Adipose Tissue Metabolism and Cardiometabolic Health in Obesity
Adipose Tissue Metabolism and Cardiometabolic Health in Obesity
health in obesity
Citation for published version (APA):
Stinkens, R. M. E. (2017). Adipose tissue metabolism and cardiometabolic health in obesity: effects of
pharmacological and lifestyle interventions. [Doctoral Thesis, Maastricht University]. Maastricht University.
https://doi.org/10.26481/dis.20171005rs
DOI:
10.26481/dis.20171005rs
Document Version:
Publisher's PDF, also known as Version of record
• A submitted manuscript is the version of the article upon submission and before peer-review. There can
be important differences between the submitted version and the official published version of record.
People interested in the research are advised to contact the author for the final version of the publication,
or visit the DOI to the publisher's website.
• The final author version and the galley proof are versions of the publication after peer review.
• The final published version features the final layout of the paper including the volume, issue and page
numbers.
Link to publication
General rights
Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright
owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these
rights.
• Users may download and print one copy of any publication from the public portal for the purpose of private study or research.
• You may not further distribute the material or use it for any profit-making activity or commercial gain
• You may freely distribute the URL identifying the publication in the public portal.
If the publication is distributed under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license above,
please follow below link for the End User Agreement:
www.umlib.nl/taverne-license
ISBN: 978-94-6233-699-5
For articles published or accepted for publication, the copyright has been
transferred to the respective publisher. No parts of this thesis may be reproduced,
stored in a retrieval system, or transmitted in any form or by any means without the
permission of the author, or, when appropriate, from the publishers of the
manuscript.
   Adipose tissue metabolism and
  cardiometabolic health in obesity
    Effects of pharmacological and lifestyle interventions
PROEFSCHRIFT
 ter verkrijging van de graad van doctor aan de Universiteit Maastricht, op gezag
van de Rector Magnificus, Prof. dr. Rianne M. Letschert volgens het besluit van het
               College van Decanen, in het openbaar te verdedigen
                  op Donderdag 5 Oktober 2017 om 12.00 uur.
door
CO-PROMOTOR
Dr. G.H. Goossens
BEOORDELINGSCOMMISSIE
Prof. dr. C.D.A. Stehouwer   (Chair)
Prof. dr. J.F.C. Glatz
Prof. dr. A.H. Kersten       (Wageningen University & Research)
Prof. dr. L.J.C. van Loon
Prof. dr. B.M. Stallknecht   (University of Copenhagen)
Voor papa en mama
TABLE OF CONTENTS
10    Chapter 1
           General introduction
38    Chapter 2
           Targeting fatty acid metabolism to improve glucose
           metabolism
116   Chapter 3
           Improved insulin sensitivity with angiotensin receptor
           neprilysin inhibition in individuals with obesity and
           hypertension
142   Chapter 4
           Effect of sacubitril/valsartan on exercise induced lipid
           metabolism in individuals with obesity and hypertension
162   Chapter 5
           The effects of angiotensin receptor neprilysin inhibition
           by sacubitril/valsartan on adipose tissue transcriptome
           and protein expression in obese hypertensive patients
180   Chapter 6
           Exercise training-induced effects on the abdominal
           subcutaneous adipose tissue phenotype in obese
           humans
202   Chapter 7
           Coordinated regulation of adipose tissue adrenergic-
           and non-adrenergic-mediated lipolysis during exercise
           in lean and obese individuals: the effect of exercise
           training
234   Chapter 8
           General discussion
254   Chapter 9
           Addenda
CHAPTER 1
GENERAL INTRODUCTION
Chapter 1
10
                                                                          Chapter 1
                                                                                  11
Chapter 1
12
                                                                         Chapter 1
                                                                                 13
Chapter 1
14
                                                                          Chapter 1
                                                                                  15
Chapter 1
are associated with a lower prevalence of new-onset type 2 diabetes [99, 100].
Beside changes in circulating NP concentrations, changes in the NP-receptors
have also been observed. A decreased NPRA receptor expression at the level of
abdominal subcutaneous adipose tissue, together with an increased NPRC
receptor expression at both abdominal subcutaneous and omental adipose tissue
has been observed in obese subjects with or without type 2 diabetes as compared
to lean individuals, thereby altering the NPRA/NPRC ratio [80, 101-104]. This
altered receptor expression, in combination with an increased NEP expression in
adipose tissue of obese and insulin resistant subjects [94], may cause an
increased NP clearance, leading to reduced NP bioavailability, thereby contributing
to the development of cardiometabolic disturbances.
16
                                                                        Chapter 1
chronic heart failure patients [121] and healthy men [122], and to reduce systemic
leptin concentrations in healthy men [123].
                                                                               17
Chapter 1
LIFESTYLE INTERVENTION
The management and treatment of obesity and obesity-related disorders is more
complex than only achieving weight loss. The rational should not only focus on the
maintenance of fat mass loss, but also on prolonged cardiometabolic risk reduction
and health improvement. Lifestyle characteristics such as diet, physical inactivity,
smoking, alcohol consumption and stress are important factors that influence the
development of obesity and related comorbidities, and guidelines recommend
changes in these lifestyle characteristics for both prevention and management of
metabolic disease [135].
Manipulation of diet and physical activity levels are the first-choice interventions to
reverse metabolic disturbances. Even though several large lifestyle intervention
trials did not show significant beneficial effects of intensive exercise or combined
diet and exercise lifestyle interventions on reducing the risk for cardiovascular
outcomes [136, 137], other trials did observe beneficial effects on modifiable risk
factors for cardiovascular disease [138], type 2 diabetes incidence and the
metabolic syndrome [139]. This indicates that lifestyle interventions do have
beneficial effects. Several important large lifestyle intervention trials that used
dietary advice, exercise strategies and/or combinations have been performed to
prevent type 2 diabetes. In this respect, the Malmö-study [140, 141], the Chinese
18
                                                                             Chapter 1
Da Qing IGT and Diabetes study [142], the Finnish Diabetes Prevention Study
(DPS) [143], the Diabetes Prevention Program in the USA (DPP) [144] and the
Study on Lifestyle intervention in Impaired glucose tolerance Maastricht (SLIM)
[145] indicated that the incidence of type 2 diabetes was reduced by 40-60%
following intensive (combined) diet and exercise intervention program. Importantly,
long-term follow-up of these trials revealed that diabetes risk reduction still existed
after 3 to 14 years after cessation of the intervention program [146-149]. Thus,
lifestyle interventions clearly show improvements in a variety of health outcomes
related to cardiometabolic health and can reduce the incidence of diseases [150].
However, the exact effects of the different components of the lifestyle program are
not fully elucidated. While one study revealed that the combination of diet and
exercise was more beneficial in reducing insulin resistance [151], another study
showed that diet and exercise were equally effective compared to exercise alone to
prevent the progression towards type 2 diabetes [142]. Moreover, another study
found that the combination of diet and exercise was more effective than either
treatment alone [145]. Nevertheless, combined dietary and exercise interventions
have been shown to be most effective in reducing body weight [152].
Clearly, both prevention and management of disease progression can be achieved
by strategies such as increased physical activity as well as dietary manipulation via
a hypocaloric diet or an improved nutritional composition [153]. Interestingly,
several dietary components, such as fatty acids, polyphenols and fibers may
modulate fatty acid metabolism in tissues like skeletal muscle, liver and pancreas,
both acute (i.e. postprandial phase) and more long-term, ultimately improving
glucose metabolism, as extensively reviewed in chapter 2 [34]. Physical activity
strategies include increased habitual physical activity (e.g. sitting less) and physical
exercise levels (e.g. cycling) which lead to increased energy expenditure, improved
physical fitness [154, 155] and contribute to an improved metabolic health. The
exercise-induced improvements in metabolic risk profile have largely been
attributed to changes in skeletal muscle metabolism and function, but physical
exercise is likely to induce alterations in almost all metabolically active tissues,
including the adipose tissue, as will be discussed below.
                                                                                     19
Chapter 1
20
                                                                               Chapter 1
have shown that exercise training may beneficially alter adipokine expression [195,
196], but human data are conflicting. Although there is consistent evidence that
exercise increases adipose tissue interleukin-6 (IL-6) expression and secretion
after a single bout of exercise [197-199], results of other adipokines, such as
adiponectin, leptin, TNF-α after a single bout of exercise or after exercise training
are currently limited and conflicting [181, 200-206].
Collectively, these data may suggest that exercise improves adipose tissue
metabolism and function and can ultimately contribute to a reduced disease
progression and improved peripheral insulin sensitivity. Indeed, transplantation of
white adipose tissue from trained animals to untrained recipients markedly
improved skeletal muscle glucose uptake [187], suggesting that improvement of
adipose tissue function may contribute to the increased peripheral insulin sensitivity
after exercise training.
In chapters 6 and 7 of this thesis, the exercise training-induced effects on
abdominal subcutaneous adipose tissue metabolism and lipolysis were
investigated in obese and lean subjects.
THESIS OUTLINE
This thesis describes the effects of a pharmacological intervention as well as
physical exercise interventions to improve metabolic health in obese individuals,
with a focus on adipose tissue metabolism.
As described earlier in this thesis, adipose tissue dysfunction contributes to the
development of insulin resistance and impaired glucose metabolism. By targeting
fatty acid metabolism in the adipose tissue, liver, skeletal muscle or even the
pancreas and the intestine, insulin sensitivity and glucose homeostasis may be
improved. In Chapter 2, an extensive overview is provided of the fatty acid
metabolism-related pathways in several metabolically active organs that can be
targeted by dietary interventions, thereby improving whole-body glucose
metabolism and insulin sensitivity.
Targeting the renin angiotensin system and the natriuretic peptide system by
cardiovascular medication may improve adipose tissue and metabolic dysfunction,
next to its effect on the cardiovascular system and hypertension. To investigate
this, we conducted in Chapter 3, a multicenter, randomized, double-blind, double-
dummy, parallel-group study to examine the metabolic effects of
sacubitril/valsartan, which is a first-in-class angiotensin receptor neprilysin inhibitor,
in obese hypertensive patients. By means of a hyperinsulinemic-euglycemic
glucose clamp, we investigated the effects on peripheral insulin sensitivity, while
                                                                                     2
whole-body lipolysis was determined using a stable isotope tracer ([1,1,2,3,3- H]-
glycerol) and abdominal subcutaneous adipose tissue lipolysis was measured with
the microdialysis technique. In addition to the collection of adipose tissue biopsies,
we also performed indirect calorimetry measurements to assess energy
expenditure and substrate utilization in these patients.
Chapter 4 extends the outcomes from the previous chapter and describes the
effects of the 8-weeks treatment with sacubitril/valsartan compared to amlodipine
on whole-body and adipose tissue lipolysis and lipid oxidation during a single bout
of exercise.
                                                                                       21
Chapter 1
To obtain more detailed insight into possible mechanisms underlying the findings
described in chapters 3 and 4, we assessed adipose tissue gene expression
patterns using microarray analysis and protein expression profiles of enzymes
involved in lipolysis, the natriuretic peptide signaling pathway and mitochondrial
oxidative phosphorylation complexes. The results from these analyses are
provided in Chapter 5.
There is some evidence that exercise training may improve adipose tissue function,
which may contribute to the reduced risk for developing obesity-related insulin
resistance and other comorbidities. However, human studies that investigated the
effects of exercise training on the adipose tissue function are limited. Chapter 6
addresses the results of a supervised, progressive, combined endurance and
resistance exercise training intervention for 12 weeks in well-phenotyped, obese
subjects. In this study, we investigated exercise training-induced effects on adipose
tissue by measuring abdominal subcutaneous adipocyte morphology, gene and
protein expression of markers related to adipose tissue function. Moreover, we
determined the exercise training-induced effects on ex vivo adipocyte lipolysis.
The aim of the study, described in Chapter 7, was to elucidate the physiological
role of ANP-mediated lipolysis in abdominal subcutaneous adipose tissue of
middle-aged obese insulin sensitive, obese insulin resistant and age-matched lean
insulin sensitive men. By means of local combined blockade of the α- and β-
adrenergic receptors (using a microdialysis approach), abdominal subcutaneous
adipose tissue lipolysis was investigated during a single bout of low-intensity
endurance exercise. In addition, we examined whether a combined endurance and
resistance exercise training intervention for 12 weeks could improve abdominal
subcutaneous adipose tissue lipolysis in obese insulin resistant individuals.
The main conclusions from the studies described in this thesis are discussed in
Chapter 8 and placed into a broader perspective, accompanied by suggestions for
further research.
22
                                                                                  Chapter 1
REFERENCES
1.    World Health Organization. Fact sheet: Obesity and overweight. Updated June 2016.
      2016.
2.    World Health Organization. World Health Statistics 2015, Geneva,2015 [Available
      from: www.who.int/gho/publications/world_health_statistics/2015/en/.
3.    Di Cesare M, Bentham J, Stevens GA, Zhou B, Danaei G, Lu Y, Bixby H, Cowan MJ,
      Riley LM, Hajifathalian K, Fortunato L, Taddei C, Bennett JE, Ikeda N, Zhu D,
      Zimmermann E, J. ZC. Trends in adult body-mass index in 200 countries from 1975
      to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2
      million participants. Lancet. 2016;387(10026):1377-96.
4.    Despres JP, Lemieux I. Abdominal obesity and metabolic syndrome. Nature.
      2006;444(7121):881-7.
5.    Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance
      and type 2 diabetes. Nature. 2006;444(7121):840-6.
6.    Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease.
      Diabetes. 1988;37(12):1595-607.
7.    Porte D, Jr. Banting lecture 1990. Beta-cells in type II diabetes mellitus. Diabetes.
      1991;40(2):166-80.
8.    Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with
      cardiovascular disease. Nature. 2006;444(7121):875-80.
9.    Li L, Liu DW, Yan HY, Wang ZY, Zhao SH, Wang B. Obesity is an independent risk
      factor for non-alcoholic fatty liver disease: evidence from a meta-analysis of 21 cohort
      studies. Obes Rev. 2016;17(6):510-9.
10.   Preiss K, Brennan L, Clarke D. A systematic review of variables associated with the
      relationship between obesity and depression. Obes Rev. 2013;14(11):906-18.
11.   Guh DP, Zhang W, Bansback N, Amarsi Z, Birmingham CL, Anis AH. The incidence
      of co-morbidities related to obesity and overweight: a systematic review and meta-
      analysis. BMC Public Health. 2009;9:88.
12.   Bhaskaran K, Douglas I, Forbes H, dos-Santos-Silva I, Leon DA, Smeeth L. Body-
      mass index and risk of 22 specific cancers: a population-based cohort study of 5.24
      million UK adults. Lancet. 2014;384(9945):755-65.
13.   Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K,
      International Agency for Research on Cancer Handbook Working G. Body Fatness
      and Cancer--Viewpoint of the IARC Working Group. N Engl J Med. 2016;375(8):794-
      8.
14.   Hruschka DJ, Hadley C. How much do universal anthropometric standards bias the
      global monitoring of obesity and undernutrition? Obes Rev. 2016;17(11):1030-39.
15.   Tomiyama AJ, Hunger JM, Nguyen-Cuu J, Wells C. Misclassification of
      cardiometabolic health when using body mass index categories in NHANES 2005-
      2012. Int J Obes (Lond). 2016;40(5):883-6.
16.   Karpe F, Pinnick KE. Biology of upper-body and lower-body adipose tissue--link to
      whole-body phenotypes. Nat Rev Endocrinol. 2015;11(2):90-100.
17.   Rush EC, Goedecke JH, Jennings C, Micklesfield L, Dugas L, Lambert EV, Plank LD.
      BMI, fat and muscle differences in urban women of five ethnicities from two countries.
      Int J Obes (Lond). 2007;31(8):1232-9.
18.   Unnikrishnan R, Anjana RM, Mohan V. Diabetes in South Asians: is the phenotype
      different? Diabetes. 2014;63(1):53-5.
19.   Blundell JE, Dulloo AG, Salvador J, Fruhbeck G, BMI ESWGo. Beyond BMI--
      phenotyping the obesities. Obes Facts. 2014;7(5):322-8.
20.   Nicholson JK, Holmes E, Kinross JM, Darzi AW, Takats Z, Lindon JC. Metabolic
      phenotyping in clinical and surgical environments. Nature. 2012;491(7424):384-92.
                                                                                           23
Chapter 1
24
                                                                                  Chapter 1
40.   Rey-Lopez JP, de Rezende LF, Pastor-Valero M, Tess BH. The prevalence of
      metabolically healthy obesity: a systematic review and critical evaluation of the
      definitions used. Obes Rev. 2014;15(10):781-90.
41.   Bell JA, Kivimaki M, Hamer M. Metabolically healthy obesity and risk of incident type
      2 diabetes: a meta-analysis of prospective cohort studies. Obes Rev.
      2014;15(6):504-15.
42.   Kramer CK, Zinman B, Retnakaran R. Are metabolically healthy overweight and
      obesity benign conditions?: A systematic review and meta-analysis. Ann Intern Med.
      2013;159(11):758-69.
43.   Eckel N, Meidtner K, Kalle-Uhlmann T, Stefan N, Schulze MB. Metabolically healthy
      obesity and cardiovascular events: A systematic review and meta-analysis. Eur J
      Prev Cardiol. 2016;23(9):956-66.
44.   Appleton SL, Seaborn CJ, Visvanathan R, Hill CL, Gill TK, Taylor AW, Adams RJ,
      North West Adelaide Health Study T. Diabetes and cardiovascular disease outcomes
      in the metabolically healthy obese phenotype: a cohort study. Diabetes Care.
      2013;36(8):2388-94.
45.   Ryden M, Hrydziuszko O, Mileti E, Raman A, Bornholdt J, Boyd M, Toft E, Qvist V,
      Naslund E, Thorell A, Andersson DP, Dahlman I, Gao H, Sandelin A, Daub CO,
      Arner P. The Adipose Transcriptional Response to Insulin Is Determined by Obesity,
      Not Insulin Sensitivity. Cell Rep. 2016;16(9):2317-26.
46.   Munoz-Garach A, Cornejo-Pareja I, Tinahones FJ. Does Metabolically Healthy
      Obesity Exist? Nutrients. 2016;8(6).
47.   Goossens GH. The Metabolic Phenotype in Obesity: Fat Mass, Body Fat Distribution,
      and Adipose Tissue Function. Obes Facts. 2017;10(3):207-15.
48.   Bell JA, Shipley MJ, Kivimaki M. Healthy obesity is not safe obesity. Int J Obes
      (Lond). 2016;40(8):1333.
49.   Berard E, Niel O, Rubio A. Is the renin-angiotensin system actually hypertensive?
      Pediatr Nephrol. 2014;29(6):951-60.
50.   Goossens GH. The renin-angiotensin system in the pathophysiology of type 2
      diabetes. Obes Facts. 2012;5(4):611-24.
51.   Frigolet ME, Torres N, Tovar AR. The renin-angiotensin system in adipose tissue and
      its metabolic consequences during obesity. J Nutr Biochem. 2013;24(12):2003-15.
52.   Ramalingam L, Menikdiwela K, LeMieux M, Dufour JM, Kaur G, Kalupahana N,
      Moustaid-Moussa N. The renin angiotensin system, oxidative stress and
      mitochondrial function in obesity and insulin resistance. Biochim Biophys Acta. 2016.
53.   Borghi F, Seva-Pessoa B, Grassi-Kassisse DM. The adipose tissue and the
      involvement of the renin-angiotensin-aldosterone system in cardiometabolic
      syndrome. Cell Tissue Res. 2016;366(3):543-8.
54.   Goossens GH, Blaak EE, van Baak MA. Possible involvement of the adipose tissue
      renin-angiotensin system in the pathophysiology of obesity and obesity-related
      disorders. Obes Rev. 2003;4(1):43-55.
55.   Unger T. The role of the renin-angiotensin system in the development of
      cardiovascular disease. Am J Cardiol. 2002;89(2A):3A-9A; discussion 10A.
56.   Danser AH. Local renin-angiotensin systems. Mol Cell Biochem. 1996;157(1-2):211-
      6.
57.   Darimont C, Vassaux G, Gaillard D, Ailhaud G, Negrel R. In situ microdialysis of
      prostaglandins in adipose tissue: stimulation of prostacyclin release by angiotensin II.
      Int J Obes Relat Metab Disord. 1994;18(12):783-8.
58.   Jones BH, Standridge MK, Moustaid N. Angiotensin II increases lipogenesis in 3T3-
      L1 and human adipose cells. Endocrinology. 1997;138(4):1512-9.
59.   Janke J, Engeli S, Gorzelniak K, Luft FC, Sharma AM. Mature adipocytes inhibit in
      vitro differentiation of human preadipocytes via angiotensin type 1 receptors.
      Diabetes. 2002;51(6):1699-707.
                                                                                           25
Chapter 1
26
                                                                                  Chapter 1
                                                                                           27
Chapter 1
92.    Ralat LA, Guo Q, Ren M, Funke T, Dickey DM, Potter LR, Tang WJ. Insulin-
       degrading enzyme modulates the natriuretic peptide-mediated signaling response. J
       Biol Chem. 2011;286(6):4670-9.
93.    Potter LR. Natriuretic peptide metabolism, clearance and degradation. FEBS J.
       2011;278(11):1808-17.
94.    Standeven KF, Hess K, Carter AM, Rice GI, Cordell PA, Balmforth AJ, Lu B, Scott
       DJ, Turner AJ, Hooper NM, Grant PJ. Neprilysin, obesity and the metabolic
       syndrome. Int J Obes (Lond). 2011;35(8):1031-40.
95.    Walford GA, Ma Y, Christophi CA, Goldberg RB, Jarolim P, Horton E, Mather KJ,
       Barrett-Connor E, Davis J, Florez JC, Wang TJ, Diabetes Prevention Program
       Research G. Circulating natriuretic peptide concentrations reflect changes in insulin
       sensitivity over time in the Diabetes Prevention Program. Diabetologia.
       2014;57(5):935-9.
96.    Jujic A, Nilsson PM, Persson M, Holst JJ, Torekov SS, Lyssenko V, Groop L,
       Melander O, Magnusson M. Atrial Natriuretic Peptide in the High Normal Range Is
       Associated With Lower Prevalence of Insulin Resistance. J Clin Endocrinol Metab.
       2016;101(4):1372-80.
97.    Magnusson M, Jujic A, Hedblad B, Engstrom G, Persson M, Struck J, Morgenthaler
       NG, Nilsson P, Newton-Cheh C, Wang TJ, Melander O. Low plasma level of atrial
       natriuretic peptide predicts development of diabetes: the prospective Malmo Diet and
       Cancer study. J Clin Endocrinol Metab. 2012;97(2):638-45.
98.    Lazo M, Young JH, Brancati FL, Coresh J, Whelton S, Ndumele CE, Hoogeveen R,
       Ballantyne CM, Selvin E. NH2-terminal pro-brain natriuretic peptide and risk of
       diabetes. Diabetes. 2013;62(9):3189-93.
99.    Pfister R, Sharp S, Luben R, Welsh P, Barroso I, Salomaa V, Meirhaeghe A, Khaw
       KT, Sattar N, Langenberg C, Wareham NJ. Mendelian randomization study of B-type
       natriuretic peptide and type 2 diabetes: evidence of causal association from
       population studies. PLoS Med. 2011;8(10):e1001112.
100.   Jujic A, Nilsson PM, Engstrom G, Hedblad B, Melander O, Magnusson M. Atrial
       natriuretic peptide and type 2 diabetes development--biomarker and genotype
       association study. PLoS One. 2014;9(2):e89201.
101.   Pivovarova O, Gogebakan O, Kloting N, Sparwasser A, Weickert MO, Haddad I,
       Nikiforova VJ, Bergmann A, Kruse M, Seltmann AC, Bluher M, Pfeiffer AF, Rudovich
       N. Insulin up-regulates natriuretic peptide clearance receptor expression in the
       subcutaneous fat depot in obese subjects: a missing link between CVD risk and
       obesity? J Clin Endocrinol Metab. 2012;97(5):E731-9.
102.   Ryden M, Backdahl J, Petrus P, Thorell A, Gao H, Coue M, Langin D, Moro C, Arner
       P. Impaired atrial natriuretic peptide-mediated lipolysis in obesity. Int J Obes (Lond).
       2016;40(4):714-20.
103.   Kovacova Z, Tharp WG, Liu D, Wei W, Xie H, Collins S, Pratley RE. Adipose tissue
       natriuretic peptide receptor expression is related to insulin sensitivity in obesity and
       diabetes. Obesity (Silver Spring). 2016;24(4):820-8.
104.   Verboven K, Hansen D, Moro C, Eijnde BO, Hoebers N, Knol J, Bouckaert W, Dams
       A, Blaak EE, Jocken JW. Attenuated atrial natriuretic peptide-mediated lipolysis in
       subcutaneous adipocytes of obese type 2 diabetic men. Clin Sci (Lond).
       2016;130(13):1105-14.
105.   Clerico A, Vittorini S. The Cardiac Natriuretic Hormone System. In: Clerico A, Emdin
       M, editors. Natriuretic peptides: The Hormones of the Heart: Springer-Verlag Italia;
       2006. p. 21-64.
106.   Kerkela R, Ulvila J, Magga J. Natriuretic Peptides in the Regulation of Cardiovascular
       Physiology and Metabolic Events. J Am Heart Assoc. 2015;4(10):e002423.
28
                                                                                   Chapter 1
                                                                                            29
Chapter 1
30
                                                                                 Chapter 1
       KA, McKelvie RS, Zannad F, Pina IL, Investigators H-A. Efficacy and safety of
       exercise training in patients with chronic heart failure: HF-ACTION randomized
       controlled trial. JAMA. 2009;301(14):1439-50.
138.   Gaede P, Vedel P, Larsen N, Jensen GV, Parving HH, Pedersen O. Multifactorial
       intervention and cardiovascular disease in patients with type 2 diabetes. N Engl J
       Med. 2003;348(5):383-93.
139.   den Boer AT, Herraets IJ, Stegen J, Roumen C, Corpeleijn E, Schaper NC, Feskens
       E, Blaak EE. Prevention of the metabolic syndrome in IGT subjects in a lifestyle
       intervention: results from the SLIM study. Nutr Metab Cardiovasc Dis.
       2013;23(11):1147-53.
140.   Eriksson KF, Lindgarde F. Prevention of type 2 (non-insulin-dependent) diabetes
       mellitus by diet and physical exercise. The 6-year Malmo feasibility study.
       Diabetologia. 1991;34(12):891-8.
141.   Eriksson KF, Lindgarde F. No excess 12-year mortality in men with impaired glucose
       tolerance who participated in the Malmo Preventive Trial with diet and exercise.
       Diabetologia. 1998;41(9):1010-6.
142.   Pan XR, Li GW, Hu YH, Wang JX, Yang WY, An ZX, Hu ZX, Lin J, Xiao JZ, Cao HB,
       Liu PA, Jiang XG, Jiang YY, Wang JP, Zheng H, Zhang H, Bennett PH, Howard BV.
       Effects of diet and exercise in preventing NIDDM in people with impaired glucose
       tolerance. The Da Qing IGT and Diabetes Study. Diabetes Care. 1997;20(4):537-44.
143.   Tuomilehto J, Lindstrom J, Eriksson JG, Valle TT, Hamalainen H, Ilanne-Parikka P,
       Keinanen-Kiukaanniemi S, Laakso M, Louheranta A, Rastas M, Salminen V,
       Uusitupa M, Finnish Diabetes Prevention Study G. Prevention of type 2 diabetes
       mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N
       Engl J Med. 2001;344(18):1343-50.
144.   Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA,
       Nathan DM, Diabetes Prevention Program Research G. Reduction in the incidence of
       type 2 diabetes with lifestyle intervention or metformin. N Engl J Med.
       2002;346(6):393-403.
145.   Mensink M, Blaak EE, Corpeleijn E, Saris WH, de Bruin TW, Feskens EJ. Lifestyle
       intervention according to general recommendations improves glucose tolerance.
       Obes Res. 2003;11(12):1588-96.
146.   Lindstrom J, Ilanne-Parikka P, Peltonen M, Aunola S, Eriksson JG, Hemio K,
       Hamalainen H, Harkonen P, Keinanen-Kiukaanniemi S, Laakso M, Louheranta A,
       Mannelin M, Paturi M, Sundvall J, Valle TT, Uusitupa M, Tuomilehto J, Finnish
       Diabetes Prevention Study G. Sustained reduction in the incidence of type 2 diabetes
       by lifestyle intervention: follow-up of the Finnish Diabetes Prevention Study. Lancet.
       2006;368(9548):1673-9.
147.   Li G, Zhang P, Wang J, Gregg EW, Yang W, Gong Q, Li H, Li H, Jiang Y, An Y,
       Shuai Y, Zhang B, Zhang J, Thompson TJ, Gerzoff RB, Roglic G, Hu Y, Bennett PH.
       The long-term effect of lifestyle interventions to prevent diabetes in the China Da
       Qing Diabetes Prevention Study: a 20-year follow-up study. Lancet.
       2008;371(9626):1783-9.
148.   Lindstrom J, Peltonen M, Eriksson JG, Ilanne-Parikka P, Aunola S, Keinanen-
       Kiukaanniemi S, Uusitupa M, Tuomilehto J, Finnish Diabetes Prevention S. Improved
       lifestyle and decreased diabetes risk over 13 years: long-term follow-up of the
       randomised Finnish Diabetes Prevention Study (DPS). Diabetologia. 2013;56(2):284-
       93.
149.   Knowler WC, Fowler SE, Hamman RF, Christophi CA, Hoffman HJ, Brenneman AT,
       Brown-Friday JO, Goldberg R, Venditti E, Nathan DM. 10-year follow-up of diabetes
       incidence and weight loss in the Diabetes Prevention Program Outcomes Study.
       Lancet. 2009;374(9702):1677-86.
                                                                                          31
Chapter 1
150.   Roumen C, Blaak EE, Corpeleijn E. Lifestyle intervention for prevention of diabetes:
       determinants of success for future implementation. Nutr Rev. 2009;67(3):132-46.
151.   Torjesen PA, Birkeland KI, Anderssen SA, Hjermann I, Holme I, Urdal P. Lifestyle
       changes may reverse development of the insulin resistance syndrome. The Oslo Diet
       and Exercise Study: a randomized trial. Diabetes Care. 1997;20(1):26-31.
152.   Chin SH, Kahathuduwa CN, Binks M. Physical activity and obesity: what we know
       and what we need to know. Obes Rev. 2016;17(12):1226-44.
153.   Blaak EE. Carbohydrate quantity and quality and cardio-metabolic risk. Curr Opin
       Clin Nutr Metab Care. 2016;19(4):289-93.
154.   Lin X, Zhang X, Guo J, Roberts CK, McKenzie S, Wu WC, Liu S, Song Y. Effects of
       Exercise Training on Cardiorespiratory Fitness and Biomarkers of Cardiometabolic
       Health: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J
       Am Heart Assoc. 2015;4(7).
155.   Cassidy S, Thoma C, Houghton D, Trenell MI. High-intensity interval training: a
       review of its impact on glucose control and cardiometabolic health. Diabetologia.
       2017;60(1):7-23.
156.   Caspersen CJ, Powell KE, Christenson GM. Physical activity, exercise, and physical
       fitness: definitions and distinctions for health-related research. Public Health Rep.
       1985;100(2):126-31.
157.   Thompson D, Karpe F, Lafontan M, Frayn K. Physical activity and exercise in the
       regulation of human adipose tissue physiology. Physiol Rev. 2012;92(1):157-91.
158.   Stanford KI, Middelbeek RJ, Goodyear LJ. Exercise Effects on White Adipose Tissue:
       Beiging and Metabolic Adaptations. Diabetes. 2015;64(7):2361-8.
159.   Stanford KI, Goodyear LJ. Exercise regulation of adipose tissue. Adipocyte.
       2016;5(2):153-62.
160.   Hodgetts V, Coppack SW, Frayn KN, Hockaday TD. Factors controlling fat
       mobilization from human subcutaneous adipose tissue during exercise. J Appl
       Physiol (1985). 1991;71(2):445-51.
161.   Romijn JA, Coyle EF, Sidossis LS, Gastaldelli A, Horowitz JF, Endert E, Wolfe RR.
       Regulation of endogenous fat and carbohydrate metabolism in relation to exercise
       intensity and duration. Am J Physiol. 1993;265(3 Pt 1):E380-91.
162.   Karpe F, Fielding BA, Ilic V, Humphreys SM, Frayn KN. Monitoring adipose tissue
       blood flow in man: a comparison between the (133)xenon washout method and
       microdialysis. Int J Obes Relat Metab Disord. 2002;26(1):1-5.
163.   Brenner BM, Ballermann BJ, Gunning ME, Zeidel ML. Diverse biological actions of
       atrial natriuretic peptide. Physiol Rev. 1990;70(3):665-99.
164.   Moro C, Pillard F, De Glisezinski I, Harant I, Rivi??Re D, Stich V, Lafontan MAX,
       Crampes FO, Berlan M. Training Enhances ANP Lipid-Mobilizing Action in Adipose
       Tissue of Overweight Men. Medicine & Science in Sports & Exercise.
       2005;37(7):1126-32.
165.   Samra JS, Simpson EJ, Clark ML, Forster CD, Humphreys SM, Macdonald IA, Frayn
       KN. Effects of epinephrine infusion on adipose tissue: interactions between blood
       flow and lipid metabolism. Am J Physiol. 1996;271(5 Pt 1):E834-9.
166.   Berggren JR, Hulver MW, Houmard JA. Fat as an endocrine organ: influence of
       exercise. J Appl Physiol (1985). 2005;99(2):757-64.
167.   Mulla NA, Simonsen L, Bulow J. Post-exercise adipose tissue and skeletal muscle
       lipid metabolism in humans: the effects of exercise intensity. J Physiol. 2000;524 Pt
       3:919-28.
168.   Moro C, Pillard F, de Glisezinski I, Crampes F, Thalamas C, Harant I, Marques MA,
       Lafontan M, Berlan M. Sex differences in lipolysis-regulating mechanisms in
       overweight subjects: effect of exercise intensity. Obesity (Silver Spring).
       2007;15(9):2245-55.
32
                                                                                 Chapter 1
169.   Stallknecht B, Kiens B, Helge JW, Richter EA, Galbo H. Interstitial glycerol
       concentrations in human skeletal muscle and adipose tissue during graded exercise.
       Acta Physiol Scand. 2004;180(4):367-77.
170.   Ryden M, Jocken J, van Harmelen V, Dicker A, Hoffstedt J, Wiren M, Blomqvist L,
       Mairal A, Langin D, Blaak E, Arner P. Comparative studies of the role of hormone-
       sensitive lipase and adipose triglyceride lipase in human fat cell lipolysis. Am J
       Physiol Endocrinol Metab. 2007;292(6):E1847-55.
171.   Jocken JW, Blaak EE. Catecholamine-induced lipolysis in adipose tissue and skeletal
       muscle in obesity. Physiol Behav. 2008;94(2):219-30.
172.   Mauriege P, Despres JP, Prud'homme D, Pouliot MC, Marcotte M, Tremblay A,
       Bouchard C. Regional variation in adipose tissue lipolysis in lean and obese men. J
       Lipid Res. 1991;32(10):1625-33.
173.   Reynisdottir S, Wahrenberg H, Carlstrom K, Rossner S, Arner P. Catecholamine
       resistance in fat cells of women with upper-body obesity due to decreased
       expression of beta 2-adrenoceptors. Diabetologia. 1994;37(4):428-35.
174.   Crampes F, Beauville M, Riviere D, Garrigues M. Effect of physical training in
       humans on the response of isolated fat cells to epinephrine. J Appl Physiol (1985).
       1986;61(1):25-9.
175.   Crampes F, Riviere D, Beauville M, Marceron M, Garrigues M. Lipolytic response of
       adipocytes to epinephrine in sedentary and exercise-trained subjects: sex-related
       differences. Eur J Appl Physiol Occup Physiol. 1989;59(4):249-55.
176.   Riviere D, Crampes F, Beauville M, Garrigues M. Lipolytic response of fat cells to
       catecholamines in sedentary and exercise-trained women. J Appl Physiol (1985).
       1989;66(1):330-5.
177.   Despres JP, Bouchard C, Savard R, Prud'homme D, Bukowiecki L, Theriault G.
       Adaptive changes to training in adipose tissue lipolysis are genotype dependent. Int J
       Obes. 1984;8(1):87-95.
178.   Despres JP, Bouchard C, Savard R, Tremblay A, Marcotte M, Theriault G. The effect
       of a 20-week endurance training program on adipose-tissue morphology and lipolysis
       in men and women. Metabolism. 1984;33(3):235-9.
179.   De Glisezinski I, Crampes F, Harant I, Berlan M, Hejnova J, Langin D, Riviere D,
       Stich V. Endurance training changes in lipolytic responsiveness of obese adipose
       tissue. Am J Physiol. 1998;275(6 Pt 1):E951-6.
180.   van Aggel-Leijssen DP, Saris WH, Wagenmakers AJ, Senden JM, van Baak MA.
       Effect of exercise training at different intensities on fat metabolism of obese men. J
       Appl Physiol (1985). 2002;92(3):1300-9.
181.   Richterova B, Stich V, Moro C, Polak J, Klimcakova E, Majercik M, Harant I, Viguerie
       N, Crampes F, Langin D, Lafontan M, Berlan M. Effect of endurance training on
       adrenergic control of lipolysis in adipose tissue of obese women. J Clin Endocrinol
       Metab. 2004;89(3):1325-31.
182.   de Glisezinski I, Moro C, Pillard F, Marion-Latard F, Harant I, Meste M, Berlan M,
       Crampes F, Riviere D. Aerobic training improves exercise-induced lipolysis in SCAT
       and lipid utilization in overweight men. Am J Physiol Endocrinol Metab.
       2003;285(5):E984-90.
183.   Stich V, de Glisezinski I, Galitzky J, Hejnova J, Crampes F, Riviere D, Berlan M.
       Endurance training increases the beta-adrenergic lipolytic response in subcutaneous
       adipose tissue in obese subjects. Int J Obes Relat Metab Disord. 1999;23(4):374-81.
184.   Sutherland LN, Bomhof MR, Capozzi LC, Basaraba SA, Wright DC. Exercise and
       adrenaline increase PGC-1{alpha} mRNA expression in rat adipose tissue. J Physiol.
       2009;587(Pt 7):1607-17.
185.   Trevellin E, Scorzeto M, Olivieri M, Granzotto M, Valerio A, Tedesco L, Fabris R,
       Serra R, Quarta M, Reggiani C, Nisoli E, Vettor R. Exercise training induces
                                                                                          33
Chapter 1
34
                                                                                 Chapter 1
                                                                                         35
CHAPTER 2
Targeting fatty acid metabolism to
improve glucose metabolism
ABSTRACT
Disturbances in fatty acid metabolism in adipose tissue, liver, skeletal muscle, gut
and pancreas play an important role in the development of insulin resistance,
impaired glucose metabolism and type 2 diabetes mellitus. Alterations in diet
composition may contribute to prevent and/or reverse these disturbances through
modulation of fatty acid metabolism.
Besides an increased fat mass, adipose tissue dysfunction, characterized by an
altered capacity to store lipids and an altered secretion of adipokines, may result in
lipid overflow, systemic inflammation and excessive lipid accumulation in non-
adipose tissues like liver, skeletal muscle and the pancreas. These impairments
together promote the development of impaired glucose metabolism, insulin
resistance and type 2 diabetes mellitus. Furthermore, intrinsic functional
impairments in either of these organs may contribute to lipotoxicity and insulin
resistance. The present review provides an overview of fatty acid metabolism-
related pathways in adipose tissue, liver, skeletal muscle, pancreas and gut, which
can be targeted by diet or food components, thereby improving glucose
metabolism.
38
                                                                           Chapter 2
INTRODUCTION
Obesity is considered a global health problem, since it is closely associated with
the development of chronic metabolic diseases, including cardiovascular disease,
type 2 diabetes mellitus (T2D) and certain types of cancer [1]. The pathogenesis of
T2D is characterized by the development of both insulin resistance in peripheral
tissues and pancreatic β-cell failure [2, 3]. Disturbances in fatty acid metabolism
play a crucial role in the development of an impaired glucose metabolism and
diabetes. Combined dietary and physical activity intervention may reduce the
incidence of T2D by 30-60% [4, 5], which may for a considerable part be explained
through modulation of fatty acid metabolism [6]. A better understanding of the
interaction between diet, fatty acid metabolism, insulin resistance and β-cell
dysfunction is needed to develop novel strategies to prevent impairments in
glucose metabolism and, consequently, the development of T2D.
A tight interplay between adipose tissue, skeletal muscle, liver, pancreas and the
gut regulate fatty acid metabolism in the human body. Besides an increased fat
mass, adipose tissue dysfunction, characterized by an altered capacity to store
lipids and low-grade inflammation, plays a major role in the development of insulin
resistance and impaired glucose metabolism by promoting excessive fat storage in
non-adipose tissues like liver, skeletal muscle, pancreas, the heart and kidneys [7-
10] (Figure 1).
Furthermore, intrinsic functional impairments in either of these organs may
contribute to lipotoxicity and insulin resistance. Skeletal muscle is generally
considered the most important organ in peripheral insulin resistance [11]. Beside
an increased fatty acid (FA) supply [12], a reduced skeletal muscle oxidative
capacity [13, 14] may contribute to the accumulation of triacylglycerol (TAG) and
bioactive lipid metabolites [15-17]. An increased lipid supply to the liver may result
in an increased hepatic TAG content and very low-density lipoprotein (VLDL-TAG)
output [18], a higher glucose production and output [19-21] and a reduced insulin
clearance by the liver [22-24], leading to hyperlipidemia, insulin resistance and
glucose intolerance [25]. Furthermore, hyperglycemia may, together with
hyperlipidemia-related lipotoxicity in the pancreas, result in decreased glucose-
stimulated insulin secretion by the pancreatic β-cells [26].
Recent evidence also indicates that the gut microbiota and its products may
contribute to the development of insulin resistance and a disturbed glucose
metabolism (Figure 1). There is accumulating evidence that alterations in the gut
microbiota composition and function may affect adipose tissue, liver and skeletal
muscle lipid and glucose metabolism, at least partly through effects on bile acid
metabolism [27-29].
Energy intake and diet composition (i.e. dietary fatty acids, polyphenols, fibers)
may have a significant impact on many aspects of fatty acid metabolism in different
tissues. These effects can be both acute (i.e. postprandial phase) and more long-
term, ultimately affecting health status. The aim of the present review is to provide
an overview of fatty acid metabolism related pathways in adipose tissue, liver,
skeletal muscle, pancreas and gut that can be targeted by diet or food components
and, as a consequence, improve whole-body glucose metabolism and insulin
sensitivity. The focus of the present review will be on the major tissues involved in
inter-organ substrate metabolism and lipid-induced insulin resistance. Associations
                                                                                   39
Chapter 2
40
                                                                                                Chapter 2
                                                                                                          41
Chapter 2
42
                                                                            Chapter 2
A considerable part of the variation in LPL activity during fasting, exercise and in
the postprandial state may be explained by post-translational regulation by
apolipoproteins (APO-C1, -2, -3, -5, APO-E) and the angiopoietin-like proteins
(ANGPTL-3, -4, -8), as reviewed in detail elsewhere [62]. ANGPTL4, in particular,
has been identified as an inhibitor of LPL activity and may be regulated by diet and
fasting, since its expression and secretion changes in parallel with circulating FFA
concentrations [62-66].
In obese conditions, basal LPL activity has been reported to be increased, whereas
FA spillover from LPL-mediated TAG hydrolysis across adipose tissue during
hyperinsulinemia is less suppressed as compared to lean healthy individuals [67,
68]. Indeed, the removal of TAG across adipose tissue was found to be impaired in
obesity, insulin resistance and T2D, due to a reduced insulin-mediated stimulation
of LPL activity [68-73], suggesting less efficient removal of dietary lipids by adipose
tissue in these subjects. In line, a recent study has demonstrated that the relative
quantity of meal fat stored in adipose tissue after the intake of subsequent meals
was significantly reduced in (abdominally) obese versus lean individuals [69], which
in turn may contribute to increased lipid spillover and ectopic fat deposition. To
define nutritional targets affecting the regulation of LPL activity, which may
subsequently reduce lipid spillover, ectopic fat deposition and improve insulin
sensitivity, additional studies to determine the role of different LPL modulators in
the regulation of its activity in vivo are clearly needed.
In addition to impaired LPL action in obese and insulin resistant conditions,
disturbances in the uptake of the liberated FFAs (lipid spillover) may contribute to
impaired lipid buffering. FAs are able to cross the endothelial barrier via passive
diffusion, which is dependent on the concentration gradient of FAs across the
membrane of the adipocyte, or via active transport [41, 43, 74, 75]. Active FA
transport is facilitated by several enzymes and proteins, of which numerous FA-
transport proteins have been identified in adipocytes [76], including fatty acid
translocase/CD36 (CD36), membrane-bound and cytosolic fatty acid binding
protein (FABPpm and FABPc, respectively) and fatty acid transporter protein
(FATP) [77]. It has been shown that the translocation of CD36 from the intracellular
stores to the plasma membrane is regulated by various stimuli, including insulin
and increased activation of AMP-activated protein kinase (AMPK) [78-81]. Although
CD36 protein expression is higher in the subcutaneous adipose tissue of obese,
overweight and T2D compared to lean subjects [82], the net removal of TAG and
FAs by adipose tissue is reduced in the former groups, suggestive of impairments
in intracellular CD36 trafficking. Taken together, strategies to reduce spillover and
increase FFA trapping in adipose tissue, mainly in the postprandial phase, will lead
to a reduced lipid spillover and may consequently prevent or at least reduce
ectopic fat deposition. On the one hand, this may be achieved by an increased
FFA concentration gradient across the adipocyte membrane and a subsequent
decreased intracellular FFA concentration. This might be accomplished by
increasing FA reesterification and/or oxidation, which will be discussed later in this
section. On the other hand, an increased facilitated diffusion by fatty acid
transporters like CD36 may possibly increase fatty acid trapping, thereby reducing
FFA spillover. Indeed, it has been shown that tissue-specific CD36 transcripts
differentially influence fatty acid homeostasis and insulin sensitivity [83]. Adipocyte
CD36 appears to be metabolically protective, and its selective upregulation might
                                                                                    43
Chapter 2
44
                                                                                               Chapter 2
Adipose tissue lipid uptake (1) is impaired in obesity, insulin resistance and T2D, due to a reduced
insulin-mediated stimulation of LPL activity and, possibly, decreased facilitated FFA uptake via CD36 in
adipocytes, which contributes to lipid overflow in the circulation. TAG and DAG are broken down by the
lipases ATGL and HSL. Fasting lipolysis, expressed per unit fat mass, may be decreased in obese
subjects (2). Nevertheless, insulin-mediated suppression of adipose tissue lipolysis per unit fat mass is
attenuated in obese individuals, resulting in an increase in whole-body lipolysis during postprandial
conditions. A decreased adipose tissue mitochondrial function (3) may lead to impaired scavenging of
fatty acids, resulting in increased intracellular FFA concentrations, reducing FFA trapping and an
enhanced release of FFA into the circulation. Furthermore, dynamic changes occur in the adipose
tissue immune cell populations during the development of obesity, causing a shift from an anti-
inflammatory towards a more pro-inflammatory phenotype, resulting in a state of low-grade inflammation
(4), affecting insulin sensitivity through different mechanisms. Finally, a reduced adipose tissue
differentiation and expandability (5), leading to a reduced lipid storage capacity (6), contributes to lipid
overflow into the circulation.
Abbreviations: LPL: Lipoprotein lipase; FFA: Free fatty acid; CD36: Fatty acid translocase CD36; TAG:
Triacylglycerol; DAG: Diacylglycerol; MAG: monoacylglycerol; ATGL: Adipose triglyceride lipase; HSL:
Hormone sensitive lipase; ANGPTL4: Angiopoietin-like protein 4; PUFA: Poly unsaturated fatty acid;
SCFA: Short chain fatty acid; FGF21: Fibroblast growth factor 21.
Dashed lines indicate inhibition. Solid lines indicate stimulation. Green lines indicate beneficial effects.
