0% found this document useful (0 votes)
54 views208 pages

Biochemistry Textbook

Uploaded by

Desalegn Amenu
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
54 views208 pages

Biochemistry Textbook

Uploaded by

Desalegn Amenu
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 208

BIOCHEMISTRY TEXTBOOK

Desalegn Amenu and Ayantu Nugusa, 2024

Page i of 208
Preface

The study of biochemistry is an essential cornerstone of understanding the intricate molecular


mechanisms that drive all living organisms. As a bridge between biology and chemistry,
biochemistry allows us to delve deep into the chemical processes that underpin cellular
functions, from the simplest unicellular organisms to the most complex multicellular beings,
including humans.

Biochemistry's historical development is marked by groundbreaking discoveries and the


relentless pursuit of knowledge. From early explorations of fermentation and digestion to the
revolutionary elucidation of DNA's double-helix structure, biochemistry has continually
expanded our understanding of life at the molecular level. This discipline has evolved
significantly over the past century, driven by technological advancements and the integration
of interdisciplinary approaches, solidifying its importance in the scientific world.

The core concepts of biochemistry encompass the study of biomolecules—proteins, nucleic


acids, carbohydrates, and lipids—that are the building blocks of life. Understanding these
molecules' structures and functions is crucial for deciphering the complex web of reactions
that sustain life. Metabolism, the sum of all chemical reactions within an organism, is another
fundamental aspect, encompassing both the breakdown of molecules for energy (catabolism)
and the synthesis of necessary compounds (anabolism).

Enzymology, the study of enzymes as biological catalysts, reveals how these proteins
accelerate biochemical reactions, ensuring the efficient functioning of metabolic pathways.
Genetics, focusing on the storage, transmission, and expression of genetic information,
provides insights into the blueprint of life encoded in DNA. Cell signaling, exploring the
communication between cells, sheds light on how cells coordinate their activities in response
to internal and external stimuli. Structural biology, using advanced techniques like X-ray

Page ii of 208
crystallography and NMR spectroscopy, allows us to visualize the intricate architecture of
biomolecules.

The importance of biochemistry extends far beyond academic curiosity. In medicine, it


underpins our understanding of diseases at the molecular level, guiding the development of
new treatments and diagnostic tools. Pharmacology relies on biochemical principles to design
and develop new drugs, while agricultural advancements, such as enhanced crop yields and
pest resistance, are grounded in biochemical research. Environmental science also benefits
from biochemistry, with applications in developing biofuels and assessing the impact of
pollutants on ecosystems.

This text aims to introduce the principles and applications of biochemistry. It is designed for
students, researchers, and professionals who seek to understand the molecular basis of life.
By exploring the fundamental concepts and current trends in biochemistry, readers will gain
a solid foundation in this dynamic field and appreciate its pivotal role in advancing science
and medicine.

We embark on this journey into the world of biochemistry with the hope that it will inspire a
deeper appreciation of the molecular marvels that constitute life and encourage further
exploration and discovery in this ever-evolving discipline.

Page iii of 208


Table of contents

Table of Contents
Preface .......................................................................................................................... ii
1. Introduction .................................................................................................................... 1
2. An Overview of the Cellular Foundation of Life .............................................................. 4
3. The Molecules of Life ..................................................................................................... 4
3.1.1. Amino acids, Peptides and Proteins .................................................................... 8
3.1.2. Protein Sequence and Evolution ........................................................................ 19
3.1.3. Protein Sequence and Evolution: Structural and Functional Insights ................. 22
3.1.4. The Three-Dimensional Structure of Proteins .................................................... 25
3.1.5. Protein Folding and Denaturation ...................................................................... 29
3.1.6. Methods to Study Proteins ................................................................................. 32
3.1.7. Determining protein structure............................................................................. 35
4. Enzymes ...................................................................................................................... 37
4.1. Enzymes classification .......................................................................................... 37
4.2. How enzymes work ............................................................................................... 41
4.3. Enzyme kinetics ....................................................................................................... 44
4.4. Factors affecting rate of enzyme catalyzed reaction ................................................. 46
4.5. Regulation of enzyme catalyzed reactions ............................................................... 49
5. Carbohydrates ............................................................................................................. 52
5.1. Monosaccharides .................................................................................................. 55
5.2. Oligosaccharides................................................................................................... 57
5.2. Polysaccharides .................................................................................................... 59
5.3. Polysaccharides .................................................................................................... 61
5.4. Glycoconjugates.................................................................................................... 63
6. Lipids ........................................................................................................................... 67

