. Lin et al.
13. van Dijk M, Smith TH, Suglihara S, Burger EH, Wuisman PI. The effect of cage stiffness on
the rate of lumbar interbody fusion: an in vivo model using poly(L-lactic acid) and titanium
cages. Spine. 2002;27:682–8.
14. van Dijk M, Smit TH, Burger EH, Wuisman PI. Bioabsorbable poly-L-lactic acid cages for
lumbar interbody fusion: three-year follow-up radiographic, histologic, and histomorphomet-
ric analysis in goats. Spine. 2002;27(23):2706–14.
15. van Dijk M, Tunc DC, Smit TH, Higham P, Burger EH, Wuisman PI. In vitro and in vivo deg-
radation of bioabsorbable PLLA spinal fusion cages. J Biomed Mater Res. 2002;63(6):752–9.
16. Huiskes R, Weinan H, Grootenboer HJ, Dalstra M, Fudala B, Slooff TJ. Adaptive bone-
remodeling theory applied to prosthetic-design analysis. J Biomech. 1987;20:1135–50.
17. Smith L. Ceramic plastic material as a bone substitute. Arch Surg. 1963;87:653.
18. Buser D, Schenk RK, Steinemann S, Fiorellini JP, Fox CH, Stich H. Influence of surface
characteristics on bone integration of titanium implants. A histomorphometric study in min-
iature pigs. J Biomed Mater Res. 1991;25:889–902.
19. Carter DR, Giori NJ. Effect of mechanical stress on tissue differentiation in the bony implant
bed. In: Davies JE, editor. The bone-biomaterial interface. Toronto: U. Toronto Press; 1989.
20. Maniatopoulos C, Pilliar RM, Smith DC. Threaded versus porous-surfaced designs for implant
stabilization in bone-endodontic implant model. J Biomed Mater Res. 1986;20:1309–33.
21. Bobyn J, Pilliar R, Cameron H, Weatherly G, Kent G. The effect of porous surface configura-
tion on the tensile strength of fixation of implants by bone ingrowth. Clin Orthop Relat Res.
1980;149:291–8.
22. Bobyn JD, Pilliar RM, Cameron HU, Weatherly GC. The optimum pore size for the fixa-
tion of porous-surfaced metal implants by the ingrowth of bone. Clin Orthop Relat Res.
1980;150:263–70.
23. Hulbert SF, Morrison SJ, Klawitter JJ. Tissue reaction to three ceramics of porous and non-
porous structures. J Biomed Mater Res. 1972;6:347–74.
24. Hacking SA, Bobyn JD, Toh KK, Tanzer M, Krygier JJ. Fibrous tissue ingrowth and attach-
ment to porous tantalum. J Biomed Mater Res. 2000;52:631–8.
25. Bensoe MP, Kikuchi N. Generating optimal yopologies in structural design using a homog-
enization method. Comput Methods Appl Mech Eng. 1988;71:197.
26. Neves M, Rodrigues H, Guedes J. Optimal design of periodic linear elastic microstructures.
Comput Struct. 2000;76:421.
27. Kikuchi N. Design optimization method for compliant mechanisms and material microstruc-
ture. Comput Math App Mech Eng. 1998;151:401.
28. Cioranescu D, Donato P. An introduction to homogenization. Oxford: Oxford University
Press; 1999.
29. Scachez-Palencia E, Zaoui A. In: Araki H, Kyoto J, et al., editors. Homogenization tech-
niques for composite media. New York: Springer; 1987.
30. Guedes JM, Kikuchi N. Preprocess and postprocess for material based on the homogeni-
zation method with adaptive finite element methods. Comput Methods Appl Mech Eng.
1989;83:143.
31. Laakman RW, Kaufman B. Han JS. MRI imaging in patients with metallic implants.
Radiology. 1985;157:711–4.
32. Mechlin M, Thickman D, Kressel HY, Gefter W, Joseph P. Magmetic resonance imaging of
postoperative patients with metallic implants. Am J Radiol. 1984;143(6):1281–4.
33. Levi AD, Choi WG, Keller PJ, Heiserman JE, Sonntag VK, Dickman CA. The radiographic
and imaging characteristics of porous tantalum implants within the human cervical spine.
Spine. 1998;23(11):1245–50; discussion 51
34. Wang JC, Yu WD, Sandhu HS, Tam V, Delamarter RB. A comparison of magnetic resonance
and computed tomographic image quality after the implantation of tantalum and titanium
spinal instrumentation. Spine. 1998;23(15):1684–8.
35. Farahani K, Sinha U, Sinha S. Effect of field strength onsusceptibility artifacts in magnetic
resonance imaging. Comput Med Imaging Graph. 1990;14:409–13.