Symbols: (=): unchanged; (): Increased; (): Decreased.
                                                                                                        45
Chapter 2
46
                                                                          Chapter 2
adipose tissue of obese subjects, which persists after significant weight loss [89,
94, 95]. These data suggest that impaired cathecholamine-induced lipolysis may
be an important primary factor in the development of obesity. Data are limited with
respect to NP-induced lipolysis. However, reduced circulating NP levels [104] and
a defective ANP-mediated lipolytic response in subcutaneous adipocytes and
adipose tissue from obese subjects have been observed [105]. Therefore, in
contrast to the anti-lipolytic approach with selective lipase inhibition, improving
catecholamine- and NP-sensitivity has been extensively investigated for the
treatment of obesity and obesity-related complications but has, so far, not shown
promising results due to cardiovascular side effects and receptor desensitization
[106-108].
In addition to classical lipolysis, three recent studies have implicated autophagy, a
homeostatic mechanism functioning as a ‘self-digestion’ system, in selective lipid
hydrolysis under basal and catecholamine-stimulated conditions in adipocytes,
termed lipophagy [109-111]. These data indicate that activation of cytosolic lipases
(i.e. ATGL and HSL) is no longer the sole molecular mechanism to liberate FA from
adipocyte TAG stores. Since ATGL and HSL are expressed at much higher level in
adipocytes than other cell types it is plausible that under normal physiological
conditions adipocytes rely mainly on classical cytosolic lipolysis, while the
alternative pathway for lipid breakdown, lipophagy, may become more important in
pathophysiological conditions, with a reduced ATGL and HSL activity, to maintain
lipid homeostasis. In line, autophagy markers and fluxes appear to be elevated in
adipose tissue of obese insulin resistant and T2D subjects [112-115]. In addition,
nutritional and hormonal regulation of adipose tissue autophagy is impaired in
obese rodents [116].
To summarize, obesity is characterized by an increased basal and a blunted
catecholamine and NP-stimulated lipolysis in subcutaneous adipocytes. This
altered lipid turnover may be an early factor in the development of increased fat
stores and obesity-associated metabolic complications. Modulation of classical
lipolysis recently regained interest in the treatment of obesity-related insulin
resistance, indicated by the development of selective ATGL and HSL inhibitors.
However, to prevent excessive gain in body weight, tissue FFA turnover (uptake,
esterification and oxidation) should be adapted accordingly. Furthermore, lipohagy
might be increased in adipose tissue of obese subjects as compensatory
mechanism to deal with the increased lipid availability due to an attenuated
classical lipolysis. Importantly, before considering manipulation of the classical
and/or alternative pathway of adipose tissue lipolysis for therapeutic purposes, a
better insight into its role in pathophysiology as well as hormonal and nutritional
regulation is warranted.
Lipid droplet formation and fatty acid storage in the adipose tissue
In humans, adipocytes, the major cell type in white and brown adipose tissue
(BAT), are specialized for storing lipids in LD. Several proteins bind the LD surface
and regulate LD size, fusion and number. They include PAT proteins (i.e. perilipin
1, perilipin 2/adipophilin/ADRP, perilipin 3/TIP47 and perilipin 4/S3-12), CIDE (Cell
Death inducing DNA Fragmentation Factor) proteins, Soluble NSF Attachment
                                                                                  47
Chapter 2
Protein Receptor (SNARE), caveolins and several lipases. LDs are dynamic
organelles, constantly forming, growing or shrinking. In recent years, our
knowledge of LD dynamics and biogenesis has increased, as reviewed extensively
elsewhere [117-123]. Impaired LD expansion and TAG storage capacity may play a
role in lipid storage diseases, including obesity and T2D. In obese conditions, the
storage capacity of the LD in adipocytes is exceeded. CIDE-C/FSP27 (fat-specific
protein 27) and PLIN-1 largely regulate TAG storage in human adipocytes by
facilitating lipid transfer from smaller to larger LDs and by regulating intracellular
lipolysis. Studies examining the expression of CIDE proteins and PLIN-1 as a
function of insulin sensitivity found that mRNA levels of these LD-associated
proteins correlate positively with insulin sensitivity in subjects with similar body
mass index [124, 125]. These findings in humans contrast with findings in mice, in
which the lack of CIDEC/FSP27 or PLIN-1 protected against high fat diet-induced
obesity and insulin resistance [126-128], highlighting the difficulties in extrapolating
results from mice to human pathologies.
In summary, although human data are limited, high levels of protein that promote
TAG storage, including CIDEC/FSP27 and PLIN-1, might help to sequester lipids in
the adipose tissue and to protect against insulin resistance. Furthermore, impaired
LD expandability may prevent the recruitment of new adipocytes by either initiating
a pro-inflammatory response or by preventing the secretion of yet unidentified
factors that promotes recruitment of adipocyte progenitors.
48
                                                                           Chapter 2
                                                                                   49
Chapter 2
50
                                                                           Chapter 2
                                                                                   51
Chapter 2
may in turn lead to ectopic fat storage and insulin resistance [188]. In addition,
lipid-induced impairments in adipokine secretion may also exert direct effects on
peripheral insulin sensitivity [189]. It has been demonstrated that adiponectin
increases skeletal muscle fat oxidation and therefore, the decreased adiponectin
concentrations in obese conditions may impact fat oxidation, and as such, affect
lipid accumulation and insulin sensitivity [190, 191].
As reviewed by Ohashi et al [192], several anti-inflammatory adipokines, such as
adiponectin, the family of the C1q/TNF-related Proteins (CTRP3, -6, -9), adipolin
and omentin-1 may exert beneficial effects on obesity-related complications.
CTRP3 is a novel adipokine, expressed at the AT and found in circulating plasma
[193, 194], which regulates hepatic glucose output [195], suppresses chemokine
production in response to lauric acid, LPS or TLR stimulation in macrophages and
adipocytes [196] and stimulates the expression of adiponectin in primary human
adipocytes and cultured 3T3-L1 adipocytes [197]. CTRP6 has been found to
increase the expression of the anti-inflammatory cytokine IL-10 in human
monocyte-derived macrophages [198], to stimulate activation of AMPK and
enhance fatty acid oxidation in skeletal muscle cells [199]. In cultured myocytes,
CTRP9 activates phosphorylation of AMPK and protein kinase B (PKB) and
promotes insulin stimulated glucose uptake [194]. It also reduced diet-induced
weight gain, decreased insulin resistance and hepatic steatosis, with enhanced
AMPK activation and fat oxidation in skeletal muscle in CTRP9 transgenic mice
[200]. CTRP12, also known as Adipolin (adipose-derived insulin-sensitizing factor)
[201], has been reported to activate insulin signaling in the liver and adipose tissue
of obese mice [202]. Furthermore, omentin-1, also known as intelectin-1, is
abundantly expressed in visceral fat tissue [203], is decreased in obese individuals
[204] and increased insulin stimulated glucose uptake in cultured adipocytes in
vitro [203].
In addition, dipeptidyl peptidase-4 (DPP4) is released by mature adipocytes and
inhibits skeletal muscle insulin signaling [205, 206]. Beside lipolysis and insulin
signaling, pro-inflammatory cytokines may also regulate adipocyte mitochondrial
function [207]. Of note, not only adipose tissue but also other organs such as the
liver, skeletal muscle, heart and pancreas may contribute to lipid-induced systemic
inflammation in obesity via secretion of cytokines [208, 209].
Taken together, lipids are important triggers for adipose tissue inflammation and
consequently insulin resistance in obesity. Therefore, interventions aimed at
improving lipid metabolism may improve glucose metabolism via reduction of
adipose tissue and systemic inflammation.
52
                                                                             Chapter 2
Presently, there is a clear need for additional studies to determine the regulators of
LPL activity, including the involvement of proteins affecting post-translational
mechanisms, like ANGPTL4, which may be regulated by diet [63].
Secondly, as discussed above partial HSL or ATGL inhibition may reduce lipid
overflow and possibly improve metabolic profile. Interestingly, intravenous acetate
infusion [210] or colonic acetate administration may reduce systemic FFA
concentrations, through an effect on HSL phosphorylation [211]. Dietary
manipulation of colonic acetate and short chain fatty acid (SCFA) concentration
through pre- and probiotics may be a promising target in this respect.
Thirdly, modulation of the adipose tissue lipophagic pathway might be a potential
target pathway as well. Recently, we have shown that dietary polyphenols including
resveratrol and epigallocatechin-gallate (EGCG), found naturally in red wine and
green tea have caloric restriction-like effects in overweight humans [212].
Interestingly, our microarray data showed that an improved adipose tissue lipolysis
and function, is associated with selectively targeting of the master regulator of
lipophagy, TFEB, in human adipose tissue following resveratrol supplementation
[213]. However, it remains to be determined whether lipophagy-mediated lipid
catabolism in adipose tissue is directly involved in the potential beneficial effects of
polyphenols.
As indicated above, nutritional strategies to improve mitochondrial function, like
specific polyphenols or a combination of polyphenols, may be effective in balancing
lipid supply to utilization [212, 213], improving thereby adipose tissue function.
Moreover, recent data from our group also suggest that a dysbalance between
oxygen supply and oxygen utilization leading to an increased adipose tissue
oxygen tension may induced adipose tissue dysfunction [214, 215], again
illustrating the importance of a normal mitochondrial mass and function.
Factors affecting the activity and recruitment of BAT, may have positive effect with
respect to adipose tissue function, body weight control and insulin sensitivity.
Currently, most promising may be dietary components affecting our microbial bile
acid metabolism, leading to increased circulating FGF21, a mediator of lipid and
carbohydrate metabolism, also inducing recruitment of brown adipocytes [216].
The effect of dietary quality on adipose tissue function and ectopic fat accumulation
remains an area of particular interest in composing optimal diets that minimize
ectopic and abdominal fat accumulation. There is strong evidence that avoidance
of high saturated fatty acids (SFA) diet contributes to lower health risks among
obese, metabolic syndrome and diabetic patients [217]. There are indications that
the monounsaturated fatty acids (MUFA) and/or PUFA have more beneficial effects
compared to SFA on the action of insulin [218-221]. Research showed that n-3
PUFA (eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA) and
docosahexaenoic acid (DHA)) reduced LD formation in 3T3-L1 cells compared to
SFA [222]. Further, long chain n-3 PUFA may increase fatty acid oxidation and
mitochondrial biogenesis in adipose tissue [223, 224], may inhibit fat cell
proliferation [225] and may limit fat cell hypertrophy and hyperplasia [226].
Additionally, positive effect have been ascribed to specifically n-6 fatty acids in
reducing abdominal fat area, improving insulin sensitivity and in reducing visceral
fat/subcutaneous fat ratio compared with a SFA diet [227, 228].
Dietary fat quality not only modulates lipid metabolism, it may also affect low-grade
inflammation, as for instance seen by exposure of myotubes or adipocytes to SFA
                                                                                     53
Chapter 2
which increased IL-6 mRNA and protein expression, possibly via activation of
nuclear factor-κB (NF-κB) [229, 230]. A human intervention study with a SFA rich
diet for 5 weeks showed higher concentrations of C-reactive protein (CRP),
fibrinogen and IL-6 compared to a diet enriched in MUFA [231]. Substantial
increase in the PUFA intake can also decrease low-grade inflammation as
indicated by circulating CRP, IL-6 or soluble adhesion molecules [232, 233].
In summary, polyphenols, specific dietary fatty acids and pre- and probiotics may
be promising nutritional components in improving the balance between lipolysis,
autophagy and mitochondrial function and in stimulation of adipose tissue
browning, thereby improving adipose tissue function and whole-body glucose
homeostasis. For an overview of putative nutritional targets see Table 1.
54
                                                                              Chapter 2
In the liver, beside LPL, hepatic lipase (HL) plays an important role in fatty acid
metabolism, as it is both a phospholipase and a TAG lipase [253]. An increased HL
activity promotes FFA uptake into hepatocytes and has been shown to be
increased in obese humans [254-256], insulin resistant rodents [257], type 1
diabetic patients [258] and in NAFLD patients [259].
The rate of plasma FFA uptake by the liver is determined by both the plasma FA
concentration and the hepatocellular capacity for FA uptake [260], which depends
on the number and activity of transporter proteins on the plasma membrane of the
hepatocyte. The main proteins in the hepatic transmembrane FA transport are
FATP-2 [261], FATP-5 [262], caveolins, and to a lower extent fatty acid translocase
CD36 [263, 264]. FATP-5 knockout mice show resistance to diet-induced obesity
and hepatic lipid accumulation, but there is no evidence for involvement of this
isoform in human obesity [265]. Also, silencing of hepatic FATP-5 in hyperglycemic
mice showed a rapid (2-3 weeks) reduction in liver FFA uptake and a reduction in
serum glucose levels, reaching normal glycaemia after 5 weeks [262]. Additionally,
expression of FABP-4 and -5 correlated with hepatic fatty acid infiltration in NAFLD
patients [266]. Interestingly, it was shown that specific bile acids (BAs)
(ursodeoxycholic acid (UDCA) and deoxycholic acid (DCA)) inhibit liver-specific
FATP-5 in mice [267], suggesting that specific bile acids can impact hepatic lipid
metabolism. Caveolin proteins consisting out of caveolin-1, -2 and -3, play an
important role in protein trafficking and the formation of lipid droplets. Caveolin-1
knockout mice have lower TAG accumulation in the liver and are resistant to the
development of diet-induced obesity, suggesting that this protein may be of
importance in liver fat accumulation and TAG synthesis [268]. Furthermore, rodent
in vivo and in vitro data show that overexpression of caveolin-3 in liver resulted in
an improved insulin receptor signaling, insulin sensitivity and glucose metabolism
[269]. Normally, CD36 is not highly expressed in liver, but its expression was
positively correlated with hepatic TAG content in NAFLD patients, underscoring the
importance of this protein in lipid accumulation [270]. Moreover, CD36 deficiency
increased insulin sensitivity in muscle, but resulted in an insulin resistant state in
the liver of mice [271]. It is evident that further research is required to elucidate the
role of these FA transporters in normal physiology as well as pathological
conditions.
Hepatic lipolysis
Rodent in vivo and in vitro data showed that hepatic ATGL (also known as PNPLA2
and desnutrin) knockdown enhances glucose tolerance by increasing hepatic
glucose utilization and improved insulin action from hepatic TAG accumulation
[272]. Furthermore, HSL-knockout mice also showed reduced hepatic TAG stores
and increased hepatic insulin sensitivity [273], but results are not consistent [274].
In adipose tissue, ATGL and HSL are highly expressed and active [275], but in the
liver, the contribution of other lipases may also play a role in the catabolism of
stored lipids. Several members of the carboxylesterase/lipase family and the
patatin-like phospholipase domain-containing protein (PNPLA) family have been
suggested as potential TAG hydrolases [276]. One of them, carboxyl esterase-
3/triglyceride hydrolase-1 (Ces-3/Tgh-1, ortholog of human Ces-1), has gained
                                                                                      55
Chapter 2
56
                                                                                             Chapter 2
When circulating levels of FFA and lipoprotein remnants are increased, uptake and storage (1) of TAG
and lipid metabolites in hepatocytes occur via increased LPL, HL activity and number and activity of
transporter proteins (such as FATP2-5, FABP4) leading to hepatic insulin resistance. Increased insulin-
stimulated de novo lipogenesis (2) contributes to hepatic lipid accumulation. In addition, under insulin
resistant conditions hepatic mitochondrial function is decreased (3) and may inhibit FA oxidation,
contributing to hepatic lipid accumulation, increased VLDL production (4) and an inflammatory state (5).
During conditions of hepatic lipid accumulation, an overproduction of VLDL occurs and together with an
increased de novo lipogenesis results in hypertriglyceridemia, a condition often seen in insulin
resistance and T2D. Finally, circulating bile acid composition (6) is altered in insulin resistant and T2D
patients and might affect liver lipid metabolism via membrane (e.g. TGR5) and nuclear (e.g. RXR)
receptor signaling. Interestingly, plant sterols have been shown to be effective nuclear receptor
activators. Abbreviations: FFA: Free fatty acids; TAG: Triacylglycerol; DAG: Diacylglycerol; LPL:
Lipoprotein lipase; HL: Hepatic lipase; FATP2-5: Fatty acid transport proteins 2-5; FABP4: Fatty acid
binding protein 4; VLDL: Very-low-density lipoprotein; TGR5: G-protein coupled receptor TGR5; FXR:
Farnesoid X receptor; LXR: Liver X receptor; RXR: Retinoid X receptor.
Solid lines indicate stimulation. Green lines indicate beneficial effects. Symbols: (): Altered; ():
Increased; (): Decreased
                                                                                                      57
Chapter 2
De novo lipogenesis
On the other hand, increased insulin concentrations stimulate de novo lipogenesis
(DNL), a process that is characterized by converting carbohydrates towards FAs
and generating “new lipids” [294, 295]. The overproduction of VLDL together with
an increased DNL, leads to hypertriglyceridemia, a condition often seen in insulin
resistance and T2D [245]. During DNL, the conversion of glucose to fatty acids
includes a coordinated series of enzymatic reactions of which fatty acid synthase
(FAS) is the key rate-limiting enzyme that regulates the conversion of malonyl-CoA
into palmitate, which is, thereafter, converted into complex fatty acids [296].
Knockout of FAS has been shown in mice to provoke a fatty liver phenotype upon
high carbohydrate feeding, perhaps due to an increase in hepatic malonyl-CoA
[297]. Another key enzyme is stearoyl-CoA desaturase 1 (SCD1), which plays a
role in the desaturation of fatty acids and is predominantly expressed in liver.
Depletion of SCD1 in mice showed a decreased lipogenesis and an increased β-
oxidation [298]. During DNL, the final step of TAG synthesis is catalyzed by
diacylglyceroltransferase (DGAT), which consists of DGAT1 and DGAT2. DGAT1
deficient mice are resistant to diet-induced obesity [299], have decreased levels of
tissue TAG and are more insulin and leptin sensitive [300]. Furthermore, glucose
and insulin regulate the expression of lipogenic enzymes via carbohydrate
responsive element binding protein (ChREBP) [301] and sterol regulatory element
binding protein-1 (SREBP1c) [302, 303], respectively. Both SREBP1c and
ChREBP are involved in the transcriptional regulation of lipogenic genes and have
been associated with increased DNL in NAFLD [304]. DNL is a highly regulated
process and can lead to adverse metabolic consequences when dysregulated.
Therapeutic targeting of this pathway may open a new window of opportunity for
combating various lipogenesis-driven pathological conditions like obesity and
insulin resistance.
58
                                                                             Chapter 2
FXR-receptor
The FXR-receptor is highly expressed in the liver, intestine, kidney and adrenal
gland [321, 323, 324, 328] and is also shown to play a role in liver regeneration,
inflammation and tumorgenesis [321, 324]. Both conjugated and unconjugated BAs
can activate FXR and the order of potency is CDCA > LCA = DCA > CA. CA
feeding increased hepatic expression of ApoC-2, a LPL activator, specifically
                                                   -/-
through FXR as this effect was not observed in FXR mice [329]. In addition, FXR
activation increased the expression of VLDL receptor [330] and syndecan-1 [331],
which are responsible for increased clearance of TAG-rich lipoprotein and remnant
particles, respectively. Furthermore, BA sequestrants, such as cholestyramine,
                                                                                     59
Chapter 2
were found to increase plasma TAG, mediated at least in part, through FXR [332],
which increased plasma lipoprotein clearance. However, other mechanisms may
also be involved in FXR-mediated lipid lowering effects. For example, PPAR-α was
induced by CDCA and GW4064 (a synthetic FXR agonist) treatment in HepG2
cells and primary hepatocytes [333]. GW4064 treatment also increased the mRNA
expression of PDK4, a PPAR-α target gene involved in the regulation of substrate
metabolism, in both rat hepatoma cells, human primary hepatocytes and also
resulted in a reduced plasma TAG concentration in vivo in mice [334]. BA or
GW4064 induced activation of FXR also increased the expression and secretion of
FGF21 [335], a cytokine modulating systematic carbohydrate and lipid metabolism
and reducing hepatic TAG levels [336-338]. FGF21 has been reported to inhibit
lipogenesis through suppressing the transcriptional activity of SREBP-1c [339] and
therefore, the FGF21 pathway may play an important role in FXR-mediated
decrease in hepatic TAG levels, which may be of importance in decreasing hepatic
insulin resistance [340, 341]. Besides the nuclear receptor FXR, the liver X
receptor (LXR-α and LXR-β) and retinoid x receptor (RXR) play an important role in
regulating carbohydrate and lipid metabolism in humans [342, 343]. Interestingly,
sitosterol, campesterol and certain oxidized derivatives of phytosterols
(oxyphytosterols) are effective LXR activators [344]. A rodent study showed that
LXR-α and LXR-α/β knockout mice remained glucose tolerant and insulin sensitive,
while LXR-β knockout mice became highly insulin resistant after a high-fat diet
[345]. Treatment of lean and ob/ob mice with the pharmacological LXR activator
(GW3695) resulted in lower blood glucose levels and significantly improved whole
body insulin sensitivity in the ob/ob mice, but no changes were found in the lean
mice [346]. Furthermore, the synthetic LXR-α/β activator (T0901317) augmented
diet-induced hyperlipidemia, normalized glucose tolerance and improved insulin-
stimulated glucose uptake in isolated soleus muscle and completely restored
glucose transporter 4 (GLUT4) expression and insulin-stimulated AS160
phosphorylation in rat muscle [347]. Although the vast majority of studies have
been performed in non-adipose cells/tissues, results in recent years suggest that
LXRs may have important modulatory roles on adipose lipid and glucose
metabolism [348].
In addition, PPAR-γ and RXR agonists have complementary effects on glucose
and lipid metabolism in human skeletal muscle [349], as well as in diabetic and
obese rodent models [350]. It has also been shown that a RXR ligand (LG100754)
improved insulin resistance in vivo in db/db mice [351] and a recent study showed
that a novel RXR partial agonist (CBt-PMN (11b)) has a glucose-lowering effect
                                                                            y
and improved insulin secretion and glucose tolerance in the liver of KK-A mice
[352].
TGR5-receptor
TGR5 is a family member of the G-protein coupled receptors (also known as
GPR19) and is highly expressed in gallbladder, ileum and colon and in lower
concentrations in BAT, liver, muscle and the central nervous system [336]. The
activation of TGR5 is highest with the bile acid LCA, followed by DCA, CDCA and
CA [323, 336] and results in the activation of PKA [336]. Furthermore, TGR5
activation regulates the expression of genes involved in inflammation [353],
60
                                                                              Chapter 2
increases energy expenditure in skeletal muscle and BAT [354] through stimulation
of mitochondrial function [355-357] and modulates plasma glucose and lipid
concentrations [336, 355, 356, 358]. BA mediated activation of TGR5 has
beneficial metabolic effects and the BA composition is altered in patients with
insulin resistance [359] and T2D [360]. Since gram-positive bacteria have a more
pronounced effect on the transformation of primary to secondary BAs [361, 362],
compared to most of the gram-negative bacteria [363], an alteration in the gut
microbiota might have a distinct effect on bile acid metabolism and might be an
effective strategy to improve insulin sensitivity. Indeed, in the study of Vrieze et al.,
a vancomycin-induced decrease in gram-positive bacteria was associated with a
reduced conversion of primary to secondary BAs and a tendency towards a
reduced peripheral insulin sensitivity [364]. Also, transgenic liver overexpression of
cholesterol 7α-hydroxylase, protected mice against high-fat diet induced obesity,
fatty liver and insulin resistance [365]. Furthermore, glucose metabolism can also
be improved in diabetic patients by administration of BA sequestrants, such as
colesevelam [366, 367]. Thus, a promising nutritional strategy may be to alter the
gut microbiota by modifying the diet by either prebiotics or probiotics and
modulating thereby BA metabolism [368, 369].
Hepatic inflammation
Since insulin resistance and obesity are characterized by a low-grade inflammatory
status [370], inflammation induced by the increased lipid accumulation, could also
be an underlying cause for the development of hepatic insulin resistance. Of
interest, ER stress has been shown to be involved in the development of hepatic
inflammation and insulin resistance. First, ER stress can directly modify key hepatic
enzymes involved in gluconeogenesis and lipogenesis, and stimulate stress
kinases that interfere with insulin signaling. Secondly, ER stress may indirectly
induce inflammation and lipotoxicity by promoting fat accumulation in hepatocytes
[179, 371, 372].
Furthermore, hepatic peroxisomal FA oxidation causes an increased concentration
of reactive oxygen species [373], which in excess cause a decrease in the natural
antioxidant concentrations, leading to oxidative stress in hepatocytes [374, 375]. In
turn, this oxidative stress causes hepatocyte degeneration and death [374] evoking
an inflammatory response [376]. Specialized liver macrophages, called Kupffer
cells, play a central role in this inflammatory process since in vitro stimulation of
these cells by endotoxin (e.g. lipopolysaccharides (LPS)) or specific FAs (e.g. SFA)
and their metabolites, lead to toll-like receptor signal transduction and the
production of inflammatory cytokines, including TNF-α and IL-6 [377].
Furthermore, in analogy to the adipokines, the liver-derived proteins (known as
hepatokines) [378] such as leukocyte cell-derived chemotaxin 2 (LECT2) regulate
cross talk with other tissues and link obesity with skeletal muscle insulin resistance
[379].
Taken together, the oversupply of lipids to the liver, together with impaired
clearance of circulating remnants, increased de novo lipogenesis and decreased
FA oxidation contribute to lipid accumulation in the liver, referred to as hepatic
steatosis, which can progress to NAFLD, NASH and liver cancer [380].
                                                                                      61
Chapter 2
62
                                                                             Chapter 2
                                                                                     63
Chapter 2
64
                                                                            Chapter 2
                                                                                    65
Chapter 2
66
                                                                                             Chapter 2
Skeletal muscle fatty acid uptake, which is regulated via LPL and fatty acid transport proteins (FATP1-6,
CD36), might be higher in subjects with impaired glucose metabolism compared to control subjects (1).
In addition, a reduced mitochondrial mass and/or mitochondrial function (2) have been proposed as
underlying mechanisms for reduced muscle fat oxidation, contributing to the accumulation of TAG and
lipid-intermediates (LCFA-CoA, DAG, ceramides, acylcarnitines) in myocytes (3), interfering with insulin
signaling (4) in the obese insulin resistant state. Finally, the capacity to increase intramyocellular fat
oxidation during conditions of high FA supply has shown to be impaired in obese, T2D subjects.
PUFA’s, polyphenols, plant sterols and bile acids may have beneficial effects on skeletal muscle lipid
metabolism.
Abbreviations: LPL: Lipoprotein lipase; FATP1-6: Fatty acid transport protein 1-6; CD36: Fatty acid
translocase CD36; TAG: Triacylglycerol; DAG: Diacylglycerol; LCFA-CoA: long chain fatty acyl co-
enzyme A; PUFA: Poly unsaturated fatty acids. Dashed lines indicate inhibition. Solid lines indicate
stimulation. Green lines indicate beneficial effects. Symbols: (): Increased; (): Decreased
                                                                                                      67
Chapter 2
68
                                                                              Chapter 2
                                                                                      69
Chapter 2
obese insulin resistance state. Since there is increasing support for the notion that
lipid overflow in insulin resistance under physiological conditions is rather
characterized by increased circulating TAG concentrations than by increased FFA
concentrations, it is a plausible option that increased TAG-derived FA oxidation
may impair IMTAG utilization in the obese insulin resistant state. However, this
option remains to be investigated.
Furthermore, differences in IMTAG oxidation may be caused by differences in
IMTAG lipolysis. Interestingly, lipolysis from IMTAG might be necessary for the
maintenance of oxidative gene expression and FA oxidation. Recent findings
indicate that in particular ATGL-mediated lipolysis generates lipid ligands for PPAR
activation and the subsequent transcription of oxidative genes [90]. It was shown
that a cycle of FA esterification and rehydrolysis is required for activation of PPAR-
α and normal mitochondrial and oxidative phosphorylation, at leasat in
cardiomyocytes [467]. In vivo tracer data show that in patients with NLSDM
(caused by mutations in the ATGL gene) fat oxidative capacity is blunted and can
be partly reversed by PPAR-α treatment [468, 469]. These data highlight the
importance of ATGL activity in regulating muscle fat oxidation. Furthermore, it was
shown recently that activation of NP signaling in human skeletal muscle enhances
mitochondrial oxidative metabolism and fat oxidation [470]. However, further
studies are required linking lipolysis and NP signaling to muscle oxidative
metabolism and insulin resistance in human.
Finally, a reduced FA transport across the mitochondrial membrane and a reduced
mitochondrial mass and/or mitochondrial function have been proposed as
underlying causes of a reduced muscle fat oxidation in insulin resistance. In rodent
models of obesity and insulin resistance an increased content of malonyl-CoA, an
allosteric inhibitor of mitochondrial FA transport, has been shown in skeletal muscle
in combination with hyperglycaemia and hyperinsulinaemia and a reduced lipid
oxidation [471]. Also, in human skeletal muscle it has been shown that a
combination of hyperglycaemia and hyperinsulinaemia increases malonyl-CoA,
inhibits functional carnitine palmitoyl transferase 1 (CPT1b, muscle isoform) activity
and shunts long chain FA away from oxidation and towards storage in human
muscle [472]. Furthermore, two studies have shown that an improvement in fat
oxidation by exercise or by lifestyle intervention was accompanied by decreased
acetyl-CoA carboxylase (ACC) mRNA expression, involved in the formation of
malonyl-CoA, suggesting that a reduced inhibition of mitochondrial FA transport
through malonyl-CoA may have contributed to the improved fat oxidative capacity
[473, 474]. Furthermore, a lowered mitochondrial transport through CPT1 may
contribute to the reduced fat oxidation. In line, heterozygous CPT1b deficiency
results in mitochondrial abnormalities and lipid accumulation with elevated TAG
and ceramide content at least in mice cardiomyocytes [475]. Moreover, it has been
shown that an increased CPT1b activity is sufficient to improve high-fat diet
induced insulin resistance [476]. Peroxisome proliferator-activated receptor gamma
coactivator-1alpha (PGC-1α) is involved in mitochondrial biogenesis by supporting
the transcriptional activity of nuclear respiration factor 1 (NRF1), thereby regulating
the transcription of genes involved in oxidative metabolism. The expression of
genes responsible for oxidative phosphorylation is coordinately downregulated in
muscle of T2D individuals [477, 478] and muscle specific overexpression of PGC-
1α in rodents has been shown to improve lipid utilization, insulin signaling and
70
                                                                          Chapter 2
glucose transport [479]. In vivo mitochondrial capacity and function have been
reported to be reduced in T2D, independent of mitochondrial content [388, 480,
481], although data are not consistent [482, 483]. Nevertheless, a reduced
mitochondrial capacity and a reduced activity of TCA cycle [484] and citrate
synthase (-14%) [485] have also been reported in primary myotubes from T2D
subjects, suggesting that skeletal muscle mitochondrial function is intrinsically
impaired in insulin resistance. Moreover, a marked reduction in mitochondrial
oxidative and phosphorylation activities, together with intramyocellular lipid
accumulation, were found in skeletal muscle in insulin resistant offspring of T2D
subjects [486, 487]. Furthermore, the definition of mitochondrial dysfunction may
also include a dysbalance balance between β-oxidation and TCA cycle activity
resulting in an accumulation of incompletely oxidized, fatty acid products (i.e.
acylcarnitines), which can impair skeletal muscle insulin sensitivity, as extensively
reviewed elsewhere [488].
In summary, skeletal muscle fat oxidation is reduced in the obese insulin resistant
state. This impaired fat oxidation may contribute to the accumulation of lipid
metabolites and may be driven by lipid supply from either extracellular or
intracellular sources as well as intrinsic impairments in mitochondrial function.
However, whether reduced mitochondrial function is causally related to insulin
resistance or rather a consequence of the sedentary lifestyle needs to be
investigated in more detail in future research.
                                                                                  71
Chapter 2
                                             .
metabolism and insulin sensitivity [218, 495] Indeed, we recently showed that a
PUFA rich meal acutely improved insulin sensitivity in obese insulin resistant
subjects as compared to a high-SFA meal, which was accompanied by a reduced
TAG-derived skeletal muscle FA uptake, an altered intramyocellular lipid
partitioning and a more oxidative transcriptional phenotype [496]. Additionally,
studies in human myotubes showed that palmitate induced a lower lipolysis rate
[497] and follows a different metabolic pathway as compared to oleic acid
[497,498]. Also, a lower palmitate oxidation has been shown in myotubes of T2D
subjects as compared to control, whilst oleate oxidation was not significantly
different [499]. These studies indicate that dietary fat quality may modulate the
nature of lipid-induced insulin resistance through affecting pathways of fatty acid
partitioning. Further studies are required to investigate the impact of gender and
dietary fat quality on FA dynamics and to investigate the causative role of an
impaired TAG synthesis capacity in the etiology of insulin resistance. In summary,
polyphenols and modulation of dietary fat quality may affect storage of bioactive
lipid metabolites in skeletal muscle, thereby improving insulin sensitivity and
glucose homeostasis. Beside that, manipulation the nuclear FXR and LXR
receptors by modulating bile acid metabolism or by ingestion of plant sterols may
also have positive effects on muscle lipid metabolism and insulin sensitivity, but
apparently more human evidence is required. For a summary see Table 1.
72
     Tabel 1: Summary of pathways, possible targets and (dietary) components in adipose tissue, liver and gut and skeletal muscle
Adipose tissue Fatty acid trapping Stimulation of LPL Dietary modulation of ANGPTL4 [63] [425] Rodent - in vivo/ex vivo
                                                  Selective HSL and/or ATGL         Pre- and probiotics affecting SCFA metabolism                         Rodent - in vitro/in vivo
                             Lipolysis                                                                                                  [100, 101]
                                                           inhibition                                 [210], [211]                                        Human - in vitro/in vivo
                                                                                                                                                             Rodent - in vivo
                       Bile acid manipulation             Activate LXR                           Plant sterols [344]                    [346, 347]
                                                                                                                                                          Rodent - in vivo/ex vivo
TAG extraction Reduce TAG-derived- FA uptake PUFA [496] [496] Human - in vivo
                                                                                                                                                             Rodent - in vivo
                          Gut microbiota                                                    Plant sterols, Pre-, probiotics
                                                      Activate LXR, TGR5                                                              [346, 347] [356]    Rodent - in vivo/ex vivo
                           manipulation                                                              [344] [369]
                                                                                                                                                             Rodent - in vivo
                                                                                                                                                                                      Chapter 2
73
Chapter 2
74
                                                                                            Chapter 2
Lipid accumulation in pancreatic islets can result in inflammation, ER-stress, mitochondrial overload and
lipotoxicity. These mechanisms damage the β-cell function and lower β-cell mass, resulting in a
decreased insulin secretion and ultimately leading to the development of T2D. Polyphenols, vitamins
and carotenoids may counteract these negative effects.
Abbreviations: TAG: Triacylglycerol; DAG: Diacylglycerol; MAG: monoacylglycerol; FFA: Free fatty
acids; ER-stress: Endoplasmic reticulum stress. Dashed lines indicate inhibition. Solid lines indicate
stimulation. Green lines indicate beneficial effects. Symbols: (): Increased; (): Decreased
                                                                                                     75
Chapter 2
76
                                                                           Chapter 2
respectively [569-572], although conflicting results have also been reported [570,
573-575]. Interestingly, germ-free mice infected with the gut microbiota of
conventionally raised mice demonstrated increased body weight, insulin resistance
and glucose intolerance together with reduced food intake and increased oxygen
consumption [576]. The body fat gain was related to suppressed gut expression of
the LPL inhibitor ANGPTL4, leading to increased fat accumulation in adipocytes
and increased energy harvest [570, 577-579].
In obese men, seven days treatment with the antibiotic vancomycin modulated their
gut microbiota and decreased peripheral insulin sensitivity [364]. Finally, the
transfer of intestinal microbiota from human lean donors to individuals with the
metabolic syndrome increased insulin sensitivity, along with increase of intestinal
butyrate-producing microbiota [580].
Several mechanisms link our gut microbiota to obesity and diabetes (diabesity), as
extensively reviewed elsewhere [581]. Firstly, alterations in gut microbiota
composition can increase the amount of LPS through increased production,
reduced breakdown or increased translocation across the gut wall, thereby
inducing a strong immune response to protect the organism from bacterial infection
[582]. This LPS-induced inflammatory state is referred to as ‘metabolic
endotoxemia’ and is accompanied by body weight gain and insulin resistance in
animal models [583]. In addition, major microbial products from the fermentation of
indigestible carbohydrates, like the SCFAs acetate, butyrate and propionate may
affect energy and fat metabolism through various mechanisms [584-587]. The type
of substrate is the primary determinant of the SCFA production rate and SCFA
ratio, compared to the composition of the intestinal microbiota [584] and the level of
substrate fermentability varies with fiber solubility. Soluble fibers are fermented
more completely than insoluble fibers [588] and highly fermentable substrates
produce higher amounts of SCFA [589], which have physiological effects in
different tissues [590]. At first, it has been proposed that they may play a role in
harvesting extra energy from the diet, which would be a promoting factor for body
weight gain. However, evidence that this largely contributes to human energy
balance is not yet convincing [591]. On the other hand, increased SCFA
concentrations have rather been associated with distinct generally positive,
metabolic effects, affecting hormonal release of glucagon-like peptide 1 (GLP1)
and peptide YY (PYY), cell proliferation and differentiation [584, 592-595], regulate
adipocyte lipolysis [210, 211], modulate inflammation, affecting adipose tissue fat
storage and ectopic fat storage, as has been extensively reviewed elsewhere
[596]. Effects of SCFAs may be related to the activation of their related G-protein
coupled receptors GPR41 (also known as free fatty acid receptor 3 (FFAR3)) and
GPR43 (FFAR2) [596], which are present in the gut epithelium but also in
peripheral tissues like adipose tissue and skeletal muscle [595, 597]. Thus,
manipulation of SCFA production by prebiotics and probiotics (or a combination of
both) may, among other processes, differentially affect human fat and glucose
metabolism.
Furthermore, besides converting non-digestible fibers, the microbiota are also
closely linked to BA metabolism. On the one hand, microbiota can transform
primary to the so-called secondary BAs [598], while on the other hand BAs by
themselves exert an anti-microbial effect on the microbiota [599, 600]. As
discussed above, the secondary BAs can affect host metabolism via binding to
                                                                                   77
Chapter 2
several nuclear receptors and GPRs and altering the intestinal microbiota could be
a potential therapeutic target to reduce obesity and insulin resistance.
78
                                                                                            Chapter 2
An altered gut microbiota composition, as seen in obesity and T2D, may increase local and systemic
lipopolysaccharide (LPS) concentrations, referred to as metabolic endotoxemia (1) that can lead to
systemic inflammation. Besides LPS, also bile acid composition and SCFA concentrations are altered.
Pre- and probiotics, plant sterols and PUFAs exert a beneficial effect on gut microbiota composition (2),
leading to an altered SCFA production, bile acid composition and a decreased LPS concentration,
thereby affecting gut inflammation (3) and gut lipid metabolism (4). In addition, changes in systemic
SCFA, bile acids and LPS affect host lipid metabolism and inflammation in adipose tissue, liver and
skeletal muscle (5).
Abbreviations: PUFA: Poly unsaturated fatty acids; SCFA: short chain fatty acid; LPS:
Lipopolysaccharide. Dashed lines indicate inhibition. Solid lines indicate stimulation. Green lines
indicate beneficial effects. Symbols: (): Altered; (): Increased; (): Decreased
                                                                                                     79
Chapter 2
SUMMARY
This review focuses on the disturbances in fatty acid metabolism at both the tissue
and whole-body level that play a role in the etiology of insulin resistance, β-cell
dysfunction and an impaired glucose metabolism. The most important pathways
related to lipid metabolism in adipose tissue, liver, skeletal muscle, pancreas and
gut have been discussed, and targets to improve lipid metabolism and glycaemia,
which may be modulated by diet, have been described. A detailed overview of
pathways and potential targets to improve lipid metabolism and glycaemia in
adipose tissue, liver, skeletal muscle, pancreas and gut, which can be modulated
by dietary interventions and food components, is provided in Table 1, and will be
explained in more detail below.
Within adipose tissue, the balance between fatty acid extraction, lipolysis,
adipocyte differentiation and mitochondrial function is important to maintain
adequate lipid storage capacity. Improving the lipid storage capacity of adipose
tissue prevents lipid overflow in the circulation and subsequent ectopic fat
deposition, and therefore has high potential to improve glucose tolerance and
insulin sensitivity. Increasing adipose tissue fatty acid uptake, possibly via
stimulation of LPL, might reduce lipid overflow. On the other hand, a reduced lipid
overflow might also be achieved by partial inhibition of lipolysis (partial lipase
inhibition) and/or by modulation of the alternative pathway of lipid breakdown
(lipophagy). There is some evidence that partial lipolysis inhibition may be
achieved by modulation of SCFA metabolism through pro- and prebiotics and that
the lipophagic pathway may be modulated by specific polyphenols, like resveratrol.
Furthermore, adipose tissue mitochondrial function seems important in balancing
lipid supply and utilization, and thereby affects adipose tissue function. Improved
adipose tissue mitochondrial function may be achieved by nutritional strategies
such as supplementation with specific polyphenols (or a combination of
polyphenols) and altering dietary fatty acid composition. Dietary fat quality not only
modulates lipid metabolism but may also affect low-grade inflammation, which in
turn may lower the risk of developing insulin resistance and T2D.
An attractive target to tackle obesity and insulin resistance could be manipulation of
the bile acid metabolism and/or gut microbiota composition. On the one hand, bile
acids influence energy expenditure and glucose homeostasis via their effects on
gluconeogenesis, insulin secretion and insulin sensitivity, and an altered
concentrations of secondary bile acids can affect host metabolism via binding to
several nuclear receptors (e.g. FXR, LXR, RXR), some of which can be activated
by sitosterol, campesterol and certain oxidized derivatives of phytosterols
(oxyphytosterols). On the other hand, an alteration of the gut microbiota by
modulation of the dietary fat quality or pre- and probiotics might have a distinct
effect on bile acid composition and metabolism and may also affect fermentation
products from dietary fibers (e.g. SCFA and monosaccharides), affecting thereby
liver gluconeogenesis and lipogenesis, pathways directly or indirectly affecting
glucose homeostasis.
Targeting skeletal muscle lipid turnover and the balance between lipolysis, FA
uptake and mitochondrial function/fat oxidation may constitute an interesting
strategy to improve insulin sensitivity in obesity and T2D by limiting the
accumulation of bioactive lipid-intermediates interfering with insulin sensitivity and
80
                                                                            Chapter 2
                                                                                    81
Chapter 2
REFERENCES
1.    Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease.
      Diabetes 1988; 37: 1595-1607.
2.    Relimpio F. "The relative contributions of insulin resistance and beta-cell dysfunction
      to the pathophysiology of Type 2 diabetes", by Kahn SE. Diabetologia 2003; 46:
      1707.
3.    Bergman RN, Ader M, Huecking K, Van Citters G. Accurate assessment of beta-cell
      function: the hyperbolic correction. Diabetes 2002; 51 Suppl 1: S212-220.
4.    Roumen C, Blaak EE, Corpeleijn E. Lifestyle intervention for prevention of diabetes:
      determinants of success for future implementation. Nutr Rev 2009; 67: 132-146.
5.    Tuomilehto J. Nonpharmacologic therapy and exercise in the prevention of type 2
      diabetes. Diabetes Care 2009; 32 Suppl 2: S189-193.
6.    Mensink M, Blaak EE, Wagenmakers AJ, Saris WH. Lifestyle intervention and fatty
      acid metabolism in glucose-intolerant subjects. Obes Res 2005; 13: 1354-1362.
7.    Goossens GH. The role of adipose tissue dysfunction in the pathogenesis of obesity-
      related insulin resistance. Physiol Behav 2008; 94: 206-218.
8.    Unger RH, Clark GO, Scherer PE, Orci L. Lipid homeostasis, lipotoxicity and the
      metabolic syndrome. Biochim Biophys Acta 2010; 1801: 209-214.
9.    Virtue S, Vidal-Puig A. Adipose tissue expandability, lipotoxicity and the Metabolic
      Syndrome--an allostatic perspective. Biochim Biophys Acta 2010; 1801: 338-349.
10.   Snel M, Jonker JT, Schoones J, et al. Ectopic fat and insulin resistance:
      pathophysiology and effect of diet and lifestyle interventions. Int J Endocrinol 2012;
      2012: 983814.
11.   DeFronzo RA, Jacot E, Jequier E, Maeder E, Wahren J, Felber JP. The effect of
      insulin on the disposal of intravenous glucose. Results from indirect calorimetry and
      hepatic and femoral venous catheterization. Diabetes 1981; 30: 1000-1007.
12.   Perseghin G, Scifo P, De Cobelli F, et al. Intramyocellular triglyceride content is a
      determinant of in vivo insulin resistance in humans: a 1H-13C nuclear magnetic
      resonance spectroscopy assessment in offspring of type 2 diabetic parents. Diabetes
      1999; 48: 1600-1606.
13.   Schrauwen P, Hesselink MK. Oxidative capacity, lipotoxicity, and mitochondrial
      damage in type 2 diabetes. Diabetes 2004; 53: 1412-1417.
14.   Phielix E, Mensink M. Type 2 diabetes mellitus and skeletal muscle metabolic
      function. Physiol Behav 2008; 94: 252-258.
15.   Goodpaster BH, Theriault R, Watkins SC, Kelley DE. Intramuscular lipid content is
      increased in obesity and decreased by weight loss. Metabolism 2000; 49: 467-472.
16.   Krssak M, Falk Petersen K, Dresner A, et al. Intramyocellular lipid concentrations are
      correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study.
      Diabetologia 1999; 42: 113-116.
17.   Pan DA, Lillioja S, Kriketos AD, et al. Skeletal muscle triglyceride levels are inversely
      related to insulin action. Diabetes 1997; 46: 983-988.
18.   Byrne CD, Brindle NP, Wang TW, Hales CN. Interaction of non-esterified fatty acid
      and insulin in control of triacylglycerol secretion by Hep G2 cells. Biochem J 1991;
      280 ( Pt 1): 99-104.
19.   Fanelli C, Calderone S, Epifano L, et al. Demonstration of a critical role for free fatty
      acids in mediating counterregulatory stimulation of gluconeogenesis and suppression
      of glucose utilization in humans. J Clin Invest 1993; 92: 1617-1622.
20.   Ferrannini E, Barrett EJ, Bevilacqua S, DeFronzo RA. Effect of fatty acids on glucose
      production and utilization in man. J Clin Invest 1983; 72: 1737-1747.
21.   Lewis GF, Vranic M, Harley P, Giacca A. Fatty acids mediate the acute extrahepatic
      effects of insulin on hepatic glucose production in humans. Diabetes 1997; 46: 1111-
      1119.
82
                                                                                 Chapter 2
22.   Hennes MM, Dua A, Kissebah AH. Effects of free fatty acids and glucose on
      splanchnic insulin dynamics. Diabetes 1997; 46: 57-62.
23.   Svedberg J, Bjorntorp P, Smith U, Lonnroth P. Free-fatty acid inhibition of insulin
      binding, degradation, and action in isolated rat hepatocytes. Diabetes 1990; 39: 570-
      574.
24.   Wiesenthal SR, Sandhu H, McCall RH, et al. Free fatty acids impair hepatic insulin
      extraction in vivo. Diabetes 1999; 48: 766-774.
25.   Frayn KN, Williams CM, Arner P. Are increased plasma non-esterified fatty acid
      concentrations a risk marker for coronary heart disease and other chronic diseases?
      Clin Sci (Lond) 1996; 90: 243-253.
26.   Tikellis C, Wookey PJ, Candido R, Andrikopoulos S, Thomas MC, Cooper ME.
      Improved islet morphology after blockade of the renin- angiotensin system in the ZDF
      rat. Diabetes 2004; 53: 989-997.
27.   Cani PD, Delzenne NM. Interplay between obesity and associated metabolic
      disorders: new insights into the gut microbiota. Curr Opin Pharmacol 2009; 9: 737-
      743.