15 Biomechanics of Osteo-Synthetics 423
36. Ortiz O, Pait TG, McAllister P. Postoperative magnetic resonance imaging with titanium
implants of the thoracic and lumbar spine. Neurosurgery. 1996;38:741–5.
37. Tartaglino LM, Flanders AE, Vinitski S. Metallic artifactson MR images of the postoperative
spine: reduction with fastspin-echo techniques. Radiology. 1994;190:565–9.
38. Lange M, Philipp A, Fink U. Anterior cervical spine fusion using RABEA-Titan-Cages avoid-
ing iliac crest spongiosa: first experiences and results. Neurol Neurochir Pol. 2000;34(suppl
6):64–9.
39. Carvi y Nievas MN, Pollath A, Haas E. Cervical discectomy: bone graft or cage fusion? In:
Brock M, Schwarz W, Wille C, editors. First interdisciplinary world congress on spinal sur-
gery and related disciplines. Monduzzi Editore: Bologna; 2000. p. 123–8.
40. Craven TG, Carson WL, Asher MA, Robinson RG. The effects of implant stiffness on
the bypassed bone mineral density and facet fusion stiffness of the canine spine. Spine.
1994;19(15):1664–73.
41. Dalenberg DD, Asher MA, Robinson RG, Jayaraman G. The effect of a stiff spinal implant
and its loosening on bone mineral content in canines. Spine. 1993;18(13):1862–6.
42. Farey ID, McAfee PC, Gurr KR, Randolph MA. Quantitative histologic study of the influence
of spinal instrumentation on lumbar fusions: a canine model. J Orthop Res. 1989;7(5):709–22.
43. Kandziora F, Kerschbaumer F, Starker M, Mittlmeier T. Biomechanical assessment of tran-
soral plate fixation for atlantoaxial instability. Spine. 2000;25(12):1555–61.
44. McAfee PC, Farey ID, Sutterlin CE, Gurr KR, Warden KE, Cunningham BW. The effect
of spinal implant rigidity on vertebral bone density. A canine model. Spine. 1991;16(6
Suppl):S190–7.
45. Shirado O, Zdeblick TA, McAfee PC, Cunningham BW, DeGroot H, Warden KE. Quantitative
histologic study of the influence of anterior spinal instrumentation and biodegradable polymer
on lumbar interbody fusion after corpectomy. A canine model. Spine. 1992;17(7):795–803.
46. Kanayama M, Cunningham BW, Haggerty CJ, Abumi K, Kaneda K, McAfee PC. In vitro
biomechanical investigation of the stability and stress-shielding effect of lumbar interbody
fusion devices. J Neurosurg. 2000;93:259–65.
47. Hodgson AR, Stock FE. Anterior spinal fusion. Br J Surg. 1956;44:226–75.
48. Hodgson AR, Stock FE. Anterior spine fusion for the treatment of tuberculosis of the spine.
J Bone Joint Surg. 1960;42:295–310.
49. Harmon PH. Anterior excision and vertebral body fusion operation for intervertebral disc
syndromes of the lower lumbar spine. Clin Orthop Relat Res. 1963;26:107–27.
50. Sacks S. Anterior interbody fusion of the lumbar spine. J Bone Joint Surg. 1965;47B:211–23.
51. O’Brien JP, Dawson MH, Heard CW. Simultaneous combined anterior and posterior fusion.
Clin Orthop Relat Res. 1986;203:191–5.
52. Lowery G, Kulkarni S, Pennisi AE. Use of autologous growth factors in lumbar spinal fusion.
Bone. 1999;25:47S–50S.
53. Brantigan JW, Steffee AD, Lewis ML, Quinn LM, Persenaire JM. Lumbar interbody
fusion using the Brantigan I/F cage for PLIF and the VSP pedicle screw system: two year
results of a Food and Drug Administration IDE clinical trial. In: Intersomatique du Rachis
Lumbaire. 1996.
54. Hashimoto T, Shigenobu K, Kanayama M, Harada M, Oha F, Ohkoshi Y, et al. Clinical
results of single-level posterior lumbar interbody fusion using the Brantigan I/F carbon
cage filled with a mixture of local morselized bone and bioactive ceramic granules. Spine.
2002;27:258–62.
55. Molinari RW, Gerlinger T. Functional outcomes of instrumented posterior lumbar interbody
fusion in active-duty US servicemen: a comparison with nonoperative management. Spine
J. 2001;1:215–24.
56. Ciccone WJ, Motz C, Bentley C, Tasto JP. Bioabsorbable implants in orthopaedics: new
developments and clinical applications. J Am Acad Orthop Surg. 2001;9:280–8.
57. Bergsma JE, de Bruijn WC, Rozema FR, Bos RRM, Boering G. Late degradation tissue
response to poly (l-lactide) bone plates and screws. Biomaterials. 1995;16:25–31.