28.   Delzenne NM, Cani PD. Gut microbiota and the pathogenesis of insulin resistance.
      Curr Diab Rep 2011; 11: 154-159.
29.   Delzenne NM, Neyrinck AM, Cani PD. Modulation of the gut microbiota by nutrients
      with prebiotic properties: consequences for host health in the context of obesity and
      metabolic syndrome. Microb Cell Fact 2011; 10 Suppl 1: S10.
30.   Iacobellis G, Leonetti F. Epicardial adipose tissue and insulin resistance in obese
      subjects. J Clin Endocrinol Metab 2005; 90: 6300-6302.
31.   Wang CP, Hsu HL, Hung WC, et al. Increased epicardial adipose tissue (EAT)
      volume in type 2 diabetes mellitus and association with metabolic syndrome and
      severity of coronary atherosclerosis. Clin Endocrinol (Oxf) 2009; 70: 876-882.
32.   Hammer S, van der Meer RW, Lamb HJ, et al. Short-term flexibility of myocardial
      triglycerides and diastolic function in patients with type 2 diabetes mellitus. Am J
      Physiol Endocrinol Metab 2008; 295: E714-718.
33.   Kim MK, Tanaka K, Kim MJ, et al. Comparison of epicardial, abdominal and regional
      fat compartments in response to weight loss. Nutr Metab Cardiovasc Dis 2009; 19:
      760-766.
34.   Iacobellis G, Singh N, Wharton S, Sharma AM. Substantial changes in epicardial fat
      thickness after weight loss in severely obese subjects. Obesity (Silver Spring) 2008;
      16: 1693-1697.
35.   Hammer S, van der Meer RW, Lamb HJ, et al. Progressive caloric restriction induces
      dose-dependent changes in myocardial triglyceride content and diastolic function in
      healthy men. J Clin Endocrinol Metab 2008; 93: 497-503.
36.   de Vries AP, Ruggenenti P, Ruan XZ, et al. Fatty kidney: emerging role of ectopic
      lipid in obesity-related renal disease. Lancet Diabetes Endocrinol 2014; 2: 417-426.
37.   Guebre-Egziabher F, Alix PM, Koppe L, et al. Ectopic lipid accumulation: A potential
      cause for metabolic disturbances and a contributor to the alteration of kidney
      function. Biochimie 2013; 95: 1971-1979.
38.   Wei J, Ferron M, Clarke CJ, et al. Bone-specific insulin resistance disrupts whole-
      body glucose homeostasis via decreased osteocalcin activation. J Clin Invest 2014;
      124: 1-13.
39.   Veldhuis-Vlug AG, Fliers E, Bisschop PH. Bone as a regulator of glucose
      metabolism. Neth J Med 2013; 71: 396-400.
40.   Gimble JM, Nuttall ME. The relationship between adipose tissue and bone
      metabolism. Clin Biochem 2012; 45: 874-879.
41.   Frayn KN. Adipose tissue as a buffer for daily lipid flux. Diabetologia 2002; 45: 1201-
      1210.
                                                                                          83
Chapter 2
42.   Evans K, Burdge GC, Wootton SA, Clark ML, Frayn KN. Regulation of dietary fatty
      acid entrapment in subcutaneous adipose tissue and skeletal muscle. Diabetes 2002;
      51: 2684-2690.
43.   Frayn KN, Coppack SW, Fielding BA, Humphreys SM. Coordinated regulation of
      hormone-sensitive lipase and lipoprotein lipase in human adipose tissue in vivo:
      implications for the control of fat storage and fat mobilization. Adv Enzyme Regul
      1995; 35: 163-178.
44.   Yost TJ, Jensen DR, Haugen BR, Eckel RH. Effect of dietary macronutrient
      composition on tissue-specific lipoprotein lipase activity and insulin action in normal-
      weight subjects. Am J Clin Nutr 1998; 68: 296-302.
45.   Brunzell JD, Schwartz RS, Eckel RH, Goldberg AP. Insulin and adipose tissue
      lipoprotein lipase activity in humans. Int J Obes 1981; 5: 685-694.
46.   Goldberg IJ, Eckel RH, Abumrad NA. Regulation of fatty acid uptake into tissues:
      lipoprotein lipase- and CD36-mediated pathways. J Lipid Res 2009; 50 Suppl: S86-
      90.
47.   Ong JM, Kern PA. Effect of feeding and obesity on lipoprotein lipase activity,
      immunoreactive protein, and messenger RNA levels in human adipose tissue. J Clin
      Invest 1989; 84: 305-311.
48.   Olivecrona T, Bergo M, Hultin M, Olivecrona G. Nutritional regulation of lipoprotein
      lipase. Can J Cardiol 1995; 11 Suppl G: 73G-78G.
49.   Ong JM, Simsolo RB, Saffari B, Kern PA. The regulation of lipoprotein lipase gene
      expression by dexamethasone in isolated rat adipocytes. Endocrinology 1992; 130:
      2310-2316.
50.   Koliwad SK, Kuo T, Shipp LE, et al. Angiopoietin-like 4 (ANGPTL4, fasting-induced
      adipose factor) is a direct glucocorticoid receptor target and participates in
      glucocorticoid-regulated triglyceride metabolism. J Biol Chem 2009; 284: 25593-
      25601.
51.   Fried SK, Russell CD, Grauso NL, Brolin RE. Lipoprotein lipase regulation by insulin
      and glucocorticoid in subcutaneous and omental adipose tissues of obese women
      and men. J Clin Invest 1993; 92: 2191-2198.
52.   Ottosson M, Marin P, Karason K, Elander A, Bjorntorp P. Blockade of the
      glucocorticoid receptor with RU 486: effects in vitro and in vivo on human adipose
      tissue lipoprotein lipase activity. Obes Res 1995; 3: 233-240.
53.   Kim SJ, Nian C, McIntosh CH. GIP increases human adipocyte LPL expression
      through CREB and TORC2-mediated trans-activation of the LPL gene. J Lipid Res
      2010; 51: 3145-3157.
54.   Kim SJ, Nian C, McIntosh CH. Resistin knockout mice exhibit impaired adipocyte
      glucose-dependent insulinotropic polypeptide receptor (GIPR) expression. Diabetes
      2013; 62: 471-477.
55.   Beigneux AP, Davies BS, Gin P, et al. Glycosylphosphatidylinositol-anchored high-
      density lipoprotein-binding protein 1 plays a critical role in the lipolytic processing of
      chylomicrons. Cell Metab 2007; 5: 279-291.
56.   Davies BS, Beigneux AP, Barnes RH, 2nd, et al. GPIHBP1 is responsible for the
      entry of lipoprotein lipase into capillaries. Cell Metab 2010; 12: 42-52.
57.   Young SG, Davies BS, Voss CV, et al. GPIHBP1, an endothelial cell transporter for
      lipoprotein lipase. J Lipid Res 2011; 52: 1869-1884.
58.   Davies BS, Waki H, Beigneux AP, et al. The expression of GPIHBP1, an endothelial
      cell binding site for lipoprotein lipase and chylomicrons, is induced by peroxisome
      proliferator-activated receptor-gamma. Mol Endocrinol 2008; 22: 2496-2504.
59.   Pei-Ling Chiu A, Wang F, Lal N, et al. Endothelial cells respond to hyperglycemia by
      increasing the LPL transporter GPIHBP1. Am J Physiol Endocrinol Metab 2014; 306:
      E1274-1283.
84
                                                                                       Chapter 2
60.   Adeyo O, Goulbourne CN, Bensadoun A, Beigneux AP, Fong LG, Young SG.
      Glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 and
      the intravascular processing of triglyceride-rich lipoproteins. J Intern Med 2012; 272:
      528-540.
61.   Weinstein MM, Goulbourne CN, Davies BS, et al. Reciprocal metabolic perturbations
      in the adipose tissue and liver of GPIHBP1-deficient mice. Arterioscler Thromb Vasc
      Biol 2012; 32: 230-235.
62.   Kersten S. Physiological regulation of lipoprotein lipase. Biochim Biophys Acta 2014;
      1841: 919-933.
63.   Jonker JT, Smit JW, Hammer S, et al. Dietary modulation of plasma angiopoietin-like
      protein 4 concentrations in healthy volunteers and in patients with type 2 diabetes.
      Am J Clin Nutr 2013; 97: 255-260.
64.   Brands M, Sauerwein HP, Ackermans MT, Kersten S, Serlie MJ. Omega-3 long-chain
      fatty acids strongly induce angiopoietin-like 4 in humans. J Lipid Res 2013; 54: 615-
      621.
65.   Amri EZ, Teboul L, Vannier C, Grimaldi PA, Ailhaud G. Fatty acids regulate the
      expression of lipoprotein lipase gene and activity in preadipose and adipose cells.
      Biochem J 1996; 314 ( Pt 2): 541-546.
66.   Nagashima K, Lopez C, Donovan D, et al. Effects of the PPARgamma agonist
      pioglitazone on lipoprotein metabolism in patients with type 2 diabetes mellitus. J Clin
      Invest 2005; 115: 1323-1332.
67.   Sadur CN, Yost TJ, Eckel RH. Insulin responsiveness of adipose tissue lipoprotein
      lipase is delayed but preserved in obesity. J Clin Endocrinol Metab 1984; 59: 1176-
      1182.
68.   Riemens SC, Sluiter WJ, Dullaart RP. Enhanced escape of non-esterified fatty acids
      from tissue uptake: its role in impaired insulin-induced lowering of total rate of
      appearance in obesity and Type II diabetes mellitus. Diabetologia 2000; 43: 416-426.
69.   McQuaid SE, Hodson L, Neville MJ, et al. Downregulation of adipose tissue fatty acid
      trafficking in obesity: a driver for ectopic fat deposition? Diabetes 2011; 60: 47-55.
70.   Panarotto D, Remillard P, Bouffard L, Maheux P. Insulin resistance affects the
      regulation of lipoprotein lipase in the postprandial period and in an adipose tissue-
      specific manner. Eur J Clin Invest 2002; 32: 84-92.
71.   Coppack SW, Evans RD, Fisher RM, et al. Adipose tissue metabolism in obesity:
      lipase action in vivo before and after a mixed meal. Metabolism 1992; 41: 264-272.
72.   Potts JL, Coppack SW, Fisher RM, Humphreys SM, Gibbons GF, Frayn KN. Impaired
      postprandial clearance of triacylglycerol-rich lipoproteins in adipose tissue in obese
      subjects. Am J Physiol 1995; 268: E588-594.
73.   Annuzzi G, Giacco R, Patti L, et al. Postprandial chylomicrons and adipose tissue
      lipoprotein lipase are altered in type 2 diabetes independently of obesity and whole-
      body insulin resistance. Nutr Metab Cardiovasc Dis 2008; 18: 531-538.
74.   Frayn KN, Shadid S, Hamlani R, et al. Regulation of fatty acid movement in human
      adipose tissue in the postabsorptive-to-postprandial transition. Am J Physiol 1994;
      266: E308-317.
75.   Ruge T, Hodson L, Cheeseman J, et al. Fasted to fed trafficking of Fatty acids in
      human adipose tissue reveals a novel regulatory step for enhanced fat storage. J Clin
      Endocrinol Metab 2009; 94: 1781-1788.
76.   Brasaemle DL. Thematic review series: adipocyte biology. The perilipin family of
      structural lipid droplet proteins: stabilization of lipid droplets and control of lipolysis. J
      Lipid Res 2007; 48: 2547-2559.
77.   Glatz JF, Luiken JJ, Bonen A. Involvement of membrane-associated proteins in the
      acute regulation of cellular fatty acid uptake. J Mol Neurosci 2001; 16: 123-132;
      discussion 151-127.
                                                                                                85
Chapter 2
78.   Schwenk RW, Luiken JJ, Bonen A, Glatz JF. Regulation of sarcolemmal glucose and
      fatty acid transporters in cardiac disease. Cardiovasc Res 2008; 79: 249-258.
79.   Buque X, Cano A, Miquilena-Colina ME, Garcia-Monzon C, Ochoa B, Aspichueta P.
      High insulin levels are required for FAT/CD36 plasma membrane translocation and
      enhanced fatty acid uptake in obese Zucker rat hepatocytes. Am J Physiol Endocrinol
      Metab 2012; 303: E504-514.
80.   Chabowski A, Coort SL, Calles-Escandon J, et al. Insulin stimulates fatty acid
      transport by regulating expression of FAT/CD36 but not FABPpm. Am J Physiol
      Endocrinol Metab 2004; 287: E781-789.
81.   Luiken JJ, Dyck DJ, Han XX, et al. Insulin induces the translocation of the fatty acid
      transporter FAT/CD36 to the plasma membrane. Am J Physiol Endocrinol Metab
      2002; 282: E491-495.
82.   Bonen A, Tandon NN, Glatz JF, Luiken JJ, Heigenhauser GJ. The fatty acid
      transporter FAT/CD36 is upregulated in subcutaneous and visceral adipose tissues in
      human obesity and type 2 diabetes. Int J Obes (Lond) 2006; 30: 877-883.
83.   Pietka TA, Schappe T, Conte C, et al. Adipose and muscle tissue profile of CD36
      transcripts in obese subjects highlights the role of CD36 in fatty acid homeostasis
      and insulin resistance. Diabetes Care 2014; 37: 1990-1997.
84.   Bush NC, Triay JM, Gathaiya NW, Hames KC, Jensen MD. Contribution of very low-
      density lipoprotein triglyceride fatty acids to postabsorptive free fatty acid flux in
      obese humans. Metabolism 2014; 63: 137-140.
85.   Lafontan M, Langin D. Lipolysis and lipid mobilization in human adipose tissue. Prog
      Lipid Res 2009; 48: 275-297.
86.   Sengenes C, Berlan M, De Glisezinski I, Lafontan M, Galitzky J. Natriuretic peptides:
      a new lipolytic pathway in human adipocytes. FASEB J 2000; 14: 1345-1351.
87.   Sengenes C, Bouloumie A, Hauner H, et al. Involvement of a cGMP-dependent
      pathway in the natriuretic peptide-mediated hormone-sensitive lipase phosphorylation
      in human adipocytes. J Biol Chem 2003; 278: 48617-48626.
88.   Birkenfeld AL, Boschmann M, Moro C, et al. Lipid mobilization with physiological
      atrial natriuretic peptide concentrations in humans. J Clin Endocrinol Metab 2005; 90:
      3622-3628.
89.   Jocken JW, Blaak EE. Catecholamine-induced lipolysis in adipose tissue and skeletal
      muscle in obesity. Physiol Behav 2008; 94: 219-230.
90.   Zechner R, Zimmermann R, Eichmann TO, et al. FAT SIGNALS--lipases and lipolysis
      in lipid metabolism and signaling. Cell Metab 2012; 15: 279-291.
91.   Jocken JW, Langin D, Smit E, et al. Adipose triglyceride lipase and hormone-
      sensitive lipase protein expression is decreased in the obese insulin-resistant state. J
      Clin Endocrinol Metab 2007; 92: 2292-2299.
92.   Bickerton AS, Roberts R, Fielding BA, et al. Adipose tissue fatty acid metabolism in
      insulin-resistant men. Diabetologia 2008; 51: 1466-1474.
93.   Groop LC, Bonadonna RC, Simonson DC, Petrides AS, Shank M, DeFronzo RA.
      Effect of insulin on oxidative and nonoxidative pathways of free fatty acid metabolism
      in human obesity. Am J Physiol 1992; 263: E79-84.
94.   Langin D, Dicker A, Tavernier G, et al. Adipocyte lipases and defect of lipolysis in
      human obesity. Diabetes 2005; 54: 3190-3197.
95.   Ryden M, Jocken J, van Harmelen V, et al. Comparative studies of the role of
      hormone-sensitive lipase and adipose triglyceride lipase in human fat cell lipolysis.
      Am J Physiol Endocrinol Metab 2007; 292: E1847-1855.
96.   Ray H, Pinteur C, Frering V, Beylot M, Large V. Depot-specific differences in perilipin
      and hormone-sensitive lipase expression in lean and obese. Lipids Health Dis 2009;
      8: 58.
97.   Wang Y, Sullivan S, Trujillo M, et al. Perilipin expression in human adipose tissues:
      effects of severe obesity, gender, and depot. Obes Res 2003; 11: 930-936.
86
                                                                                  Chapter 2
                                                                                           87
Chapter 2
120.   Coleman RA, Lewin TM, Muoio DM. Physiological and nutritional regulation of
       enzymes of triacylglycerol synthesis. Annu Rev Nutr 2000; 20: 77-103.
121.   Stone SJ, Levin MC, Farese RV, Jr. Membrane topology and identification of key
       functional amino acid residues of murine acyl-CoA:diacylglycerol acyltransferase-2. J
       Biol Chem 2006; 281: 40273-40282.
122.   Gong J, Sun Z, Wu L, et al. Fsp27 promotes lipid droplet growth by lipid exchange
       and transfer at lipid droplet contact sites. J Cell Biol 2011; 195: 953-963.
123.   Jambunathan S, Yin J, Khan W, Tamori Y, Puri V. FSP27 promotes lipid droplet
       clustering and then fusion to regulate triglyceride accumulation. PLoS One 2011; 6:
       e28614.
124.   Puri V, Ranjit S, Konda S, et al. Cidea is associated with lipid droplets and insulin
       sensitivity in humans. Proc Natl Acad Sci U S A 2008; 105: 7833-7838.
125.   Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions linking obesity
       to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol 2008; 9: 367-377.
126.   Nishino N, Tamori Y, Tateya S, et al. FSP27 contributes to efficient energy storage in
       murine white adipocytes by promoting the formation of unilocular lipid droplets. J Clin
       Invest 2008; 118: 2808-2821.
127.   Martinez-Botas J, Anderson JB, Tessier D, et al. Absence of perilipin results in
       leanness and reverses obesity in Lepr(db/db) mice. Nat Genet 2000; 26: 474-479.
128.   Tansey JT, Sztalryd C, Gruia-Gray J, et al. Perilipin ablation results in a lean mouse
       with aberrant adipocyte lipolysis, enhanced leptin production, and resistance to diet-
       induced obesity. Proc Natl Acad Sci U S A 2001; 98: 6494-6499.
129.   Krotkiewski M, Bjorntorp P, Sjostrom L, Smith U. Impact of obesity on metabolism in
       men and women. Importance of regional adipose tissue distribution. J Clin Invest
       1983; 72: 1150-1162.
130.   Konige M, Wang H, Sztalryd C. Role of adipose specific lipid droplet proteins in
       maintaining whole body energy homeostasis. Biochim Biophys Acta 2014; 1842: 393-
       401.
131.   Tchoukalova YD, Votruba SB, Tchkonia T, Giorgadze N, Kirkland JL, Jensen MD.
       Regional differences in cellular mechanisms of adipose tissue gain with overfeeding.
       Proc Natl Acad Sci U S A 2010; 107: 18226-18231.
132.   Mariman EC, Wang P. Adipocyte extracellular matrix composition, dynamics and role
       in obesity. Cell Mol Life Sci 2010; 67: 1277-1292.
133.   Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin
       Invest 2011; 121: 2094-2101.
134.   Chun TH. Peri-adipocyte ECM remodeling in obesity and adipose tissue fibrosis.
       Adipocyte 2012; 1: 89-95.
135.   Strissel KJ, Stancheva Z, Miyoshi H, et al. Adipocyte death, adipose tissue
       remodeling, and obesity complications. Diabetes 2007; 56: 2910-2918.
136.   Divoux A, Tordjman J, Lacasa D, et al. Fibrosis in human adipose tissue:
       composition, distribution, and link with lipid metabolism and fat mass loss. Diabetes
       2010; 59: 2817-2825.
137.   Alligier M, Meugnier E, Debard C, et al. Subcutaneous adipose tissue remodeling
       during the initial phase of weight gain induced by overfeeding in humans. J Clin
       Endocrinol Metab 2012; 97: E183-192.
138.   Kim JY, van de Wall E, Laplante M, et al. Obesity-associated improvements in
       metabolic profile through expansion of adipose tissue. J Clin Invest 2007; 117: 2621-
       2637.
139.   Danforth E, Jr. Failure of adipocyte differentiation causes type II diabetes mellitus?
       Nat Genet 2000; 26: 13.
140.   Cawthorn WP, Scheller EL, MacDougald OA. Adipose tissue stem cells: the great
       WAT hope. Trends Endocrinol Metab 2012; 23: 270-277.
88
                                                                                   Chapter 2
141.   Macotela Y, Emanuelli B, Mori MA, et al. Intrinsic differences in adipocyte precursor
       cells from different white fat depots. Diabetes 2012; 61: 1691-1699.
142.   Spalding KL, Arner E, Westermark PO, et al. Dynamics of fat cell turnover in humans.
       Nature 2008; 453: 783-787.
143.   Wilson-Fritch L, Nicoloro S, Chouinard M, et al. Mitochondrial remodeling in adipose
       tissue associated with obesity and treatment with rosiglitazone. J Clin Invest 2004;
       114: 1281-1289.
144.   Kusminski CM, Scherer PE. Mitochondrial dysfunction in white adipose tissue.
       Trends Endocrinol Metab 2012; 23: 435-443.
145.   Klimcakova E, Roussel B, Marquez-Quinones A, et al. Worsening of obesity and
       metabolic status yields similar molecular adaptations in human subcutaneous and
       visceral adipose tissue: decreased metabolism and increased immune response. J
       Clin Endocrinol Metab 2011; 96: E73-82.
146.   Chen L, Na R, Gu M, et al. Reduction of mitochondrial H2O2 by overexpressing
       peroxiredoxin 3 improves glucose tolerance in mice. Aging Cell 2008; 7: 866-878.
147.   Qatanani M, Tan Y, Dobrin R, et al. Inverse regulation of inflammation and
       mitochondrial function in adipose tissue defines extreme insulin sensitivity in morbidly
       obese patients. Diabetes 2013; 62: 855-863.
148.   Bogacka I, Xie H, Bray GA, Smith SR. Pioglitazone induces mitochondrial biogenesis
       in human subcutaneous adipose tissue in vivo. Diabetes 2005; 54: 1392-1399.
149.   Dahlman I, Forsgren M, Sjogren A, et al. Downregulation of electron transport chain
       genes in visceral adipose tissue in type 2 diabetes independent of obesity and
       possibly involving tumor necrosis factor-alpha. Diabetes 2006; 55: 1792-1799.
150.   Yin X, Lanza IR, Swain JM, Sarr MG, Nair KS, Jensen MD. Adipocyte mitochondrial
       function is reduced in human obesity independent of fat cell size. J Clin Endocrinol
       Metab 2014; 99: E209-216.
151.   Saris WH, Heymsfield SB. All metabolic roads lead to mitochondrial (dys)-function.
       Curr Opin Clin Nutr Metab Care 2007; 10: 661-663.
152.   Mottillo EP, Bloch AE, Leff T, Granneman JG. Lipolytic products activate peroxisome
       proliferator-activated receptor (PPAR) alpha and delta in brown adipocytes to match
       fatty acid oxidation with supply. J Biol Chem 2012; 287: 25038-25048.
153.   van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, et al. Cold-activated
       brown adipose tissue in healthy men. N Engl J Med 2009; 360: 1500-1508.
154.   Virtanen KA, Lidell ME, Orava J, et al. Functional brown adipose tissue in healthy
       adults. N Engl J Med 2009; 360: 1518-1525.
155.   Saito M, Okamatsu-Ogura Y, Matsushita M, et al. High incidence of metabolically
       active brown adipose tissue in healthy adult humans: effects of cold exposure and
       adiposity. Diabetes 2009; 58: 1526-1531.
156.   Vijgen GH, Bouvy ND, Teule GJ, et al. Increase in brown adipose tissue activity after
       weight loss in morbidly obese subjects. J Clin Endocrinol Metab 2012; 97: E1229-
       1233.
157.   Chondronikola M, Volpi E, Borsheim E, et al. Brown Adipose Tissue Improves Whole
       Body Glucose Homeostasis and Insulin Sensitivity in Humans. Diabetes 2014;
158.   Wu J, Bostrom P, Sparks LM, et al. Beige adipocytes are a distinct type of
       thermogenic fat cell in mouse and human. Cell 2012; 150: 366-376.
159.   Ishibashi J, Seale P. Medicine. Beige can be slimming. Science 2010; 328: 1113-
       1114.
160.   Petrovic N, Walden TB, Shabalina IG, Timmons JA, Cannon B, Nedergaard J.
       Chronic peroxisome proliferator-activated receptor gamma (PPARgamma) activation
       of epididymally derived white adipocyte cultures reveals a population of
       thermogenically competent, UCP1-containing adipocytes molecularly distinct from
       classic brown adipocytes. J Biol Chem 2010; 285: 7153-7164.
                                                                                            89
Chapter 2
161.   Walden TB, Hansen IR, Timmons JA, Cannon B, Nedergaard J. Recruited vs.
       nonrecruited molecular signatures of brown, "brite," and white adipose tissues. Am J
       Physiol Endocrinol Metab 2012; 302: E19-31.
162.   van Marken Lichtenbelt W. Brown adipose tissue and the regulation of nonshivering
       thermogenesis. Curr Opin Clin Nutr Metab Care 2012; 15: 547-552.
163.   Yoneshiro T, Aita S, Kawai Y, Iwanaga T, Saito M. Nonpungent capsaicin analogs
       (capsinoids) increase energy expenditure through the activation of brown adipose
       tissue in humans. Am J Clin Nutr 2012; 95: 845-850.
164.   Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell 2014;
       156: 20-44.
165.   Fisher FM, Kleiner S, Douris N, et al. FGF21 regulates PGC-1alpha and browning of
       white adipose tissues in adaptive thermogenesis. Genes Dev 2012; 26: 271-281.
166.   Schulz TJ, Huang P, Huang TL, et al. Brown-fat paucity due to impaired BMP
       signalling induces compensatory browning of white fat. Nature 2013; 495: 379-383.
167.   Cao H, Gerhold K, Mayers JR, Wiest MM, Watkins SM, Hotamisligil GS. Identification
       of a lipokine, a lipid hormone linking adipose tissue to systemic metabolism. Cell
       2008; 134: 933-944.
168.   Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW, Jr.
       Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest
       2003; 112: 1796-1808.
169.   Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat plays a crucial role in the
       development of obesity-related insulin resistance. J Clin Invest 2003; 112: 1821-
       1830.
170.   Mraz M, Haluzik M. The role of adipose tissue immune cells in obesity and low-grade
       inflammation. J Endocrinol 2014; 222: R113-127.
171.   Mathis D. Immunological goings-on in visceral adipose tissue. Cell Metab 2013; 17:
       851-859.
172.   Sun S, Ji Y, Kersten S, Qi L. Mechanisms of inflammatory responses in obese
       adipose tissue. Annu Rev Nutr 2012; 32: 261-286.
173.   Lu M, Patsouris D, Li P, et al. A new antidiabetic compound attenuates inflammation
       and insulin resistance in Zucker diabetic fatty rats. Am J Physiol Endocrinol Metab
       2010; 298: E1036-1048.
174.   Samuel VT, Petersen KF, Shulman GI. Lipid-induced insulin resistance: unravelling
       the mechanism. Lancet 2010; 375: 2267-2277.
175.   Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity
       and fatty acid-induced insulin resistance. J Clin Invest 2006; 116: 3015-3025.
176.   Caricilli AM, Saad MJ. The role of gut microbiota on insulin resistance. Nutrients
       2013; 5: 829-851.
177.   Chambrier C, Bastard JP, Rieusset J, et al. Eicosapentaenoic acid induces mRNA
       expression of peroxisome proliferator-activated receptor gamma. Obes Res 2002; 10:
       518-525.
178.   Bassaganya-Riera J, Misyak S, Guri AJ, Hontecillas R. PPAR gamma is highly
       expressed in F4/80(hi) adipose tissue macrophages and dampens adipose-tissue
       inflammation. Cell Immunol 2009; 258: 138-146.
179.   Flamment M, Hajduch E, Ferre P, Foufelle F. New insights into ER stress-induced
       insulin resistance. Trends Endocrinol Metab 2012; 23: 381-390.
180.   Sopasakis VR, Sandqvist M, Gustafson B, et al. High local concentrations and effects
       on differentiation implicate interleukin-6 as a paracrine regulator. Obes Res 2004; 12:
       454-460.
181.   Petruschke T, Hauner H. Tumor necrosis factor-alpha prevents the differentiation of
       human adipocyte precursor cells and causes delipidation of newly developed fat
       cells. J Clin Endocrinol Metab 1993; 76: 742-747.
90
                                                                                   Chapter 2
182.   Xing H, Northrop JP, Grove JR, Kilpatrick KE, Su JL, Ringold GM. TNF alpha-
       mediated inhibition and reversal of adipocyte differentiation is accompanied by
       suppressed expression of PPARgamma without effects on Pref-1 expression.
       Endocrinology 1997; 138: 2776-2783.
183.   Prins JB, Niesler CU, Winterford CM, et al. Tumor necrosis factor-alpha induces
       apoptosis of human adipose cells. Diabetes 1997; 46: 1939-1944.
184.   Hauner H, Petruschke T, Russ M, Rohrig K, Eckel J. Effects of tumour necrosis factor
       alpha (TNF alpha) on glucose transport and lipid metabolism of newly-differentiated
       human fat cells in cell culture. Diabetologia 1995; 38: 764-771.
185.   Souza SC, Palmer HJ, Kang YH, et al. TNF-alpha induction of lipolysis is mediated
       through activation of the extracellular signal related kinase pathway in 3T3-L1
       adipocytes. J Cell Biochem 2003; 89: 1077-1086.
186.   Zhang HH, Halbleib M, Ahmad F, Manganiello VC, Greenberg AS. Tumor necrosis
       factor-alpha stimulates lipolysis in differentiated human adipocytes through activation
       of extracellular signal-related kinase and elevation of intracellular cAMP. Diabetes
       2002; 51: 2929-2935.
187.   van Hall G, Steensberg A, Sacchetti M, et al. Interleukin-6 stimulates lipolysis and fat
       oxidation in humans. J Clin Endocrinol Metab 2003; 88: 3005-3010.
188.   Kahn BB, Flier JS. Obesity and insulin resistance. J Clin Invest 2000; 106: 473-481.
189.   Dyck DJ, Heigenhauser GJ, Bruce CR. The role of adipokines as regulators of
       skeletal muscle fatty acid metabolism and insulin sensitivity. Acta Physiol (Oxf) 2006;
       186: 5-16.
190.   Yamauchi T, Kamon J, Ito Y, et al. Cloning of adiponectin receptors that mediate
       antidiabetic metabolic effects. Nature 2003; 423: 762-769.
191.   Yamauchi T, Kamon J, Minokoshi Y, et al. Adiponectin stimulates glucose utilization
       and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med 2002; 8:
       1288-1295.
192.   Ohashi K, Shibata R, Murohara T, Ouchi N. Role of anti-inflammatory adipokines in
       obesity-related diseases. Trends Endocrinol Metab 2014; 25: 348-355.
193.   Wong GW, Krawczyk SA, Kitidis-Mitrokostas C, Revett T, Gimeno R, Lodish HF.
       Molecular, biochemical and functional characterizations of C1q/TNF family members:
       adipose-tissue-selective expression patterns, regulation by PPAR-gamma agonist,
       cysteine-mediated oligomerizations, combinatorial associations and metabolic
       functions. Biochem J 2008; 416: 161-177.
194.   Wong GW, Krawczyk SA, Kitidis-Mitrokostas C, et al. Identification and
       characterization of CTRP9, a novel secreted glycoprotein, from adipose tissue that
       reduces serum glucose in mice and forms heterotrimers with adiponectin. FASEB J
       2009; 23: 241-258.
195.   Peterson JM, Wei Z, Wong GW. C1q/TNF-related protein-3 (CTRP3), a novel
       adipokine that regulates hepatic glucose output. J Biol Chem 2010; 285: 39691-
       39701.
196.   Kopp A, Bala M, Buechler C, et al. C1q/TNF-related protein-3 represents a novel and
       endogenous lipopolysaccharide antagonist of the adipose tissue. Endocrinology
       2010; 151: 5267-5278.
197.   Wolfing B, Buechler C, Weigert J, et al. Effects of the new C1q/TNF-related protein
       (CTRP-3) "cartonectin" on the adipocytic secretion of adipokines. Obesity (Silver
       Spring) 2008; 16: 1481-1486.
198.   Kim MJ, Lee W, Park EJ, Park SY. C1qTNF-related protein-6 increases the
       expression of interleukin-10 in macrophages. Mol Cells 2010; 30: 59-64.
199.   Lee W, Kim MJ, Park EJ, Choi YJ, Park SY. C1qTNF-related protein-6 mediates fatty
       acid oxidation via the activation of the AMP-activated protein kinase. FEBS Lett 2010;
       584: 968-972.
                                                                                            91
Chapter 2
200.   Peterson JM, Wei Z, Seldin MM, Byerly MS, Aja S, Wong GW. CTRP9 transgenic
       mice are protected from diet-induced obesity and metabolic dysfunction. Am J
       Physiol Regul Integr Comp Physiol 2013; 305: R522-533.
201.   Enomoto T, Ohashi K, Shibata R, et al. Adipolin/C1qdc2/CTRP12 protein functions
       as an adipokine that improves glucose metabolism. J Biol Chem 2011; 286: 34552-
       34558.
202.   Wei Z, Peterson JM, Lei X, et al. C1q/TNF-related protein-12 (CTRP12), a novel
       adipokine that improves insulin sensitivity and glycemic control in mouse models of
       obesity and diabetes. J Biol Chem 2012; 287: 10301-10315.
203.   Yang RZ, Lee MJ, Hu H, et al. Identification of omentin as a novel depot-specific
       adipokine in human adipose tissue: possible role in modulating insulin action. Am J
       Physiol Endocrinol Metab 2006; 290: E1253-1261.
204.   de Souza Batista CM, Yang RZ, Lee MJ, et al. Omentin plasma levels and gene
       expression are decreased in obesity. Diabetes 2007; 56: 1655-1661.
205.   Lamers D, Famulla S, Wronkowitz N, et al. Dipeptidyl peptidase 4 is a novel
       adipokine potentially linking obesity to the metabolic syndrome. Diabetes 2011; 60:
       1917-1925.
206.   Giannocco G, Oliveira KC, Crajoinas RO, et al. Dipeptidyl peptidase IV inhibition
       upregulates GLUT4 translocation and expression in heart and skeletal muscle of
       spontaneously hypertensive rats. Eur J Pharmacol 2013; 698: 74-86.
207.   Sakamoto T, Takahashi N, Sawaragi Y, et al. Inflammation induced by RAW
       macrophages suppresses UCP1 mRNA induction via ERK activation in 10T1/2
       adipocytes. Am J Physiol Cell Physiol 2013; 304: C729-738.
208.   Cao X, Gao Z, Robert CE, et al. Pancreatic-derived factor (FAM3B), a novel islet
       cytokine, induces apoptosis of insulin-secreting beta-cells. Diabetes 2003; 52: 2296-
       2303.
209.   Shimano M, Ouchi N, Walsh K. Cardiokines: recent progress in elucidating the
       cardiac secretome. Circulation 2012; 126: e327-332.
210.   Fernandes J, Vogt J, Wolever TM. Intravenous acetate elicits a greater free fatty acid
       rebound in normal than hyperinsulinaemic humans. Eur J Clin Nutr 2012; 66: 1029-
       1034.
211.   Aberdein N, Schweizer M, Ball D. Sodium acetate decreases phosphorylation of
       hormone sensitive lipase in isoproterenol-stimulated 3T3-L1 mature adipocytes.
       Adipocyte 2014; 3: 121-125.
212.   Most J, Goossens GH, Jocken JW, Blaak EE. Short-term supplementation with a
       specific combination of dietary polyphenols increases energy expenditure and alters
       substrate metabolism in overweight subjects. Int J Obes (Lond) 2014; 38: 698-706.
213.   Konings E, Timmers S, Boekschoten MV, et al. The effects of 30 days resveratrol
       supplementation on adipose tissue morphology and gene expression patterns in
       obese men. Int J Obes (Lond) 2014; 38: 470-473.
214.   Goossens GH, Bizzarri A, Venteclef N, et al. Increased adipose tissue oxygen
       tension in obese compared with lean men is accompanied by insulin resistance,
       impaired adipose tissue capillarization, and inflammation. Circulation 2011; 124: 67-
       76.
215.   Goossens GH, Blaak EE. Adipose tissue oxygen tension: implications for chronic
       metabolic and inflammatory diseases. Curr Opin Clin Nutr Metab Care 2012; 15: 539-
       546.
216.   Lee P, Linderman JD, Smith S, et al. Irisin and FGF21 are cold-induced endocrine
       activators of brown fat function in humans. Cell Metab 2014; 19: 302-309.
217.   Nettleton JA, Jebb S, Riserus U, Koletzko B, Fleming J. Role of dietary fats in the
       prevention and treatment of the metabolic syndrome. Ann Nutr Metab 2014; 64: 167-
       178.
92
                                                                                   Chapter 2
218.   Riserus U, Willett WC, Hu FB. Dietary fats and prevention of type 2 diabetes. Prog
       Lipid Res 2009; 48: 44-51.
219.   Storlien LH, Jenkins AB, Chisholm DJ, Pascoe WS, Khouri S, Kraegen EW. Influence
       of dietary fat composition on development of insulin resistance in rats. Relationship to
       muscle triglyceride and omega-3 fatty acids in muscle phospholipid. Diabetes 1991;
       40: 280-289.
220.   Jebb SA, Lovegrove JA, Griffin BA, et al. Effect of changing the amount and type of
       fat and carbohydrate on insulin sensitivity and cardiovascular risk: the RISCK
       (Reading, Imperial, Surrey, Cambridge, and Kings) trial. Am J Clin Nutr 2010; 92:
       748-758.
221.   Tierney AC, McMonagle J, Shaw DI, et al. Effects of dietary fat modification on insulin
       sensitivity and on other risk factors of the metabolic syndrome--LIPGENE: a
       European randomized dietary intervention study. Int J Obes (Lond) 2011; 35: 800-
       809.
222.   Barber E, Sinclair AJ, Cameron-Smith D. Comparative actions of omega-3 fatty acids
       on in-vitro lipid droplet formation. Prostaglandins Leukot Essent Fatty Acids 2013; 89:
       359-366.
223.   Flachs P, Rossmeisl M, Kuda O, Kopecky J. Stimulation of mitochondrial oxidative
       capacity in white fat independent of UCP1: a key to lean phenotype. Biochim Biophys
       Acta 2013; 1831: 986-1003.
224.   Flachs P, Horakova O, Brauner P, et al. Polyunsaturated fatty acids of marine origin
       upregulate mitochondrial biogenesis and induce beta-oxidation in white fat.
       Diabetologia 2005; 48: 2365-2375.
225.   Hensler M, Bardova K, Jilkova ZM, et al. The inhibition of fat cell proliferation by n-3
       fatty acids in dietary obese mice. Lipids Health Dis 2011; 10: 128.
226.   Ruzickova J, Rossmeisl M, Prazak T, et al. Omega-3 PUFA of marine origin limit diet-
       induced obesity in mice by reducing cellularity of adipose tissue. Lipids 2004; 39:
       1177-1185.
227.   Summers LK, Fielding BA, Bradshaw HA, et al. Substituting dietary saturated fat with
       polyunsaturated fat changes abdominal fat distribution and improves insulin
       sensitivity. Diabetologia 2002; 45: 369-377.
228.   Bjermo H, Iggman D, Kullberg J, et al. Effects of n-6 PUFAs compared with SFAs on
       liver fat, lipoproteins, and inflammation in abdominal obesity: a randomized controlled
       trial. Am J Clin Nutr 2012; 95: 1003-1012.
229.   Weigert C, Brodbeck K, Staiger H, et al. Palmitate, but not unsaturated fatty acids,
       induces the expression of interleukin-6 in human myotubes through proteasome-
       dependent activation of nuclear factor-kappaB. J Biol Chem 2004; 279: 23942-
       23952.
230.   Ajuwon KM, Spurlock ME. Palmitate activates the NF-kappaB transcription factor and
       induces IL-6 and TNFalpha expression in 3T3-L1 adipocytes. J Nutr 2005; 135: 1841-
       1846.
231.   Baer DJ, Judd JT, Clevidence BA, Tracy RP. Dietary fatty acids affect plasma
       markers of inflammation in healthy men fed controlled diets: a randomized crossover
       study. Am J Clin Nutr 2004; 79: 969-973.
232.   Thies F, Miles EA, Nebe-von-Caron G, et al. Influence of dietary supplementation
       with long-chain n-3 or n-6 polyunsaturated fatty acids on blood inflammatory cell
       populations and functions and on plasma soluble adhesion molecules in healthy
       adults. Lipids 2001; 36: 1183-1193.
233.   Itariu BK, Zeyda M, Hochbrugger EE, et al. Long-chain n-3 PUFAs reduce adipose
       tissue and systemic inflammation in severely obese nondiabetic patients: a
       randomized controlled trial. Am J Clin Nutr 2012; 96: 1137-1149.
234.   Reaven GM. Pathophysiology of insulin resistance in human disease. Physiol Rev
       1995; 75: 473-486.
                                                                                            93
Chapter 2
235.   McGarry JD. Banting lecture 2001: dysregulation of fatty acid metabolism in the
       etiology of type 2 diabetes. Diabetes 2002; 51: 7-18.
236.   Kowal RC, Herz J, Goldstein JL, Esser V, Brown MS. Low density lipoprotein
       receptor-related protein mediates uptake of cholesteryl esters derived from
       apoprotein E-enriched lipoproteins. Proc Natl Acad Sci U S A 1989; 86: 5810-5814.
237.   Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis of nonalcoholic
       steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology
       2010; 52: 774-788.
238.   Kabir M, Catalano KJ, Ananthnarayan S, et al. Molecular evidence supporting the
       portal theory: a causative link between visceral adiposity and hepatic insulin
       resistance. Am J Physiol Endocrinol Metab 2005; 288: E454-461.
239.   Jensen MD. Role of body fat distribution and the metabolic complications of obesity.
       J Clin Endocrinol Metab 2008; 93: S57-63.
240.   Catalano KJ, Stefanovski D, Bergman RN. Critical role of the mesenteric depot
       versus other intra-abdominal adipose depots in the development of insulin resistance
       in young rats. Diabetes 2010; 59: 1416-1423.
241.   Rytka JM, Wueest S, Schoenle EJ, Konrad D. The portal theory supported by venous
       drainage-selective fat transplantation. Diabetes 2011; 60: 56-63.
242.   Fromenty B, Robin MA, Igoudjil A, Mansouri A, Pessayre D. The ins and outs of
       mitochondrial dysfunction in NASH. Diabetes Metab 2004; 30: 121-138.
243.   Ferre P, Foufelle F. Hepatic steatosis: a role for de novo lipogenesis and the
       transcription factor SREBP-1c. Diabetes Obes Metab 2010; 12 Suppl 2: 83-92.
244.   Wiggins D, Gibbons GF. The lipolysis/esterification cycle of hepatic triacylglycerol. Its
       role in the secretion of very-low-density lipoprotein and its response to hormones and
       sulphonylureas. Biochem J 1992; 284 ( Pt 2): 457-462.
245.   Kissebah AH, Alfarsi S, Adams PW, Wynn V. Role of insulin resistance in adipose
       tissue and liver in the pathogenesis of endogenous hypertriglyceridaemia in man.
       Diabetologia 1976; 12: 563-571.
246.   Abbasi F, McLaughlin T, Lamendola C, et al. Fasting remnant lipoprotein cholesterol
       and triglyceride concentrations are elevated in nondiabetic, insulin-resistant, female
       volunteers. J Clin Endocrinol Metab 1999; 84: 3903-3906.
247.   Watanabe N, Taniguchi T, Taketoh H, et al. Elevated remnant-like lipoprotein
       particles in impaired glucose tolerance and type 2 diabetic patients. Diabetes Care
       1999; 22: 152-156.
248.   Chan DC, Watts GF, Barrett PH, Mamo JC, Redgrave TG. Markers of triglyceride-
       rich lipoprotein remnant metabolism in visceral obesity. Clin Chem 2002; 48: 278-
       283.
249.   Lewis GF. Fatty acid regulation of very low density lipoprotein production. Curr Opin
       Lipidol 1997; 8: 146-153.
250.   Malmstrom R, Packard CJ, Caslake M, et al. Defective regulation of triglyceride
       metabolism by insulin in the liver in NIDDM. Diabetologia 1997; 40: 454-462.
251.   Taskinen MR. Diabetic dyslipidaemia: from basic research to clinical practice.
       Diabetologia 2003; 46: 733-749.
252.   Adeli K, Taghibiglou C, Van Iderstine SC, Lewis GF. Mechanisms of hepatic very
       low-density lipoprotein overproduction in insulin resistance. Trends Cardiovasc Med
       2001; 11: 170-176.
253.   Chatterjee C, Sparks DL. Hepatic lipase, high density lipoproteins, and
       hypertriglyceridemia. Am J Pathol 2011; 178: 1429-1433.
254.   Despres JP, Ferland M, Moorjani S, et al. Role of hepatic-triglyceride lipase activity in
       the association between intra-abdominal fat and plasma HDL cholesterol in obese
       women. Arteriosclerosis 1989; 9: 485-492.
94
                                                                                       Chapter 2
255.   Carr MC, Hokanson JE, Zambon A, et al. The contribution of intraabdominal fat to
       gender differences in hepatic lipase activity and low/high density lipoprotein
       heterogeneity. J Clin Endocrinol Metab 2001; 86: 2831-2837.
256.   Pardina E, Baena-Fustegueras JA, Catalan R, et al. Increased expression and
       activity of hepatic lipase in the liver of morbidly obese adult patients in relation to lipid
       content. Obes Surg 2009; 19: 894-904.
257.   Lewis GF, Murdoch S, Uffelman K, et al. Hepatic lipase mRNA, protein, and plasma
       enzyme activity is increased in the insulin-resistant, fructose-fed Syrian golden
       hamster and is partially normalized by the insulin sensitizer rosiglitazone. Diabetes
       2004; 53: 2893-2900.
258.   Sibley SD, Palmer JP, Hirsch IB, Brunzell JD. Visceral obesity, hepatic lipase activity,
       and dyslipidemia in type 1 diabetes. J Clin Endocrinol Metab 2003; 88: 3379-3384.
259.   Miksztowicz V, Lucero D, Zago V, et al. Hepatic lipase activity is increased in non-
       alcoholic fatty liver disease beyond insulin resistance. Diabetes Metab Res Rev
       2012; 28: 535-541.
260.   Bradbury MW. Lipid metabolism and liver inflammation. I. Hepatic fatty acid uptake:
       possible role in steatosis. Am J Physiol Gastrointest Liver Physiol 2006; 290: G194-
       198.
261.   Falcon A, Doege H, Fluitt A, et al. FATP2 is a hepatic fatty acid transporter and
       peroxisomal very long-chain acyl-CoA synthetase. Am J Physiol Endocrinol Metab
       2010; 299: E384-393.
262.   Doege H, Grimm D, Falcon A, et al. Silencing of hepatic fatty acid transporter protein
       5 in vivo reverses diet-induced non-alcoholic fatty liver disease and improves
       hyperglycemia. J Biol Chem 2008; 283: 22186-22192.
263.   Coburn CT, Hajri T, Ibrahimi A, Abumrad NA. Role of CD36 in membrane transport
       and utilization of long-chain fatty acids by different tissues. J Mol Neurosci 2001; 16:
       117-121; discussion 151-117.
264.   Su X, Abumrad NA. Cellular fatty acid uptake: a pathway under construction. Trends
       Endocrinol Metab 2009; 20: 72-77.
265.   Doege H, Baillie RA, Ortegon AM, et al. Targeted deletion of FATP5 reveals multiple
       functions in liver metabolism: alterations in hepatic lipid homeostasis.
       Gastroenterology 2006; 130: 1245-1258.
266.   Westerbacka J, Kolak M, Kiviluoto T, et al. Genes involved in fatty acid partitioning
       and binding, lipolysis, monocyte/macrophage recruitment, and inflammation are
       overexpressed in the human fatty liver of insulin-resistant subjects. Diabetes 2007;
       56: 2759-2765.
267.   Nie B, Park HM, Kazantzis M, et al. Specific bile acids inhibit hepatic fatty acid uptake
       in mice. Hepatology 2012; 56: 1300-1310.
268.   Fruhbeck G, Lopez M, Dieguez C. Role of caveolins in body weight and insulin
       resistance regulation. Trends Endocrinol Metab 2007; 18: 177-182.
269.   Otsu K, Toya Y, Oshikawa J, et al. Caveolin gene transfer improves glucose
       metabolism in diabetic mice. Am J Physiol Cell Physiol 2010; 298: C450-456.
270.   Greco D, Kotronen A, Westerbacka J, et al. Gene expression in human NAFLD. Am J
       Physiol Gastrointest Liver Physiol 2008; 294: G1281-1287.
271.   Goudriaan JR, Dahlmans VE, Teusink B, et al. CD36 deficiency increases insulin
       sensitivity in muscle, but induces insulin resistance in the liver in mice. J Lipid Res
       2003; 44: 2270-2277.
272.   Ong KT, Mashek MT, Bu SY, Mashek DG. Hepatic ATGL knockdown uncouples
       glucose intolerance from liver TAG accumulation. FASEB J 2013; 27: 313-321.
273.   Voshol PJ, Haemmerle G, Ouwens DM, et al. Increased hepatic insulin sensitivity
       together with decreased hepatic triglyceride stores in hormone-sensitive lipase-
       deficient mice. Endocrinology 2003; 144: 3456-3462.
                                                                                                95
Chapter 2
96
                                                                                   Chapter 2
294.   Hellerstein MK. De novo lipogenesis in humans: metabolic and regulatory aspects.
       Eur J Clin Nutr 1999; 53 Suppl 1: S53-65.
295.   Schwarz JM, Linfoot P, Dare D, Aghajanian K. Hepatic de novo lipogenesis in
       normoinsulinemic and hyperinsulinemic subjects consuming high-fat, low-
       carbohydrate and low-fat, high-carbohydrate isoenergetic diets. Am J Clin Nutr 2003;
       77: 43-50.
296.   Jensen-Urstad AP, Semenkovich CF. Fatty acid synthase and liver triglyceride
       metabolism: housekeeper or messenger? Biochim Biophys Acta 2012; 1821: 747-
       753.
297.   Chakravarthy MV, Pan Z, Zhu Y, et al. "New" hepatic fat activates PPARalpha to
       maintain glucose, lipid, and cholesterol homeostasis. Cell Metab 2005; 1: 309-322.
298.   Miyazaki M, Kim YC, Ntambi JM. A lipogenic diet in mice with a disruption of the
       stearoyl-CoA desaturase 1 gene reveals a stringent requirement of endogenous
       monounsaturated fatty acids for triglyceride synthesis. J Lipid Res 2001; 42: 1018-
       1024.
299.   Smith SJ, Cases S, Jensen DR, et al. Obesity resistance and multiple mechanisms of
       triglyceride synthesis in mice lacking Dgat. Nat Genet 2000; 25: 87-90.
300.   Chen HC, Smith SJ, Ladha Z, et al. Increased insulin and leptin sensitivity in mice
       lacking acyl CoA:diacylglycerol acyltransferase 1. J Clin Invest 2002; 109: 1049-
       1055.
301.   Herman MA, Peroni OD, Villoria J, et al. A novel ChREBP isoform in adipose tissue
       regulates systemic glucose metabolism. Nature 2012; 484: 333-338.
302.   Strable MS, Ntambi JM. Genetic control of de novo lipogenesis: role in diet-induced
       obesity. Crit Rev Biochem Mol Biol 2010; 45: 199-214.
303.   Kersten S. Mechanisms of nutritional and hormonal regulation of lipogenesis. EMBO
       Rep 2001; 2: 282-286.
304.   Postic C, Girard J. Contribution of de novo fatty acid synthesis to hepatic steatosis
       and insulin resistance: lessons from genetically engineered mice. J Clin Invest 2008;
       118: 829-838.
305.   Moczulski D, Majak I, Mamczur D. An overview of beta-oxidation disorders. Postepy
       Hig Med Dosw (Online) 2009; 63: 266-277.
306.   Wanders RJ, Waterham HR. Biochemistry of mammalian peroxisomes revisited.
       Annu Rev Biochem 2006; 75: 295-332.
307.   Schrader M, Fahimi HD. The peroxisome: still a mysterious organelle. Histochem Cell
       Biol 2008; 129: 421-440.
308.   Kerner J, Hoppel C. Fatty acid import into mitochondria. Biochim Biophys Acta 2000;
       1486: 1-17.
309.   Park EA, Mynatt RL, Cook GA, Kashfi K. Insulin regulates enzyme activity, malonyl-
       CoA sensitivity and mRNA abundance of hepatic carnitine palmitoyltransferase-I.
       Biochem J 1995; 310 ( Pt 3): 853-858.
310.   Verhoeven NM, Roe DS, Kok RM, Wanders RJ, Jakobs C, Roe CR. Phytanic acid
       and pristanic acid are oxidized by sequential peroxisomal and mitochondrial reactions
       in cultured fibroblasts. J Lipid Res 1998; 39: 66-74.
311.   Hashimoto T. Peroxisomal beta-oxidation enzymes. Neurochem Res 1999; 24: 551-
       563.
312.   Reddy JK, Mannaerts GP. Peroxisomal lipid metabolism. Annu Rev Nutr 1994; 14:
       343-370.
313.   Jakobs BS, Wanders RJ. Fatty acid beta-oxidation in peroxisomes and mitochondria:
       the first, unequivocal evidence for the involvement of carnitine in shuttling propionyl-
       CoA from peroxisomes to mitochondria. Biochem Biophys Res Commun 1995; 213:
       1035-1041.
314.   Kim JA, Wei Y, Sowers JR. Role of mitochondrial dysfunction in insulin resistance.
       Circ Res 2008; 102: 401-414.
                                                                                            97
Chapter 2
98
                                                                                   Chapter 2
                                                                                            99
Chapter 2
357.   Ockenga J, Valentini L, Schuetz T, et al. Plasma bile acids are associated with
       energy expenditure and thyroid function in humans. J Clin Endocrinol Metab 2012;
       97: 535-542.
358.   Prawitt J, Caron S, Staels B. Bile acid metabolism and the pathogenesis of type 2
       diabetes. Curr Diab Rep 2011; 11: 160-166.
359.   Haeusler RA, Astiarraga B, Camastra S, Accili D, Ferrannini E. Human insulin
       resistance is associated with increased plasma levels of 12alpha-hydroxylated bile
       acids. Diabetes 2013; 62: 4184-4191.
360.   Andersen E, Karlaganis G, Sjovall J. Altered bile acid profiles in duodenal bile and
       urine in diabetic subjects. Eur J Clin Invest 1988; 18: 166-172.
361.   Begley M, Hill C, Gahan CG. Bile salt hydrolase activity in probiotics. Appl Environ
       Microbiol 2006; 72: 1729-1738.
362.   Batta AK, Salen G, Arora R, Shefer S, Batta M, Person A. Side chain conjugation
       prevents bacterial 7-dehydroxylation of bile acids. J Biol Chem 1990; 265: 10925-
       10928.
363.   Jones BV, Begley M, Hill C, Gahan CG, Marchesi JR. Functional and comparative
       metagenomic analysis of bile salt hydrolase activity in the human gut microbiome.
       Proc Natl Acad Sci U S A 2008; 105: 13580-13585.
364.   Vrieze A, Out C, Fuentes S, et al. Impact of oral vancomycin on gut microbiota, bile
       acid metabolism, and insulin sensitivity. J Hepatol 2014; 60: 824-831.
365.   Li T, Owsley E, Matozel M, Hsu P, Novak CM, Chiang JY. Transgenic expression of
       cholesterol 7alpha-hydroxylase in the liver prevents high-fat diet-induced obesity and
       insulin resistance in mice. Hepatology 2010; 52: 678-690.
366.   Beysen C, Murphy EJ, Deines K, et al. Effect of bile acid sequestrants on glucose
       metabolism, hepatic de novo lipogenesis, and cholesterol and bile acid kinetics in
       type 2 diabetes: a randomised controlled study. Diabetologia 2012; 55: 432-442.
367.   Smushkin G, Sathananthan M, Piccinini F, et al. The effect of a bile acid sequestrant
       on glucose metabolism in subjects with type 2 diabetes. Diabetes 2013; 62: 1094-
       1101.
368.   Kootte RS, Vrieze A, Holleman F, et al. The therapeutic potential of manipulating gut
       microbiota in obesity and type 2 diabetes mellitus. Diabetes Obes Metab 2012; 14:
       112-120.
369.   Sato S, Nagai H, Igarashi Y. Effect of probiotics on serum bile acids in patients with
       ulcerative colitis. Hepatogastroenterology 2012; 59: 1804-1808.
370.   Lazar MA. The humoral side of insulin resistance. Nat Med 2006; 12: 43-44.
371.   Hotamisligil GS. Inflammation and endoplasmic reticulum stress in obesity and
       diabetes. Int J Obes (Lond) 2008; 32 Suppl 7: S52-54.
372.   Pagliassotti MJ. Endoplasmic reticulum stress in nonalcoholic fatty liver disease.
       Annu Rev Nutr 2012; 32: 17-33.
373.   Rao MS, Reddy JK. Peroxisomal beta-oxidation and steatohepatitis. Semin Liver Dis
       2001; 21: 43-55.
374.   Garcia-Monzon C, Martin-Perez E, Iacono OL, et al. Characterization of pathogenic
       and prognostic factors of nonalcoholic steatohepatitis associated with obesity. J
       Hepatol 2000; 33: 716-724.
375.   Sanyal AJ, Campbell-Sargent C, Mirshahi F, et al. Nonalcoholic steatohepatitis:
       association of insulin resistance and mitochondrial abnormalities. Gastroenterology
       2001; 120: 1183-1192.
376.   Angulo P. Nonalcoholic fatty liver disease. N Engl J Med 2002; 346: 1221-1231.
377.   Haubrich WS. Kupffer of Kupffer cells. Gastroenterology 2004; 127: 16.
378.   Stefan N, Haring HU. The role of hepatokines in metabolism. Nat Rev Endocrinol
       2013; 9: 144-152.
379.   Lan F, Misu H, Chikamoto K, et al. LECT2 functions as a hepatokine that links
       obesity to skeletal muscle insulin resistance. Diabetes 2014;
100
                                                                                    Chapter 2
                                                                                           101
Chapter 2
398.   Simoneau JA, Veerkamp JH, Turcotte LP, Kelley DE. Markers of capacity to utilize
       fatty acids in human skeletal muscle: relation to insulin resistance and obesity and
       effects of weight loss. FASEB J 1999; 13: 2051-2060.
399.   Bosma M, Kersten S, Hesselink MK, Schrauwen P. Re-evaluating lipotoxic triggers in
       skeletal muscle: relating intramyocellular lipid metabolism to insulin sensitivity. Prog
       Lipid Res 2012; 51: 36-49.
400.   Moro C, Bajpeyi S, Smith SR. Determinants of intramyocellular triglyceride turnover:
       implications for insulin sensitivity. Am J Physiol Endocrinol Metab 2008; 294: E203-
       213.
401.   Muoio DM. Intramuscular triacylglycerol and insulin resistance: guilty as charged or
       wrongly accused? Biochim Biophys Acta 2010; 1801: 281-288.
402.   Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and
       missing links. Cell 2012; 148: 852-871.
403.   Summers SA. Ceramides in insulin resistance and lipotoxicity. Prog Lipid Res 2006;
       45: 42-72.
404.   Jocken JW, Goossens GH, Boon H, et al. Insulin-mediated suppression of lipolysis in
       adipose tissue and skeletal muscle of obese type 2 diabetic men and men with
       normal glucose tolerance. Diabetologia 2013; 56: 2255-2265.
405.   Bergman BC, Hunerdosse DM, Kerege A, Playdon MC, Perreault L. Localisation and
       composition of skeletal muscle diacylglycerol predicts insulin resistance in humans.
       Diabetologia 2012; 55: 1140-1150.
406.   Nowotny B, Zahiragic L, Krog D, et al. Mechanisms underlying the onset of oral lipid-
       induced skeletal muscle insulin resistance in humans. Diabetes 2013; 62: 2240-2248.
407.   Varma V, Yao-Borengasser A, Rasouli N, et al. Muscle inflammatory response and
       insulin resistance: synergistic interaction between macrophages and fatty acids leads
       to impaired insulin action. Am J Physiol Endocrinol Metab 2009; 296: E1300-1310.
408.   Bouzakri K, Plomgaard P, Berney T, Donath MY, Pedersen BK, Halban PA. Bimodal
       effect on pancreatic beta-cells of secretory products from normal or insulin-resistant
       human skeletal muscle. Diabetes 2011; 60: 1111-1121.
409.   Di Gregorio GB, Yao-Borengasser A, Rasouli N, et al. Expression of CD68 and
       macrophage chemoattractant protein-1 genes in human adipose and muscle tissues:
       association with cytokine expression, insulin resistance, and reduction by
       pioglitazone. Diabetes 2005; 54: 2305-2313.
410.   Bruun JM, Helge JW, Richelsen B, Stallknecht B. Diet and exercise reduce low-grade
       inflammation and macrophage infiltration in adipose tissue but not in skeletal muscle
       in severely obese subjects. Am J Physiol Endocrinol Metab 2006; 290: E961-967.
411.   Tam CS, Sparks LM, Johannsen DL, Covington JD, Church TS, Ravussin E. Low
       macrophage accumulation in skeletal muscle of obese type 2 diabetics and elderly
       subjects. Obesity (Silver Spring) 2012; 20: 1530-1533.
412.   Patsouris D, Cao JJ, Vial G, et al. Insulin resistance is associated with MCP1-
       mediated macrophage accumulation in skeletal muscle in mice and humans. PLoS
       One 2014; 9: e110653.
413.   Deldicque L, Hespel P, Francaux M. Endoplasmic reticulum stress in skeletal muscle:
       origin and metabolic consequences. Exerc Sport Sci Rev 2012; 40: 43-49.
414.   Moors CC, van der Zijl NJ, Diamant M, Blaak EE, Goossens GH. Impaired insulin
       sensitivity is accompanied by disturbances in skeletal muscle fatty acid handling in
       subjects with impaired glucose metabolism. Int J Obes (Lond) 2012; 36: 709-717.
415.   Wang H, Knaub LA, Jensen DR, et al. Skeletal muscle-specific deletion of lipoprotein
       lipase enhances insulin signaling in skeletal muscle but causes insulin resistance in
       liver and other tissues. Diabetes 2009; 58: 116-124.
416.   Kim JK, Fillmore JJ, Chen Y, et al. Tissue-specific overexpression of lipoprotein
       lipase causes tissue-specific insulin resistance. Proc Natl Acad Sci U S A 2001; 98:
       7522-7527.
102
                                                                                  Chapter 2
417.   Ladu MJ, Kapsas H, Palmer WK. Regulation of lipoprotein lipase in adipose and
       muscle tissues during fasting. Am J Physiol 1991; 260: R953-959.
418.   Sugden MC, Holness MJ, Howard RM. Changes in lipoprotein lipase activities in
       adipose tissue, heart and skeletal muscle during continuous or interrupted feeding.
       Biochem J 1993; 292 ( Pt 1): 113-119.
419.   Hamilton MT, Etienne J, McClure WC, Pavey BS, Holloway AK. Role of local
       contractile activity and muscle fiber type on LPL regulation during exercise. Am J
       Physiol 1998; 275: E1016-1022.
420.   Greiwe JS, Holloszy JO, Semenkovich CF. Exercise induces lipoprotein lipase and
       GLUT-4 protein in muscle independent of adrenergic-receptor signaling. J Appl
       Physiol (1985) 2000; 89: 176-181.
421.   Schrauwen-Hinderling VB, Hesselink MK, Moonen-Kornips E, et al. Short-term
       training is accompanied by a down regulation of ACC2 mRNA in skeletal muscle. Int
       J Sports Med 2006; 27: 786-791.
422.   Vissing K, Andersen JL, Schjerling P. Are exercise-induced genes induced by
       exercise? FASEB J 2005; 19: 94-96.
423.   Farese RV, Jr., Yost TJ, Eckel RH. Tissue-specific regulation of lipoprotein lipase
       activity by insulin/glucose in normal-weight humans. Metabolism 1991; 40: 214-216.
424.   Dijk W, Kersten S. Regulation of lipoprotein lipase by Angptl4. Trends Endocrinol
       Metab 2014; 25: 146-155.
425.   Xu A, Lam MC, Chan KW, et al. Angiopoietin-like protein 4 decreases blood glucose
       and improves glucose tolerance but induces hyperlipidemia and hepatic steatosis in
       mice. Proc Natl Acad Sci U S A 2005; 102: 6086-6091.
426.   Votruba SB, Jensen MD. Regional fat deposition as a factor in FFA metabolism.
       Annu Rev Nutr 2007; 27: 149-163.
427.   Abumrad N, Coburn C, Ibrahimi A. Membrane proteins implicated in long-chain fatty
       acid uptake by mammalian cells: CD36, FATP and FABPm. Biochim Biophys Acta
       1999; 1441: 4-13.
428.   Glatz JF, Bonen A, Luiken JJ. Exercise and insulin increase muscle fatty acid uptake
       by recruiting putative fatty acid transporters to the sarcolemma. Curr Opin Clin Nutr
       Metab Care 2002; 5: 365-370.
429.   Wu Q, Ortegon AM, Tsang B, Doege H, Feingold KR, Stahl A. FATP1 is an insulin-
       sensitive fatty acid transporter involved in diet-induced obesity. Mol Cell Biol 2006;
       26: 3455-3467.
430.   Gimeno RE, Ortegon AM, Patel S, et al. Characterization of a heart-specific fatty acid
       transport protein. J Biol Chem 2003; 278: 16039-16044.
431.   Coburn CT, Knapp FF, Jr., Febbraio M, Beets AL, Silverstein RL, Abumrad NA.
       Defective uptake and utilization of long chain fatty acids in muscle and adipose
       tissues of CD36 knockout mice. J Biol Chem 2000; 275: 32523-32529.
432.   Bonen A, Benton CR, Campbell SE, et al. Plasmalemmal fatty acid transport is
       regulated in heart and skeletal muscle by contraction, insulin and leptin, and in
       obesity and diabetes. Acta Physiol Scand 2003; 178: 347-356.
433.   Corpeleijn E, Pelsers MM, Soenen S, et al. Insulin acutely upregulates protein
       expression of the fatty acid transporter CD36 in human skeletal muscle in vivo. J
       Physiol Pharmacol 2008; 59: 77-83.
434.   Bonen A, Parolin ML, Steinberg GR, et al. Triacylglycerol accumulation in human
       obesity and type 2 diabetes is associated with increased rates of skeletal muscle fatty
       acid transport and increased sarcolemmal FAT/CD36. FASEB J 2004; 18: 1144-
       1146.
435.   Corpeleijn E, Mensink M, Kooi ME, Roekaerts PM, Saris WH, Blaak EE. Impaired
       skeletal muscle substrate oxidation in glucose-intolerant men improves after weight
       loss. Obesity (Silver Spring) 2008; 16: 1025-1032.
                                                                                         103
Chapter 2
436.   van Hees AM, Jans A, Hul GB, Roche HM, Saris WH, Blaak EE. Skeletal muscle
       fatty acid handling in insulin resistant men. Obesity (Silver Spring) 2011; 19: 1350-
       1359.
437.   Jocken JW, Goossens GH, van Hees AM, et al. Effect of beta-adrenergic stimulation
       on whole-body and abdominal subcutaneous adipose tissue lipolysis in lean and
       obese men. Diabetologia 2008; 51: 320-327.
438.   Glatz JF, Luiken JJ, van Bilsen M, van der Vusse GJ. Cellular lipid binding proteins
       as facilitators and regulators of lipid metabolism. Mol Cell Biochem 2002; 239: 3-7.
439.   Watt MJ. Triglyceride lipases alter fuel metabolism and mitochondrial gene
       expression. Appl Physiol Nutr Metab 2009; 34: 340-347.
440.   Timmers S, de Vogel-van den Bosch J, Hesselink MK, et al. Paradoxical increase in
       TAG and DAG content parallel the insulin sensitizing effect of unilateral DGAT1
       overexpression in rat skeletal muscle. PLoS One 2011; 6: e14503.
441.   Amati F, Dube JJ, Alvarez-Carnero E, et al. Skeletal muscle triglycerides,
       diacylglycerols, and ceramides in insulin resistance: another paradox in endurance-
       trained athletes? Diabetes 2011; 60: 2588-2597.
442.   Dube JJ, Amati F, Toledo FG, et al. Effects of weight loss and exercise on insulin
       resistance, and intramyocellular triacylglycerol, diacylglycerol and ceramide.
       Diabetologia 2011; 54: 1147-1156.
443.   Pinnamaneni SK, Southgate RJ, Febbraio MA, Watt MJ. Stearoyl CoA desaturase 1
       is elevated in obesity but protects against fatty acid-induced skeletal muscle insulin
       resistance in vitro. Diabetologia 2006; 49: 3027-3037.
444.   Hulver MW, Berggren JR, Carper MJ, et al. Elevated stearoyl-CoA desaturase-1
       expression in skeletal muscle contributes to abnormal fatty acid partitioning in obese
       humans. Cell Metab 2005; 2: 251-261.
445.   Bergman BC, Perreault L, Hunerdosse DM, Koehler MC, Samek AM, Eckel RH.
       Intramuscular lipid metabolism in the insulin resistance of smoking. Diabetes 2009;
       58: 2220-2227.
446.   Perreault L, Bergman BC, Hunerdosse DM, Playdon MC, Eckel RH. Inflexibility in
       intramuscular triglyceride fractional synthesis distinguishes prediabetes from obesity
       in humans. Obesity (Silver Spring) 2010; 18: 1524-1531.
447.   Moors CC, Blaak EE, van der Zijl NJ, Diamant M, Goossens GH. The effects of long-
       term valsartan treatment on skeletal muscle fatty acid handling in humans with
       impaired glucose metabolism. J Clin Endocrinol Metab 2013; 98: E891-896.
448.   Sparks LM, Bosma M, Brouwers B, et al. Reduced incorporation of fatty acids into
       triacylglycerol in myotubes from obese individuals with type 2 diabetes. Diabetes
       2014; 63: 1583-1593.
449.   Haemmerle G, Lass A, Zimmermann R, et al. Defective lipolysis and altered energy
       metabolism in mice lacking adipose triglyceride lipase. Science 2006; 312: 734-737.
450.   Badin PM, Louche K, Mairal A, et al. Altered skeletal muscle lipase expression and
       activity contribute to insulin resistance in humans. Diabetes 2011; 60: 1734-1742.
451.   Natali A, Gastaldelli A, Camastra S, et al. Metabolic consequences of adipose
       triglyceride lipase deficiency in humans: an in vivo study in patients with neutral lipid
       storage disease with myopathy. J Clin Endocrinol Metab 2013; 98: E1540-1548.
452.   Blaak EE, Schiffelers SL, Saris WH, Mensink M, Kooi ME. Impaired beta-
       adrenergically mediated lipolysis in skeletal muscle of obese subjects. Diabetologia
       2004; 47: 1462-1468.
453.   Jocken JW, Roepstorff C, Goossens GH, et al. Hormone-sensitive lipase serine
       phosphorylation and glycerol exchange across skeletal muscle in lean and obese
       subjects: effect of beta-adrenergic stimulation. Diabetes 2008; 57: 1834-1841.
454.   Gjelstad IM, Haugen F, Gulseth HL, et al. Expression of perilipins in human skeletal
       muscle in vitro and in vivo in relation to diet, exercise and energy balance. Arch
       Physiol Biochem 2012; 118: 22-30.
104
                                                                                    Chapter 2
455.   Bosma M, Hesselink MK, Sparks LM, et al. Perilipin 2 improves insulin sensitivity in
       skeletal muscle despite elevated intramuscular lipid levels. Diabetes 2012; 61: 2679-
       2690.
456.   Bosma M, Sparks LM, Hooiveld GJ, et al. Overexpression of PLIN5 in skeletal
       muscle promotes oxidative gene expression and intramyocellular lipid content without
       compromising insulin sensitivity. Biochim Biophys Acta 2013; 1831: 844-852.
457.   Minnaard R, Schrauwen P, Schaart G, et al. Adipocyte differentiation-related protein
       and OXPAT in rat and human skeletal muscle: involvement in lipid accumulation and
       type 2 diabetes mellitus. J Clin Endocrinol Metab 2009; 94: 4077-4085.
458.   MacPherson RE, Ramos SV, Vandenboom R, Roy BD, Peters SJ. Skeletal muscle
       PLIN proteins, ATGL and CGI-58, interactions at rest and following stimulated
       contraction. Am J Physiol Regul Integr Comp Physiol 2013; 304: R644-650.
459.   Corpeleijn E, Saris WH, Blaak EE. Metabolic flexibility in the development of insulin
       resistance and type 2 diabetes: effects of lifestyle. Obes Rev 2009; 10: 178-193.
460.   Kelley DE, Simoneau JA. Impaired free fatty acid utilization by skeletal muscle in
       non-insulin-dependent diabetes mellitus. J Clin Invest 1994; 94: 2349-2356.
461.   Blaak EE, Wagenmakers AJ, Glatz JF, et al. Plasma FFA utilization and fatty acid-
       binding protein content are diminished in type 2 diabetic muscle. Am J Physiol
       Endocrinol Metab 2000; 279: E146-154.
462.   Colberg SR, Simoneau JA, Thaete FL, Kelley DE. Skeletal muscle utilization of free
       fatty acids in women with visceral obesity. J Clin Invest 1995; 95: 1846-1853.
463.   Blaak EE, van Aggel-Leijssen DP, Wagenmakers AJ, Saris WH, van Baak MA.
       Impaired oxidation of plasma-derived fatty acids in type 2 diabetic subjects during
       moderate-intensity exercise. Diabetes 2000; 49: 2102-2107.
464.   Blaak EE, Van Baak MA, Kemerink GJ, Pakbiers MT, Heidendal GA, Saris WH. Beta-
       adrenergic stimulation of energy expenditure and forearm skeletal muscle
       metabolism in lean and obese men. Am J Physiol 1994; 267: E306-315.
465.   Corpeleijn E, Hessvik NP, Bakke SS, et al. Oxidation of intramyocellular lipids is
       dependent on mitochondrial function and the availability of extracellular fatty acids.
       Am J Physiol Endocrinol Metab 2010; 299: E14-22.
466.   van Loon LJ, Manders RJ, Koopman R, et al. Inhibition of adipose tissue lipolysis
       increases intramuscular lipid use in type 2 diabetic patients. Diabetologia 2005; 48:
       2097-2107.
467.   Haemmerle G, Moustafa T, Woelkart G, et al. ATGL-mediated fat catabolism
       regulates cardiac mitochondrial function via PPAR-alpha and PGC-1. Nat Med 2011;
       17: 1076-1085.
468.   Laforet P, Orngreen M, Preisler N, Andersen G, Vissing J. Blocked muscle fat
       oxidation during exercise in neutral lipid storage disease. Arch Neurol 2012; 69: 530-
       533.
469.   van de Weijer T, Havekes B, Bilet L, et al. Effects of bezafibrate treatment in a patient
       and a carrier with mutations in the PNPLA2 gene, causing neutral lipid storage
       disease with myopathy. Circ Res 2013; 112: e51-54.
470.   Engeli S, Birkenfeld AL, Badin PM, et al. Natriuretic peptides enhance the oxidative
       capacity of human skeletal muscle. J Clin Invest 2012; 122: 4675-4679.
471.   Ruderman NB, Saha AK, Vavvas D, Witters LA. Malonyl-CoA, fuel sensing, and
       insulin resistance. Am J Physiol 1999; 276: E1-E18.
472.   Rasmussen BB, Holmback UC, Volpi E, Morio-Liondore B, Paddon-Jones D, Wolfe
       RR. Malonyl coenzyme A and the regulation of functional carnitine
       palmitoyltransferase-1 activity and fat oxidation in human skeletal muscle. J Clin
       Invest 2002; 110: 1687-1693.
473.   Schrauwen P, van Aggel-Leijssen DP, Hul G, et al. The effect of a 3-month low-
       intensity endurance training program on fat oxidation and acetyl-CoA carboxylase-2
       expression. Diabetes 2002; 51: 2220-2226.
                                                                                           105
Chapter 2
474.   Mensink M, Blaak EE, Vidal H, De Bruin TW, Glatz JF, Saris WH. Lifestyle changes
       and lipid metabolism gene expression and protein content in skeletal muscle of
       subjects with impaired glucose tolerance. Diabetologia 2003; 46: 1082-1089.
475.   He L, Kim T, Long Q, et al. Carnitine palmitoyltransferase-1b deficiency aggravates
       pressure overload-induced cardiac hypertrophy caused by lipotoxicity. Circulation
       2012; 126: 1705-1716.
476.   Bruce CR, Hoy AJ, Turner N, et al. Overexpression of carnitine palmitoyltransferase-
       1 in skeletal muscle is sufficient to enhance fatty acid oxidation and improve high-fat
       diet-induced insulin resistance. Diabetes 2009; 58: 550-558.
477.   Mootha VK, Lindgren CM, Eriksson KF, et al. PGC-1alpha-responsive genes involved
       in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat
       Genet 2003; 34: 267-273.
478.   Patti ME, Butte AJ, Crunkhorn S, et al. Coordinated reduction of genes of oxidative
       metabolism in humans with insulin resistance and diabetes: Potential role of PGC1
       and NRF1. Proc Natl Acad Sci U S A 2003; 100: 8466-8471.
479.   Benton CR, Holloway GP, Han XX, et al. Increased levels of peroxisome proliferator-
       activated receptor gamma, coactivator 1 alpha (PGC-1alpha) improve lipid utilisation,
       insulin signalling and glucose transport in skeletal muscle of lean and insulin-resistant
       obese Zucker rats. Diabetologia 2010; 53: 2008-2019.
480.   Phielix E, Schrauwen-Hinderling VB, Mensink M, et al. Lower intrinsic ADP-
       stimulated mitochondrial respiration underlies in vivo mitochondrial dysfunction in
       muscle of male type 2 diabetic patients. Diabetes 2008; 57: 2943-2949.
481.   Lowell BB, Shulman GI. Mitochondrial dysfunction and type 2 diabetes. Science
       2005; 307: 384-387.
482.   Boushel R, Gnaiger E, Schjerling P, Skovbro M, Kraunsoe R, Dela F. Patients with
       type 2 diabetes have normal mitochondrial function in skeletal muscle. Diabetologia
       2007; 50: 790-796.
483.   van Tienen FH, Praet SF, de Feyter HM, et al. Physical activity is the key determinant
       of skeletal muscle mitochondrial function in type 2 diabetes. J Clin Endocrinol Metab
       2012; 97: 3261-3269.
484.   Gaster M. Reduced TCA flux in diabetic myotubes: A governing influence on the
       diabetic phenotype? Biochem Biophys Res Commun 2009; 387: 651-655.
485.   Ortenblad N, Mogensen M, Petersen I, et al. Reduced insulin-mediated citrate
       synthase activity in cultured skeletal muscle cells from patients with type 2 diabetes:
       evidence for an intrinsic oxidative enzyme defect. Biochim Biophys Acta 2005; 1741:
       206-214.
486.   Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. Impaired mitochondrial
       activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med
       2004; 350: 664-671.
487.   Befroy DE, Petersen KF, Dufour S, et al. Impaired mitochondrial substrate oxidation
       in muscle of insulin-resistant offspring of type 2 diabetic patients. Diabetes 2007; 56:
       1376-1381.
488.   Schooneman MG, Vaz FM, Houten SM, Soeters MR. Acylcarnitines: reflecting or
       inflicting insulin resistance? Diabetes 2013; 62: 1-8.
489.   Mercader J, Palou A, Bonet ML. Resveratrol enhances fatty acid oxidation capacity
       and reduces resistin and Retinol-Binding Protein 4 expression in white adipocytes. J
       Nutr Biochem 2011; 22: 828-834.
490.   Sun C, Zhang F, Ge X, et al. SIRT1 improves insulin sensitivity under insulin-
       resistant conditions by repressing PTP1B. Cell Metab 2007; 6: 307-319.
491.   Lagouge M, Argmann C, Gerhart-Hines Z, et al. Resveratrol improves mitochondrial
       function and protects against metabolic disease by activating SIRT1 and PGC-
       1alpha. Cell 2006; 127: 1109-1122.
106
                                                                                    Chapter 2
                                                                                           107
Chapter 2
511.   Marchetti P, Del Guerra S, Marselli L, et al. Pancreatic islets from type 2 diabetic
       patients have functional defects and increased apoptosis that are ameliorated by
       metformin. J Clin Endocrinol Metab 2004; 89: 5535-5541.
512.   Haataja L, Gurlo T, Huang CJ, Butler PC. Islet amyloid in type 2 diabetes, and the
       toxic oligomer hypothesis. Endocr Rev 2008; 29: 303-316.
513.   Giacca A, Xiao C, Oprescu AI, Carpentier AC, Lewis GF. Lipid-induced pancreatic
       beta-cell dysfunction: focus on in vivo studies. Am J Physiol Endocrinol Metab 2011;
       300: E255-262.
514.   Meier JJ, Breuer TG, Bonadonna RC, et al. Pancreatic diabetes manifests when beta
       cell area declines by approximately 65% in humans. Diabetologia 2012; 55: 1346-
       1354.
515.   Donath MY, Ehses JA, Maedler K, et al. Mechanisms of beta-cell death in type 2
       diabetes. Diabetes 2005; 54 Suppl 2: S108-113.
516.   Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Are oxidative stress-activated
       signaling pathways mediators of insulin resistance and beta-cell dysfunction?
       Diabetes 2003; 52: 1-8.
517.   Green K, Brand MD, Murphy MP. Prevention of mitochondrial oxidative damage as a
       therapeutic strategy in diabetes. Diabetes 2004; 53 Suppl 1: S110-118.
518.   Maedler K, Sergeev P, Ris F, et al. Glucose-induced beta cell production of IL-1beta
       contributes to glucotoxicity in human pancreatic islets. J Clin Invest 2002; 110: 851-
       860.
519.   Donath MY, Storling J, Berchtold LA, Billestrup N, Mandrup-Poulsen T. Cytokines
       and beta-cell biology: from concept to clinical translation. Endocr Rev 2008; 29: 334-
       350.
520.   Scheuner D, Kaufman RJ. The unfolded protein response: a pathway that links
       insulin demand with beta-cell failure and diabetes. Endocr Rev 2008; 29: 317-333.
521.   Back SH, Kaufman RJ. Endoplasmic reticulum stress and type 2 diabetes. Annu Rev
       Biochem 2012; 81: 767-793.
522.   Back SH, Kang SW, Han J, Chung HT. Endoplasmic reticulum stress in the beta-cell
       pathogenesis of type 2 diabetes. Exp Diabetes Res 2012; 2012: 618396.
523.   Evans-Molina C, Hatanaka M, Mirmira RG. Lost in translation: endoplasmic reticulum
       stress and the decline of beta-cell health in diabetes mellitus. Diabetes Obes Metab
       2013; 15 Suppl 3: 159-169.
524.   Muoio DM, Newgard CB. Mechanisms of disease: molecular and metabolic
       mechanisms of insulin resistance and beta-cell failure in type 2 diabetes. Nat Rev
       Mol Cell Biol 2008; 9: 193-205.
525.   Supale S, Li N, Brun T, Maechler P. Mitochondrial dysfunction in pancreatic beta
       cells. Trends Endocrinol Metab 2012; 23: 477-487.
526.   Sharma RB, Alonso LC. Lipotoxicity in the pancreatic Beta cell: not just survival and
       function, but proliferation as well? Curr Diab Rep 2014; 14: 492.
527.   Oh YS, Jun HS. Role of bioactive food components in diabetes prevention: effects on
       Beta-cell function and preservation. Nutr Metab Insights 2014; 7: 51-59.
528.   Nogueira JP, Brites FD. Role of enterocytes in dyslipidemia of insulin-resistant states.
       Endocrinol Nutr 2013; 60: 179-189.
529.   Ding S, Lund PK. Role of intestinal inflammation as an early event in obesity and
       insulin resistance. Curr Opin Clin Nutr Metab Care 2011; 14: 328-333.
530.   Duez H, Lamarche B, Uffelman KD, Valero R, Cohn JS, Lewis GF. Hyperinsulinemia
       is associated with increased production rate of intestinal apolipoprotein B-48-
       containing lipoproteins in humans. Arterioscler Thromb Vasc Biol 2006; 26: 1357-
       1363.
531.   Abumrad NA, Davidson NO. Role of the gut in lipid homeostasis. Physiol Rev 2012;
       92: 1061-1085.
108
                                                                                    Chapter 2
532.   Iqbal J, Hussain MM. Intestinal lipid absorption. Am J Physiol Endocrinol Metab 2009;
       296: E1183-1194.
533.   Duee PH, Darcy-Vrillon B, Blachier F, Morel MT. Fuel selection in intestinal cells.
       Proc Nutr Soc 1995; 54: 83-94.
534.   Fleming SE, Fitch MD, DeVries S, Liu ML, Kight C. Nutrient utilization by cells
       isolated from rat jejunum, cecum and colon. J Nutr 1991; 121: 869-878.
535.   Storch J, Zhou YX, Lagakos WS. Metabolism of apical versus basolateral sn-2-
       monoacylglycerol and fatty acids in rodent small intestine. J Lipid Res 2008; 49:
       1762-1769.
536.   Demignot S, Beilstein F, Morel E. Triglyceride-rich lipoproteins and cytosolic lipid
       droplets in enterocytes: key players in intestinal physiology and metabolic disorders.
       Biochimie 2014; 96: 48-55.
537.   Zhu J, Lee B, Buhman KK, Cheng JX. A dynamic, cytoplasmic triacylglycerol pool in
       enterocytes revealed by ex vivo and in vivo coherent anti-Stokes Raman scattering
       imaging. J Lipid Res 2009; 50: 1080-1089.
538.   Bouchoux J, Beilstein F, Pauquai T, et al. The proteome of cytosolic lipid droplets
       isolated from differentiated Caco-2/TC7 enterocytes reveals cell-specific
       characteristics. Biol Cell 2011; 103: 499-517.
539.   Rivellese AA, De Natale C, Di Marino L, et al. Exogenous and endogenous
       postprandial lipid abnormalities in type 2 diabetic patients with optimal blood glucose
       control and optimal fasting triglyceride levels. J Clin Endocrinol Metab 2004; 89:
       2153-2159.
540.   Kolovou GD, Anagnostopoulou KK, Pavlidis AN, et al. Postprandial lipemia in men
       with metabolic syndrome, hypertensives and healthy subjects. Lipids Health Dis
       2005; 4: 21.
541.   Patsch JR, Miesenbock G, Hopferwieser T, et al. Relation of triglyceride metabolism
       and coronary artery disease. Studies in the postprandial state. Arterioscler Thromb
       1992; 12: 1336-1345.
542.   Miller M, Stone NJ, Ballantyne C, et al. Triglycerides and cardiovascular disease: a
       scientific statement from the American Heart Association. Circulation 2011; 123:
       2292-2333.
543.   Kimura R, Takahashi N, Murota K, et al. Activation of peroxisome proliferator-
       activated receptor-alpha (PPARalpha) suppresses postprandial lipidemia through
       fatty acid oxidation in enterocytes. Biochem Biophys Res Commun 2011; 410: 1-6.
544.   Mori T, Kondo H, Hase T, Tokimitsu I, Murase T. Dietary fish oil upregulates intestinal
       lipid metabolism and reduces body weight gain in C57BL/6J mice. J Nutr 2007; 137:
       2629-2634.
545.   van Schothorst EM, Flachs P, Franssen-van Hal NL, et al. Induction of lipid oxidation
       by polyunsaturated fatty acids of marine origin in small intestine of mice fed a high-fat
       diet. BMC Genomics 2009; 10: 110.
546.   Murase T, Aoki M, Wakisaka T, Hase T, Tokimitsu I. Anti-obesity effect of dietary
       diacylglycerol in C57BL/6J mice: dietary diacylglycerol stimulates intestinal lipid
       metabolism. J Lipid Res 2002; 43: 1312-1319.
547.   Warnakula S, Hsieh J, Adeli K, Hussain MM, Tso P, Proctor SD. New insights into
       how the intestine can regulate lipid homeostasis and impact vascular disease:
       frontiers for new pharmaceutical therapies to lower cardiovascular disease risk. Can
       J Cardiol 2011; 27: 183-191.
548.   Xie Y, Newberry EP, Young SG, et al. Compensatory increase in hepatic lipogenesis
       in mice with conditional intestine-specific Mttp deficiency. J Biol Chem 2006; 281:
       4075-4086.
549.   Mera Y, Odani N, Kawai T, et al. Pharmacological characterization of diethyl-2-({3-
       dimethylcarbamoyl-4-[(4'-trifluoromethylbiphenyl-2-carbonyl)amino]p
                                                                                           109
Chapter 2
110
                                                                                 Chapter 2
569.   Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity
       alters gut microbial ecology. Proc Natl Acad Sci U S A 2005; 102: 11070-11075.
570.   Schwiertz A, Taras D, Schafer K, et al. Microbiota and SCFA in lean and overweight
       healthy subjects. Obesity (Silver Spring) 2010; 18: 190-195.
571.   Karlsson FH, Tremaroli V, Nookaew I, et al. Gut metagenome in European women
       with normal, impaired and diabetic glucose control. Nature 2013; 498: 99-103.
572.   Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in
       type 2 diabetes. Nature 2012; 490: 55-60.
573.   Carvalho BM, Guadagnini D, Tsukumo DM, et al. Modulation of gut microbiota by
       antibiotics improves insulin signalling in high-fat fed mice. Diabetologia 2012; 55:
       2823-2834.
574.   Henao-Mejia J, Elinav E, Jin C, et al. Inflammasome-mediated dysbiosis regulates
       progression of NAFLD and obesity. Nature 2012; 482: 179-185.
575.   Collado MC, Isolauri E, Laitinen K, Salminen S. Distinct composition of gut microbiota
       during pregnancy in overweight and normal-weight women. Am J Clin Nutr 2008; 88:
       894-899.
576.   Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that
       regulates fat storage. Proc Natl Acad Sci U S A 2004; 101: 15718-15723.
577.   Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-
       associated gut microbiome with increased capacity for energy harvest. Nature 2006;
       444: 1027-1031.
578.   Ferraris RP, Vinnakota RR. Intestinal nutrient transport in genetically obese mice. Am
       J Clin Nutr 1995; 62: 540-546.
579.   Warwick BP, Romsos DR. Energy balance in adrenalectomized ob/ob mice: effects
       of dietary starch and glucose. Am J Physiol 1988; 255: R141-148.
580.   Vrieze A, Van Nood E, Holleman F, et al. Transfer of intestinal microbiota from lean
       donors increases insulin sensitivity in individuals with metabolic syndrome.
       Gastroenterology 2012; 143: 913-916 e917.
581.   Diamant M, Blaak EE, de Vos WM. Do nutrient-gut-microbiota interactions play a role
       in human obesity, insulin resistance and type 2 diabetes? Obes Rev 2011; 12: 272-
       281.
582.   Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol 2004; 4: 499-511.
583.   Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and
       insulin resistance. Diabetes 2007; 56: 1761-1772.
584.   Mortensen PB, Clausen MR. Short-chain fatty acids in the human colon: relation to
       gastrointestinal health and disease. Scand J Gastroenterol Suppl 1996; 216: 132-
       148.
585.   Cummings JH, Englyst HN, Wiggins HS. The role of carbohydrates in lower gut
       function. Nutr Rev 1986; 44: 50-54.
586.   Cummings JH, Englyst HN. Fermentation in the human large intestine and the
       available substrates. Am J Clin Nutr 1987; 45: 1243-1255.
587.   Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT. Short chain fatty
       acids in human large intestine, portal, hepatic and venous blood. Gut 1987; 28: 1221-
       1227.
588.   Wong JM, de Souza R, Kendall CW, Emam A, Jenkins DJ. Colonic health:
       fermentation and short chain fatty acids. J Clin Gastroenterol 2006; 40: 235-243.
589.   Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: roles of
       resistant starch and nonstarch polysaccharides. Physiol Rev 2001; 81: 1031-1064.
590.   Macfarlane GT, Macfarlane S. Fermentation in the human large intestine: its
       physiologic consequences and the potential contribution of prebiotics. J Clin
       Gastroenterol 2011; 45 Suppl: S120-127.
591.   Musso G, Gambino R, Cassader M. Interactions between gut microbiota and host
       metabolism predisposing to obesity and diabetes. Annu Rev Med 2011; 62: 361-380.
                                                                                        111
Chapter 2
592.   Scheppach W, Bartram P, Richter A, et al. Effect of short-chain fatty acids on the
       human colonic mucosa in vitro. JPEN J Parenter Enteral Nutr 1992; 16: 43-48.
593.   Zaibi MS, Stocker CJ, O'Dowd J, et al. Roles of GPR41 and GPR43 in leptin
       secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett 2010;
       584: 2381-2386.
594.   Xiong Y, Miyamoto N, Shibata K, et al. Short-chain fatty acids stimulate leptin
       production in adipocytes through the G protein-coupled receptor GPR41. Proc Natl
       Acad Sci U S A 2004; 101: 1045-1050.
595.   Al-Lahham SH, Roelofsen H, Priebe M, et al. Regulation of adipokine production in
       human adipose tissue by propionic acid. Eur J Clin Invest 2010; 40: 401-407.
596.   Vinolo MA, Rodrigues HG, Hatanaka E, Sato FT, Sampaio SC, Curi R. Suppressive
       effect of short-chain fatty acids on production of proinflammatory mediators by
       neutrophils. J Nutr Biochem 2011; 22: 849-855.
597.   Brown AJ, Goldsworthy SM, Barnes AA, et al. The Orphan G protein-coupled
       receptors GPR41 and GPR43 are activated by propionate and other short chain
       carboxylic acids. J Biol Chem 2003; 278: 11312-11319.
598.   Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal
       bacteria. J Lipid Res 2006; 47: 241-259.
599.   Stacey M, Webb M. Studies on the antibacterial properties of the bile acids and some
       compounds derived from cholanic acid. Proc R Soc Med 1947; 134: 523-537.
600.   Kurdi P, Kawanishi K, Mizutani K, Yokota A. Mechanism of growth inhibition by free
       bile acids in lactobacilli and bifidobacteria. J Bacteriol 2006; 188: 1979-1986.
601.   David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the
       human gut microbiome. Nature 2014; 505: 559-563.
602.   Cani PD, Lecourt E, Dewulf EM, et al. Gut microbiota fermentation of prebiotics
       increases satietogenic and incretin gut peptide production with consequences for
       appetite sensation and glucose response after a meal. Am J Clin Nutr 2009; 90:
       1236-1243.
603.   Davis LM, Martinez I, Walter J, Hutkins R. A dose dependent impact of prebiotic
       galactooligosaccharides on the intestinal microbiota of healthy adults. Int J Food
       Microbiol 2010; 144: 285-292.
604.   Cani PD, Neyrinck AM, Fava F, et al. Selective increases of bifidobacteria in gut
       microflora improve high-fat-diet-induced diabetes in mice through a mechanism
       associated with endotoxaemia. Diabetologia 2007; 50: 2374-2383.
605.   Roberfroid M, Gibson GR, Hoyles L, et al. Prebiotic effects: metabolic and health
       benefits. Br J Nutr 2010; 104 Suppl 2: S1-63.
606.   Pouteau E, Nguyen P, Ballevre O, Krempf M. Production rates and metabolism of
       short-chain fatty acids in the colon and whole body using stable isotopes. Proc Nutr
       Soc 2003; 62: 87-93.
607.   Kellow NJ, Coughlan MT, Reid CM. Metabolic benefits of dietary prebiotics in human
       subjects: a systematic review of randomised controlled trials. Br J Nutr 2014; 111:
       1147-1161.
608.   Chen CH, Wang Y, Nakatsuji T, et al. An innate bactericidal oleic acid effective
       against skin infection of methicillin-resistant Staphylococcus aureus: a therapy
       concordant with evolutionary medicine. J Microbiol Biotechnol 2011; 21: 391-399.