424 C.-Y. J. Lin et al.
58. Bergsma JE, Rozema FR, Bos RRM, de Bruijn WC. Foreign body reactions to resorbable
poly (l-lactide) bone plates and screws used for the fixation of unstable zygomatic fractures.
J Oral Maxillofac Surg. 1993;51:666–70.
59. Bostman O, Hirvensalo E, Makinen J, Rokkanen P. Foreign-body reactions to fracture fixa-
tion implants of biodegradable synthetic polymers. J Bone Joint Surg. 1990;72:592–6.
60. Tegnander A, Engebretsen L, Bergh K, Eide E, Holen KJ, Iversen OJ. Activation of the
complement system and adverse effects of biodegradable pins of polylactic acid (Biofix) in
osteochondritis dissecans. Acta Orthop Scand. 1994;65:472–5.
61. Christel P, Chabot F, Leray JL, Morin C, Vert M. Biodegradable composites for internal fixa-
tion. Biomaterials. New York: Wiley; 1980. p. 271–80.
62. Cutright DE, Hunsuck EE. The repair of fractures of the orbital floor using biodegradable
polylactic acid. Oral Surg. 1972;33:28–34.
63. Miller RA, Brady JM, Cutright DE. Degradation rates of oral resorbable implants:
rate modification with changes in PLA/PGA copolymer ratios. J Biomed Mater Res.
1977;11:711–9.
64. Cutright DE, Hunsuck EE. Tissue reaction to the biodegradable polylactic acid suture. Oral
Surg Oral Med Oral Pathol. 1971;31:134.
65. Van der Elst M, Klein CP, Blieck-Hogervorst JM, Patka P, Haarman HJ. Bone tissue response
to biodegradable polymers used for intramedullary fracture fixation: a long-term in vivo
study in sheep femora. Biomaterials. 1999;20:121.
66. Hollinger JO, Battistone GC. Biodegradable bone repair materials. Synthetic polymers and
ceramics. Clin Orthop Relat Res. 1986;207:290.
67. Vert M, Christel P, Chabot F, Leray J. Bioresorbable plastic materials for bone surgery. In:
Hastings GW, Ducheyne P, editors. Macromolecular biomaterials. Boca Raton: CRC Press;
1984. p. 119–42.
68. Kulkarni RK, Pani KC, Neuman C, Leonard F. Polylactic acid for surgical implants. Arch
Surg. 1966;93:839.
69. Bucholz RW, Henry S, Henley MB. Fixation with bioabsorbable screws for the treatment of
fractures of the ankle. J Bone Joint Surg. 1994;76:319–24.
70. Caborn DNM, Coen M, Neef R, Hamilton D, Nyland J, Johnson DL. Quadrupled
semitendinous-gracilis autograft fixation in the femoral tunnel: a comparison between a
metal and a bioabsorbable interference screw. Arthroscopy. 1998;14:241–5.
71. Cohen B, Tasto J. Meniscal arrow. Tech Orthop. 1998;13:164–9.
72. Cordewener FW, Bos RR, Rozema FR, Houtman WA. Poly(L-lactide) implants for repair of
human orbital floor defects: clinical and magnetic resonance imaging evaluation of long-term
results. J Oral Maxillofac Surg. 1996;54:9–13.
73. Warme WJ, Arciero RA, Savoie FHI, Uhorchak JM, Walton M. Nonabsorbable versus
absorbable suture anchors for open Bankart repair: a prospective, randomized comparison.
Am J Sports Med. 1999;27:742–6.
74. Kandziora F, Pflugmacher R, Kleemann R, Duda G, Wise DL, Trantolo DJ, et al. Biomechanical
analysis of biodegradable interbody fusion cages augmented with poly(propylene glycol-co-
fumaric acid). Spine. 2002;27(15):1644–51.
75. Toth JM, Estes BT, Wang M, Seim HB 3rd, Scifert JL, Turner AS, et al. Evaluation of 70/30
poly (L-lactide-co-D,L-lactide) for use as a resorbable interbody fusion cage. J Neurosurg.
2002;97(4 Suppl):423–32.
76. Hollister SJ. Porous scaffold design for tissue engineering. Nat Mater. 2005;4(7):518–24.
77. Bensoussan A, Lions JL, Papanicolaou G. Asymptotic analysis for periodic structures.
Amsterdam: North-Holland Pub. Co sole distributors for the U.S.A. and Canada, Elsevier
North-Holland; 1978.. xxiv, 700 p
78. Sanchez-Palencia E, Zaoui A, CISM International Center for Mechanical Sciences.
In: Homogenization techniques for composite media: lectures delivered at the CISM
International Center for Mechanical Sciences, Udine, Italy, July 1–5, 1985. Berlin: Springer;
1987. ix, 397 p.