609.   Desbois AP, Smith VJ. Antibacterial free fatty acids: activities, mechanisms of action
       and biotechnological potential. Appl Microbiol Biotechnol 2010; 85: 1629-1642.
610.   Alcock J, Franklin ML, Kuzawa CW. Nutrient signaling: evolutionary origins of the
       immune-modulating effects of dietary fat. Q Rev Biol 2012; 87: 187-223.
611.   Fava F, Gitau R, Griffin BA, Gibson GR, Tuohy KM, Lovegrove JA. The type and
       quantity of dietary fat and carbohydrate alter faecal microbiome and short-chain fatty
       acid excretion in a metabolic syndrome 'at-risk' population. Int J Obes (Lond) 2013;
       37: 216-223.
112
                                                                                     Chapter 2
612.   Simoes CD, Maukonen J, Kaprio J, Rissanen A, Pietilainen KH, Saarela M. Habitual
       dietary intake is associated with stool microbiota composition in monozygotic twins. J
       Nutr 2013; 143: 417-423.
613.   Santacruz A, Marcos A, Warnberg J, et al. Interplay between weight loss and gut
       microbiota composition in overweight adolescents. Obesity (Silver Spring) 2009; 17:
       1906-1915.
614.   Brinkworth GD, Noakes M, Clifton PM, Bird AR. Comparative effects of very low-
       carbohydrate, high-fat and high-carbohydrate, low-fat weight-loss diets on bowel
       habit and faecal short-chain fatty acids and bacterial populations. Br J Nutr 2009;
       101: 1493-1502.
615.   de Wit N, Derrien M, Bosch-Vermeulen H, et al. Saturated fat stimulates obesity and
       hepatic steatosis and affects gut microbiota composition by an enhanced overflow of
       dietary fat to the distal intestine. Am J Physiol Gastrointest Liver Physiol 2012; 303:
       G589-599.
616.   Shen W, Gaskins HR, McIntosh MK. Influence of dietary fat on intestinal microbes,
       inflammation, barrier function and metabolic outcomes. J Nutr Biochem 2014; 25:
       270-280.
617.   Sayin SI, Wahlstrom A, Felin J, et al. Gut microbiota regulates bile acid metabolism
       by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR
       antagonist. Cell Metab 2013; 17: 225-235.
618.   Kuo SM, Merhige PM, Hagey LR. The effect of dietary prebiotics and probiotics on
       body weight, large intestine indices, and fecal bile acid profile in wild type and IL10-/-
       mice. PLoS One 2013; 8: e60270.
619.   Kim JH, Bae KH, Choi YK, et al. Fibroblast growth factor 21 analogue LY2405319
       lowers blood glucose in streptozotocin-induced insulin-deficient diabetic mice by
       restoring brown adipose tissue function. Diabetes Obes Metab 2015; 17: 161-169.
                                                                                            113
CHAPTER 3
Improved insulin sensitivity with
angiotensin receptor neprilysin inhibition
in individuals with obesity and
hypertension
Jordan J., Stinkens R.*, Jax T.*, Engeli S., Blaak E.E., May M.,
Havekes B., Schindler C., Pal P., Heise T., Goossens G.H.,
Langenickel T.H.
* Shared authorship
ABSTRACT
Natriuretic peptide (NP) deficiency and sustained renin-angiotensin system
activation are associated with impaired oxidative metabolism and predispose to
type-2 diabetes. We hypothesized that sacubitril/valsartan (LCZ696), which
augments NP through neprilysin inhibition while blocking angiotensin II type-1
(AT1)-receptors, improves insulin sensitivity, lipid mobilization, and oxidation. After
8 weeks of treatment of obese patients with hypertension, sacubitril/valsartan 400
mg q.d., but not amlodipine 10 mg q.d., was associated with a significant increase
from baseline in the insulin sensitivity index (hyperinsulinemic-euglycemic clamp),
and tended to be higher in patients treated with sacubitril/valsartan compared to
amlodipine. Abdominal adipose tissue interstitial glycerol concentrations increased
with sacubitril/valsartan, but decreased with amlodipine. Whole-body lipolysis and
substrate oxidation did not change with either treatment. Results confirm that
sacubitril/valsartan treatment leads to a metabolic benefit in the study population
and supports the relevance of neprilysin inhibition along with AT1-receptor
blockade in the regulation of human glucose and lipid metabolism.
116
                                                                            Chapter 3
INTRODUCTION
Obesity, type-2 diabetes mellitus (T2DM), and cardiovascular diseases are
interrelated. Patients with obesity and T2DM are at increased risk for arterial
hypertension and heart failure [1, 2], whereas patients with arterial hypertension or
heart ,failure frequently exhibit impaired muscular oxidative capacity predisposing
them to insulin resistance through accumulation of toxic lipid intermediates [3, 4].
The endopeptidase neprilysin is ubiquitously expressed, including in human
adipocytes [5]. Its plasma activity positively correlates with measures of obesity
and insulin resistance in humans, implicating neprilysin in the regulation of
cardiometabolic crosstalk presumably through cleavage of neprilysin substrates
with cardiometabolic effects, such as natriuretic peptides (NPs) and angiotensin II
[6]. Indeed, patients with obesity [7] or heart failure exhibit absolute or relative
natriuretic peptide deficiency, respectively. Circulating atrial natriuretic peptide
levels are inversely related to plasma glucose and insulin concentrations, and low
atrial natriuretic peptide levels predict the development of both arterial hypertension
and diabetes mellitus in humans [8]. Furthermore, polymorphisms in the genes
encoding atrial natriuretic peptide and B-type natriuretic peptide contribute to the
variability in the risk for type 2 diabetes after adjustment for age, gender, and body
mass index [9, 10]. NP promotes lipid mobilization from adipose tissue [11],
increases postprandial lipid oxidation [12], elicits adiponectin release, and
enhances muscular oxidative capacity [13, 14]. In contrast to atrial natriuretic
peptide, angiotensin II promotes insulin resistance, and angiotensin II type-1 (AT1)-
receptor blockade modestly improved insulin sensitivity and pancreatic beta-cell
function in humans [15].
Sacubitril/valsartan (LCZ696), a novel angiotensin receptor neprilysin inhibitor, was
recently approved in the United States, Europe, and many other countries to
reduce the risk of cardiovascular death and hospitalizations in patients with chronic
heart failure (New York Heart Association classes II–IV) and reduced ejection
fraction [16]. Upon oral administration, sacubitril/valsartan provides exposure to
sacubitril (AHU377), a prodrug that is rapidly metabolized to the biologically active
neprilysin inhibitor, sacubitrilat (LBQ657), and to the AT1-receptor blocker
valsartan. This active drug augments neprilysin substrates, such as natriuretic
peptides, while inhibiting AT 1-receptor mediated responses [17].
We hypothesized that sacubitril/valsartan, through its combined mechanism of
action, improves peripheral insulin sensitivity and increases lipid mobilization from
subcutaneous abdominal adipose tissue in patients with obesity and arterial
hypertension.
We conducted a mechanistic metabolic profiling study to investigate the effects of
8-week treatment with sacubitril/valsartan and amlodipine on peripheral insulin
sensitivity, abdominal subcutaneous adipose tissue lipolysis, whole-body lipolysis,
and energy expenditure and substrate oxidation in patients with obesity and
elevated blood pressure. In this study, the patient population served as an
exemplary human model for relative natriuretic peptide deficiency and impaired
insulin sensitivity, and was specifically selected to support the comparison of
sacubitril/valsartan with a metabolically neutral comparator, amlodipine.
                                                                                   117
Chapter 3
RESULTS
Subjects
Of the 98 patients enrolled, (sacubitril/valsartan=50; amlodipine=48), 92 patients
(93.9%) completed the study. All patients were included in the safety analysis set
(Figure 1). Patients with missing baseline or post-treatment assessments were
excluded from the pharmacodynamic analysis set, resulting in a maximum of 96
patients (sacubitril/valsartan=50; amlodipine=46) included in the pharmacodynamic
analysis set.
Patient demographics and baseline characteristics are summarized in Table 1 and
were comparable between the groups.
*Indicates those who had to discontinue antihypertensive medication. # Indicates those who had to
discontinue before the first dose. § Indicates subcutaneous adipose tissue microdialysis.
‡ Indicates indirect calorimetry by Spirometry. EOS, end of study; HEGC, hyperinsulinemic-euglycemic
glucose clamp.
118
                                        Table 1. Patient demographics and baseline characteristics
                         Parameter                                  LCZ696 (400 mg) N = 50                         Amlodipine (10 mg) N = 48
      Data are mean (SD) unless specified otherwise. BMI, body mass index; bpm, beats per minute; GIR, glucose infusion rate; msSBP, mean seated
                                                                                                                                                    Chapter 3
119
      systolic blood pressure; msDBP, mean seated diastolic blood pressure; SI, sensitivity index.   * N = 45; † N = 48; ‡ N = 42.
Chapter 3
120
                                                                                        Chapter 3
Figure 2. Mean changes from baseline in insulin sensitivity variables after 8-week
treatment with sacubitril/valsartan vs. amlodipine.
Insulin sensitivity index (A); glucose infusion rate (GIR) (B); GIR by body weight (C); and metabolic
clearance rate (D). CI, confidence interval.
                                                                                                121
Chapter 3
Figure 4. Adjusted geometric mean ratios of local adipose tissue lipolysis variables
after 8-week treatment with sacubitril/valsartan and amlodipine.
Ethanol ratio (A); glycerol (B); glucose (C); and lactate (D). Data were analyzed for repeated
measurements on log transformed values with treatment, visit, time and treatment*visit*time interaction
as fixed effect assuming an unstructured covariance. Subjects with missing data were excluded from
the analysis. The number of missing values ranged from 3 to 6 and 3 to 7 across all variables in the
sacubitril/valsartan and amlodipine groups, respectively. CI, confidence interval.
122
                                                                         Chapter 3
Whole-body lipolysis
The rate of appearance (95% CI) of endogenous plasma glycerol at baseline on
                                                                           -1
day 1 was 172.8 (158.0–189.0) and 200.6 (182.4–220.6) µmol/kg.min in the
sacubitril/valsartan and amlodipine groups, respectively. Whole-body glycerol rate
of appearance was not significantly affected by amlodipine treatment, but was
significantly lower at week 8 compared to baseline in the sacubitril/valsartan group
at the 30 + 45-minute time point (geometric mean ratio (95% CI), 0.93 (0.87–1.00),
p=0.045; Figure 5). There was no change from baseline in the sacubitril/valsartan
group at the 45-minute time point. There were no differences between treatment
groups in whole-body lipolysis after 8 weeks of treatment.
                                                                               123
Chapter 3
Blood pressure
Baseline data for mean seated blood pressure is presented in Table 1. At week 8,
mean seated blood pressure had decreased more in the sacubitril/valsartan group
compared with the amlodipine group (systolic blood pressure: -21.0 ± 16.1 mmHg
vs. -12.4 ± 14.7 mmHg; diastolic blood pressure: -12.4 ± 9.1 mmHg vs. -10.0 ± 7.8
mmHg; Figure 6).
Safety Assessments
Of the 98 patients, 67 patients (68.4%) experienced at least one adverse effect
(AE). Most AEs were mild or moderate in intensity. The overall AE incidence was
lower in the sacubitril/valsartan compared with the amlodipine group (60.0% vs.
77.1%; Supplementary Table S2). Five patients discontinued due to AEs. Two
patients discontinued due to a serious adverse event, which was unrelated to the
study drug (ruptured cerebral aneurysm in one patient in the sacubitril/valsartan
group and nephrolithiasis in one patient in the amlodipine group). Three patients
discontinued due to AEs (pruritus in one patient in the sacubitril/valsartan group,
and a single case of hypertension and peripheral edema in the amlodipine group).
The most commonly occurring AEs were nasopharyngitis, peripheral edema, and
headache, each of which had an overall incidence of >10% (Supplementary Table
S2). AEs suspected to be related to the study drug by the investigators occurred
more frequently in the amlodipine group than in patients receiving
sacubitril/valsartan (46% vs. 24%). Peripheral edema was more common in the
amlodipine group, whereas pruritus occurred only in the sacubitril/valsartan group
(Supplementary Table S2). Two patients in the sacubitril/valsartan group reported
mild orthostatic hypertension, both of which resolved by the end of the study. Both
of these events were suspected to be related to the study drug. No deaths were
reported in the study.
124
                                                                      Chapter 3
Figure 6. Arithmetic mean (SD) of mean seated systolic blood pressure (SBP) (A);
and mean seated diastolic blood pressure (DBP) (B) after 8-week treatment with
sacubitril/valsartan and amlodipine.
                                                                            125
Chapter 3
DISCUSSION
Treatment with sacubitril/valsartan compared with amlodipine for 8 weeks improved
peripheral insulin sensitivity in obese patients with elevated blood pressure. This
improvement was not explained by changes in body weight or waist circumference.
Furthermore, sacubitril/valsartan significantly increased abdominal subcutaneous
adipose tissue lipolysis without changing whole-body lipolysis and plasma free fatty
acid concentrations. Therefore, this study is the first to show that simultaneous
neprilysin inhibition and AT 1-receptor blockade regulates glucose and lipid
metabolism in humans, supporting the concept that neprilysin substrates and AT 1-
receptor blockade have an important role in the crosstalk between the
cardiovascular system and metabolism in humans. These findings imply that
sacubitril/valsartan may improve cardiovascular and metabolic health in patients
with cardiovascular disease.
The metabolic effects of sacubitril/valsartan observed here could be mediated
through increased availability of neprilysin substrates, AT 1-receptor inhibition, or
both mechanisms combined. Indeed, individuals with obesity exhibit increased
renin angiotensin system (RAS) activity [18, 19] both systemically and at the tissue
level. Although genetically modified animals overexpressing RAS components are
characterized by insulin resistance that improved with pharmacological RAS
inhibition [20], the improvement in insulin resistance after RAS inhibition in humans
has been less consistent [16]. Among patients with impaired glucose tolerance and
cardiovascular disease or risk factors, the use of valsartan for 5 years, along with
lifestyle modification, led to a 14% relative reduction in T2DM incidence [21].
However, ramipril compared with placebo did not reduce the risk of new onset
T2DM in patients with impaired fasting glucose levels [22]. Overall, the
improvement of glucose metabolism after RAS inhibition seems to be modest and
may not solely explain the improvement in insulin sensitivity observed with
sacubitril/valsartan treatment. Moreover, AT1-receptor blockade does not increase
abdominal subcutaneous adipose tissue lipolysis, which was observed with
sacubitril/valsartan treatment [23, 24].
The present study suggests that neprilysin inhibition contributed to the metabolic
effect of sacubitril/valsartan treatment. Neprilysin degrades multiple peptides that
have the potential to modulate lipid and glucose metabolism, such as natriuretic
peptides, bradykinin, endothelin-1, and glucagon-like peptide 1. Therefore, we
cannot distinguish the contribution of individual neprilysin substrates to the
observed metabolic response. Eventually, the pharmacological effect of neprilysin
inhibition will depend on the net effect on all biologically relevant neprilysin
substrates. For example, bradykinin has been suggested to improve insulin
sensitivity and attenuate lipolysis, whereas endothelin-1 promotes insulin
resistance and increases lipolysis [25, 26]. Given the minor contribution of
neprilysin relative to angiotensin converting enzyme and aminopeptidase [27] and
the proven clinical safety of sacubitril/valsartan [16], it seems unlikely that an
increase in bradykinin explains the observed metabolic response. Likewise,
sacubitril/valsartan was shown to decrease, not increase, endothelin-1 plasma
concentrations in patients with heart failure and reduced ejection fraction. Indeed,
neprilysin contributes to both endothelin-1 formation from its precursors and
endothelin-1 degradation [28]. Hence, an involvement of endothelin-1 in mediation
126
                                                                             Chapter 3
                                                                                   127
Chapter 3
128
                                                                          Chapter 3
CONCLUSION
In conclusion, our study demonstrated that sacubitril/valsartan treatment compared
with amlodipine resulted in improved peripheral insulin sensitivity and increased
abdominal subcutaneous adipose tissue lipid mobilization, but did not show
significant effects on whole-body lipolysis, energy expenditure, and substrate
oxidation. Moreover, our study highlights the utility of mechanistic profiling studies
in discerning metabolic drug actions that could go undetected in clinical
development.
                                                                                 129
Chapter 3
METHODS
130
                                                                          Chapter 3
                                                               2
of insulin (a priming dose over the first 10 minutes (103 mU/m /min at 0–5 minutes
                 2
and 57 mU/m /min at 5–10 minutes) followed by a continuous infusion at 40
       2
mU/m /min thereafter until 2 hours) and a variable glucose infusion to achieve
steady-state plasma insulin levels while maintaining blood glucose levels at 90.1
mg/dL (5.0 mmol/L). Blood samples were collected continuously (automated
clamp) or at approximately 5 minute intervals (manual clamp) to determine glucose
levels during hyperinsulinemic- euglycemic clamp testing. The last 30 minutes
(minutes 90 - 120) of the clamp were considered as the steady-state period and the
mean GIR was calculated for this period.
Insulin SI was calculated from steady-state GIRs and plasma insulin and glucose
concentrations (SI: glucose infusion rate/(plasma glucose x plasma insulin),
µg/kg*min/(mmol/L*pmol/L)). Whole-body glucose disposal rate (M, mg/min) was
calculated from mean GIRs at steady state to assess peripheral insulin sensitivity.
Finally, the metabolic clearance rate was calculated from the M-value and the
mean blood glucose concentration at steady state (metabolic clearance rate:
M/blood glucose, min*mmol). The glucose disposal rate was expressed per unit of
insulin at steady state, calculated from M-value and plasma insulin concentrations
(M/I, U/min).
Lipolysis
Lipolysis was assessed on day 1 before the first dosage and on day 57. One
microdialysis probe was placed in the subcutaneous adipose tissue 6–8 cm lateral
from the umbilicus with the patient under local anesthesia and sterile conditions, as
described previously [38]. The process comprised a recovery phase of 60 minutes
during which the probes were infused with perfusion solution (50 mM ethanol + T1
perfusion solution; CMA Microdialysis AB, Stockholm, Sweden) at a flow rate of 0.3
µL/min followed by a flow calibration phase of 60 minutes. During the flow
calibration phase, the perfusion rate was maintained at 0.3 µL/min for the first 30
minutes and increased thereafter to 2.0 µL/min for the remaining 30 minutes.
Microdialysate samples were collected at 30 minutes and 45 minutes in the flow
calibration phase.
Glycerol (an indicator of local lipolysis), glucose, and lactate concentrations were
measured in microdialysates. The ethanol ratio (ratio of ethanol concentration in
dialysate to ethanol concentration in perfusate) was measured as an indicator of
adipose tissue blood flow.
                                                       2
Whole-body lipolysis was assessed using [1,1,2,3,3- H]-glycerol tracer kinetics. A
glycerol tracer bolus (3.0 µg/kg) was injected at the start of the microdialysis flow
calibration phase, followed by a continuous infusion (0.1 µg/kg/min) until the end of
the sampling period. Blood samples were collected at 0, 30, 40, 50, and 60
minutes during the flow calibration phase and at 15, 30, and 45 minutes thereafter.
The rate of appearance of endogenous glycerol was calculated as the ratio of the
glycerol tracer infusion rate to the plasma glycerol tracer enrichment. At steady
state, glycerol rate of appearance was calculated from glycerol tracer enrichment
using Steele’s equation.
                                                                                 131
Chapter 3
Statistical analysis
Based on 80% power to detect a difference of 0.1 in SI at week 8 between the two
treatment groups at 2.5% significance level with two-sided alternative, 90 subjects
(45 in each group) were needed to complete the study. SI at 8 weeks was analyzed
using the analysis of covariance model with treatment as a fixed effect and
baseline insulin sensitivity as a covariate for assessing the difference in the mean
effect of sacubitril/valsartan vs. amlodipine. For the purpose of a two-sided
hypothesis of equality of treatment effects at 2.5% level of significance after the
completion of the study, the point estimate and 97.5% CI for the difference along
with the p-value for equality of the two treatments were reported. The smaller level
of significance (2.5%) was chosen to account for the conduct of an interim analysis.
Differences between treatments for change in SI from baseline were analyzed as
described above. Similar analyses at 5% level of significance were performed for
all other clamp-derived measures of insulin sensitivity.
For abdominal subcutaneous adipose tissue microdialysate data (ethanol ratio,
dialysate lactate, dialysate glucose, and plasma glycerol, glycerol, NEFA, glucose,
insulin, adrenalin, and noradrenalin) at rest, data were analyzed using repeated
measures analysis on log transformed values with treatment, visit, time and
treatment*visit*time interaction as fixed effects. Ratio to day 1 (day 57 vs. day 1) at
each treatment and ratio of sacubitril/valsartan vs. amlodipine for ratio to day 1
along with the corresponding 95% CIs and p-values are presented for 30 and 45-
minute microdialysis time points combined. Comparison of 30 and 45-minute
microdialysis time points demonstrated steady state of the microdialysis
experiment.
Oxidative metabolism was analyzed using the analysis of covariance with
treatment as fixed effect and baseline as covariate and the treatment mean
difference with 95% CIs and p-values are reported.
For biomarkers, data were analyzed using repeated measures of analysis on log
transformed values with treatment, visit, time and treatment*visit*time interaction as
fixed effects. Ratio to day 1 (day 57 vs. day 1) at each treatment and ratio of
sacubitril/valsartan vs. amlodipine for ratio to day 1 along with the corresponding
95% CIs and p-values are presented.
132
                                                                                  Chapter 3
REFERENCES
1.    Jordan J, Yumuk V, Schlaich M, Nilsson PM, Zahorska-Markiewicz B, Grassi G, et al.
      Joint statement of the European Association for the Study of Obesity and the
      European Society of Hypertension: obesity and difficult to treat arterial hypertension.
      J Hypertens. 2012;30(6):1047-55.
2.    Del Gobbo LC, Kalantarian S, Imamura F, Lemaitre R, Siscovick DS, Psaty BM, et al.
      Contribution of Major Lifestyle Risk Factors for Incident Heart Failure in Older Adults:
      The Cardiovascular Health Study. JACC Heart Fail. 2015;3(7):520-8.
3.    Samuel VT, Petersen KF, Shulman GI. Lipid-induced insulin resistance: unravelling
      the mechanism. Lancet. 2010;375(9733):2267-77.
4.    van Hees AM, Jans A, Hul GB, Roche HM, Saris WH, Blaak EE. Skeletal muscle
      fatty acid handling in insulin resistant men. Obesity (Silver Spring). 2011;19(7):1350-
      9.
5.    Schling P, Schafer T. Human adipose tissue cells keep tight control on the
      angiotensin II levels in their vicinity. J Biol Chem. 2002;277(50):48066-75.
6.    Standeven KF, Hess K, Carter AM, Rice GI, Cordell PA, Balmforth AJ, et al.
      Neprilysin, obesity and the metabolic syndrome. Int J Obes (Lond). 2011;35(8):1031-
      40.
7.    Wang TJ, Larson MG, Levy D, Benjamin EJ, Leip EP, Wilson PW, et al. Impact of
      obesity on plasma natriuretic peptide levels. Circulation. 2004;109(5):594-600.
8.    Magnusson M, Jujic A, Hedblad B, Engstrom G, Persson M, Struck J, et al. Low
      plasma level of atrial natriuretic peptide predicts development of diabetes: the
      prospective Malmo Diet and Cancer study. J Clin Endocrinol Metab. 2012;97(2):638-
      45.
9.    Meirhaeghe A, Sandhu MS, McCarthy MI, de Groote P, Cottel D, Arveiler D, et al.
      Association between the T-381C polymorphism of the brain natriuretic peptide gene
      and risk of type 2 diabetes in human populations. Hum Mol Genet.
      2007;16(11):1343-50.
10.   Jujic A, Nilsson PM, Engstrom G, Hedblad B, Melander O, Magnusson M. Atrial
      natriuretic peptide and type 2 diabetes development--biomarker and genotype
      association study. PLoS One. 2014;9(2):e89201.
11.   Birkenfeld AL, Boschmann M, Moro C, Adams F, Heusser K, Franke G, et al. Lipid
      mobilization with physiological atrial natriuretic peptide concentrations in humans. J
      Clin Endocrinol Metab. 2005;90(6):3622-8.
12.   Birkenfeld AL, Budziarek P, Boschmann M, Moro C, Adams F, Franke G, et al. Atrial
      natriuretic peptide induces postprandial lipid oxidation in humans. Diabetes.
      2008;57(12):3199-204.
13.   Coue M, Badin PM, Vila IK, Laurens C, Louche K, Marques MA, et al. Defective
      Natriuretic Peptide Receptor Signaling in Skeletal Muscle Links Obesity to Type 2
      Diabetes. Diabetes. 2015;64(12):4033-45.
14.   Engeli S, Birkenfeld AL, Badin PM, Bourlier V, Louche K, Viguerie N, et al. Natriuretic
      peptides enhance the oxidative capacity of human skeletal muscle. J Clin Invest.
      2012;122(12):4675-9.
15.   van der Zijl NJ, Moors CC, Goossens GH, Hermans MM, Blaak EE, Diamant M.
      Valsartan improves {beta}-cell function and insulin sensitivity in subjects with
      impaired glucose metabolism: a randomized controlled trial. Diabetes Care.
      2011;34(4):845-51.
16.   McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, et al.
      Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med.
      2014;371(11):993-1004.
                                                                                         133
Chapter 3
134
                                                                                     Chapter 3
                                                                                            135
Chapter 3
SUPPLEMENTARY MATERIAL
Blood Pressure
Office BP was measured at screening, during washout and throughout the study at
baseline, week 4, week 8 and end of study using the same arm and the same
automated equipment with an appropriate cuff size. Measurements were performed
in triplicates at 2-minute intervals after patients have been sitting for 15 minutes
with the back supported and both feet on the floor. During the home stay period,
patients were given a home measurement device and instructed to monitor BP
twice weekly at approximately the same time each morning (7–9 AM).
Safety Assessments
Safety assessments included adverse events (AEs) and serious adverse events
(SAEs) and were regularly monitored throughout the study based on hematology,
blood chemistry and urine analysis as well as assessments of vital signs (BP and
pulse measurements), ECG, physical condition and body weight.
136
                                                                    Chapter 3
Supplementary Figure 1A. Comparison of glucose infusion rate with body weight
at baseline and Day 56.
Supplementary Figure 1B. Change from baseline in glucose infusion rate vs.
body weight following 8-week treatment with LCZ696 vs. amlodipine
                                                                          137
Chapter 3
Ratio to baseline 1.00 (0.99, 1.02) 1.00 (0.98, 1.01) 1.01 (0.98, 1.03)
Insulin (pmol/L)
    Baseline          41.09 (34.68, 48.67)       37.54 (31.45, 44.79)
      Day 57          41.07 (34.57; 48.79)       37.91 (31.61, 45.47)
Ratio to baseline       1.00 (0.88, 1.14)          1.01 (0.88; 1.16)            0.99 (0.82, 1.20)
Glycerol (µmol/L)
    Baseline          85.39 (77.97; 93.52)      91.61 (83.32, 100.73)
      Day 57          84.48 (76.85, 92.87)       87.90 (79.51, 97.18)
Ratio to baseline       0.99 (0.91, 1.08)          0.96 (0.87; 1.05)            1.03 (0.91, 1.17)
Non-esterified fatty acids (mmol/L)
Baseline, adjusted
                        0.63 (0.56, 0.70)          1.03 (0.91, 1.17)
      mean
      Day 57            0.60 (0.53, 0.68)          1.03 (0.91, 1.17)
Ratio to baseline       0.96 (0.86, 1.06)          0.92 (0.82, 1.03)            1.04 (0.89, 1.21)
Epinephrine (pg/mL)
    Baseline          25.23 (21.20, 30.03)       20.98 (17.49, 25.16)
      Day 57          20.42 (17.19, 24.25)       21.91 (18.28, 26.26)
                                            a                                                      b
Ratio to baseline       0.81 (0.72, 0.91)          1.04 (0.93, 1.18)           0.77 (0.65, 0.91)
Norepinephrine (pg/mL)
    Baseline,         217.3 (188.3, 250.7)       230.8 (198.8; 267.9)
      Day 57          236.8 (204.2, 274.5)       276.2 (236.4; 322.9)
                                                                       c
Ratio to baseline       1.09 (0.98, 1.21)          1.20 (1.07, 1.33)            0.91 (0.78, 1.06)
a
 p<0.001; b p=0.002; c p=0.001
Data was analyzed using repeated measurements on log transformed values with treatment, visit, time
and treatment*visit*time interaction as fixed effect assuming an Unstructured covariance. Values are
then log back transformed.
138
                                                                                        Chapter 3
Pruritus 5 (10.0) -
Gastroenteritis 2 (4.0) -
                                                                                             139
CHAPTER 4
Effect of sacubitril/valsartan on exercise
induced lipid metabolism in individuals
with obesity and hypertension
Engeli S., Stinkens R., Heise T., May M., Goossens G.H., Blaak E.E.,
Jax T., Albrecht D., Pal P., Tegtbur U., Haufe S., Langenickel T.H.,
Jordan J.
Submitted
Chapter 4
ABSTRACT
Background: sacubitril/valsartan (LCZ696), a novel angiotensin receptor-neprilysin
inhibitor, was recently approved for the treatment of heart failure with reduced
ejection fraction. Neprilysin degrades several peptides that modulate lipid
metabolism, including natriuretic peptides. In this study, we investigated the effects
of 8 weeks’ treatment with sacubitril/valsartan on whole body and adipose tissue
lipolysis and lipid oxidation during defined physical exercise compared with the
metabolically neutral comparator amlodipine.
Methods: This was a multicenter, randomized, double-blind, active-controlled,
parallel-group study enrolling subjects with abdominal obesity and moderate
hypertension (mean sitting systolic blood pressure [msSBP] ≥130-180 mmHg).
Lipolysis during rest and exercise was assessed by microdialysis and [1,1,2,3,3-
2
  H]-glycerol tracer kinetics. Energy expenditure and substrate oxidation were
measured simultaneously using indirect calorimetry. Plasma non-esterified fatty
acids, glycerol, insulin, glucose, adrenaline and noradrenaline concentrations,
blood pressure and heart rate were also determined.
Results: Exercise elevated plasma glycerol, free fatty acids and interstitial glycerol
concentrations and increased the rate of glycerol appearance. However, exercise-
induced stimulation of lipolysis was not augmented upon sacubitril/valsartan
treatment compared with amlodipine treatment. Furthermore, sacubitril/valsartan
did not alter energy expenditure and substrate oxidation during exercise compared
with amlodipine treatment.
Conclusion: Sacubitril/valsartan treatment for 8 weeks did not elicit clinically
relevant changes in exercise-induced lipolysis or substrate oxidation in obese
patients with hypertension, implying that its beneficial cardiovascular effects cannot
be explained by changes in lipid metabolism during exercise.
142
                                                                              Chapter 4
INTRODUCTION
Fatty acids are stored in the form of triglycerides in the adipose tissue and are
released during lipolysis to fuel lipid oxidation in energy consuming tissues.
Lipolysis and skeletal muscle lipid oxidation decrease following carbohydrate
ingestion and increase in the fasting state or during physical exercise [1]. An
imbalance between fatty acid mobilization and utilization may adversely affect
cardiovascular and metabolic health. Acute experimental increases in circulating
fatty acids in humans worsened hepatic [2] and skeletal muscle [3] insulin
sensitivity and endothelium-mediated vasodilation [4]. Chronic increase in fatty acid
availability promotes hepatic, skeletal muscle and myocardial lipotoxicity,
dyslipidemia, insulin resistance, and type 2 diabetes mellitus [5, 6]. Conversely,
interventions that reduce fatty acid levels, improve metabolic health [5]. These
observations are highly relevant for cardiovascular medications with the potential to
affect lipid turnover. Sacubitril/valsartan, comprising a novel neprilysin inhibitor pro-
drug sacubitril and angiotensin receptor blocker (valsartan) has been approved for
the treatment of chronic heart failure (NYHA Class II-IV) with reduced ejection
fraction [7]. The endopeptidase neprilysin is ubiquitously expressed, including in
human adipocytes, and degrades multiple peptides such as natriuretic peptides
(NPs), angiotensin II, bradykinin and endothelin that may modulate lipid
metabolism [8, 9]. Notably, NPs potently augment human adipose tissue lipolysis,
postprandial lipid oxidation, and skeletal muscle oxidative capacity [9], whereas,
angiotensin II elicits more subtle changes in fatty acid turnover [10].
Given the role and association of aberrant NP- and renin-angiotensin-aldosterone
signaling in cardiovascular diseases and metabolic dysfunction, we hypothesized
that simultaneous blockade of angiotensin receptor and neprilysin with
sacubitril/valsartan can potentially ameliorate metabolic dysfunction, especially
lipid-turnover, compared with amlodipine. In the present study, we investigated the
effects of 8-weeks treatment with sacubitril/valsartan compared with the
metabolically neutral comparator amlodipine on whole-body and adipose tissue
lipolysis, energy expenditure and substrate oxidation during defined physical
exercise, which is known to stimulate NP release and induces lipolysis and lipid
oxidation.
METHODS
Study design
The study design, key inclusion and exclusion criteria of the patients and the
primary results of this study have been described earlier [11]. Briefly, this was a
multicenter, randomized, double-blind, double-dummy, active-controlled, parallel-
group study enrolling adult subjects with abdominal obesity (waist circumference
≥102 cm for men and ≥88 cm for women) and moderate hypertension (mean sitting
systolic blood pressure [msSBP] ≥130 and<180 mmHg). Key exclusion criteria
were severe hypertension (msSBP>180 mmHg), type 1 or 2 diabetes (fasting
plasma glucose ≥126 mg/dL or HbA1c ≥6.5%), dyslipidemia requiring therapy with
fibrates or nicotinic acid, concomitant use of antihypertensive, anti-diabetic or other
                                                                                    143
Chapter 4
medications that affect glucose and/or lipid metabolism, and a history or current
diagnosis of heart failure (NYHA class II-IV).
The study included a screening period of up to 4-weeks followed by a 4-week
washout period and an 8-week, randomized, double-blind, double-dummy
treatment phase. Patients receiving antihypertensive medications at the time of
screening discontinued the therapy during the washout period. During the
treatment period, patients were randomized to receive either sacubitril/valsartan
400 mg every day (QD) or amlodipine 10 mg QD along with a matching placebo for
8 weeks. Patients were stratified into 4 groups based on baseline homeostatic
model assessment of insulin resistance (HOMA-IR) and statin use.
All patients provided written informed consent before screening. The clinical study
protocol was reviewed and approved by the Independent Ethics Committee or
Institutional Review Board at each center and, conducted in accordance with
declaration of Helsinki and registered at clinicaltrials.gov under the identifier
NCT01631864.
Exercise test
An incremental exercise test on a bicycle ergometer was conducted before the
start of the intervention (Day -14) to determine the maximal aerobic capacity
(VO2peak) at volitional exhaustion by measuring the individual maximum workload
prior to stopping for exhaustion or until pre-defined heart rate or blood pressure
(BP) criteria were met. At baseline (Day 1) and after 8 weeks (Day 57), subjects
exercised at 50% of VO2peak (as determined on Day -14) for a period of 60 min.
Measurement of lipolysis
Local adipose tissue and whole body lipolysis were assessed at baseline and after
8 weeks of treatment, as described previously [11]. Local adipose tissue lipolysis
was measured by microdialysis and assessed during a 45 min interval at rest,
followed by a 60 min interval during which the patients exercised at 50% of their
individual VO2peak. Dialysates were collected from abdominal subcutaneous
adipose tissue at the lower right abdominal quadrant at rest and at 15-min intervals
during exercise. Concentrations of glycerol (as an indicator of lipolysis), glucose
and lactic acid in dialysates were measured. The ethanol outflow/inflow ratio (ratio
of ethanol concentration in the dialysate and the perfusate) was measured as an
indicator of adipose tissue blood flow.
                                                        2
Whole body lipolysis was estimated using [1,1,2,3,3- H]-glycerol tracer kinetics
                                                         -1
following an intravenous glycerol bolus (2 µmol·kg ) after insertion of the
microdialysis catheter, approximately 60-min before the baseline measurements
                                                                      -1   -1
started, and subsequent infusion at an infusion rate of 0.1 µmol·kg min at rest
                   -1   -1
and 0.2 µmol·kg min during exercise. Blood samples were collected at 15-min
intervals at rest and during exercise. The rate of appearance (Ra) of endogenous
glycerol was calculated as the ratio of glycerol tracer infusion rate to plasma
glycerol tracer enrichment. At steady state, glycerol Ra was calculated from
glycerol enrichment using Steele’s equation.
144
                                                                          Chapter 4
Blood pressure
Office BP was measured at screening, during washout and throughout the study at
baseline, Week 4, Week 8 and at the end of study using the same automated
equipment with an appropriate cuff size. Measurements were performed in
triplicate at 2-min intervals after patients had been sitting for 15 minutes with the
back supported and both feet on the floor. BP was also measured during the
exercise phase. During the home stay period, patients were given a home
measurement device and instructed to monitor BP twice weekly at approximately
the same time each morning.
Statistical analysis
Following 8 weeks of treatment with sacubitril/valsartan or amlodipine,
assessments of local adipose tissue lipolysis, whole body lipolysis, oxidative
metabolism, BP and biomarkers during exercise were performed as pre-specified
study objectives.
For abdominal subcutaneous adipose tissue microdialysate data (ethanol ratio,
dialysate lactate, dialysate glucose, dialysate glycerol), plasma biomarkers
(glycerol, NEFA, glucose, insulin, adrenaline and noradrenaline) and whole-body
lipolysis (rate of glycerol appearance) data for 45 minutes at rest and 4 time-points
during exercise (15, 30, 45 and 60 minutes) were analyzed using repeated-
measures analysis on log-transformed values with treatment, visit, time and
treatment*visit*time interaction as fixed effects. Geometric mean ratios of each
exercise time-point to 45 minutes resting for each day and treatment, ratios of Day
57 to Day 1 for each treatment and each exercise time-point, and the ratio between
sacubitril/valsartan and amlodipine for Day 57 to Day 1 were calculated.
Oxidative metabolism was analyzed using analysis of covariance (ANCOVA) with
treatment as the fixed effect and baseline as the covariate. Oxidative metabolism
during exercise was analyzed using analysis of variance (ANOVA) for repeated
measurements with treatment, visit and treatment*visit interaction as fixed effects.
Mean difference to Day1 (Day 57 vs. Day 1) for each treatment along with the
corresponding 95% CIs and p-values are presented. Data for exercise and resting
phase were analyzed for each day and treatment with a mixed-effects linear model
with phase (exercise or resting) as the fixed effect and subject as the random effect
                                                                                 145
Chapter 4
to obtain the mean difference estimate and 95% CI for exercise vs. rest
comparison. Respiratory quotient (CO2/O2 ratio) was calculated at each of the Days
1 and 57 at rest and during exercise. A statistical comparison of the quotients was
made between rest and exercise within each day.
RESULTS
Exercise testing
On Day 1, 39 patients in the sacubitril/valsartan group and 24 patients randomized
to the amlodipine group completed the constant workload exercise for 60 minutes.
On Day 57, 36 patients treated with sacubitril/valsartan and 23 treated with
amlodipine completed the exercise for 60 minutes. Similar observations were made
in patients completing only 45 and 60 minutes of exercise, suggesting that 8 weeks
of treatment of patients with obesity and hypertension with sacubitril/valsartan or
amlodipine did not have any clinically relevant impact on the exercise duration.
Oxygen consumption and workload were comparable between Day 1 and Day 57
in both treatment groups (Supplementary Table 1).
146
                                                                          Chapter 4
                                                                                 147
Chapter 4
Whole-body lipolysis
Plasma glycerol concentrations increased with exercise in both treatment groups,
both on Day 1 and Day 57 [amlodipine group, Day 1 vs. Day 57: resting (89.77 vs.
88.04 µmol/L); 15 min (141.12 vs. 119.56 µmol/L); 30 min (184.78 vs. 156.03
µmol/L); 45 min (216.04 vs. 179.27 µmol/L); 60 min (224.85 vs. 191.95 µmol/L) and
sacubitril/valsartan, Day 1 vs. Day 57: resting (85.64 vs. 83.93 µmol/L); 15 min
(139.3 vs. 126.92 µmol/L); 30 min (177.65 vs. 157.29 µmol/L); 45 min (205.68 vs.
189.84 µmol/L); 60 min (225.62 vs. 205.26 µmol/L)]. Compared to baseline, plasma
glycerol levels were lower in both the treatment groups on Day 57. While the
change from baseline to Day 57 was significant at all time-points in the amlodipine
group (p<0.05), it was significant at 30 minutes in the sacubitril/valsartan group
(p=0.012). The differences in plasma glycerol levels between treatment groups
were not significant.
As compared with glycerol Ra following 45 minutes rest, a significant increase was
observed during exercise at all time-points in both treatment groups on Day 1 and
Day 57 (p<0.001). The change from baseline to Day 57 was statistically significant
in the sacubitril/valsartan group at 15 min (p=0.026), 30 min (p=0.012) and 45 min
(p=0.035), but was not significant at any time-point in the amlodipine group (Figure
2A). However, there was no significant difference between treatment groups at any
time-point.
Plasma NEFA concentrations decreased on Day 57 at 15 minutes in the
sacubitril/valsartan group (p=0.018) and at 15 min and 30 min (p<0.05) in the
amlodipine group. No significant differences were observed between treatment
groups. When compared with NEFA levels at rest (45 minutes), the levels were
lower during the initial phases of exercise, but increased gradually with increasing
exercise duration in both treatment groups (Figure 2B).
148
                                                                           Chapter 4
                                                                                149
Chapter 4
150
                                                                                         Chapter 4
                                                                                              151
Chapter 4
Blood pressure
After 8-weeks of treatment, systolic BP, diastolic BP and pulse pressure decreased
from baseline in both treatment groups at rest. Systolic and diastolic BP and pulse
rate values increased during exercise in both treatment groups on both Day 1 and
Day 57 without clinically relevant differences between treatment groups (Table 1).
152
                                                                              Chapter 4
DISCUSSION
The present study demonstrated that treatment with sacubitril/valsartan compared
with amlodipine for 8 weeks did not elicit relevant changes in exercise-induced
lipolysis and substrate oxidation in obese patients with hypertension. The exercise-
induced increase in abdominal subcutaneous adipose tissue and whole-body
lipolysis was not augmented following sacubitril/valsartan treatment compared with
amlodipine treatment. Moreover, the shift in substrate oxidation towards
carbohydrate catabolism during exercise was comparable in both treatment
groups, implying that sacubitril/valsartan did not significantly affect lipid utilization
during acute exercise. We have previously observed significantly improved whole-
body insulin sensitivity and a modest increase in resting abdominal subcutaneous
adipose tissue lipolysis with no marked changes in whole-body lipolysis with
sacubitril/valsartan compared with amlodipine treatment [11]. Overall, these
findings imply that the beneficial cardiometabolic effects of sacubitril/valsartan may
not be explained by changes in lipid mobilization or oxidation.
                                                                              2
In this study, we used state-of-the-art methodology including [1,1,2,3,3- H]-glycerol
tracer kinetics and abdominal subcutaneous adipose tissue microdialysis to assess
whole-body and local lipolysis, respectively, in a large patient sample. Furthermore,
we treated patients with a total daily dose of sacubitril/valsartan which provided
superior BP control in patients with arterial hypertension (400 mg QD) [12] and
reduced cardiovascular mortality and heart failure hospitalizations in patients with
heart failure and reduced ejection fraction (HFrEF) (200 mg twice daily) compared
with standard-of-care renin-angiotensin system (RAS) inhibition [7]. This study,
therefore, was appropriately designed to study the effect of sacubitril/valsartan on
lipid turnover.
Our study extends previous investigations on the role of neprilysin substrates and
angiotensin II type-1 (AT1)-receptors in the regulation of lipid turnover. All
components of the RAS are expressed in adipose tissue, and AT 1-receptors have
been implicated in the regulation of adipose tissue differentiation, inflammation,
and metabolism [10]. Conflicting findings have been reported with respect to the
effects of angiotensin II on adipose tissue lipolysis. More specifically, both
increased [13, 14] and decreased [15] subcutaneous adipose tissue lipolysis have
been demonstrated [14]. Moreover, intravenous angiotensin II infusions and
angiotensin converting enzyme inhibition did not elicit major changes in whole-body
lipolysis as determined by glycerol tracer kinetics [16]. AT1-receptor blockade in
human subjects did not increase lipolytic gene expression or lipolysis in abdominal
subcutaneous adipose tissue [17, 18]. However, long-term AT1-receptor blockade
altered intramuscular lipid partitioning, manifested by decreased saturation of
skeletal muscle triacylglycerol and diacylglycerol stores, reduced postprandial fatty
acid spillover and lipolysis [19]. Overall, angiotensin II actions on AT 1-receptors
appear to have modest effects on lipid turnover. Although postprandial fatty acid
handling has not been examined in this study, the present findings suggest that
AT1-receptor blockade in the context of neprilysin inhibition by sacubitril/valsartan
does not have clinically relevant effects on lipid mobilization or utilization.
Neprilysin degrades multiple peptides potentially modulating lipid metabolism such
as natriuretic peptides (NPs), bradykinin, endothelin-1, and glucagon-like peptide 1
(GLP-1) [20]. We cannot discern contributions of individual neprilysin substrates to
                                                                                    153
Chapter 4
154
                                                                            Chapter 4
                                                                                   155
Chapter 4
REFERENCES
1.    Spriet LL. New insights into the interaction of carbohydrate and fat metabolism during
      exercise. Sports Med. 2014;44 Suppl 1:S87-96.
2.    Bevilacqua S, Bonadonna R, Buzzigoli G, Boni C, Ciociaro D, Maccari F, Giorico MA,
      Ferrannini E. Acute elevation of free fatty acid levels leads to hepatic insulin
      resistance in obese subjects. Metabolism. 1987;36(5):502-6.
3.    Roden M, Price TB, Perseghin G, Petersen KF, Rothman DL, Cline GW, Shulman GI.
      Mechanism of free fatty acid-induced insulin resistance in humans. J Clin Invest.
      1996;97(12):2859-65.
4.    Steinberg HO, Tarshoby M, Monestel R, Hook G, Cronin J, Johnson A, Bayazeed B,
      Baron AD. Elevated circulating free fatty acid levels impair endothelium-dependent
      vasodilation. J Clin Invest. 1997;100(5):1230-9.
5.    Shulman GI. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic
      disease. N Engl J Med. 2014;371(23):2237-8.
6.    Stinkens R, Goossens GH, Jocken JW, Blaak EE. Targeting fatty acid metabolism to
      improve glucose metabolism. Obes Rev. 2015;16(9):715-57.
7.    McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL,
      Shi VC, Solomon SD, Swedberg K, Zile MR, Investigators P-H, Committees.
      Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med.
      2014;371(11):993-1004.
8.    Schling P, Schafer T. Human adipose tissue cells keep tight control on the
      angiotensin II levels in their vicinity. J Biol Chem. 2002;277(50):48066-75.
9.    Moro C. Natriuretic peptides and fat metabolism. Curr Opin Clin Nutr Metab Care.
      2013;16(6):645-9.
10.   Goossens GH. The renin-angiotensin system in the pathophysiology of type 2
      diabetes. Obes Facts. 2012;5(4):611-24.
11.   Jordan J, Stinkens R, Jax T, Engeli S, Blaak EE, May M, Havekes B, Schindler C,
      Albrecht D, Pal P, Heise T, Goossens GH, Langenickel TH. Improved Insulin
      Sensitivity With Angiotensin Receptor Neprilysin Inhibition in Individuals With Obesity
      and Hypertension. Clin Pharmacol Ther. 2017;101(2):254-63.
12.   Kario K, Sun N, Chiang FT, Supasyndh O, Baek SH, Inubushi-Molessa A, Zhang Y,
      Gotou H, Lefkowitz M, Zhang J. Efficacy and safety of LCZ696, a first-in-class
      angiotensin receptor neprilysin inhibitor, in Asian patients with hypertension: a
      randomized, double-blind, placebo-controlled study. Hypertension. 2014;63(4):698-
      705.
13.   Boschmann M, Adams F, Schaller K, Franke G, Sharma AM, Klaus S, Luft FC,
      Jordan J. Hemodynamic and metabolic responses to interstitial angiotensin II in
      normal weight and obese men. J Hypertens. 2006;24(6):1165-71.
14.   Boschmann M, Jordan J, Adams F, Christensen NJ, Tank J, Franke G, Stoffels M,
      Sharma AM, Luft FC, Klaus S. Tissue-specific response to interstitial angiotensin II in
      humans. Hypertension. 2003;41(1):37-41.
15.   Goossens GH, Blaak EE, Saris WH, van Baak MA. Angiotensin II-induced effects on
      adipose and skeletal muscle tissue blood flow and lipolysis in normal-weight and
      obese subjects. J Clin Endocrinol Metab. 2004;89(6):2690-6.
16.   Townsend RR. The effects of angiotensin-II on lipolysis in humans. Metabolism.
      2001;50(4):468-72.
17.   Goossens GH, Moors CC, van der Zijl NJ, Venteclef N, Alili R, Jocken JW, Essers Y,
      Cleutjens JP, Clement K, Diamant M, Blaak EE. Valsartan improves adipose tissue
      function in humans with impaired glucose metabolism: a randomized placebo-
      controlled double-blind trial. PLoS One. 2012;7(6):e39930.
156
                                                                                    Chapter 4
18.   Wang TJ, Larson MG, Keyes MJ, Levy D, Benjamin EJ, Vasan RS. Association of
      plasma natriuretic peptide levels with metabolic risk factors in ambulatory individuals.
      Circulation. 2007;115(11):1345-53.
19.   Moors CC, Blaak EE, van der Zijl NJ, Diamant M, Goossens GH. The effects of long-
      term valsartan treatment on skeletal muscle fatty acid handling in humans with
      impaired glucose metabolism. J Clin Endocrinol Metab. 2013;98(5):E891-6.
20.   Mangiafico S, Costello-Boerrigter LC, Andersen IA, Cataliotti A, Burnett JC, Jr.
      Neutral endopeptidase inhibition and the natriuretic peptide system: an evolving
      strategy in cardiovascular therapeutics. Eur Heart J. 2013;34(12):886-93c.
21.   Kobalava Z, Kotovskaya Y, Averkov O, Pavlikova E, Moiseev V, Albrecht D, Chandra
      P, Ayalasomayajula S, Prescott MF, Pal P, Langenickel TH, Jordaan P, Rajman I.
      Pharmacodynamic and Pharmacokinetic Profiles of Sacubitril/Valsartan (LCZ696) in
      Patients with Heart Failure and Reduced Ejection Fraction. Cardiovasc Ther.
      2016;34(4):191-8.
22.   Eriksson AK, van Harmelen V, Stenson BM, Astrom G, Wahlen K, Laurencikiene J,
      Ryden M. Endothelin-1 stimulates human adipocyte lipolysis through the ET A
      receptor. Int J Obes (Lond). 2009;33(1):67-74.
23.   Mori MA, Sales VM, Motta FL, Fonseca RG, Alenina N, Guadagnini D, Schadock I,
      Silva ED, Torres HA, dos Santos EL, Castro CH, D'Almeida V, Andreotti S, Campana
      AB, Sertie RA, Saad MJ, Lima FB, Bader M, Pesquero JB. Kinin B1 receptor in
      adipocytes regulates glucose tolerance and predisposition to obesity. PLoS One.
      2012;7(9):e44782.
24.   Bertin E, Arner P, Bolinder J, Hagstrom-Toft E. Action of glucagon and glucagon-like
      peptide-1-(7-36) amide on lipolysis in human subcutaneous adipose tissue and
      skeletal muscle in vivo. J Clin Endocrinol Metab. 2001;86(3):1229-34.
25.   Villanueva-Penacarrillo ML, Marquez L, Gonzalez N, Diaz-Miguel M, Valverde I.
      Effect of GLP-1 on lipid metabolism in human adipocytes. Horm Metab Res.
      2001;33(2):73-7.
26.   Birkenfeld AL, Boschmann M, Moro C, Adams F, Heusser K, Franke G, Berlan M,
      Luft FC, Lafontan M, Jordan J. Lipid mobilization with physiological atrial natriuretic
      peptide concentrations in humans. J Clin Endocrinol Metab. 2005;90(6):3622-8.
27.   Sengenes C, Zakaroff-Girard A, Moulin A, Berlan M, Bouloumie A, Lafontan M,
      Galitzky J. Natriuretic peptide-dependent lipolysis in fat cells is a primate specificity.
      Am J Physiol Regul Integr Comp Physiol. 2002;283(1):R257-65.
28.   Birkenfeld AL, Adams F, Schroeder C, Engeli S, Jordan J. Metabolic actions could
      confound advantageous effects of combined angiotensin II receptor and neprilysin
      inhibition. Hypertension. 2011;57(2):e4-5.
29.   Toal CB, Meredith PA, Elliott HL. Long-acting dihydropyridine calcium-channel
      blockers and sympathetic nervous system activity in hypertension: a literature review
      comparing amlodipine and nifedipine GITS. Blood Press. 2012;21 Suppl 1:3-10.
30.   Stankovic S, Panz V, Klug E, Di Nicola G, Joffe BI. Amlodipine and physiological
      responses to brisk exercise in healthy subjects. Cardiovasc Drugs Ther.
      1999;13(6):513-7.
31.   de Champlain J, Karas M, Assouline L, Nadeau R, LeBlanc AR, Dube B, Larochelle
      P. Effects of valsartan or amlodipine alone or in combination on plasma
      catecholamine levels at rest and during standing in hypertensive patients. J Clin
      Hypertens (Greenwich). 2007;9(3):168-78.
32.   Latini R, Masson S, Anand I, Judd D, Maggioni AP, Chiang YT, Bevilacqua M, Salio
      M, Cardano P, Dunselman PH, Holwerda NJ, Tognoni G, Cohn JN, Valsartan Heart
      Failure Trial I. Effects of valsartan on circulating brain natriuretic peptide and
      norepinephrine in symptomatic chronic heart failure: the Valsartan Heart Failure Trial
      (Val-HeFT). Circulation. 2002;106(19):2454-8.
                                                                                           157
Chapter 4
33.   Anand IS, Fisher LD, Chiang YT, Latini R, Masson S, Maggioni AP, Glazer RD,
      Tognoni G, Cohn JN, Val-He FTI. Changes in brain natriuretic peptide and
      norepinephrine over time and mortality and morbidity in the Valsartan Heart Failure
      Trial (Val-HeFT). Circulation. 2003;107(9):1278-83.
34.   Moro C, Klimcakova E, Lafontan M, Berlan M, Galitzky J. Phosphodiesterase-5A and
      neutral endopeptidase activities in human adipocytes do not control atrial natriuretic
      peptide-mediated lipolysis. Br J Pharmacol. 2007;152(7):1102-10.
35.   Seferovic JP, Claggett B, Seidelmann SB, Seely EW, Packer M, Zile MR, Rouleau
      JL, Swedberg K, Lefkowitz M, Shi VC, Desai AS, McMurray JJV, Solomon SD. Effect
      of sacubitril/valsartan versus enalapril on glycaemic control in patients with heart
      failure and diabetes: a post-hoc analysis from the PARADIGM-HF trial. Lancet
      Diabetes Endocrinol. 2017;5(5):333-40.
158
Chapter 4
     159
CHAPTER 5
The effects of angiotensin receptor
neprilysin inhibition by sacubitril/valsartan
on adipose tissue transcriptome and
protein expression in obese hypertensive
patients
Stinkens R., van der Kolk B.W., Jordan J., Jax T., Engeli S.,
Heise T., Jocken J.W., May M., Schindler C., Havekes B.,
Schaper N., Albrecht D., Kaiser S., Hartmann N., Letzkus M.,
Langenickel T.H., Goossens G.H., Blaak E.E.
Submitted
Chapter 5
ABSTRACT
Increased activation of the renin-angiotensin system is involved in the onset and
progression of cardiometabolic diseases, while natriuretic peptides (NP) may exert
protective effects. We have recently demonstrated that sacubitril/valsartan
(LCZ696), a first-in-class angiotensin receptor neprilysin inhibitor, which blocks the
angiotensin II type-1 receptor and augments natriuretic peptide levels, improved
peripheral insulin sensitivity in obese hypertensive patients. Here, we investigated
the effects of sacubitril/valsartan (400mg QD) treatment for 8 weeks on the
abdominal subcutaneous adipose tissue (AT) phenotype compared to the
metabolically neutral comparator amlodipine (10mg QD) in 70 obese hypertensive
patients. Abdominal subcutaneous AT biopsies were collected before and after
intervention to determine the AT transcriptome and expression of proteins involved
in lipolysis, NP signaling and mitochondrial oxidative metabolism. Both
sacubitril/valsartan and amlodipine treatment did not significantly induce AT
transcriptional changes in pathways related to lipolysis, NP signaling and oxidative
metabolism. Furthermore, protein expression of adipose triglyceride lipase (ATGL)
(Ptime*group=0.195), hormone-sensitive lipase (HSL) (Ptime*group=0.458), HSL-ser660
phosphorylation (Ptime*group=0.340), NP receptor-A (NPRA) (Ptime*group=0.829) and
OXPHOS complexes (Ptime*group=0.964) remained unchanged. In conclusion,
sacubitril/valsartan treatment for 8 weeks did not alter the abdominal subcutaneous
AT transcriptome and expression of proteins involved in lipolysis, NP signaling and
oxidative metabolism in obese hypertensive patients.
162
                                                                                Chapter 5
INTRODUCTION
Obesity is strongly associated with cardiometabolic risk factors [1], which is
reflected by an increased risk for arterial hypertension, heart failure and type 2
diabetes mellitus (T2DM) [2]. An impaired adipose tissue function and excessive fat
mass in obesity represent key factors in the development of insulin resistance and
related chronic diseases, including cardiovascular disease and T2DM [3]. Evidence
suggests that impaired insulin sensitivity in obesity might be related to an altered
renin-angiotensin system (RAS) and natriuretic peptide (NP) signaling in adipose
tissue. Blockade of the RAS using angiotensin-converting enzyme (ACE) inhibitors
or angiotensin type-1 receptor blockers (ARB) has been shown to improve insulin
sensitivity and beta-cell function [4] and reduces the incidence of T2DM [5] as
reviewed elsewhere [6]. However, results are not consistent [7]. In addition, NPs
are positively associated with insulin sensitivity and low atrial natriuretic peptide
(ANP) concentrations are associated with an increased risk of developing arterial
hypertension and T2DM [8]. In accordance, neprilysin (NEP), which is involved in
the degradation and inactivation of NP, is linked to insulin resistance, increased
blood pressure and impaired lipid metabolism [9]. Therefore, combined RAS
blockade and NEP inhibition might have synergistic beneficial effects on blood
pressure and peripheral insulin sensitivity. We recently demonstrated that
combined ARB and NEP inhibition, using sacubitril/valsartan (LCZ696), improved
peripheral insulin sensitivity following 8 weeks of treatment compared to amlodipine
(AMLO) in obese hypertensive patients [10]. However, the mechanisms underlying
these beneficial effects remain to be established.
Evidence suggests that both the RAS and ANP affect adipose tissue metabolism,
thereby determining insulin sensitivity [6, 11]. It has been shown that valsartan
(ARB) reduced adipocyte size, increased adipose tissue blood flow and decreased
gene expression of angiogenesis, adipogenesis and macrophage infiltration
markers [12], which may have contributed to the valsartan-induced increased
insulin sensitivity [4]. Furthermore, angiotensin II inhibited lipolysis in vitro in mature
human adipocytes [13], although conflicting findings on adipose tissue lipolysis in
vivo in humans have been reported [14, 15]. ANP has been shown to increase
adipose tissue lipid mobilization and oxidation [11] and we and others have
recently demonstrated that ANP-mediated lipolysis is impaired in subcutaneous
mature adipocytes from obese men with and without T2DM [16, 17] .
Therefore, it is hypothesized that ARB and NEP inhibition with sacubitril/valsartan
may affect adipose tissue function, thereby contributing to the observed improved
peripheral insulin sensitivity in obese individuals [10]. The present study
investigated the effects of sacubitril/valsartan compared to amlodipine treatment for
8 weeks on the abdominal subcutaneous adipose tissue transcriptome and protein
expression profiles in obese hypertensive individuals.
                                                                                      163
Chapter 5
METHODS
Study design
Ninety-eight obese hypertensive patients participated in a multicenter, randomized,
double-blind, parallel-group study to investigate the effects of sacubitril/valsartan
(400mg QD) compared with amlodipine (10mg QD) treatment for 8 weeks. A
detailed description of the inclusion and exclusion criteria is described elsewhere
(clinicaltrials.gov - NCT01631864). Before and after treatment, abdominal
subcutaneous adipose tissue biopsies were collected by needle aspiration under
local anesthesia after an overnight fast. We determined adipose tissue gene
expression profiles using microarray analysis in a subgroup of 70 patients who had
a RNA Integrity Number (RIN) ≥5.0, as described in detail below. Next, in a
subgroup (N=12-13), we determined the expression of proteins involved in the
lipolytic pathway, the natriuretic peptide signaling pathway and mitochondrial
oxidative phosphorylation. Subjects showing a treatment-induced increase in
adipose tissue lipolysis in vivo following sacubitril/valsartan and unchanged
lipolysis after amlodipine treatment were included in the latter analyses.
All patients gave written informed consent before participation. The Independent
Ethical Committee or Institutional Review Board of each center reviewed the study
protocol. The study was performed in accordance with the Declaration of Helsinki.
164
                                                                         Chapter 5
Statistics
Statistical analyses of gene expression data was performed with R v.3.2.2 (R
Development Core Team, Vienna, http://www.R-project.org) and the Bioconductor
limma R package v.3.26.8. Statistical significance was set at nominal P≤0.05 and
P-values were corrected for multiple testing using the Bonferroni method. A total of
8319 transcripts (out of assessed 18898 transcripts/microarray) fulfilled the
expression level filtering criteria and the Bonferroni adjusted threshold was
                                  -6
therefore defined as P<6.01·10 . Longitudinal treatment-induced transcriptional
changes were expressed as Ratio Change from baseline (RC; (post-treatment
                            -1
level)·(pre-treatment level) ). Thresholds for minimum relevant treatment-induced
effects were defined as 0.66≥RC≥1.50 (i.e. RC range equivalent to absolute fold
changes≥1.5).
Protein expression data are expressed as mean±SEM. All variables were checked
for normal distribution by Shapiro-Wilk test and variables were Ln-transformed to
satisfy conditions of normality (HSL, HSL S660 phosphorylation, NPRA and
OXPHOS). Data was analyzed using two-way repeated measures ANOVA, with
time (pre, post) and treatment (sacubitril/valsartan, amlodipine) as factors.
Bonferroni post-hoc correction was applied when a significant time*treatment
interaction was found. Calculations were performed using SPSS v.21 for Mac OSX
(IBM, Chicago, IL, USA) and P≤0.05 was considered statistically significant.
RESULTS
Subject characteristics
Baseline characteristics of patients contributing to the present study are shown in
Table 1. Importantly, the two groups were well matched and there were no major
differences in clinical characteristics between groups.
Transcriptomics
At baseline, no significant differences in the abdominal subcutaneous adipose
tissue transcriptome were found between treatment arms. Unsupervised
assessments revealed 1443 longitudinally modulated transcripts (nominal P<0.05),
but none of these fulfilled both thresholds for Bonferroni adjusted P-value and
minimum treatment-induced RC effect. A set of selected transcripts is shown in
Supplementary Table S1.
Supervised analyses identified no significant treatment-induced changes (defined
based on the two thresholds stated above) in gene expression levels of transcripts
involved in lipolysis, NP signaling, oxidative metabolism and adipokines (Figure 1).
                                                                                165
Chapter 5
The heatmap plot shows per patient longitudinal treatment-induced gene expression changes [defined
as log2(post treatment expression) - log2(pre-treatment expression)] for selected transcripts related to
natriuretic peptide signaling, lipolytic pathway, oxidative pathway and adipokines in abdominal
subcutaneous adipose tissue (n=70 patients; grouped per treatment arm). Color in the heatmap reflects
transcript change from baseline per subject. Blue: downregulated transcripts; yellow: upregulated
transcripts. The adjacent table depicts the corresponding transcript ratio change from baseline (RC) and
P-value per treatment arm.
166
               Table 1. Baseline clinical characteristics of patients involved in transcriptomic and protein expression analyses
Transcriptomic analyses
       Glucose infusion rate (μmol·kg bw-1·min-1)         362.6 (141.71)             401.1 (104.46)                379.7 (125.74)
                                                                                                                                    Chapter 5
167
      Data are expressed as mean (SD).
Chapter 5
Protein expression
Sacubitril/valsartan treatment did not significantly change protein expression of
ATGL (LCZ696: 1.00±0.21 vs. 0.85±0.19 AU; AMLO: 1.00±0.26 vs. 1.36±0.30 AU;
Ptime=0.583, Ptime*treatment=0.195; Figure 2A), HSL (LCZ696: 1.00±0.23 vs. 1.38±0.24
AU; AMLO: 1.00±0.26 vs. 1.00±0.18 AU; Ptime=0.141, Ptime*treatment=0.458; Figure
2B), HSL serine 660 phosphorylation (LCZ696: 1.00±0.16 vs. 1.04±0.14 AU;
AMLO: 1.00±0.25 vs. 0.71±0.19 AU; Ptime=0.551, Ptime*treatment=0.340; Figure 2C) or
NPRA (LCZ696: 1.00±0.24 vs. 0.99±0.29 AU; AMLO: 1.00±0.36 vs. 0.96±0.30 AU;
Ptime=0.775, Ptime*treatment=0.829; Figure 2D). Furthermore, total OXPHOS protein
expression (LCZ696: 1.00±0.22 vs. 1.76±0.48 AU; AMLO: 1.00±0.15 vs. 1.74±0.50
AU; Ptime=0.125, Ptime*treatment=0.964; Figure 2E) remained unchanged following
treatment. More specifically, OXPHOS complexes I-V were not affected (data not
shown). Representative Western Blots are presented in Supplementary Figure S1.
168
                                                                                        Chapter 5
Expression of proteins involved in the lipolytic pathway (ATGL, N=12, A; HSL, N=13, B; HSL serine 660
phosphorylation, N=13, C), the natriuretic peptide signaling pathway (NPR-A, N=12, D) and
mitochondrial oxidative metabolism (total OXPHOS, N=13, E) expressed as fold change relative to
baseline for each treatment arm (Sacubitril/valsartan: LCZ696 or Amlodipine: AMLO). Data are
expressed as mean ± SEM. White bars: baseline values; black bars: post-treatment values.
Representative Western Blots are presented in Supplementary Figure S2.
                                                                                                169
Chapter 5
DISCUSSION
Here, we investigated the effects of sacubitril/valsartan versus amlodipine
treatment for 8 weeks on the abdominal subcutaneous adipose tissue
transcriptome and protein expression in obese hypertensive patients. We
demonstrated that sacubitril/valsartan treatment did not significantly alter adipose
tissue gene and protein expression of factors related to lipolysis, natriuretic peptide
signaling and oxidative metabolism.
We recently demonstrated that in obese hypertensive patients sacubitril/valsartan
treatment, which provides simultaneous ARB blockade and NEP inhibition,
significantly increased peripheral insulin sensitivity [10]. Furthermore,
sacubitril/valsartan slightly but significantly increased abdominal subcutaneous
adipose tissue lipolysis, although no changes in whole-body lipolysis were
observed [10]. Therefore, we hypothesized that RAS blockade and NEP inhibition
has synergistic beneficial effects on abdominal subcutaneous adipose tissue
metabolism and might underlie the observed improvement in insulin sensitivity. We
showed that the abdominal adipose tissue phenotype was not significantly affected
by 8 weeks of sacubitril/valsartan treatment in obese hypertensive patients. First,
sacubitril/valsartan treatment did not reveal any transcriptional changes in
abdominal subcutaneous adipose tissue, as determined by an unsupervised
analysis. More specific, no treatment-induced changes in expression of genes
involved in lipolysis, the NP signaling pathway and mitochondrial oxidative pathway
were found. Secondly, the expression of proteins involved in these pathways, as
well as post-translational modification of HSL, remained unchanged after the
intervention.
Evidence suggests that both the RAS and NP system may affect adipose tissue
metabolism, thereby contributing to improved insulin sensitivity [6, 11]. It has
previously been shown that angiotensin II decreased adipose tissue lipolysis in vivo
in humans [14, 19, 20] and in human isolated adipocytes [13], which seems to be
mediated via the angiotensin II type-1 receptor. However, increased adipose tissue
lipolysis has also been reported [15]. In accordance with our results, long-term
ARB treatment with valsartan improved insulin sensitivity in subjects with impaired
glucose metabolism [4], but adipose tissue gene and protein expression of several
lipolytic enzymes remained unchanged [12].
Several studies have shown that ANP promotes adipose tissue lipid mobilization
and oxidation in healthy individuals [11] via cGMP-mediated phosphorylation of
HSL [21, 22]. Furthermore, ANP induced mitochondrial biogenesis and uncoupling
in human adipocytes from healthy, non-diabetic women [23]. Here, we did not find
significant changes in gene and protein expression of markers involved in lipolysis,
phosphorylation of HSL and mitochondrial oxidative metabolism, which may be
explained by reduced ANP-mediated signaling in the study population. Indeed, an
impaired ANP-mediated lipolysis has recently been described in situ in
subcutaneous adipose tissue and in vitro in subcutaneous adipocytes from obese
individuals [16, 17]. Moreover, it has been shown that NPRC (NP clearance
receptor) is increased in adipose tissue of obese hypertensive patients compared
to lean and normotensive individuals [24], together with increased NEP expression
in obesity [9]. These data suggest reduced NP signaling and increased NP
clearance in adipose tissue in obesity. This may explain the unaltered
170
                                                                          Chapter 5
CONCLUSION
The present study demonstrated that simultaneous RAS blockade and NEP
inhibition following sacubitril/valsartan treatment for 8 weeks did not significantly
alter the adipose tissue metabolic phenotype in obese hypertensive patients. More
specifically, abdominal subcutaneous adipose tissue gene and protein expression
of factors involved in lipolysis, natriuretic peptide signaling and mitochondrial
oxidative metabolism remained unchanged. Therefore, alterations in the adipose
tissue metabolic phenotype do not seem to contribute to the improved peripheral
insulin sensitivity following 8 weeks of sacubitril/valsartan treatment [10].
                                                                                 171
Chapter 5
REFERENCES
1.    Reaven GM. Insulin resistance: the link between obesity and cardiovascular disease.
      Med Clin North Am. 2011;95(5):875-92.
2.    Jordan J, Yumuk V, Schlaich M, Nilsson PM, Zahorska-Markiewicz B, Grassi G,
      Schmieder RE, Engeli S, Finer N. Joint statement of the European Association for the
      Study of Obesity and the European Society of Hypertension: obesity and difficult to
      treat arterial hypertension. J Hypertens. 2012;30(6):1047-55.
3.    Stinkens R, Goossens GH, Jocken JW, Blaak EE. Targeting fatty acid metabolism to
      improve glucose metabolism. Obes Rev. 2015;16(9):715-57.
4.    van der Zijl NJ, Moors CC, Goossens GH, Hermans MM, Blaak EE, Diamant M.
      Valsartan improves {beta}-cell function and insulin sensitivity in subjects with
      impaired glucose metabolism: a randomized controlled trial. Diabetes Care.
      2011;34(4):845-51.
5.    McMurray JJ, Holman RR, Haffner SM, Bethel MA, Holzhauer B, Hua TA, Belenkov
      Y, Boolell M, Buse JB, Buckley BM, Chacra AR, Chiang FT, Charbonnel B, Chow
      CC, Davies MJ, Deedwania P, Diem P, Einhorn D, Fonseca V, Fulcher GR, Gaciong
      Z, Gaztambide S, Giles T, Horton E, Ilkova H, Jenssen T, Kahn SE, Krum H, Laakso
      M, Leiter LA, Levitt NS, Mareev V, Martinez F, Masson C, Mazzone T, Meaney E,
      Nesto R, Pan C, Prager R, Raptis SA, Rutten GE, Sandstroem H, Schaper F, Scheen
      A, Schmitz O, Sinay I, Soska V, Stender S, Tamas G, Tognoni G, Tuomilehto J,
      Villamil AS, Vozar J, Califf RM. Effect of valsartan on the incidence of diabetes and
      cardiovascular events. N Engl J Med. 2010;362(16):1477-90.
6.    Goossens GH. The renin-angiotensin system in the pathophysiology of type 2
      diabetes. Obes Facts. 2012;5(4):611-24.
7.    Bosch J, Yusuf S, Gerstein HC, Pogue J, Sheridan P, Dagenais G, Diaz R, Avezum
      A, Lanas F, Probstfield J, Fodor G, Holman RR. Effect of ramipril on the incidence of
      diabetes. N Engl J Med. 2006;355(15):1551-62.
8.    Magnusson M, Jujic A, Hedblad B, Engstrom G, Persson M, Struck J, Morgenthaler
      NG, Nilsson P, Newton-Cheh C, Wang TJ, Melander O. Low plasma level of atrial
      natriuretic peptide predicts development of diabetes: the prospective Malmo Diet and
      Cancer study. J Clin Endocrinol Metab. 2012;97(2):638-45.
9.    Standeven KF, Hess K, Carter AM, Rice GI, Cordell PA, Balmforth AJ, Lu B, Scott
      DJ, Turner AJ, Hooper NM, Grant PJ. Neprilysin, obesity and the metabolic
      syndrome. Int J Obes (Lond). 2011;35(8):1031-40.
10.   Jordan J, Stinkens R, Jax T, Engeli S, Blaak EE, May M, Havekes B, Schindler C,
      Albrecht D, Pal P, Heise T, Goossens GH, Langenickel TH. Improved Insulin
      Sensitivity with Angiotensin Receptor Neprilysin Inhibition in Individuals with Obesity
      and Hypertension. Clin Pharmacol Ther. 2016.
11.   Moro C. Natriuretic peptides and fat metabolism. Curr Opin Clin Nutr Metab Care.
      2013;16(6):645-9.
12.   Goossens GH, Moors CC, van der Zijl NJ, Venteclef N, Alili R, Jocken JW, Essers Y,
      Cleutjens JP, Clement K, Diamant M, Blaak EE. Valsartan improves adipose tissue
      function in humans with impaired glucose metabolism: a randomized placebo-
      controlled double-blind trial. PLoS One. 2012;7(6):e39930.
13.   Goossens GH, Blaak EE, Arner P, Saris WH, van Baak MA. Angiotensin II: a
      hormone that affects lipid metabolism in adipose tissue. Int J Obes (Lond).
      2007;31(2):382-4.
14.   Goossens GH, Blaak EE, Saris WH, van Baak MA. Angiotensin II-induced effects on
      adipose and skeletal muscle tissue blood flow and lipolysis in normal-weight and
      obese subjects. J Clin Endocrinol Metab. 2004;89(6):2690-6.
172
                                                                                  Chapter 5
                                                                                         173
Chapter 5
SUPPLEMENTARY MATERIAL
174
                                                                                          Chapter 5
A total of 1443 transcripts were modulated by either sacubitril/valsartan (n=916) or amlodipine (n=668)
treatment (nominal P≤0.05, no multiple correction applied). A set of selected genes with corresponding
ratio changes from baseline (RC) and P-value per treatment arm is shown.
                                                                                                  175
Chapter 5
176
                                                                                    Chapter 5
Membranes were probed with antibodies directed against total ATGL, total HSL, phosphorylated HSL
(p-HSL) on Ser660, NPRA (A) and OXPHOS protein expression (B).
                                                                                           177
CHAPTER 6
Exercise training-induced effects on the
abdominal subcutaneous adipose tissue
phenotype in obese humans
* Shared authorship
To be submitted
Chapter 6
ABSTRACT
Aims/hypothesis: Rodent studies have indicated that physical exercise may
improve adipose tissue (AT) function. We investigated the effects of a 12-weeks
supervised, progressive exercise training program on adipocyte morphology and
abdominal subcutaneous AT function in well-phenotyped, obese subjects.
Methods: 21 obese men (14 metabolically compromised - non-alcoholic fatty liver
and/or type 2 diabetes - and 7 metabolically healthy controls) participated in a 12-
weeks supervised, progressive, combined exercise training program. At baseline
and after intervention, abdominal subcutaneous AT biopsies were collected to
determine 1) adipocyte morphology, 2) gene expression of markers for lipolysis,
inflammation, browning, adipokines and mitochondrial biogenesis/function, 3)
protein expression of mitochondrial oxidative phosphorylation (OXPHOS)
complexes and 4) ex vivo basal and β2-adrenergic stimulated lipolysis.
Results: At baseline, AT gene expression of HSL (P=0.005), CGI-58 (P<0.001)
and PGC-1α (P=0.037) were significantly lower in the metabolically compromised
as compared to metabolically healthy obese subjects. Mean adipocyte diameter
and total OXPHOS protein content in AT were comparable between groups. The
exercise training program, which increased maximal aerobic capacity (P time<0.001)
and muscle strength (Ptime<0.001), slightly reduced AT mass (~ 0.7 kg, Ptime=0.037)
but did not affect abdominal subcutaneous adipocyte size (Ptime=0.860), AT gene
expression of markers for mitochondrial biogenesis and function, browning,
lipolysis, inflammation and adipokines, total OXPHOS protein content (P time=0.826)
and β2-adrenergic sensitivity of lipolysis (Ptime=0.555), irrespective of baseline
metabolic status.
Conclusions/interpretation: A 12-weeks supervised, progressive exercise
training program did neither alter abdominal subcutaneous adipocyte morphology
and AT gene and protein expression of markers related to adipose tissue function,
nor β2-adrenergic sensitivity of lipolysis in obese subjects, irrespective of baseline
metabolic status.
180
                                                                           Chapter 6
INTRODUCTION
The obesity epidemic is paralleled by a tremendous increase in the prevalence of
obesity-related diseases, including type 2 diabetes (T2DM), non-alcoholic fatty liver
(NAFL), cardiovascular disease and certain types of cancer [1]. A sedentary
lifestyle is a major contributor to obesity and related complications. In line,
increased habitual physical activity and exercise training may have beneficial
effects on insulin sensitivity and glucose homeostasis in obese, insulin resistant
and T2DM patients [2-4]. Therefore, increasing physical activity is a recommended
lifestyle modification in the prevention and treatment of obesity-related disorders,
including T2DM [5].
Since skeletal muscle is responsible for the majority of glucose disposal,
adaptations in skeletal muscle metabolism are thought to play a central role in the
exercise training-induced improvement of insulin sensitivity. Adipose tissue
dysfunction in obesity, however, represents a key step in the development of
obesity-related insulin resistance and chronic diseases [6, 7]. The reason for this is
that adipocyte hypertrophy in obesity promotes low-grade inflammation and
decreases the adipose tissue lipid buffering capacity. Consequently, lipids
accumulate in non-adipose tissues (e.g. skeletal muscle and liver) when lipid
supply exceeds fat oxidation, thereby accelerating the development and
progression of insulin resistance and chronic metabolic diseases [6-8].
Interestingly, there is evidence that exercise training may improve white adipose
tissue function [9]. Several rodent studies demonstrated that exercise training
increased adipose tissue mitochondrial biogenesis [10, 11] and function [12-14],
induced browning of white adipose tissue [10, 11, 13, 15, 16] and altered adipokine
expression [17, 18]. Furthermore, transplantation of white adipose tissue from
trained animals to untrained recipients markedly improved skeletal muscle glucose
uptake [13], suggesting that improvement of adipose tissue function may contribute
to the increased peripheral insulin sensitivity after exercise training. However,
human studies that investigated the effects of exercise training on the adipose
tissue phenotype are scarce. Exercise training has been shown to increase gene
expression of peroxisome proliferator-activated receptor-gamma coactivator-1
alpha (PGC-1α) [19] and oxidative metabolism markers [20], yet conflicting data
regarding the expression of adipokines and markers of lipolysis in human adipose
tissue have been reported [21-29]. Furthermore, the evidence that exercise training
enhances adipose tissue lipolysis, assessed either ex vivo in isolated adipocytes or
in vivo at rest and in response to a lipolytic stimulus, is inconsistent and
complicated by confounding factors such as recent energy balance, as reviewed
[30]. Importantly, most human studies that have investigated the exercise training-
induced effects on adipose tissue metabolism did not perform detailed metabolic
phenotyping. Therefore, it remains to be established whether the metabolic
phenotype at baseline determines study outcomes.
The aim of the present study was to investigate the effects of a 12-weeks
supervised, progressive, combined exercise training program on abdominal
subcutaneous adipocyte morphology, adipose tissue gene expression of markers
related to mitochondrial biogenesis/function, browning, lipolysis, inflammation and
adipokines and protein expression of mitochondrial oxidative phosphorylation
(OXPHOS) in obese, metabolically healthy and metabolically compromised
                                                                                  181
Chapter 6
individuals, matched for age and BMI. Furthermore, using isolated adipocytes from
these subjects, we determined the exercise training-induced effects on ex vivo
basal and β2-adrenergic stimulation of lipolysis.
METHODS
Study design
Twenty-one sedentary, middle-aged (40–70 yrs), overweight/obese men (14
NAFL/T2DM and 7 age and BMI-matched metabolically healthy control subjects)
participated in the present study, which was conducted within the framework of a
larger clinical trial designed to primarily investigate the effects of exercise training
on liver fat content, hepatic, adipose tissue and peripheral insulin sensitivity [31].
Control subjects (n=7) had low liver fat content (all ≤4%), as measured with proton
                                       1
magnetic resonance spectroscopy ( H-MRS), in the absence of liver dysfunction
(defined as alanine aminotransferase (ALAT) >2.5 times normal values) and had to
be normoglycemic according to the WHO criteria. Subjects were defined as NAFL
                                                                            1
patients (n=7) when having a liver fat content ≥5% as measured with H-MRS, in
addition to a fasting plasma glucose concentration <7.0 mmol/l. Furthermore, at
screening, T2DM patients (n=7) were allowed to be on sulphonyl urea, metformin,
dipeptidyl peptidase-4 inhibitors therapy (or a combination) for at least 6 months
with stable dosage for at least 2 months or on a dietary treatment for 6 months,
with fasting plasma glucose concentrations ≥7.0 and <10.0 mmol/l. Liver fat
content was not a selection criteria for T2DM patients. All subjects gave written
informed consent before participation in the study. The Medical Ethical Committee
                                            +
of Maastricht University Medical Center approved the study protocol, which was
performed according the principles expressed in the Declaration of Helsinki.
All participants were asked not to change their habitual dietary intake during the
study period. General exclusion criteria were unstable body weight, cardiovascular
disease, impaired renal function, hemoglobin <7.5 mmol/l, blood pressure
>160/100 mmHg, participation in a weight-loss or exercise program, history of
substantial alcohol use (>3 units/day), history of drug abuse, use of beta-blockers,
anti-thrombotic medication, insulin therapy and use of medication known to
interfere with glucose homeostasis (except for T2DM patients).
At screening, routine laboratory analyses and physical examinations were
performed, medical history was checked and a resting electrocardiogram (ECG)
was taken. Maximal power output (W max) and maximal aerobic capacity (VO2max)
were assessed during a graded cycling test with concurrent ECG until exhaustion.
Body composition was determined using DEXA (Hologic Discovery A, Waltham,
MA, USA). Furthermore, a two-step hyperinsulinemic-euglycemic clamp with
                  2
primed D-[6,6- H2]-glucose was performed to assess peripheral, hepatic and
adipose tissue insulin sensitivity, as described elsewhere [31].
182
                                                                           Chapter 6
Adipocyte morphology
A part of the adipose tissue biopsy was fixed overnight in 4% paraformaldehyde
and embedded in paraffin for histological sections (8 μm). Sections were cut from
paraffin-embedded tissue, mounted on microscope glass slides and dried overnight
in an incubator at 37°C. The sections were stained with hematoxylin (VWR,
Radnor, PA, USA) and eosin (Klinipath BV, Duiven, The Netherlands). Digital
images were captured with a Leica DFC320 digital camera (Leica, Rijswijk, The
Netherlands) at x20 magnification (Leica DM3000 microscope, Leica, Rijswijk, The
Netherlands) and computerized morphometric analysis (Leica QWin V3,
Cambridge, UK) of individual adipocytes was performed in a blinded manner.
Approximately 400 adipocytes per sample were measured.
Gene expression
Total RNA was extracted from frozen adipose tissue biopsies (~500 mg) using
Trizol chloroform extraction (Invitrogen, Cergy Pontoise, France) and 300ng RNA
was reversed transcribed using iScript cDNA synthesis kit (BIO-RAD). Gene
expression for markers of lipolysis (ATGL [PNPLA2], HSL [LIPE], CGI-58 and
PLIN1 [perilipin 1]), inflammation (TNFα, IL-6, MCP-1 [CCL2], CD68), browning
(CIDEA, PRDM16), mitochondrial biogenesis (PGC-1α [PPARGC1A]) and
adipokine expression (ADIPOQ and LEP) (supplementary table 1 for primer
sequences) was determined in a total volume of 25 μL containing 12.5 ng cDNA
using SYBR-Green based qPCR (iCycler/MyIQ, BIO-RAD). Results were
                                                                                  183
Chapter 6
                       -ΔCT
calculated via the 2          method and normalized for 18S (housekeeping gene)
ribosomal RNA.
Protein expression
A detailed description can be found in the supplementary material. Briefly,
subcutaneous adipose tissue (~500mg) was ground to a fine powder under liquid
nitrogen and homogenized in radioimmunoprecipitation assay buffer. The
homogenate was lysed, vortexed and the supernatant was collected and stored at -
80°C. The protein concentration was determined by the Bradford-based protein
assay (Santa Cruz Biotechnology, Dallas, TX, USA).
Next, solubilized proteins (15 µg) were separated on a precast gel (Criterion™
TGX any kD, Bio-Rad Laboratories Inc, Hercules, CA, USA) and transferred onto a
nitrocellulose membrane (Trans Blot® Turbo™ transfer system; Bio-Rad).
Thereafter, quantitative Western Blot analysis was performed to determine the
levels of OXPHOS proteins. OXPHOS blots were probed with Total OXPHOS
Antibody Cocktail (Mitoscience/Abcam, Cambridge, MA, USA) and a secondary
horseradish     peroxidase     (HRP)-conjugated      Rabbit-anti-Mouse antibody
(DakoCytomation, Glostrup, Denmark). Antigen-antibody complexes were
visualized using chemiluminescence by a ChemiDoc™ XRS apparatus (Bio-Rad)
                                 ®
and analyzed with Quantity One software (Bio-Rad), which calculated the optical
density units that are expressed as average intensity.
Biochemistry
Arterialized blood samples were collected and immediately centrifuged at 4°C for
10 min at 1000 g and plasma was snap frozen in liquid nitrogen and stored at -
80°C until further analysis. Plasma non-esterified fatty acid (NEFA; Wako NEFA C
test kit; Wako Chemicals, Neuss, Germany) and glucose (hexokinase method;
LaRoche, Basel, Switzerland) concentrations were measured with enzymatic
assays, whereas triacylglycerol (TAG) concentrations were measured
colorimetrically (Roche, Vienna, Austria), automated on a Cobas Fara/Mira.
Plasma insulin and serum liver function parameters (aspartate aminotransferase
184
                                                                              Chapter 6
Statistics
Student’s unpaired t-test was used for baseline comparisons between groups. The
effects of exercise training in metabolically healthy and metabolically compromised
obese subjects were compared by two-way repeated measures ANOVA, using
time (baseline and post-intervention) as within-subject factor and group as
between-subject factor. When a significant time*treatment interaction was
observed, post-hoc analysis with Bonferroni correction was applied to identify
significant within-group effects. All variables were checked for normal distribution
and were Ln-transformed to satisfy conditions of normality. All data are presented
as means ± SEM. Calculations were done using SPSS 21 for Mac OS X (IBM,
Chicago, IL, USA). P<0.05 was considered statistically significant.
RESULTS
                                                                                    185
Chapter 6
Adipocyte morphology
At baseline, no differences in mean adipocyte size and adipocyte size distribution
were observed between groups (Figure 1A). The training intervention did not affect
mean adipocyte size, neither in the total group (62.9 ± 1.4 vs. 63.3 ± 1.4 μm,
P=0.860, Figure 1A) nor in both groups separately (Figure 1A). In line, adipocyte
size distribution was not affected by the exercise training in the total group (Figure
1B) and subgroups (Figure 1C).
Mean adipocyte size (A) and adipocyte size distribution in the total group (B); adipocyte size distribution
in the metabolically healthy and metabolically compromised subjects (C).
Panel A and B: white bars, baseline values; black bars, post-intervention values.
Panel C: white and grey bars, baseline values of the metabolically healthy and compromised subjects,
respectively; black and striped bars, post-intervention values of the metabolically healthy and
compromised subjects, respectively.
186
         Table 1. Anthropometric and clinical subject characteristics before and after the 12-weeks supervised, progressive exercise training program.
                                                                                                        Post
                                                     Baseline           Post        Baseline                            Baseline               Post            Ptime           Pgroup       Ptime*group
                                                                                                    intervention
                                                                    intervention                                                           intervention
      Age (years)                                   58.1 ± 1.6            -        60.1 ± 2.3            -              57.1 ± 2.2              -                -               -               -
Body weight (kg) 95.4 ± 2.6 95.4 ± 2.6 93.6 ± 5.5 93.9 ± 5.6 96.3 ± 3.6 96.2 ± 3.8 0.866 0.676 0.707
                -2
      BMI (kg·m )                                   30.0 ± 0.6       30.0 ± 0.6    29.7 ± 1.2        29.8 ± 1.2         30.1 ± 0.8          30.0 ± 0.8         0.890           0.849          0.684
Fat mass (kg) 27.9 ± 1.2 27.2 ± 1.3 27.4 ± 3.0 26.8 ± 3.0 28.3 ± 1.4 27.4 ± 1.6 0.037 0.806 0.838
Fat free mass (kg) 65.7 ± 1.5 66.0 ± 1.6 64.4 ± 3.0 64.8 ± 3.2 66.4 ± 2.2 66.6 ± 2.4 0.309 0.602 0.772
Body fat percentage (%) 28.8 ± 0.7 28.2 ± 0.7 28.6 ± 1.5 28.0 ± 1.6 28.9 ± 0.8 28.3 ± 0.9 0.008 0.888 0.907
                          -1        -1
      VO2max (ml·min ·kg )                          26.9 ± 0.9       29.5 ± 1.0    26.9 ± 1.8        29.3 ± 2.0         27.0 ± 1.0          29.6 ± 1.1        <0.001           0.929          0.910
Wmax (W·kg-1) 2.0 ± 0.8 2.4 ± 1.0 2.1 ± 0.2 2.4 ± 0.2 2.0 ± 0.1 2.3 ± 0.1 <0.001 0.805 0.927
Strength (kg) 83.4 ± 3.8 97.1 ± 4.7 78.2 ± 6.8 91.2 ± 9.6 88.5 ± 5.2 101.8 ± 5.8 <0.001 0.355 0.822
                                               -1
      Fasting plasma glucose (mmol·l )               6.6 ± 0.5        6.6 ± 0.5     5.2 ± 0.2        5.2 ± 0.1          7.2 ± 0.8           7.4 ± 0.9          0.959           0.040          0.982
                                     -1
      Fasting insulin (mU·l )                       13.1 ± 1.5       12.4 ± 1.4     9.8 ± 1.3        9.8 ± 1.1          13.7 ± 2.1          13.7 ± 2.0         0.353           0.188          0.748
HOMA-IR 3.7 ± 0.5 3.4 ± 0.4 2.3 ± 0.3 2.3 ± 0.2 4.1 ± 0.6 4.0 ± 0.6 0.399 0.048 0.751
Fasting plasma NEFA (μmol·l-1) 702 ± 31.7 648 ± 38.8 669 ± 46.6 633 ± 90.4 735 ± 44.3 670 ± 36.2 0.184 0.566 0.691
                                          -1
      Fasting plasma TAG (mmol·l )                   1.6 ± 0.1        1.6 ± 0.2     1.4 ± 0.2        1.4 ± 0.3          1.6 ± 0.2           1.7 ± 0.2          0.712           0.302          0.709
                     -1        -1
      Rd (μmol·kg ·min )                            11.5 ± 2.2       14.2 ± 2.8    22.1 ± 2.9        27.8 ± 2.5         6.7 ± 1.1           8.0 ± 1.9          0.015          <0.001          0.106
EGP suppression (%) -48.6 ± 4.5 -53.4 ± 5.6 -71.0 ± 3.6 -78.3 ± 10.6 -38.4 ± 2.8 -42.0 ± 2.5 0.213 <0.001 0.663
NEFA suppression (%) -61.6 ± 3.2 -62.1 ± 3.2 -69.8 ± 2.4 -68.9 ± 3.0 -56.8 ± 4.4 -58.1 ± 4.4 0.943 0.036 0.736
      Data are expressed as mean ± SEM (n=21). VO2max: maximal aerobic capacity; Wmax: maximal power output; TAG: triacylglycerol; Rd: glucose rate of disappearance (reflects peripheral insulin
      sensitivity); EGP: endogenous glucose production (EGP suppression reflects hepatic insulin sensitivity); NEFA: non-esterified fatty acid (NEFA suppression reflects adipose tissue insulin
                                                                                                                                                                                                          Chapter 6
187
      sensitivity)
Chapter 6
Gene expression
At baseline, gene expression of HSL (0.35 ± 0.08 vs. 0.45 ± 0.11, P=0.005), CGI-
58 (0.39 ± 0.04 vs. 0.75 ± 0.19, P<0.001) and PGC-1α (-0.35 ± 0.22 vs. 0.00 ±
0.13, P=0.037) was significantly lower in the obese metabolically compromised as
compared to obese metabolically healthy individuals. In the total group, the
exercise training did not alter adipose tissue gene expression of ATGL
(Ptime=0.332, Figure 2A), HSL (Ptime=0.862, Figure 2B), PLIN1 (Ptime=0.614, Figure
2C) and CGI-58 (Ptime=0.546, Figure 2D). Furthermore, the inflammatory markers
TNFα (Ptime=0.604, Figure 2E), IL-6 (Ptime=0.507, Figure 2F), MCP-1 (Ptime=0.222,
Figure 2G) and CD68 (Ptime=0.688, Figure 2H) were unchanged after the
intervention. Next, gene expression of the browning markers CIDEA (Ptime=0.943,
Figure 2I) and PRDM16 (Ptime=0.839, Figure 2J) and PGC-1α (Ptime=0.835, Figure
2K), a major regulator of mitochondrial biogenesis and function, remained
unchanged following the exercise training. Finally, gene expression of LEP
(Ptime=0.840, Figure 2L) and ADIPOQ (Ptime=0.413, Figure 2M) was also not
significantly altered after the 12-weeks training program. In line, no significant
differences in exercise-induced alterations in these parameters were observed
between groups, except for a slight but significant change in CGI-58
(Ptime*group=0.037).
188
                                                                                        Chapter 6
Genes involved in lipolysis (A-D), inflammation (E-H), browning (I-J), mitochondrial biogenesis and
function (K) and adipokine expression (L-M) are expressed as fold change relative to the baseline
values of the total group and of the obese metabolically healthy control group.
* Ptime*group<0.05; #P<0.05 compared to baseline value of obese metabolically healthy control group.
White bars, baseline values; black bars, post-intervention values.