15 Biomechanics of Osteo-Synthetics 425
79. Hollister SJ, Maddox RD, Taboas JM. Optimal design and fabrication of scaffolds to mimic
tissue properties and satisfy biological constraints. Biomaterials. 2002;23(20):4095–103.
80. Bendsoe MP, Kikuchi N. Generating optimal topologies in structural design using a homog-
enization method. Comput Methods Appl Mech. 1988;71(2):197–224.
81. Sigmund O. Tailoring materials for specific needs. J Intel Mater Syst Struct. 1994;5(6):736–42.
82. Sigmund O. Tailoring materials with prescribed elastic properties. Mech Mater.
1995;20(4):351–68.
83. Lin CY, Kikuchi N, Hollister SJ. A novel method for biomaterial scaffold internal architecture
design to match bone elastic properties with desired porosity. J Biomech. 2004;37(5):623–36.
84. Hollister SJ, Lin CY. Computational design of tissue engineering scaffolds. Comput Methods
Appl Mech. 2007;196(31–32):2991–8.
85. Guest JK, Prevost JH. Optimizing multifunctional materials: design of microstructures for
maximized stiffness and fluid permeability. Int J Solids Struct. 2006;43(22–23):7028–47.
86. de Kruijf N, Zhou SW, Li Q, Mai YW. Topological design of structures and composite materi-
als with multiobjectives. Int J Solids Struct. 2007;44(22–23):7092–109.
87. Challis VJ, Roberts AP, Wilkins AH. Design of three dimensional isotropic microstructures
for maximized stiffness and conductivity. Int J Solids Struct. 2008;45(14–15):4130–46.
88. Kemppainen JM, Hollister SJ. Differential effects of designed scaffold permeability on chon-
drogenesis by chondrocytes and bone marrow stromal cells. Biomaterials. 2010;31(2):279–87.
89. Bendsoe MP, Sigmund O. Material interpolation schemes in topology optimization. Arch
Appl Mech. 1999;69(9–10):635–54.
90. Sigmund O, Petersson J. Numerical instabilities in topology optimization: a survey on pro-
cedures dealing with checkerboards, mesh-dependencies and local minima. Struct Optimiz.
1998;16(1):68–75.
91. Sigmund O. Design of material structures using topology optimization. Technical University
of Denmark; 1994.
92. Svanberg K. The method of moving asymptotes—a new method for structural optimization.
Int J Numer Methods Eng. 1987;24(2):359–73.
93. Hollister SJ, Liao EE, Moffitt EN, Jeong CG, Kemppainen JM. Defining design targets for
tissue engineering scaffolds. Berlin: Springer; 2009. p. 521–37.
94. Thomson RC, Yaszemski MJ, Powers JM, Mikos AG. Fabrication of biodegradable polymer
scaffolds to engineer trabecular bone. J Biomater Sci Polym Ed. 1995;7(1):23–38.
95. Hutmacher DW. Scaffold design and fabrication technologies for engineering tissues—state
of the art and future perspectives. J Biomater Sci Polym Ed. 2001;12(1):107–24.
96. Simmons CA, Meguid SA, Pilliar RM. Differences in osseointegration rate due to implant sur-
face geometry can be explained by local tissue strains. J Orthopaed Res. 2001;19(2):187–94.
97. Malda J, Woodfield TBF, van der Vloodt F, Kooy FK, Martens DE, Tramper J, et al. The
effect of PEGT/PBT scaffold architecture on oxygen gradients in tissue engineered cartilagi-
nous constructs. Biomaterials. 2004;25(26):5773–80.
98. Malda J, Martens DE, Tramper J, van Blitterswijk CA, Riesle J. Cartilage tissue engineering:
controversy in the effect of oxygen. Crit Rev Biotechnol. 2003;23(3):175–94.
99. Guest JK, Prevost JH, Belytschko T. Achieving minimum length scale in topology optimi-
zation using nodal design variables and projection functions. Int J Numer Methods Eng.
2004;61(2):238–54.
100. Wang MY, Zhou S, Ding H. Nonlinear diffusions in topology optimization. Struct Multidiscip
Optim. 2004;28(4):262–76.
101. Adachi T, Osako Y, Tanaka M, Hojo M, Hollister SJ. Framework for optimal design of porous
scaffold microstructure by computational simulation of bone regeneration. Biomaterials.
2006;27(21):3964–72.
102. Anderson ARA, Chaplain MAJ. A mathematical model for capillary network formation in
the absence of endothelial cell proliferation. Appl Math Lett. 1998;11(3):109–14.
103. Hollister SJ, Lin C-Y, Kang H,AdachiT. Computational design and simulation of tissue engineering
scaffolds; 2008. p. 113–27. https://link.springer.com/chapter/10.1007/978-0-387-68831-2_6.