                                                                                               189
Chapter 6
Protein expression
At baseline, total OXPHOS protein content was not significantly different between
groups (P=0.176). In the total group, total OXPHOS protein expression remained
unchanged following the training program (27.0 ± 9.7 vs. 24.9 ± 6.9 AU,
Ptime=0.826, Figure 3A). More specific, OXPHOS complex I (2.9 ± 1.2 vs. 2.9 ± 1.0
AU, Ptime=0.857, Figure 3B), complex II (9.2 ± 2.4 vs. 8.9 ± 1.7 AU, P time=0.804,
Figure 3C), complex III (4.7 ± 2.7 vs. 2.1 ± 1.0 AU, Ptime=0.549, Figure 3D),
complex IV (0.8 ± 0.2 vs. 0.9 ± 0.3 AU, Ptime=0.870, Figure 3E) and complex V
(14.5 ± 6.8 vs. 13.3 ± 5.3 AU, Ptime=0.666, Figure 3F) were not affected. In line, no
significant differences in exercise-induced alterations in OXPHOS protein
complexes were observed between groups (Figure 3A-F).
Protein content of total OXPHOS (A), OXPHOS complex I (B), OXPHOS complex II (C), OXPHOS
complex III (D), OXPHOS complex IV (E) and OXPHOS complex V (F), expressed as fold change
relative to baseline values of the total group (time effect) and of the obese metabolically healthy group
(time*treatment interaction).
White bars, baseline values; black bars, post-intervention values.
190
                                                                          Chapter 6
DISCUSSION
The aim of the present study was to investigate the effects of a 12-weeks
supervised, progressive exercise training program on the abdominal subcutaneous
adipose tissue phenotype in metabolically healthy and metabolically compromised,
well-phenotyped obese individuals. Here, we demonstrate that exercise training did
neither alter abdominal subcutaneous adipocyte morphology and adipose tissue
gene expression of markers for mitochondrial biogenesis/function, browning,
lipolysis, inflammation and adipokines, adipose tissue OXPHOS protein content,
nor β2-adrenergic stimulation of adipocyte lipolysis in obese subjects, irrespective
of baseline metabolic status. These data suggest that alterations in the phenotype
of abdominal subcutaneous adipose tissue do not significantly contribute to the
exercise-induced improvement in peripheral insulin sensitivity in obese men when
adipose tissue mass is only slightly reduced (~0.7 kg).
The training program induced a significant increase in aerobic capacity, maximal
power output and maximal muscle strength, indicating that the supervised,
progressive nature of the program was successful regarding enhancement of
physical fitness. This was accompanied by a slight but significant decrease in fat
mass and body fat percentage. In agreement with the findings in a larger study
population [31], we observed that peripheral insulin sensitivity was significantly
increased, whereas hepatic and adipose tissue insulin sensitivity remained
unchanged after the training program.
Exercise training interventions may affect adipocyte morphology in humans [30].
We demonstrated that a 12-weeks exercise training intervention did not
significantly alter mean adipocyte size or adipocyte size distribution, despite a 0.7
kg decrease in total fat mass. In contrast, Despres et al. [32] demonstrated that 20
weeks of endurance training decreased mean adipocyte size in young men, but not
in women. Importantly, they observed a more pronounced reduction in body weight
(~3.0 kg). Thus, a more prolonged intervention period, leading to a more
pronounced decrease in adipose tissue mass, seems necessary to induce
beneficial changes in adipocyte morphology.
Since an altered rate of lipolysis is one of the characteristics of adipose tissue
dysfunction and relates to peripheral insulin resistance [8], we determined adipose
tissue gene expression of lipolytic enzymes and genes encoding lipid droplet-
associated proteins. At baseline, gene expression of HSL and CGI-58 was lower in
obese metabolically compromised as compared to obese metabolically healthy
subjects. These findings are in line with previous studies from our group and
                                                                                 191
Chapter 6
192
                                                                          Chapter 6
                                                                                 193
Chapter 6
REFERENCES
1.    Kopelman PG. Obesity as a medical problem. Nature. 2000;404(6778):635-43.
2.    Mann S, Beedie C, Balducci S, Zanuso S, Allgrove J, Bertiato F, Jimenez A.
      Changes in insulin sensitivity in response to different modalities of exercise: a review
      of the evidence. Diabetes Metab Res Rev. 2014;30(4):257-68.
3.    Roberts CK, Little JP, Thyfault JP. Modification of insulin sensitivity and glycemic
      control by activity and exercise. Med Sci Sports Exerc. 2013;45(10):1868-77.
4.    Meex RC, Schrauwen-Hinderling VB, Moonen-Kornips E, Schaart G, Mensink M,
      Phielix E, van de Weijer T, Sels JP, Schrauwen P, Hesselink MK. Restoration of
      muscle mitochondrial function and metabolic flexibility in type 2 diabetes by exercise
      training is paralleled by increased myocellular fat storage and improved insulin
      sensitivity. Diabetes. 2010;59(3):572-9.
5.    Colberg SR, Sigal RJ, Fernhall B, Regensteiner JG, Blissmer BJ, Rubin RR, Chasan-
      Taber L, Albright AL, Braun B, American College of Sports M, American Diabetes A.
      Exercise and type 2 diabetes: the American College of Sports Medicine and the
      American Diabetes Association: joint position statement. Diabetes Care.
      2010;33(12):e147-67.
6.    Goossens GH. The role of adipose tissue dysfunction in the pathogenesis of obesity-
      related insulin resistance. Physiol Behav. 2008;94(2):206-18.
7.    Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell.
      2014;156(1-2):20-44.
8.    Stinkens R, Goossens GH, Jocken JW, Blaak EE. Targeting fatty acid metabolism to
      improve glucose metabolism. Obes Rev. 2015;16(9):715-57.
9.    Stanford KI, Middelbeek RJ, Goodyear LJ. Exercise Effects on White Adipose Tissue:
      Beiging and Metabolic Adaptations. Diabetes. 2015;64(7):2361-8.
10.   Sutherland LN, Bomhof MR, Capozzi LC, Basaraba SA, Wright DC. Exercise and
      adrenaline increase PGC-1{alpha} mRNA expression in rat adipose tissue. J Physiol.
      2009;587(Pt 7):1607-17.
11.   Trevellin E, Scorzeto M, Olivieri M, Granzotto M, Valerio A, Tedesco L, Fabris R,
      Serra R, Quarta M, Reggiani C, Nisoli E, Vettor R. Exercise training induces
      mitochondrial biogenesis and glucose uptake in subcutaneous adipose tissue
      through eNOS-dependent mechanisms. Diabetes. 2014;63(8):2800-11.
12.   Stallknecht B, Vinten J, Ploug T, Galbo H. Increased activities of mitochondrial
      enzymes in white adipose tissue in trained rats. Am J Physiol. 1991;261(3 Pt
      1):E410-4.
13.   Stanford KI, Middelbeek RJ, Townsend KL, Lee MY, Takahashi H, So K, Hitchcox
      KM, Markan KR, Hellbach K, Hirshman MF, Tseng YH, Goodyear LJ. A novel role for
      subcutaneous adipose tissue in exercise-induced improvements in glucose
      homeostasis. Diabetes. 2015;64(6):2002-14.
14.   Vernochet C, Mourier A, Bezy O, Macotela Y, Boucher J, Rardin MJ, An D, Lee KY,
      Ilkayeva OR, Zingaretti CM, Emanuelli B, Smyth G, Cinti S, Newgard CB, Gibson
      BW, Larsson NG, Kahn CR. Adipose-specific deletion of TFAM increases
      mitochondrial oxidation and protects mice against obesity and insulin resistance. Cell
      Metab. 2012;16(6):765-76.
15.   Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, Rasbach KA, Bostrom
      EA, Choi JH, Long JZ, Kajimura S, Zingaretti MC, Vind BF, Tu H, Cinti S, Hojlund K,
      Gygi SP, Spiegelman BM. A PGC1-alpha-dependent myokine that drives brown-fat-
      like development of white fat and thermogenesis. Nature. 2012;481(7382):463-8.
16.   Cao L, Choi EY, Liu X, Martin A, Wang C, Xu X, During MJ. White to brown fat
      phenotypic switch induced by genetic and environmental activation of a
      hypothalamic-adipocyte axis. Cell Metab. 2011;14(3):324-38.
194
                                                                                Chapter 6
17.   Zachwieja JJ, Hendry SL, Smith SR, Harris RB. Voluntary wheel running decreases
      adipose tissue mass and expression of leptin mRNA in Osborne-Mendel rats.
      Diabetes. 1997;46(7):1159-66.
18.   Bradley RL, Jeon JY, Liu FF, Maratos-Flier E. Voluntary exercise improves insulin
      sensitivity and adipose tissue inflammation in diet-induced obese mice. Am J Physiol
      Endocrinol Metab. 2008;295(3):E586-94.
19.   Ruschke K, Fishbein L, Dietrich A, Kloting N, Tonjes A, Oberbach A, Fasshauer M,
      Jenkner J, Schon MR, Stumvoll M, Bluher M, Mantzoros CS. Gene expression of
      PPARgamma and PGC-1alpha in human omental and subcutaneous adipose tissues
      is related to insulin resistance markers and mediates beneficial effects of physical
      training. Eur J Endocrinol. 2010;162(3):515-23.
20.   Ronn T, Volkov P, Tornberg A, Elgzyri T, Hansson O, Eriksson KF, Groop L, Ling C.
      Extensive changes in the transcriptional profile of human adipose tissue including
      genes involved in oxidative phosphorylation after a 6-month exercise intervention.
      Acta Physiol (Oxf). 2014;211(1):188-200.
21.   Bluher M, Williams CJ, Kloting N, Hsi A, Ruschke K, Oberbach A, Fasshauer M,
      Berndt J, Schon MR, Wolk A, Stumvoll M, Mantzoros CS. Gene expression of
      adiponectin receptors in human visceral and subcutaneous adipose tissue is related
      to insulin resistance and metabolic parameters and is altered in response to physical
      training. Diabetes Care. 2007;30(12):3110-5.
22.   Sjogren P, Sierra-Johnson J, Kallings LV, Cederholm T, Kolak M, Halldin M, Brismar
      K, de Faire U, Hellenius ML, Fisher RM. Functional changes in adipose tissue in a
      randomised controlled trial of physical activity. Lipids Health Dis. 2012;11:80.
23.   Trachta P, Drapalova J, Kavalkova P, Touskova V, Cinkajzlova A, Lacinova Z,
      Matoulek M, Zelinka T, Widimsky J, Jr., Mraz M, Haluzik M. Three months of regular
      aerobic exercise in patients with obesity improve systemic subclinical inflammation
      without major influence on blood pressure and endocrine production of subcutaneous
      fat. Physiol Res. 2014;63 Suppl 2:S299-308.
24.   Moghadasi M, Mohebbi H, Rahmani-Nia F, Hassan-Nia S, Noroozi H, Pirooznia N.
      High-intensity endurance training improves adiponectin mRNA and plasma
      concentrations. Eur J Appl Physiol. 2012;112(4):1207-14.
25.   Klimcakova E, Polak J, Moro C, Hejnova J, Majercik M, Viguerie N, Berlan M, Langin
      D, Stich V. Dynamic strength training improves insulin sensitivity without altering
      plasma levels and gene expression of adipokines in subcutaneous adipose tissue in
      obese men. J Clin Endocrinol Metab. 2006;91(12):5107-12.
26.   Polak J, Klimcakova E, Moro C, Viguerie N, Berlan M, Hejnova J, Richterova B,
      Kraus I, Langin D, Stich V. Effect of aerobic training on plasma levels and
      subcutaneous abdominal adipose tissue gene expression of adiponectin, leptin,
      interleukin 6, and tumor necrosis factor alpha in obese women. Metabolism.
      2006;55(10):1375-81.
27.   Hulver MW, Zheng D, Tanner CJ, Houmard JA, Kraus WE, Slentz CA, Sinha MK,
      Pories WJ, MacDonald KG, Dohm GL. Adiponectin is not altered with exercise
      training despite enhanced insulin action. Am J Physiol Endocrinol Metab.
      2002;283(4):E861-5.
28.   Richterova B, Stich V, Moro C, Polak J, Klimcakova E, Majercik M, Harant I, Viguerie
      N, Crampes F, Langin D, Lafontan M, Berlan M. Effect of endurance training on
      adrenergic control of lipolysis in adipose tissue of obese women. J Clin Endocrinol
      Metab. 2004;89(3):1325-31.
29.   Covington JD, Bajpeyi S, Moro C, Tchoukalova YD, Ebenezer PJ, Burk DH, Ravussin
      E, Redman LM. Potential effects of aerobic exercise on the expression of perilipin 3
      in the adipose tissue of women with polycystic ovary syndrome: a pilot study. Eur J
      Endocrinol. 2015;172(1):47-58.
                                                                                      195
Chapter 6
30.   Thompson D, Karpe F, Lafontan M, Frayn K. Physical activity and exercise in the
      regulation of human adipose tissue physiology. Physiol Rev. 2012;92(1):157-91.
31.   Brouwers B, Schrauwen-Hinderling VB, Jelenik T, Gemmink A, Havekes B, Bruls YM,
      Dahlmans D, Roden M, Hesselink MK, Schrauwen P. Metabolic disturbances of non-
      alcoholic fatty liver resemble the alterations typical for overt type 2 diabetes. Clin Sci
      (Lond). 2017;131(15):1905-17.
32.   Despres JP, Bouchard C, Savard R, Tremblay A, Marcotte M, Theriault G. The effect
      of a 20-week endurance training program on adipose-tissue morphology and lipolysis
      in men and women. Metabolism. 1984;33(3):235-9.
33.   Jocken JW, Langin D, Smit E, Saris WH, Valle C, Hul GB, Holm C, Arner P, Blaak
      EE. Adipose triglyceride lipase and hormone-sensitive lipase protein expression is
      decreased in the obese insulin-resistant state. J Clin Endocrinol Metab.
      2007;92(6):2292-9.
34.   Watt MJ, Carey AL, Wolsk-Petersen E, Kraemer FB, Pedersen BK, Febbraio MA.
      Hormone-sensitive lipase is reduced in the adipose tissue of patients with type 2
      diabetes mellitus: influence of IL-6 infusion. Diabetologia. 2005;48(1):105-12.
35.   Schiffelers SL, Saris WH, Boomsma F, van Baak MA. beta(1)- and beta(2)-
      Adrenoceptor-mediated thermogenesis and lipid utilization in obese and lean men. J
      Clin Endocrinol Metab. 2001;86(5):2191-9.
36.   De Glisezinski I, Crampes F, Harant I, Berlan M, Hejnova J, Langin D, Riviere D,
      Stich V. Endurance training changes in lipolytic responsiveness of obese adipose
      tissue. Am J Physiol. 1998;275(6 Pt 1):E951-6.
37.   Reynisdottir S, Langin D, Carlstrom K, Holm C, Rossner S, Arner P. Effects of weight
      reduction on the regulation of lipolysis in adipocytes of women with upper-body
      obesity. Clin Sci (Lond). 1995;89(4):421-9.
38.   Alvehus M, Boman N, Soderlund K, Svensson MB, Buren J. Metabolic adaptations in
      skeletal muscle, adipose tissue, and whole-body oxidative capacity in response to
      resistance training. Eur J Appl Physiol. 2014;114(7):1463-71.
39.   Harms M, Seale P. Brown and beige fat: development, function and therapeutic
      potential. Nat Med. 2013;19(10):1252-63.
40.   Elsen M, Raschke S, Eckel J. Browning of white fat: does irisin play a role in
      humans? J Endocrinol. 2014;222(1):R25-38.
41.   Bordicchia M, Liu D, Amri EZ, Ailhaud G, Dessi-Fulgheri P, Zhang C, Takahashi N,
      Sarzani R, Collins S. Cardiac natriuretic peptides act via p38 MAPK to induce the
      brown fat thermogenic program in mouse and human adipocytes. J Clin Invest.
      2012;122(3):1022-36.
42.   Vosselman MJ, Hoeks J, Brans B, Pallubinsky H, Nascimento EB, van der Lans AA,
      Broeders EP, Mottaghy FM, Schrauwen P, van Marken Lichtenbelt WD. Low brown
      adipose tissue activity in endurance-trained compared with lean sedentary men. Int J
      Obes (Lond). 2015;39(12):1696-702.
43.   Camera DM, Anderson MJ, Hawley JA, Carey AL. Short-term endurance training
      does not alter the oxidative capacity of human subcutaneous adipose tissue. Eur J
      Appl Physiol. 2010;109(2):307-16.
44.   Norheim F, Langleite TM, Hjorth M, Holen T, Kielland A, Stadheim HK, Gulseth HL,
      Birkeland KI, Jensen J, Drevon CA. The effects of acute and chronic exercise on
      PGC-1alpha, irisin and browning of subcutaneous adipose tissue in humans. FEBS J.
      2014;281(3):739-49.
45.   Heinonen S, Buzkova J, Muniandy M, Kaksonen R, Ollikainen M, Ismail K,
      Hakkarainen A, Lundbom J, Lundbom N, Vuolteenaho K, Moilanen E, Kaprio J,
      Rissanen A, Suomalainen A, Pietilainen KH. Impaired Mitochondrial Biogenesis in
      Adipose Tissue in Acquired Obesity. Diabetes. 2015;64(9):3135-45.
46.   Semple RK, Crowley VC, Sewter CP, Laudes M, Christodoulides C, Considine RV,
      Vidal-Puig A, O'Rahilly S. Expression of the thermogenic nuclear hormone receptor
196
                                                                                Chapter 6
                                                                                      197
Chapter 6
SUPPLEMENTARY MATERIAL
Glycerol release into the medium (µmol glycerol·107 cells-1·2h incubation-1) was used as an indicator of
lipolysis.
Open circles represent baseline values; closed circles represent post-intervention values.
198
                                                                                       Chapter 6
The following RT-qPCR primer sequences were used for gene expression analysis.
Upper sequences represent forward primers (5' - 3'), while lower sequences represent reverse primer
(5' - 3').
Genes Sequences
                                                      GTGTCAGACGGCGAGAATG
                  ATGL
                                                      TGGAGGGAGGGAGGGATG
                                                    GCGGATCACACAGAACCTGGAC
                   HSL
                                                     AGCAGGCGGCTTACCCTCAC
                                                    CAGCATCCAGTCCTTACGACCA
                 CGI-58
                                                    GTTCAGTCCACAGTGTCGCAGA
                                                      CTCTCGATACACCGTGCAGA
                  PLIN1
                                                      TGGTCCTCATGATCCTCCTC
                                                     CCGAGTGACAAGCCTGTAGC
                  TNFα
                                                    GAGGACCTGGGAGTAGATGAG
                                                   AAATTCGGTACATCCTCGACGG
                   IL-6
                                                   GGAAGGTTCAGGTTGTTTTCTGC
                                                      CCCCAGTCACCTGCTGTTAT
                  MCP-1
                                                      TCCTGAACCCACTTCTGCTT
                                                     CCCTATGGACACCTCAGCTTT
                  CD68
                                                    GAAGGACACATTGTACTCCACC
                                                    TCAGACCTTGGGAGACAACACG
                  CIDEA
                                                    CGAAGGTGACTCTCGCTATTCC
                                                    CAGCCAATCTCACCAGACACCT
                PRDM16
                                                    GTGGCACTTGAAAGGCTTCTCC
                                                   TCTGAGTCTGTATGGAGTGACAT
                 PGC-1α
                                                   CCAAGTCGTTCACATCTAGTTCA
                                                       TGGTGAGAAGGGTGAGAA
                ADIPOQ
                                                      AGATCTTGGTAAAGCGAATG
                                                    GAACCCTGTGCGGATTCTTGT
                   LEP
                                                   TCCATCTTGGATAAGGTCAGGAT
                                                                                              199
CHAPTER 7
Coordinated regulation of adipose tissue
adrenergic- and non-adrenergic-
mediated lipolysis during exercise in lean
and obese individuals:
the effect of exercise training
Stinkens R.*, Verboven K.*, Hansen D., Wens I., Frederix I.,
Eijnde B.O., Jocken J.W., Goossens G.H.#, Blaak E.E.#
Submitted
Chapter 7
ABSTRACT
Background: Adipose tissue dysfunction, which includes impairments in (adipose
tissue) lipolysis, contributes to insulin resistance. Subcutaneous adipose tissue
(SCAT) lipolysis in obesity is characterized by catecholamine resistance and an
impaired ANP responsiveness. It remains to be established whether exercise
training improves non-adrenergically-mediated lipolysis, next to the adrenergic
pathway, in metabolically compromised conditions. The aim of the present study
was to investigate the effect of local combined α- and β-adrenergic receptor
blockade on SCAT lipolysis in obese insulin sensitive (IS), obese insulin resistant
(IR) and age-matched lean IS men. Moreover, obese individuals underwent
endurance and resistance exercise training to improve metabolic profile and (non-)
adrenergically-mediated SCAT lipolysis.
Methods: Abdominal SCAT lipolysis was investigated in 10 obese IS, 10 obese IR
and 10 age-matched lean IS men using microdialysis in the presence or absence
of local combined α- and β-adrenergic receptor blockade at rest, during 60 min of
low-intense (40% VO2max) endurance-type exercise and recovery. Systemic
responses were investigated using venous blood sampling. Obese individuals
participated in a supervised, endurance and resistance exercise training
intervention for 12 weeks (3 sessions/week) after which the microdialysis
measurements were repeated in obese IR men.
Results: Exercise-induced increase in abdominal SCAT lipolysis (expressed as
total area under the curve) was more pronounced in obese IS (81%) and IR (34%)
as compared to lean individuals (Pgroup=0.012). Abdominal SCAT lipolysis was
significantly reduced (~40%) following local combined α-/β-adrenoceptor blockade
in obese IS individuals only. Despite improvements in body composition, physical
fitness and exercise-induced changes in circulating free fatty acids, lactate and
adrenalin, exercise intervention did not significantly affect (non-)adrenergically-
mediated lipolysis in abdominal SCAT of obese IR individuals.
Conclusion: Our findings indicate a major contribution of non-adrenergically
mediated lipolysis during exercise in abdominal SCAT of lean and obese
individuals. Furthermore, a 12-week exercise training program improved metabolic
profile and body composition in obese individuals, but did not affect abdominal
SCAT lipolysis.
202
                                                                            Chapter 7
INTRODUCTION
Adipose tissue (AT) dysfunction is commonly observed in human obesity and
contributes to insulin resistance (IR) and chronic metabolic diseases, including
cardiovascular disease, type 2 diabetes mellitus (T2D) and certain types of cancer
[1, 2]. Disturbances in AT lipid metabolism, including a decreased lipid uptake and
impairments in lipid mobilization are closely linked to ectopic fat deposition and
obesity-related IR [3]. An important function of the AT is to release fatty acids
through lipolysis [4, 5], especially during fasting and increased energy demanding
conditions such as exercise. Multiple endocrine factors affect the activity of lipid
droplet-associated proteins and lipases, thereby regulating the release of free fatty
acids (FFA) and glycerol [6]. However, impairments in the regulation of lipolysis
have been identified in subcutaneous AT (SCAT) of obese humans [7], including a
blunted catecholamine-mediated lipolysis [8, 9]. More specific, β-adrenergically-
mediated lipolysis is reduced [9] and inhibitory α2-adrenoceptors become
predominant on adipocytes in the obese insulin resistant state [10] [11], leading to
a blunted adrenergically-mediated lipolysis [9, 12]. Of interest, local β-adrenergic
blockade (alone or in combination with α2-adrenergic blockade) in SCAT, inhibits
exercise-induced lipolysis only to a minor extent at low-to-moderate intensities in
healthy lean [13-15] and overweight individuals [16]. In this respect, Moro et al. [16]
demonstrated that non-adrenergically-mediated lipolysis in SCAT substantially
contributes to lipid mobilization during exercise in healthy young lean men [15] and
healthy young overweight men [16]. Other key regulators of lipolysis are insulin [17]
and lactate [18], which both exert an inhibitory role in the physiological control of
AT during exercise [17-19].
More recently, evidence has emerged that natriuretic peptides (NP) not only affect
the cardiovascular system, but also have pronounced effects in several key
metabolic organs such as AT and skeletal muscle [20]. Interestingly, several
studies have indicated that the circulating NP concentrations are reduced in human
obesity and T2D [21-23]. The latter findings, together with evidence that reduced
systemic NP concentrations increase the risk of developing T2D [24, 25], highlight
the importance of NP in metabolic disease. Of the NP family, atrial natriuretic
peptide (ANP) has been shown to be the most potent stimulator of human AT
lipolysis [26], via guanylyl cyclase-coupled natriuretic peptide receptor type A
(NPRA)-mediated activation of hormone-sensitive lipase (HSL) [27, 28].
Interestingly, we have recently found that maximal ANP responsiveness is impaired
in isolated abdominal subcutaneous adipocytes of obese non-diabetic and T2D
men [29]. In line, Rydén and colleagues [30] have recently shown a blunted
lipolytic effect of ANP in isolated abdominal subcutaneous adipocytes of obese
women and in situ (microdialysis) in abdominal SCAT of overweight men under
resting conditions. Importantly, however, the physiological role of exercise-induced
ANP-mediated lipolysis in human obesity remains to be established.
It has been shown that endurance exercise training can partly improve β-
adrenoceptor activity, reduce anti-lipolytic α2-adrenoceptor sensitivity in human
SCAT [31-33], and alleviate ANP-mediated lipolysis in subcutaneous adipocytes in
young, metabolically healthy overweight individuals [28, 34]. However, to date, it
remains elusive if endurance and resistance exercise intervention improves ANP-
induced activation of lipolysis in metabolically compromised conditions.
                                                                                   203
Chapter 7
The aim of the present study was to compare the effect of local combined α- and β-
adrenoceptor blockade on local SCAT lipolysis at rest, during low-intensity
endurance-type exercise and during recovery from exercise in middle-aged obese
insulin sensitive (IS), obese insulin resistant (IR) and age-matched lean IS men. In
addition, we investigated whether a 12-week endurance and resistance exercise
training improved the metabolic profile in obese men and (non-)adrenergically-
mediated abdominal SCAT lipolysis in obese IR men.
METHODS
Subjects
Ten middle-aged healthy lean insulin sensitive (IS), 10 obese IS and 10 obese
insulin resistant (IR) men, matched for age and BMI (obese groups) participated in
the present study. Subjects were included when they had a stable body weight for
at least 3 months prior to the start of the intervention and had no contraindications
for participation in an exercise training intervention based on their medical history.
Major exclusion criteria were a history, or clinical symptoms, of heart, lung or
kidney disease, presence of endocrine anomalies and/or the use of beta-blockers,
glucose or lipid-lowering medication. Insulin sensitivity was assessed via
homeostasis assessment of insulin resistance (HOMA-IR) [35]. Subjects were
classified as insulin sensitive or insulin resistant when HOMA-IR was ≤ 2.3 [36] or ≥
3.8 [37], respectively. Height, weight, waist and hip circumference and blood
pressure were measured during screening. Body composition was measured using
a Dual Energy X-ray Absorptiometry scan (Hologic Series Delphi-A Fan Beam X-
ray Bone Densitometer). One week before the investigational protocol, peak
oxygen uptake (VO2peak) was determined during a maximal cardiopulmonary
exercise test performed on an electrical braked cycle ergometer (Gymna Ergofit
Cycle 400, Bilzen, Belgium) by using an incremental procedure (work rate
increased by 15W/min until volitional exhaustion). Heart rate (electrocardiography)
was monitored continuously and VO2peak was measured using a Metalyzer II
(Cortex Medical, Leipzig, Germany). The study was approved by the Medical
Ethical Committee of the Jessa Hospital and Hasselt University, Hasselt, Belgium,
and performed in accordance with the declaration of Helsinki (2008). All individuals
gave written informed consent prior to the start of the study.
Experimental protocol
Subjects arrived at the hospital at 07:30 AM after an overnight fast. They were
instructed to consume a standardized meal and snack the evening before the test
day (total energy: 2628 kJ (626 kcal); 23.4g fat (10.4g saturated fat); 73.8g
carbohydrates (of which 6.8g sugar); 28.8g protein; 2.9g salt; 2.3g fibres) and to
abstain from exhausting activities 48 hours prior to the experimental protocol. On
arrival, a catheter was inserted into the antecubital vein for blood sampling. Two
microdialysis catheters (CMA 63, CMA Microdialysis AB, Stockholm, Sweden)
                                                                                 ®
were inserted percutaneously into SCAT after epidermal anesthesia (EMLA
crème: lidocaine 2.5% and prilocaine 2.5%, AstraZeneca AB) at a distance of 6-8
204
                                                                           Chapter 7
cm from the umbilicus (one probe on the left side and one probe on the right side of
the umbilicus). The probes were connected to a microinfusion pump (Harvard
apparatus, Plato BV, Diemen, The Netherlands) and perfused with Ringer solution
(in mmol/l: 147 sodium, 4 potassium, 2.25 calcium and 156 chloride; Fresenius
Kabi BV, ‘s Hertogenbosch, The Netherlands) at a perfusion rate of 2.0 µl/min.
Ethanol (50 mmol/l) was added to the perfusate to semi-qualitatively estimate
changes in local adipose tissue blood flow (ATBF), using the ethanol outflow/inflow
(out/in) ratio [38]. A higher ethanol out/in ratio, corresponding to a lower ethanol
wash-out, reflects a lower regional ATBF.
One microdialysis catheter was perfused with Ringer solution (control), while the
contralateral catheter was perfused with Ringer, supplemented with 100 μmol/l
phentolamine (α1,2-adrenergic receptor antagonist) (Regitin 10 mg/ml; Novartis
Pharma BV, The Netherlands) and 100 μmol/l propranolol (nonselective β-
adrenergic receptor antagonist) (propranolol hydrochloride, Dociton 1 mg/ml, Mibe
GmbH, Germany), concentrations that completely suppress lipolysis [28, 39, 40].
After a 60-min equilibration period (recovery from insertion), two 30-min fraction of
dialysate were collected at a flow rate of 0.3 µL/min after which the perfusion rate
was increased to 2.0 µL/min for the remaining of the experiment. During the resting
phase, three 15-min fractions of the outgoing dialysate were collected from both
sites to determine the extracellular glycerol concentration (reflecting basal
lipolysis). Next, subjects performed a single bout of endurance exercise for 60 min
at 40% of their VO2max on a cycle ergometer while heart rate was monitored
continuously (Polar, Kempele, Finland). Exercise was followed by a 60-min
recovery period in supine position. During exercise and recovery, dialysate
samples were collected at 15 min intervals without disconnecting the microdialysis
probes from the microinfusion pumps.
Ethanol concentrations were determined both in the ingoing (perfusate) and
outgoing (dialysate) fluid to assess the ethanol out/in ratio as an indicator of local
nutritive blood flow. Ethanol concentrations were determined at the same day,
whereas dialysate samples for measurement of extracellular glycerol, glucose and
lactate concentrations were immediately frozen and stored at -80 °C until analysis.
Venous blood samples were taken at rest, during exercise and recovery in
prechilled 20 mL tubes at 15 min intervals throughout the study protocol.
Indirect calorimetry
Substrate utilization and energy expenditure were determined at rest and during
submaximal exercise via indirect calorimetry using a Metalyzer II (Cortex Medical,
Leipzig, Germany). Substrate oxidation rates (g/min) and energy expenditure were
calculated from VO2 and VCO2 [41, 42]. Water intake was allowed ad libitum during
the exercise and recovery period.
                                                                                  205
Chapter 7
Biochemical analysis
Microdialysate samples were analyzed for glycerol, glucose and lactate
concentrations by means of bioluminescence on an ISCUS clinical microdialysis
analyzer (M dialysis AB, Stockholm, Sweden). Ethanol concentrations in dialysate
(out) and perfusate (in) were measured spectrophotometrically using a COBAS
FARA semi-automatic analyzer (Roche Diagnostics, Basal, Switzerland) and using
a standard ethanol assay kit (Boehringer Mannheim, Germany).
Blood samples were centrifuged at 4°C for 10 min at 1200 g and plasma and
serum was stored at -80°C until further analysis. Plasma free glycerol was
                                                                  TM
measured after precipitation with an enzymatic assay (Enzytec Glycerol, Roche
Biopharm, Switzerland), automated on a Cobas Fara spectrophotometric
autoanalyzer (Roche Diagnostics, Basel, Switzerland). Plasma FFA, glucose and
lactate concentrations were measured with enzymatic assays on an automated
spectrophotometer (ABX Pentra 400 autoanalyzer, Horiba ABX, Montpellier,
France). Plasma ANP concentrations were measured using an enzyme
immunoassay (RayBiotech, Norcross GA, USA). Catecholamine concentrations
(adrenalin and noradrenalin) were determined using high performance liquid
chromatography with electrochemical detection (ClinRep® Complete Kit for
Catecholamines in Plasma, RECIPE chemicals & Instruments GmbH, Munich,
Germany). Serum insulin concentrations were determined with radioimmunoassay
kits (Human Insulin specific RIA Kit, Millipore Corporation, MA, USA).
Statistical analysis
All data are expressed as means ± SEM. Normal distribution was tested by the
Kolmogorov-Smirnov test. Subjects were excluded from analyses when dialysate
samples of 2 subsequent time points were missing, in order to maintain paired
samples. Dialysate and systemic exercise responses were expressed as the area
under the curve (AUC) and the incremental area under the curve (iAUC),
calculated by the trapezoid method. Cross-sectional analyses (differences between
groups and conditions) for the microdialysis lipolysis data were analyzed with a
two-way repeated-measures ANOVA. In case of significance, post-hoc analyses
with Bonferroni correction were applied to identify significant within-group effects.
Differences in plasma concentrations and substrate metabolism between groups
were tested with a one-way ANOVA and differences within groups were analyzed
by means of paired t-test. Intervention effects in the obese groups were analyzed
with a two-way repeated-measures ANOVA (with pre- and post-intervention as
conditions), with Bonferroni post-hoc correction to detect within-group effects.
206
                                                                             Chapter 7
Three subjects dropped out of the exercise intervention, due to medical (n=1) or
motivational reasons (n=2) and were therefore excluded from the intervention (pre
vs. post) analyses. SPSS 21 for Macintosh OS X was used to perform all
calculations (IBM Corporation, Armonk, NY, USA). The level of statistical
significance was set at p<0.05 (2-tailed), while p<0.10 was considered a tendency.
RESULTS
BASELINE
Anthropometric and clinical characteristics
Subjects’ characteristics are presented in Table 1. By design, there was a
significant difference between the lean and the obese IS and/or obese IR group
with respect to body weight, BMI, WH-ratio, whole-body fat percentage, android
(i.e. trunk region) and gynoid (including hip and leg regions) fat mass (all p<0.05).
Furthermore, HOMA-IR and fasting serum insulin concentrations were significantly
higher in obese IR compared to the lean and obese IS individuals (p<0.001 for both
parameters in both groups). Obese IS and obese IR individuals only differed in
android fat mass, which was higher in the obese IR group (p=0.017) (Table 1).
With respect to physical fitness, VO2peak/FFM and W peak/FFM were significantly lower in
the obese IS and obese IR group as compared to the lean group (p<0.01 and
p<0.001, respectively) (Table 1), while maximal heart rate and maximal respiratory
quotient (RQ) were comparable between groups.
                                                                                    207
Chapter 7
   Table 1. Characteristics of obese insulin sensitive, obese insulin resistant individuals and healthy
                                              lean controls
                                                        Obese insulin        Obese insulin
                                         Lean
                                                          sensitive             resistant
                                       (n=10)              (n=10)                (n=10)          P ANOVA
           Age, years                   45 ± 2              47 ± 2                43 ± 1         0.527
 Cardiometabolic risk
 Fasting plasma glucose
                                       5.5 ± 0.0          5.2 ± 0.1           5.8 ± 0.2 †        0.050
 (mmol/L)
 Fasting serum insulin (mU/L)          7.3 ± 0.6          9.2 ± 0.6         19.6 ± 1.6 *** †    <0.001
 HOMA-IR                               1.8 ± 0.1          2.1 ± 0.1         5.0 ± 0.4 *** †     <0.001
 Systolic BP (mmHg)                     122 ± 2            135 ± 6              143 ± 6         0.066
 Diastolic BP (mmHg)                    72 ± 1             81 ± 4               86 ± 5          0.108
 Body composition
 Body weight (kg)                     79.9 ± 2.9       101.5 ± 3.2 **       109.6 ± 4.7 ***     <0.001
 Body mass index (kg/m²)              23.7 ± 0.4       32.6 ± 0.4 ***        33.9 ± 0.7 ***     <0.001
 Waist-to-hip ratio                  1.00 ± 0.00       1.04 ± 0.01 *         1.05 ± 0.01 *      0.010
 Fat mass (kg)                        16.4 ± 1.1       30.0 ± 1.7 ***        34.2 ± 1.8 ***     <0.001
 Fat percentage (%)                   22.0 ± 0.8       31.4 ± 0.9 ***        33.2 ± 1.1 ***     <0.001
 Fat free mass (kg)                   57.4 ± 1.8        65.0 ± 1.5           68.4 ± 2.8 **      0.004
 Exercise capacity
 VO2 peak (ml*min-1*kg-1 (FFM))         62 ± 3            48 ± 2 **            48 ± 1 **        0.002
 Wmax (Watt*kg-1 (FFM))                4.9 ± 0.1        3.7 ± 0.2 ***        3.4 ± 0.1 ***      <0.001
Data are mean ± SE. * Significantly different from lean group p < 0.05; ** p < 0.01; *** p < 0.001.
† Significantly different from obese insulin sensitive group (p < 0.05). FFM: fat free mass; HR: heart
rate; RER: respiratory exchange ratio; VO2 peak: maximum oxygen uptake; Wmax: maximum power
output
208
                                                                                        Chapter 7
Figure 1. Plasma glycerol, FFA, glucose, lactate, ANP, adrenalin, noradrenalin and
serum insulin concentrations at rest, during exercise and recovery.
Systemic glycerol (A), FFA (B), glucose (C), lactate (D), ANP (E), insulin (F), adrenalin (G) and
noradrenalin (H) responses in lean (white circles), obese insulin sensitive (white squares) and obese
insulin resistant (black triangles) individuals. Data are presented as mean ± SEM. P ANOVA values
represent differences in exercise-induced systemic responses between groups.
                                                                                                209
Chapter 7
Microdialysis
210
                                                                                             Chapter 7
                                                                                                     211
Chapter 7
Mean changes in subcutaneous adipose tissue extracellular glycerol concentration during 1 h of low-
intense exercise (40% VO2peak). Extracellular glycerol concentrations are determined in control probe
(white bars) and the probe perfused with phentolamine and propranolol (black bars). Changes were
calculated by the difference between the mean glycerol concentrations during exercise and the baseline
concentration (pgroup=0.009, ptreatment=0.069, ptreatment*group=0.035). Data are presented as mean ± SEM.
* Significantly (p<0.05) different compared to the control probe from the lean group; N.S.: not significant.
212
                                                                             Chapter 7
                                                                                    213
Chapter 7
214
                                                                                            Chapter 7
Systemic glycerol (A-B), FFA (C-D), glucose (E-F), lactate (G-H), insulin (I-J), ANP (K-L), adrenalin (M-
N) and noradrenalin (O-P) of obese insulin sensitive and obese insulin resistant individuals at baseline
(white circles) and after 12 weeks of exercise training intervention (black squares). Data are presented
as mean ± SEM.
                                                                                                    215
Chapter 7
Subcutaneous adipose tissue ethanol ratio (A) and extracellular glycerol concentration (B) in the obese
insulin resistant individuals at rest, during exercise and recovery after 12 weeks of exercise training in
control probe (white circles) and the probe perfused with phentolamine and propranolol (black squares).
Data are presented as mean ± SEM.
216
           Table 2. Body composition and exercise capacity before and after 12 weeks of supervised endurance and
                                                            resistance training
Body weight (kg) 104.5 ± 3.2 102.8 ± 2.6 110.6 ± 5.1 108.3 ± 5.0 0.002 0.360 0.585
Body mass index (kg/m²) 32.7 ± 0.5 32.2 ± 0.3 34.1 ± 0.8 33.3 ± 0.8 0.002 0.225 0.550
Waist-to-hip ratio 108.8 ± 1.6 108.8 ± 1.5 117.6 ± 3.3 113.5 ± 1.3 0.039 0.004 0.076
Fat mass (kg) 31.22 ± 1.91 29.11 ± 1.69 35.00 ± 1.92 33.07 ± 2.02 <0.001 0.168 0.833
Fat percentage (%) 31.6 ± 1.1 30.0 ± 1.0 33.7 ± 1.1 32.8 ± 1.2 <0.001 0.150 0.267
Fat free mass (kg) 66.8 ± 1.4 67.2 ± 1.3 68.5 ± 3.1 67.5 ± 3.3 0.377 0.779 0.037
Exercise capacity
Wmax (Watt*kg-1 (FFM)) 3.8 ± 0.2 4.1 ± 0.1 3.4 ± 0.1 4.0 ± 0.1 <0.001 0.382 0.069
      Data are mean ± SE. FFM: fat free mass; VO2 peak: maximum oxygen uptake; Wmax: maximum power output.
                                                                                                                                          Chapter 7
217
Chapter 7
DISCUSSION
The present study is the first to investigate (non-)adrenergically-mediated lipolysis
during low-intensity endurance-type exercise in abdominal subcutaneous adipose
tissue (SCAT) in middle-aged obese IS and obese IR men as compared to age-
matched lean IS men. Furthermore, we examined whether a 12-week exercise
intervention altered resting and exercise-induced (non-)adrenergically-mediated
SCAT lipolysis in obese IR individuals. Here, we demonstrated that the exercise-
induced increase in abdominal SCAT extracellular glycerol concentration (reflecting
local lipolysis) was more pronounced in obese as compared to lean individuals,
which may at least partly be explained by the higher adipose tissue blood flow
(ATBF) in lean individuals. Exercise-induced SCAT lipolysis was substantially
reduced (~40%) following local combined α-/β-adrenergic blockade in obese IS
individuals, but not in obese IR or lean IS individuals. Finally, the exercise training
intervention improved body composition, physical fitness and exercise-induced
systemic responses in both obese groups, and insulin sensitivity in the obese IR
group. However, this was not accompanied by changes in adrenergically- and non-
adrenergically-mediated lipolysis in the SCAT of obese IR individuals. Collectively,
our findings indicate that exercise-induced lipolysis is predominantly mediated by
non-adrenergic factors, most likely mediated by natriuretic peptides (NP) in middle-
aged lean IS, obese IS and obese IR individuals.
The present study showed a more pronounced increase in exercise-mediated
SCAT extracellular glycerol concentration in obese IS as compared to lean
individuals. Although the lipolytic response in abdominal SCAT is often blunted in
human obesity [9], the higher exercise-induced extracellular glycerol levels in both
obese groups is likely explained by the substantially lower ATBF in the obese state,
which contributes to higher extracellular glycerol concentrations due to a lower
removal of glycerol from the AT [16, 44]. Local α-/β-adrenergic blockade
substantially reduced basal ATBF in lean but not in the obese individuals. This
might suggest that adrenergic sensitivity of ATBF is reduced in the obese state,
which is in line with previous findings [39, 45]. Thus, these differences in ATBF
between lean and obese individuals should be taken into account when interpreting
local SCAT lipolysis.
Interestingly, combined α-/β-adrenergic receptor blockade reduced exercise-
induced SCAT lipolysis in obese IS men but not in lean and obese IR individuals. In
obese IR individuals, the lipolytic activity of the β-adrenergic receptors is
attenuated [9, 11, 12], while an increase in anti-lipolytic α2-adrenergic receptors in
SCAT reduce exercise-mediated lipolysis [39]. In addition, obese IR individuals
often display lower plasma catecholamine (adrenaline, noradrenaline) responses to
physical exercise [46], although the latter was not observed in the current study.
Therefore, the reduced exercise-mediated lipolytic response upon combined α-/β-
adrenergic receptor blockade in the obese IS individuals, as opposed to the obese
IR group, might suggest differences in adrenergic receptor expression and
sensitivity. The blunted lipolytic response in obese IR individuals might also be
explained by the significantly higher fasting and exercise induced serum insulin
levels. Since the anti-lipolytic effects of insulin might be normal or only slightly
impaired in obese AT [47-49], the observed hyperinsulinemia in the obese IR group
might have contributed to the attenuated adrenergically-mediated SCAT lipolysis
218
                                                                            Chapter 7
                                                                                   219
Chapter 7
220
                                                                                     Chapter 7
REFERENCES
1.    Goossens GH. The role of adipose tissue dysfunction in the pathogenesis of obesity-
      related insulin resistance. Physiol Behav. 2008;94(2):206-18.
2.    Lafontan M. Adipose tissue and adipocyte dysregulation. Diabetes Metab.
      2014;40(1):16-28.
3.    Stinkens R, Goossens GH, Jocken JW, Blaak EE. Targeting fatty acid metabolism to
      improve glucose metabolism. Obes Rev. 2015;16(9):715-57.
4.    Frayn KN. Adipose tissue as a buffer for daily lipid flux. Diabetologia.
      2002;45(9):1201-10.
5.    Jocken JW, Blaak EE. Catecholamine-induced lipolysis in adipose tissue and skeletal
      muscle in obesity. Physiol Behav. 2008;94(2):219-30.
6.    Fruhbeck G, Mendez-Gimenez L, Fernandez-Formoso JA, Fernandez S, Rodriguez
      A. Regulation of adipocyte lipolysis. Nutr Res Rev. 2014;27(1):63-93.
7.    Hansen D, Meeusen R, Mullens A, Dendale P. Effect of acute endurance and
      resistance exercise on endocrine hormones directly related to lipolysis and skeletal
      muscle protein synthesis in adult individuals with obesity. Sports Med.
      2012;42(5):415-31.
8.    Ryden M, Jocken J, van Harmelen V, Dicker A, Hoffstedt J, Wiren M, et al.
      Comparative studies of the role of hormone-sensitive lipase and adipose triglyceride
      lipase in human fat cell lipolysis. Am J Physiol Endocrinol Metab. 2007;292(6):E1847-
      55.
9.    Jocken JW, Goossens GH, van Hees AM, Frayn KN, van Baak M, Stegen J, et al.
      Effect of beta-adrenergic stimulation on whole-body and abdominal subcutaneous
      adipose tissue lipolysis in lean and obese men. Diabetologia. 2008;51(2):320-7.
10.   Mauriege P, Despres JP, Prud'homme D, Pouliot MC, Marcotte M, Tremblay A, et al.
      Regional variation in adipose tissue lipolysis in lean and obese men. J Lipid Res.
      1991;32(10):1625-33.
11.   Reynisdottir S, Wahrenberg H, Carlstrom K, Rossner S, Arner P. Catecholamine
      resistance in fat cells of women with upper-body obesity due to decreased
      expression of beta 2-adrenoceptors. Diabetologia. 1994;37(4):428-35.
12.   Lafontan M, Berlan M. Fat cell adrenergic receptors and the control of white and
      brown fat cell function. J Lipid Res. 1993;34(7):1057-91.
13.   Arner P, Kriegholm E, Engfeldt P, Bolinder J. Adrenergic regulation of lipolysis in situ
      at rest and during exercise. J Clin Invest. 1990;85(3):893-8.
14.   Hellstrom L, Blaak E, Hagstrom-Toft E. Gender differences in adrenergic regulation
      of lipid mobilization during exercise. Int J Sports Med. 1996;17(6):439-47.
15.   Moro C, Polak J, Hejnova J, Klimcakova E, Crampes F, Stich V, et al. Atrial
      natriuretic peptide stimulates lipid mobilization during repeated bouts of endurance
      exercise. Am J Physiol Endocrinol Metab. 2006;290(5):E864-9.
16.   Moro C, Pillard F, de Glisezinski I, Klimcakova E, Crampes F, Thalamas C, et al.
      Exercise-induced lipid mobilization in subcutaneous adipose tissue is mainly related
      to natriuretic peptides in overweight men. Am J Physiol Endocrinol Metab.
      2008;295(2):E505-13.
17.   Lafontan M. Fat cells: afferent and efferent messages define new approaches to treat
      obesity. Annu Rev Pharmacol Toxicol. 2005;45:119-46.
18.   Liu C, Wu J, Zhu J, Kuei C, Yu J, Shelton J, et al. Lactate inhibits lipolysis in fat cells
      through activation of an orphan G-protein-coupled receptor, GPR81. J Biol Chem.
      2009;284(5):2811-22.
19.   Langin D. Adipose tissue lipolysis revisited (again!): lactate involvement in insulin
      antilipolytic action. Cell Metab. 2010;11(4):242-3.
20.   Moro C. Targeting cardiac natriuretic peptides in the therapy of diabetes and obesity.
      Expert Opin Ther Targets. 2016;20(12):1445-52.
                                                                                            221
Chapter 7
21.   Wang TJ, Larson MG, Levy D, Benjamin EJ, Leip EP, Wilson PW, et al. Impact of
      obesity on plasma natriuretic peptide levels. Circulation. 2004;109(5):594-600.
22.   Das SR, Drazner MH, Dries DL, Vega GL, Stanek HG, Abdullah SM, et al. Impact of
      body mass and body composition on circulating levels of natriuretic peptides: results
      from the Dallas Heart Study. Circulation. 2005;112(14):2163-8.
23.   Khan AM, Cheng S, Magnusson M, Larson MG, Newton-Cheh C, McCabe EL, et al.
      Cardiac natriuretic peptides, obesity, and insulin resistance: evidence from two
      community-based studies. J Clin Endocrinol Metab. 2011;96(10):3242-9.
24.   Magnusson M, Jujic A, Hedblad B, Engstrom G, Persson M, Struck J, et al. Low
      plasma level of atrial natriuretic peptide predicts development of diabetes: the
      prospective Malmo Diet and Cancer study. J Clin Endocrinol Metab. 2012;97(2):638-
      45.
25.   Gruden G, Landi A, Bruno G. Natriuretic peptides, heart, and adipose tissue: new
      findings and future developments for diabetes research. Diabetes Care.
      2014;37(11):2899-908.
26.   Sengenes C, Berlan M, De Glisezinski I, Lafontan M, Galitzky J. Natriuretic peptides:
      a new lipolytic pathway in human adipocytes. FASEB J. 2000;14(10):1345-51.
27.   Sengenes C, Bouloumie A, Hauner H, Berlan M, Busse R, Lafontan M, et al.
      Involvement of a cGMP-dependent pathway in the natriuretic peptide-mediated
      hormone-sensitive lipase phosphorylation in human adipocytes. J Biol Chem.
      2003;278(49):48617-26.
28.   Moro C, Crampes F, Sengenes C, De Glisezinski I, Galitzky J, Thalamas C, et al.
      Atrial natriuretic peptide contributes to physiological control of lipid mobilization in
      humans. FASEB J. 2004;18(7):908-10.
29.   Verboven K, Hansen D, Moro C, Eijnde BO, Hoebers N, Knol J, et al. Attenuated
      atrial natriuretic peptide-mediated lipolysis in subcutaneous adipocytes of obese type
      2 diabetic men. Clin Sci (Lond). 2016;130(13):1105-14.
30.   Ryden M, Backdahl J, Petrus P, Thorell A, Gao H, Coue M, et al. Impaired atrial
      natriuretic peptide-mediated lipolysis in obesity. Int J Obes (Lond). 2016;40(4):714-
      20.
31.   Stich V, de Glisezinski I, Crampes F, Suljkovicova H, Galitzky J, Riviere D, et al.
      Activation of antilipolytic alpha(2)-adrenergic receptors by epinephrine during
      exercise in human adipose tissue. Am J Physiol. 1999;277(4 Pt 2):R1076-83.
32.   Stich V, de Glisezinski I, Galitzky J, Hejnova J, Crampes F, Riviere D, et al.
      Endurance training increases the beta-adrenergic lipolytic response in subcutaneous
      adipose tissue in obese subjects. Int J Obes Relat Metab Disord. 1999;23(4):374-81.
33.   Polak J, Klimcakova E, Moro C, Viguerie N, Berlan M, Hejnova J, et al. Effect of
      aerobic training on plasma levels and subcutaneous abdominal adipose tissue gene
      expression of adiponectin, leptin, interleukin 6, and tumor necrosis factor alpha in
      obese women. Metabolism. 2006;55(10):1375-81.
34.   Moro C, Pillard F, De Glisezinski I, Harant I, Rivi??Re D, Stich V, et al. Training
      Enhances ANP Lipid-Mobilizing Action in Adipose Tissue of Overweight Men.
      Medicine & Science in Sports & Exercise. 2005;37(7):1126-32.
35.   Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC.
      Homeostasis model assessment: insulin resistance and beta-cell function from
      fasting plasma glucose and insulin concentrations in man. Diabetologia.
      1985;28(7):412-9.
36.   Blaak EE, Hul G, Verdich C, Stich V, Martinez A, Petersen M, et al. Fat oxidation
      before and after a high fat load in the obese insulin-resistant state. J Clin Endocrinol
      Metab. 2006;91(4):1462-9.
37.   Wildman RP, Muntner P, Reynolds K, McGinn AP, Rajpathak S, Wylie-Rosett J, et al.
      The obese without cardiometabolic risk factor clustering and the normal weight with
      cardiometabolic risk factor clustering: prevalence and correlates of 2 phenotypes
222
                                                                                  Chapter 7
                                                                                         223
Chapter 7
224
                                                                                            Chapter 7
SUPPLEMENTARY MATERIAL
Indirect calorimetry was performed during resting conditions and during an acute exercise bout of
moderate intensity. Mean O2-consumption and CO2-production over 10 min were used for calculations
of respiratory quotient (A), energy expenditure and substrate oxidation during fasting (B + D) and during
exercise (C + E). Post intervention data are added for both obese groups (A-E). CHO, carbohydrates;
EE, energy expenditure; IR, insulin resistant; IS, insulin sensitive. Data are presented as mean± SEM.
                                                                                                    225
Chapter 7
226
                                                                                                            Supplementary Table S1. Mean plasma and serum concentrations of systemic glycerol, insulin, FFA, glucose, lactate,
                                                                                                            ANP, adrenalin and noradrenalin at rest, during low-intense exercise and recovery in obese insulin sensitive, obese insulin
                                                                                                            resistant and lean individuals
                                                                                                                                                                   Obese insulin                     Obese insulin
                                                                                                                                                  Lean (n= 10)    sensitive (n= 10)   P (vs lean)   resistant (n= 10)   P (vs lean)   P (vs OB IS)   P (ANOVA)
                                                                                                                         GLYCEROL
                                                                                                            Baseline (μmol/l)                      70.5 ± 4.8        81.8 ± 7.5         0.580          77.5 ± 5.5         1.000          1.000         0.416
                                                                                                            Exercise (AUC0-60) (μmol/l*60min)     136.73 ± 12.5     163.2 ± 19.4        0.978        168.48 ± 22.8        0.722          1.000         0.448
                                                                                                            Recovery (AUC75-105) (μmol/l*45min)    107.2 ± 6.5      124.2 ± 10.4        1.000         157.0 ± 28.4        0.183          0.660         0.159
                                                                                                                           INSULIN
                                                                                                            Baseline (μU/ml)                        7.0 ± 0.6         9.4 ± 1.1         0.563          16.9 ± 1.8        <0.001          0.001        <0.001
                                                                                                            Exercise (AUC0-60) (μU/ml*60min)        7.4±0.64          9.3±1.2           0.791          15.0±1.5          <0.001          0.005        <0.001
                                                                                                            Recovery (AUC75-105) (μU/ml*45min)      8.5 ± 1.0        11.7 ± 1.2         0.209         21.43 ± 1.3        <0.001         <0.001        <0.001
                                                                                                                                FFA
                                                                                                            Baseline (μmol/l)                     451.1 ± 30.9      528.4 ± 58.3        0.658         556.7 ± 36.3        0.292          1.000         0.224
                                                                                                            Exercise (AUC0-60) (μmol/l*60min)     466.4 ± 42.6      555.2 ± 56.0        0.628         530.9 ± 46.9        1.000          1.000         0.425
                                                                                                            Recovery (AUC75-105) (μmol/l*45min)    600.7±37.2       777.6 ± 63.8        0.189         864.5±81.9          0.019          1.000         0.020
                                                                                                                          GLUCOSE
                                                                                                            Baseline (mmol/l)                       5.2 ± 0.1         5.0 ± 0.1         0.722          5.1 ± 0.1          1.000          1.000         0.488
                                                                                                            Exercise (AUC0-60) (mmol/l*60min)       5.3 ± 0.1         5.1 ± 0.1         1.000          5.3 ± 0.2          1.000          1.000         0.679
                                                                                                            Recovery (AUC75-105) (mmol/l*45min)     5.3 ± 0.1         5.3 ± 0.2         1.000          5.3 ± 0.1          1.000          1.000         0.975
                                                                                                                          LACTATE
                                                                                                            Baseline (mmol/l)                       0.9 ± 0.1         0.9 ± 0.0         1.000          1.0 ± 0.1          0.628          0.600         0.339
      AUC75-105 (area under the curve during the recovery period (from timepoint 75 until 105min)). Group
      Data are mean ± SEM. AUC0-60 (area under the curve during exercise (from timepoint 0 till 60min)),
                                                                                                                                                                                                                                                                 Chapter 7
227
                                                                                                            Baseline (pg/ml)                      492.6 ± 84.7      437.1 ± 91.4        1.000         430.3 ± 42.2        1.000          1.000         0.816
                                                                                                            Exercise (AUC0-60) (pg/ml*60min)      1107.1 ± 80.8    1095.6 ± 83.1        1.000       1007.3 ± 110.6        1.000          1.000         0.708
                                                                                                            Recovery (AUC75-105) (pg/ml*45min)    610.1 ± 70.9      539.8 ± 48.3        1.000         580.2 ± 56.1        1.000          1.000         0.725
            Supplementary Table S3. Interstitial glucose and lactate concentrations in the control probe and the
            probe containing phentolamine and propranolol in the obese insulin sensitive, obese insulin resistant
            and lean individuals.
                                                          Obese               Obese
                                          Lean                                                             ANOVA
                                                     insulin sensitive   insulin resistant
                                         (n=10)                                              P Group   P Treatment P Treatment*Group
                                                         (n= 10)             (n= 10)
                 GLUCOSE
            Baseline (mmol/l)                                                                0.081      0.622            0.877
                            Control    3.62 ± 0.31     2.88 ± 0.39         2.56 ± 0.48
                         α/β blocker   3.72 ± 0.45     2.86 ± 0.31         2.82 ± 0.23
            Exercise (AUC0-60)                                                               0.811      0.194            0.655
                            Control    4.30 ± 0.36     4.36 ± 0.73         4.60 ± 1.06
                         α/β blocker   4.24 ± 0.55     3.43 ± 0.38         3.93 ± 0.47
            Recovery   (AUC75-120)                                                           0.925      0.624            0.482
                            Control    3.38 ± 0.30     3.45 ± 0.50         3.65 ± 0.92
                         α/β blocker   3.68 ± 0.41     3.32 ± 0.37         3.02 ± 0.16
                 LACTATE
            Baseline (mmol/l)                                                                0.121      0.091            0.709
                            Control    1.79 ± 0.35     0.98 ± 0.16         1.80 ± 0.60
                         α/β blocker   1.37 ± 0.37     0.78 ± 0.13         1.08 ± 0.14
            Exercise   (AUC0-60)                                                             0.354      0.078            0.449
                            Control    2.63 ± 0.58     1.79 ± 0.41         3.16 ± 0.95
                         α/β blocker   2.36 ± 0.62     1.46 ± 0.34         1.94 ± 0.32
            Recovery (AUC75-120)                                                             0.349      0.234            0.558
Chapter 7
                                                                                                                                       228
            Data are mean ± SEM. Control (probe containing Ringer), α/β blocker (probe containing Phentolamine
            + Propranolol). P-values were calculated via a 2-way repeated ANOVA.
      Supplemental Table S2. Interstitial glycerol concentrations and ethanol ratio in the control probe and the probe containing
      phentolamine and propranolol in obese insulin sensitive, obese insulin resistant and lean individuals
                                                                                                                                          ANOVA
                                                                          Obese                      Obese
                                           Lean                      insulin sensitive          insulin resistant
                                                                                                                         P              P                  P
                                          (n=10)                         (n= 10)                    (n= 10)
                                                                                                                       Group        Treatment       Treatment*Group
GLYCEROL
ETHANOL RATIO
      Data are expressed as mean ± SEM. Control (probe containing Ringer), α/β blocker (probe containing Phentolamine + Propranolol). P-values were calculated via a
      2-way repeated ANOVA.
                                                                                                                                                                       Chapter 7
229
Chapter 7
230
                                                                                                            Supplementary Table S4. Mean plasma and serum concentrations of circulating glycerol, insulin, FFA, glucose, lactate, ANP, adrenalin and noradrenalin
                                                                                                             at rest, during exercise and recovery, before and after 12 weeks of exercise training in the obese insulin sensitive and obese insulin resistant subjectss
                                                                                                                  GLYCEROL
                                                                                                            Baseline (μmol/l)                 74.0 ± 5.0            64.2 ± 4.8             79.9 ± 5.5            74.8 ± 6.4         0.100       0.235         0.593
                                                                                                            Exercise (AUC0-60)               157.3 ± 22.7          150.3 ± 12.7          175.7 ± 24.1          144.0 ± 23.6         0.026       0.730         0.500
                                                                                                            (μmol/l*60min)
                                                                                                            Recovery (AUC75-105)             118.9 ± 12.7           124.4 ± 6.6          164.3 ± 30.7          122.1 ± 19.3         0.316       0.405         0.198
                                                                                                            (μmol/l*45min)
                                                                                                                     INSULIN
                                                                                                            Baseline (μU/ml)                   9.5 ± 1.4             9.4 ± 1.3             16.9 ± 2.0            14.3 ± 1.3         0.196       0.008         0.263
                                                                                                            Exercise (AUC0-60)                 9.2 ± 1.5             9.1 ± 1.2             14.8 ± 1.7            13.4 ± 1.3         0.313       0.021         0.393
                                                                                                            (μU/ml*60min)
                                                                                                            Recovery (AUC75-105)              11.6 ± 1.6            13.8 ± 1.5             21.6 ± 1.5            19.4 ± 2.5         0.981       0.003         0.206
                                                                                                            (μU/ml*45min)
                                                                                                                      FFA
                                                                                                            Baseline (μmol/l)                506.2 ± 48.5          382.2 ± 30.2          571.3 ± 37.1          476.5 ± 41.1         0.016       0.060         0.723
                                                                                                            Exercise (AUC0-60)               533.0 ± 55.4          436.7 ± 42.6          540.5 ± 51.3          439.0 ± 47.7         0.040       0.930         0.954
                                                                                                            (μmol/l*60min)
                                                                                                            Recovery (AUC75-105)             759.0 ± 70.1          698.0 ± 38.5          892.5 ± 86.1          726.4 ± 76.1         0.034       0.401         0.296
                                                                                                            (μmol/l*45min)
                                                                                                                    GLUCOSE
                                                                                                            Baseline (mmol/l)                  5.0 ± 0.1             5.3 ± 0.2             5.1 ± 0.1             5.1 ± 0.1          0.030       0.671         0.021
                                                                                                            Exercise (AUC0-60)                 5.2 ± 0.1             5.3 ± 0.1             5.3 ± 0.2             5.1 ± 0.1          0.973       0.790         0.202
                                                                                                            (mmol/l*60min)
                                                                                                            Recovery (AUC75-105)               5.4 ± 0.3             5.4 ± 0.1             5.3 ± 0.2             5.1 ± 0.1          0.564       0.437         0.564
                                                                                                            (mmol/l*45min)
                                                                                                                   LACTATE
                                                                                                            Baseline (mmol/l)                  0.9 ± 0.0             0.8 ± 0.1             1.0 ± 0.1             1.0 ± 0.1          0.335       0.322         0.824
                                                                                                            Exercise (AUC0-60)                 1.1 ± 0.1             0.9 ± 0.1             1.4 ± 0.1             1.0 ± 0.1          0.016       0.127         0.609
      AUC75-105 (area under the curve during the recovery period (from timepoint 75 until 105min)). Group
                                                                                                                                                                                                                                                                          Chapter 7
231
      Data are mean ± SEM. AUC0-60 (area under the curve during exercise (from timepoint 0 till 60min)),
                                                                                                            Baseline (pg/ml)                 452.1 ± 118.1         395.6 ± 28.2          442.5 ± 45.8          400.8 ± 41.7         0.468       0.973         0.912
                                                                                                            Exercise (AUC0-60)               1078.5 ± 92.2         1002.6 ± 93.3        1043.1 ± 118.7        1045.6 ± 106.2        0.875       0.822         0.534
                                                                                                            (pg/ml*60min)
                                                                                                            Recovery (AUC75-105)             519.6 ± 50.7          552.8 ± 68.1          584.7 ± 63.4          608.1 ± 56.8         0.613       0.377         0.930
                                                                                                            (pg/ml*45min)
CHAPTER 8
GENERAL DISCUSSION
Chapter 8
The prevalence of obesity has increased enormously over the last decades and is
associated with an increased risk for metabolic impairments and chronic diseases
such as insulin resistance [1], type 2 diabetes [2] and cardiovascular diseases [3].
Obesity results from a chronic positive energy balance that leads to an increased
amount of adipose tissue mass. However, adipose tissue mass per se does not
seem to be the most important contributor to the development of obesity-related
disorders. Body fat distribution and adipose tissue dysfunction play a more
prominent role in the determination of cardiometabolic health [4, 5]. Therapies to
reduce obesity and related comorbidities can include dietary manipulation (as
extensively discussed in Chapter 2), physical activity strategies and a
pharmacological approach.
There is evidence to suggest that both the renin-angiotensin system (RAS) and the
natriuretic peptide (NP) system can influence cardiometabolic risk. Indeed, several
RAS components, which are also present in different key metabolic organs such as
the adipose tissue, skeletal muscle and the liver, are increased in obesity and
insulin resistance [6-9]. Also, reduced circulating NP concentrations have been
observed in obesity and type 2 diabetes [10]. An increased RAS activity [6] and
reduced NP concentrations [10] have detrimental metabolic effects and may
increase disease progression [11-15]. Recently, a novel dual acting drug,
sacubitril/valsartan, has been developed that facilitates the beneficial effects of the
NP system, while inhibiting the detrimental effects of the RAS [16]. This
combination therapy may have beneficial synergistic effects [17, 18] with respect to
disease progression. Indeed, sacubitril/valsartan has been shown to be superior in
reducing the risks of cardiovascular death or hospitalization for heart failure as
compared to monotherapy with enalapril [19]. Due to the common
pathophysiological impairments in individuals with cardiovascular and several
metabolic diseases (e.g. reduced oxidative capacity, altered lipolysis, increased
inflammation, insulin resistance), treatment with sacubitril/valsartan could induce
clinical benefits in both patients with cardiovascular and/or metabolic diseases.
However, it remains to be established whether sacubitril/valsartan may also have
(superior) beneficial metabolic effects as compared to other blood pressure
lowering agents.
Beside a pharmacological approach, changes in lifestyle are effective in preventing
the development of type 2 diabetes and related cardiometabolic complications [20,
21]. Increasing physical activity levels lead to increased energy expenditure,
improved physical fitness and contribute to an improved metabolic health [22, 23],
which could be attributable to changes in adipose tissue metabolism. Although
several rodent studies suggest that exercise training may improve adipose tissue
metabolism and function [24], human data are limited and need further
investigation [24-26].
Therefore, in the present thesis, the metabolic effects of pharmacological treatment
with sacubitril/valsartan, as well as exercise-training induced effects on adipose
tissue metabolism and the metabolic profile were investigated. In this chapter, our
findings will be discussed and put into a broader perspective.
234
                                                                           Chapter 8
                                                                                  235
Chapter 8
236
                                                                            Chapter 8
                                                                                   237
Chapter 8
[57, 58]. However, our data suggest that peripheral insulin sensitivity can improve
without significant changes in fat-free mass.
Although exercise training significantly increased peripheral insulin sensitivity it did
not induce changes in adipose tissue and hepatic insulin sensitivity, as described
in Chapter 6. These findings are in contrast with previous rodent [59, 60] and
human [61] studies. The unaltered adipose tissue and hepatic insulin sensitivity
may be explained by the rather minor changes in fat mass loss. Nevertheless, our
data showed that exercise training beneficially improved physical fitness, body
composition and peripheral insulin sensitivity, irrespective of the baseline metabolic
status.
238
                                                                           Chapter 8
                                                                                  239
Chapter 8
240
                                                                          Chapter 8
~65% of the exercise-mediated lipolysis in young healthy lean and overweight men
[38, 39]. We found that SCAT lipolysis in obese insulin resistant men, compared to
the obese insulin sensitive group, was not affected by local α-/β-adrenergic
blockade, which may point toward an interaction between SCAT adrenergically-
mediated lipolysis and whole-body insulin resistance. These findings support a
catecholamine-resistant phenotype of the SCAT during exercise, especially in the
obese insulin resistant state.
The 12-week combined endurance and resistance exercise training intervention did
not improve exercise-induced abdominal SCAT lipolysis. Furthermore, the
efficiency of local α-/β-adrenergic blockade was not affected and these data
combined suggest that the lipolytic disturbances in SCAT of obese insulin resistant
individuals remain unaffected after 12 weeks of exercise training. This observation
is in contrast with previous studies that showed beneficial changes in SCAT insulin
sensitivity [61], SCAT adrenergic sensitivity [61, 92, 113] as well as ANP-mediated
lipolysis [114] after exercise training in obese subjects.
Taken together, the results described in Chapter 7 show a major role for non-
adrenergically-mediated lipolysis in SCAT during low-intensity exercise, likely
involving ANP-mediated lipolysis. Furthermore, our data suggest that even after a
substantial improvement in body composition, physical fitness and insulin
sensitivity, lipolytic disturbances remain unaffected in SCAT of obese insulin
resistant individuals after a 12-week exercise training intervention.
                                                                                 241
Chapter 8
The outcomes of this thesis provide evidence for an improved metabolic health
after sacubitril/valsartan treatment and exercise training interventions, but several
questions and issues should be addressed in future research:
242
                                                                          Chapter 8
                                                                                243
Chapter 8
REFERENCES
1.    Despres JP, Lemieux I. Abdominal obesity and metabolic syndrome. Nature.
      2006;444(7121):881-7.
2.    Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance
      and type 2 diabetes. Nature. 2006;444(7121):840-6.
3.    Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with
      cardiovascular disease. Nature. 2006;444(7121):875-80.
4.    Goossens GH. The role of adipose tissue dysfunction in the pathogenesis of obesity-
      related insulin resistance. Physiol Behav. 2008;94(2):206-18.
5.    Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell.
      2014;156(1-2):20-44.
6.    Goossens GH. The renin-angiotensin system in the pathophysiology of type 2
      diabetes. Obes Facts. 2012;5(4):611-24.
7.    Frigolet ME, Torres N, Tovar AR. The renin-angiotensin system in adipose tissue and
      its metabolic consequences during obesity. J Nutr Biochem. 2013;24(12):2003-15.
8.    Ramalingam L, Menikdiwela K, LeMieux M, Dufour JM, Kaur G, Kalupahana N, et al.
      The renin angiotensin system, oxidative stress and mitochondrial function in obesity
      and insulin resistance. Biochim Biophys Acta. 2016.
9.    Borghi F, Seva-Pessoa B, Grassi-Kassisse DM. The adipose tissue and the
      involvement of the renin-angiotensin-aldosterone system in cardiometabolic
      syndrome. Cell Tissue Res. 2016;366(3):543-8.
10.   Moro C. Targeting cardiac natriuretic peptides in the therapy of diabetes and obesity.
      Expert Opin Ther Targets. 2016;20(12):1445-52.
11.   Goossens GH, Blaak EE, van Baak MA. Possible involvement of the adipose tissue
      renin-angiotensin system in the pathophysiology of obesity and obesity-related
      disorders. Obes Rev. 2003;4(1):43-55.
12.   Unger T. The role of the renin-angiotensin system in the development of
      cardiovascular disease. Am J Cardiol. 2002;89(2A):3A-9A; discussion 10A.
13.   Wang TJ, Larson MG, Levy D, Benjamin EJ, Leip EP, Wilson PW, et al. Impact of
      obesity on plasma natriuretic peptide levels. Circulation. 2004;109(5):594-600.
14.   Das SR, Drazner MH, Dries DL, Vega GL, Stanek HG, Abdullah SM, et al. Impact of
      body mass and body composition on circulating levels of natriuretic peptides: results
      from the Dallas Heart Study. Circulation. 2005;112(14):2163-8.
15.   Khan AM, Cheng S, Magnusson M, Larson MG, Newton-Cheh C, McCabe EL, et al.
      Cardiac natriuretic peptides, obesity, and insulin resistance: evidence from two
      community-based studies. J Clin Endocrinol Metab. 2011;96(10):3242-9.
16.   Langenickel T.H. DWP. Angiotensin receptor-neprilysin inhibition with LCZ696: a
      novel approach for the treatment of heart failure. Drug Discovery Today: Therapeutic
      Strategies. 2012;9(4):e131–e9.
17.   Tschop MH, Finan B, Clemmensen C, Gelfanov V, Perez-Tilve D, Muller TD, et al.
      Unimolecular Polypharmacy for Treatment of Diabetes and Obesity. Cell Metab.
      2016;24(1):51-62.
18.   Levin PA. Practical combination therapy based on pathophysiology of type 2
      diabetes. Diabetes Metab Syndr Obes. 2016;9:355-69.
19.   McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, et al. Dual
      angiotensin receptor and neprilysin inhibition as an alternative to angiotensin-
      converting enzyme inhibition in patients with chronic systolic heart failure: rationale
      for and design of the Prospective comparison of ARNI with ACEI to Determine Impact
      on Global Mortality and morbidity in Heart Failure trial (PARADIGM-HF). Eur J Heart
      Fail. 2013;15(9):1062-73.
244
                                                                                   Chapter 8
                                                                                          245
Chapter 8
246
                                                                                   Chapter 8
                                                                                          247
Chapter 8
248
                                                                                   Chapter 8
88.    Larsen S, Danielsen JH, Sondergard SD, Sogaard D, Vigelsoe A, Dybboe R, et al.
       The effect of high-intensity training on mitochondrial fat oxidation in skeletal muscle
       and subcutaneous adipose tissue. Scand J Med Sci Sports. 2015;25(1):e59-69.
89.    Ruschke K, Fishbein L, Dietrich A, Kloting N, Tonjes A, Oberbach A, et al. Gene
       expression of PPARgamma and PGC-1alpha in human omental and subcutaneous
       adipose tissues is related to insulin resistance markers and mediates beneficial
       effects of physical training. Eur J Endocrinol. 2010;162(3):515-23.
90.    Khadir A, Tiss A, Abubaker J, Abu-Farha M, Al-Khairi I, Cherian P, et al. MAP kinase
       phosphatase DUSP1 is overexpressed in obese humans and modulated by physical
       exercise. Am J Physiol Endocrinol Metab. 2015;308(1):E71-83.
91.    Morigny P, Houssier M, Mouisel E, Langin D. Adipocyte lipolysis and insulin
       resistance. Biochimie. 2016;125:259-66.
92.    De Glisezinski I, Crampes F, Harant I, Berlan M, Hejnova J, Langin D, et al.
       Endurance training changes in lipolytic responsiveness of obese adipose tissue. Am
       J Physiol. 1998;275(6 Pt 1):E951-6.
93.    Reynisdottir S, Langin D, Carlstrom K, Holm C, Rossner S, Arner P. Effects of weight
       reduction on the regulation of lipolysis in adipocytes of women with upper-body
       obesity. Clin Sci (Lond). 1995;89(4):421-9.
94.    Ryden M, Jocken J, van Harmelen V, Dicker A, Hoffstedt J, Wiren M, et al.
       Comparative studies of the role of hormone-sensitive lipase and adipose triglyceride
       lipase in human fat cell lipolysis. Am J Physiol Endocrinol Metab. 2007;292(6):E1847-
       55.
95.    Jocken JW, Goossens GH, van Hees AM, Frayn KN, van Baak M, Stegen J, et al.
       Effect of beta-adrenergic stimulation on whole-body and abdominal subcutaneous
       adipose tissue lipolysis in lean and obese men. Diabetologia. 2008;51(2):320-7.
96.    Lafontan M, Berlan M. Fat cell adrenergic receptors and the control of white and
       brown fat cell function. J Lipid Res. 1993;34(7):1057-91.
97.    Follenius M, Brandenberger G. Increase in atrial natriuretic peptide in response to
       physical exercise. Eur J Appl Physiol Occup Physiol. 1988;57(2):159-62.
98.    Verboven K, Hansen D, Moro C, Eijnde BO, Hoebers N, Knol J, et al. Attenuated
       atrial natriuretic peptide-mediated lipolysis in subcutaneous adipocytes of obese type
       2 diabetic men. Clin Sci (Lond). 2016;130(13):1105-14.
99.    Ryden M, Backdahl J, Petrus P, Thorell A, Gao H, Coue M, et al. Impaired atrial
       natriuretic peptide-mediated lipolysis in obesity. Int J Obes (Lond). 2016;40(4):714-
       20.
100.   Stich V, de Glisezinski I, Crampes F, Suljkovicova H, Galitzky J, Riviere D, et al.
       Activation of antilipolytic alpha(2)-adrenergic receptors by epinephrine during
       exercise in human adipose tissue. Am J Physiol. 1999;277(4 Pt 2):R1076-83.
101.   Stich V, de Glisezinski I, Galitzky J, Hejnova J, Crampes F, Riviere D, et al.
       Endurance training increases the beta-adrenergic lipolytic response in subcutaneous
       adipose tissue in obese subjects. Int J Obes Relat Metab Disord. 1999;23(4):374-81.
102.   Moro C, Crampes F, Sengenes C, De Glisezinski I, Galitzky J, Thalamas C, et al.
       Atrial natriuretic peptide contributes to physiological control of lipid mobilization in
       humans. FASEB J. 2004;18(7):908-10.
103.   Moro C, Pillard F, De Glisezinski I, Harant I, Rivi??Re D, Stich V, et al. Training
       Enhances ANP Lipid-Mobilizing Action in Adipose Tissue of Overweight Men.
       Medicine & Science in Sports & Exercise. 2005;37(7):1126-32.
104.   Bulow J. Human adipose tissue blood flow during prolonged exercise, III. Effect of
       beta-adrenergic blockade, nicotinic acid and glucose infusion. Scand J Clin Lab
       Invest. 1981;41(4):415-24.
105.   Stich V, De Glisezinski I, Crampes F, Hejnova J, Cottet-Emard JM, Galitzky J, et al.
       Activation of alpha(2)-adrenergic receptors impairs exercise-induced lipolysis in
                                                                                          249
Chapter 8
250
                                                                                 Chapter 8
                                                                                       251
CHAPTER 9
ADDENDA
Chapter 9
SUMMARY
254
                                                                            Chapter 9
Obesity is associated with an increased risk for metabolic impairments and chronic
diseases, including insulin resistance, type 2 diabetes and cardiovascular
diseases. Strategies to reduce body weight and obesity-related comorbidities
include dietary (as discussed in Chapter 2), pharmacological and physical activity
interventions. This thesis describes the effects of a pharmacological intervention as
well as physical exercise interventions to improve metabolic health in obese
individuals, with a focus on adipose tissue metabolism .
An increased renin-angiotensin system activity and a lower activity of the natriuretic
peptide system have been linked to the development of type 2 diabetes and
cardiovascular disease. Combination therapy with sacubitril/valsartan, a combined
angiotensin receptor blocker (ARB) and neprilysin (NEP) inhibitor, facilitates the
beneficial effects of the natriuretic peptide system, while inhibiting the detrimental
effects of the renin-angiotensin system.
To obtain more detailed insight into possible mechanisms underlying the findings
described in Chapters 3 and 4, we assessed the effects of sacubitril/valsartan on
abdominal subcutaneous adipose tissue gene expression patterns using
microarray analysis and determined adipose tissue protein expression profiles in
Chapter 5. We showed no significant changes in expression of genes and proteins
of factors involved in lipolysis, natriuretic peptide signalling and mitochondrial
oxidative metabolism.
Collectively, these data indicate that alterations in abdominal subcutaneous
adipose tissue lipolysis, whole-body lipolysis or whole-body substrate oxidation at
rest and during exercise, do not seem to contribute to the sacubitril/valsartan-
                                                                                   255
Chapter 9
Also, since atrial natriuretic peptide (ANP) increases during exercise and plays an
important role in adipose tissue lipolysis, we investigated abdominal subcutaneous
adipose tissue (non-)adrenergically-mediated lipolysis before, during and after a
single bout of endurance exercise and after 12-weeks of exercise training in
metabolically healthy and metabolically compromised individuals in Chapter 7.
Therefore, we investigated the effect of local combined α- and β-adrenoceptor
blockade on local subcutaneous adipose tissue lipolysis at rest, during low-intensity
endurance-type exercise and during recovery from exercise in sedentary, middle-
aged obese insulin sensitive, obese insulin resistant and age-matched lean insulin
sensitive men. In addition, we investigated whether a 12-week supervised,
progressive, combined endurance and resistance exercise training improved the
metabolic profile in obese men and (non-)adrenergically-mediated abdominal
subcutaneous adipose tissue lipolysis in obese insulin resistant individuals. We
demonstrated a major contribution of non-adrenergically-mediated lipolysis during
exercise in all groups. Furthermore, we showed that the exercise training
intervention improved body composition, physical fitness and exercise-induced
changes in circulating free fatty acids, lactate and adrenalin concentrations in both
obese groups and insulin sensitivity in the obese insulin resistant group. However,
256
                                                                         Chapter 9
                                                                               257
Chapter 9
VALORIZATION
258
                                                                             Chapter 9
SOCIAL RELEVANCE
The worldwide prevalence of obesity has increased enormously over the last
decades and numbers are still increasing every year. According to the World
Health Organization, 13% of the world’s adult population (11% of men and 15% of
women) was obese in 2014, while in the same year, obesity affected around 18,5%
of men and 19% of woman in The Netherlands [1]. If post-2000 trends continue,
this global prevalence of obesity is suggested to reach 18% in men and 21% in
women by 2025. Obesity is associated with an increased risk of developing chronic
diseases, including insulin resistance [2], type 2 diabetes [3], cardiovascular
diseases [4] and certain types of cancer [5, 6]. To reduce these obesity related
comorbidities, nowadays, millions of people are in need of medication, such as
glucose-, cholesterol- and/or blood pressure lowering medication, and surgical
treatments such as gastric bypass or cardiovascular surgery. Since obesity is a
major public health issue and one of the most important risk factors for the
development of metabolic diseases, it is clear that the increasing obesity
prevalence has major socioeconomic consequences [7]. National and international
guidelines recommend changes in modifiable lifestyle characteristics, such as diet
and physical activity for both prevention and management of metabolic diseases
[8]. While weight loss has been shown to be effective in reducing disease risk,
implementation of the recommended lifestyle in the long-term is often hard to
maintain by the majority of people. Therefore, to reduce the incidence of obesity
and thereby partly improving global health, it is important to obtain better insights in
the development and treatment of obesity and related metabolic diseases and to
implement new treatment strategies. The results described in this thesis contribute
to a better understanding of the role of adipose tissue metabolism in
cardiometabolic health and obesity, and provide leads for possible treatment
strategies to reduce or prevent obesity and cardiometabolic complications.
TARGET GROUPS
Scientific community
The results described in this thesis have and will become available to the scientific
community via publication of scientific articles in international peer-reviewed
journals. Additionally, results have been presented at (inter)national conferences to
scientists as well as physicians, healthcare professionals and dieticians, working in
the fields of obesity, diabetes and metabolism.
Industry
A part of this thesis was accomplished by the close collaboration between
academia and industry and research outcomes are of valuable information to the
academic community and both the nutritional and the pharmaceutical industry. The
industrial partners can translate the research outcomes to develop improved or
novel treatment strategies or products that help to prevent or reduce the
prevalence of obesity and obesity-related complications. More specific, the
nutritional industry can translate the results from chapter 2 in defining new
                                                                                    259
Chapter 9
nutritional targets. The pharmacological industry can use the results from this
thesis to develop and/or implement new or improved pharmacological therapies or
to expand the rationale for prescribing certain cardiovascular drugs in metabolically
compromised conditions, since in our studies combination therapy with
sacubitril/valsartan was shown to improve both cardiovascular and metabolic risk
factors.
260
                                                                             Chapter 9
The exercise training intervention studies showed that exercise training beneficially
affects body composition and physical fitness and is effective to improve obesity-
related disturbances like whole-body insulin resistance. Although 12 weeks of
exercise training induced a slight but significant reduction in fat mass, no significant
changes in abdominal subcutaneous adipocyte morphology, adipose tissue
function, and abdominal subcutaneous adipose tissue lipolysis were observed in
obese subjects, irrespective of their baseline metabolic status. Furthermore, we
showed that 12 weeks of exercise training did not improve disturbances in
subcutaneous adipose tissue lipolysis in obese insulin resistant individuals. It
seems that a more pronounced decrease in adipose tissue mass is needed to
induce significant changes in adipose tissue metabolism. Currently, it remains to be
established which exercise training duration and modality is most optimal to induce
beneficial effects in abdominal subcutaneous adipose tissue.
The innovative aspect of the exercise training interventions, was the investigation
of exercise training-induced effects on abdominal subcutaneous adipose tissue,
since these studies have mainly been performed in rodents whereas human
studies are scarce. Our results contribute to the knowledge and provide better
insight in exercise training-mediated metabolic changes in abdominal
subcutaneous adipose tissue.
Furthermore, metabolic phenotyping at baseline makes it possible to stratify
subjects into different subgroups and may improve the effectiveness of a particular
intervention in a specific subgroup of the population [12]. However, in this thesis we
did not find clear evidence that metabolic phenotype at baseline affected exercise
training-induced study outcomes. Therefore, before extrapolating our findings to a
larger population, more research is necessary in larger study populations following
different intervention strategies (e.g. nutritional, pharmacological as well as
                                                                                    261
Chapter 9
262
                                                                                Chapter 9
REFERENCES
1.    World Health Organization. Fact sheet: Obesity and overweight. Updated June 2016.
      2016.
2.    Despres JP, Lemieux I. Abdominal obesity and metabolic syndrome. Nature.
      2006;444(7121):881-7.
3.    Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance
      and type 2 diabetes. Nature. 2006;444(7121):840-6.
4.    Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with
      cardiovascular disease. Nature. 2006;444(7121):875-80.
5.    Bhaskaran K, Douglas I, Forbes H, dos-Santos-Silva I, Leon DA, Smeeth L. Body-
      mass index and risk of 22 specific cancers: a population-based cohort study of 5.24
      million UK adults. Lancet. 2014;384(9945):755-65.
6.    Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K,
      International Agency for Research on Cancer Handbook Working G. Body Fatness
      and Cancer--Viewpoint of the IARC Working Group. N Engl J Med. 2016;375(8):794-
      8.
7.    Di Cesare M, Bentham J, Stevens GA, Zhou B, Danaei G, Lu Y, Bixby H, Cowan MJ,
      Riley LM, Hajifathalian K, Fortunato L, Taddei C, Bennett JE, Ikeda N, Zhu D,
      Zimmermann E, J. ZC. Trends in adult body-mass index in 200 countries from 1975
      to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2
      million participants. Lancet. 2016;387(10026):1377-96.
8.    American Diabetes Association. Standards of Medical Care in Diabetes. Diabetes
      Care. 2016;39(Suppl 1).
9.    US FDA. Entresto Prescribing Information 2015 [updated July 7, 2015. Available
      from:
      http://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/207620Orig1s000Lbl.pdf.
10.   Tschop MH, Finan B, Clemmensen C, Gelfanov V, Perez-Tilve D, Muller TD,
      DiMarchi RD. Unimolecular Polypharmacy for Treatment of Diabetes and Obesity.
      Cell Metab. 2016;24(1):51-62.
11.   Levin PA. Practical combination therapy based on pathophysiology of type 2
      diabetes. Diabetes Metab Syndr Obes. 2016;9:355-69.
12.   Stefan N, Fritsche A, Schick F, Haring HU. Phenotypes of prediabetes and
      stratification of cardiometabolic risk. Lancet Diabetes Endocrinol. 2016;4(9):789-98.
                                                                                      263
Chapter 9
ACKNOWLEDGEMENTS
264
                                                                         Chapter 9
Promoveren is iets dat je zeker niet alléén kunt doen en ik ben ervan overtuigd dat
een goede samenwerking onmisbaar is tijdens het promoveren. Er zijn een
heleboel mensen die een belangrijke bijdrage hebben geleverd aan het tot stand
komen van deze thesis en ik wil deze mensen dan ook heel graag bedanken.
Allereerst wil ik mijn promotieteam, Prof. Ellen Blaak en Dr. Gijs Goossens
bedanken voor het vertrouwen en de kans die ze mij ongeveer 5 jaar geleden
hebben gegeven om aan dit promotieonderzoek te starten.
Ellen, bedankt dat je altijd voor me klaar stond wanneer dat nodig was. Dankzij
jouw wetenschappelijk inzicht werd altijd de juiste breedte en diepgang aan de
manuscripten gegeven. Ik bewonder het enorm dat je, ondanks je drukke agenda,
steeds op korte termijn mijn papers na kon kijken en met constructieve
commentaar had voorzien.
Gijs, bedankt voor de goede begeleiding. Zowel tijdens het praktisch werk op de
testdagen en de planning daarvan als op wetenschappelijk gebied heb ik veel van
je geleerd. Ik heb het enorm gewaardeerd dat ik altijd bij je terecht kon wanneer
dat nodig was en dat je altijd bereid was om (uitgebreide ;) ) raad te geven. Jouw
kritische blik op de manuscripten en de grammaticale aanpassingen (al vond ik die
soms wat te uitgebreid :) ) hebben er steeds voor gezorgd dat de manuscripten
verbeterden. Ik vond het ook leuk dat we naast het werk gezellige tijden hebben
gehad, zoals bijvoorbeeld op de congressen in Barcelona, Götenborg of in
Denemarken.
Ellen en Gijs, bedankt voor de zeer prettige samenwerking gedurende de
afgelopen jaren!
Next, I would like to thank all members of the thesis assessment committee, Prof.
C. Stehouwer, Prof. J. Glatz, Prof. S. Kersten, Prof. L. van Loon and Prof. B.
Stallknecht for taking the time and effort to review my thesis and being present at
the official dissertation.
I would also like to thank all co-authors for the pleasant cooperation and for the
excellent contributions to improve the work that is presented in this thesis.
Zonder toegewijde proefpersonen had deze thesis nooit tot stand kunnen komen.
Mijn oprechte dank gaat daarom uit naar alle mensen die hebben deelgenomen
aan de experimentele onderzoeken; voor de moeite, flexibiliteit, gemaakte
kilometers en het afstaan van bloed, vet- en spierweefsel en vooral heel veel
zweet. Ik heb veel plezierige gesprekken en momenten gehad tijdens de
verschillende test- en trainingsdagen.
                                                                               265
Chapter 9
Birgitta, ook wij hebben een aanzienlijke tijd een kamer gedeeld. We zijn ongeveer
tegelijkertijd begonnen op de universiteit en ik heb je leren kennen als een sociale,
harde werker die heel direct en eerlijk is. Naast het werk kunnen we het goed met
elkaar vinden en hebben we vele leuke gesprekken en tijden gehad, zoals
bijvoorbeeld op het congres in Barcelona en op verschillende feestjes.
Max en Birgitta, ik ben blij dat jullie mijn paranimfen willen zijn en het is voor mij
een hele geruststelling dat jullie aan mijn zijde staan!
Een groot deel van deze thesis gaat over een farmacologische interventie die
synergetische effecten kan hebben. Het is terecht om te zeggen dat ons
onderzoeksteam tijdens deze studie ook een synergetisch geheel was ;) Birgitta en
Laura, ik wil jullie alle twee heel erg bedanken voor de hulp tijdens deze toch wel
stressvolle periode, waarin we soms verschillende screeningen én VO2max testen
op één ochtend moesten combineren met een testdag. Zonder jullie hulp zou het
nooit gelukt zijn om deze studie binnen de deadline tot een goed einde te brengen!
Ook Bas, Kirsten, Bart, Irene, Nicolaas en iedereen die op één of andere manier
heeft geholpen, bedankt voor de flexibiliteit en bereidwilligheid!
Kenneth, ik wil jou ook even bedanken voor de aangename samenwerking tijdens
onze studie in Hasselt. Je hebt enorm veel werk geleverd, hele lange dagen
gemaakt (zeker wanneer er zowel in de ochtend als in de avond training sessies
plaatsvonden) en heel veel cupjes geplakt ;) Ik ben blij dat we de studie samen tot
een goed einde hebben gebracht, dat er mooie data zijn uitgekomen en dat we niet
nog één aflevering van “In de gloria” of de film “Intouchable” moeten zien ;)
Ik wens je nog veel succes met het uitbouwen van je verdere academische
carrière.
266
                                                                         Chapter 9
Ik wil alle betrokken analisten bedanken voor het uitvoeren van de vele analyses.
Zonder jullie was dit alles nooit gelukt!
De secretaresses wil ik bedanken voor alle ondersteuning en het administratieve
werk.
Mijn familie mag zeker niet ontbreken…Peter en Els (en Amber), Inge en Geert
(Stan en Sien), dank jullie voor de interesse, steun en vooral voor de gezellige en
leuke momenten die we altijd samen hebben!
Liefste mama en papa, bedankt voor alles wat jullie voor mij gedaan hebben!
Bedankt voor jullie onvoorwaardelijke liefde, om altijd voor ons klaar te staan
wanneer dat nodig was, voor jullie interesse, raad en steun en dat jullie mij de
mogelijkheden hebben gegeven om te komen waar ik nu ben! Ik hoop dat we nog
lang in goede gezondheid bij elkaar kunnen zijn! Ik kan jullie niet genoeg
bedanken, maar als teken van dank, wil ik deze thesis heel graag aan jullie
opdragen!
                                                                               267
Chapter 9
CURRICULUM VITAE
268
                                                                         Chapter 9
                                  nd
Rudi Stinkens was born on June 2 1983 in Sittard, The Netherlands and raised in
Belgium. He studied Social and Technical Sciences and completed secondary
school at the Sint-Augustinus Insituut in Bree, Belgium in 2001. Consecutively, he
started a Bachelor of Science in Nutrition and Dietetics at the Katholiek
Hogeschool Kempen in Geel, Belgium, where he graduated in 2005. During the
following years, he specialised in sports nutrition at both the HAN University of
Applied Sciences in Nijmegen, The Netherlands and the Artesis & Plantijn
Hogeschool in Antwerpen, Belgium, while working in the food industry.
In 2010 he decided to make a switch in his career and started the Master of
Science program in Physical Activity and Health at Maastricht University, The
Netherlands, where he graduated in 2011. During this Master program, he
investigated astaxanthin supplementation in endurance trained athletes during a 6-
months internship at the department of Human Movement Sciences at Maastricht
University. Following his internship, he performed several months pro deo research
towards beet root juice supplementation, under the supervision of Dr. Cermak N.
and Prof. van Loon LJ.
In November 2012, he started as a Ph.D. candidate at the department of Human
Biology at Maastricht University (NUTRIM School of Nutrition and Translational
Research in Metabolism), under supervision of Prof. Dr. Ellen Blaak and Dr. Gijs
Goossens. His research is described in this thesis and is entitled “Adipose tissue
metabolism and cardiometabolic health in obesity - Effects of pharmacological and
lifestyle interventions”. During his Ph.D., Rudi was selected by the European
                                                             st
Association for the Study of Obesity (EASO) to attend the 1 Young Investigators
United summer school in Lisbon, Portugal (2015). He was also selected as one of
the 10 best candidates from the Netherlands to present his research findings at the
North European Young Diabetologists (NEYD) meeting in cooperation with the
Danish Diabetes Academy in Snekkersten, Denmark (2016). He presented his
research findings at several national and international conferences. Furthermore,
he received a travel grant from The Netherlands Association for the Study of
                                                    st
Obesity (NASO) to present his research at the 1 European Obesity Summit in
Göteborg, Sweden (2016) as well as a travel grant from the European Association
                                                                         rd
for the Study of Diabetes (EASD) to present his research at the 53 Annual
Meeting of the European Association for the Study of Diabetes (EASD) in Lisbon,
Portugal (2017).
                                                                               269
Chapter 9
LIST OF PUBLICATIONS
270
                                                                         Chapter 9
FULL PAPERS
                                                                                271
Chapter 9
ABSTRACTS
272