Page iv of 208
6.1. Storage lipids ........................................................................................................ 69
6.2. Lipids as signal, cofactors and pigments ............................................................... 71
6.3. Working with lipds ................................................................................................. 73
7. Central metabolic pathways and energy transduction .................................................. 87
7.1. Bioenergetics ........................................................................................................ 90
7.2. Phosphoryl group transfer and ATP ...................................................................... 92
7.3. Biological oxidation -reduction reactions ............................................................... 94
7.4. Glycolysis, gluconeogenesis, and the pentose phosphate pathway ...................... 97
7.6. The pentose phosphate pathway ........................................................................ 108
7.7. Metabolic regulation (glucose and glycogen as examples).................................. 112
7.8. The citric acid cycle ............................................................................................. 115
7.8.1. Krebs cycle (TCA Cycle) Steps: ................................................................... 115
7.8.2. Regulation of Citric Acid cycles .................................................................... 116
7.8.3. TCA intermediates as precursors for biosynthesis........................................ 118
7.8.4. Glyoxylate cycle ........................................................................................... 119
7.9. Fatty Acid catabolism .......................................................................................... 122
7.9.1. Digestion, mobilization and transport of fats ................................................. 122
7.9.2. Oxidation of fatty acids ................................................................................. 124
7.9.3. Ketone bodies .............................................................................................. 126
7.10. Amino acid oxidation ........................................................................................... 128
7.10.1. Metabolic fates of amino acids ..................................................................... 128
7.10.2. Nitrogen excretion and the urea cycle .......................................................... 130
7.11. Oxidative Phosphorylation................................................................................... 135
7.11.1. The chemiosmotic theory and the mechanism of ATP synthesis .................. 137
7.11.2. The electron transport system ...................................................................... 139
8. Biosynthesis............................................................................................................... 146
8.1. Carbohydrate Biosynthesis in Plants and Bacteria .............................................. 146
Page v of 208
8.2. Photosynthetic Carbohydrate Synthesis.............................................................. 150
8.3. Photorespiration and C4 and CAM Pathways ..................................................... 154
8.5. Lipid biosynthesis ................................................................................................ 167
8.5.1. Biosynthesis of Fatty Acids and Triglycerides ............................................... 167
...................................................................................................................................... 171
...................................................................................................................................... 171
8.5.2. Biosynthesis of Membrane Phospholipids .................................................... 172
8.5.3. Biosynthesis of Cholesterol, Steroids, and Isoprenoids ................................ 176
8.6. Overview of Nitrogen Metabolism ....................................................................... 183
8.7. Biosynthesis of Amino Acids ............................................................................... 186
8.8. Biosynthesis of Nucleotides ................................................................................ 192
8.9. Regulation of nitrogen metabolism ...................................................................... 196

Page vi of 208
1. Introduction

Biochemistry is the branch of science that explores the chemical processes within and related
to living organisms. It is a laboratory-based science that combines biology and chemistry,
utilizing chemical knowledge and techniques to understand and solve biological problems.
Here are the key components of biochemistry:

Definition and Scope

Biochemistry is the study of the molecular mechanisms by which cells process and transmit
information, harness energy, and drive the myriad chemical reactions necessary for life. It
bridges the disciplines of biology and chemistry by exploring the structures and functions of
cellular components such as proteins, nucleic acids, carbohydrates, and lipids.

Core Concepts

Biomolecules: The study of the structure and function of biomolecules, including proteins,
nucleic acids, carbohydrates, and lipids. Understanding these molecules is essential as they
are the building blocks of life.

Metabolism: The chemical reactions involved in maintaining the living state of cells and
organisms. Metabolism is divided into two categories:

Catabolism: The breakdown of molecules to obtain energy.

Anabolism: The synthesis of all compounds needed by the cells.

Enzymology: Enzymes are proteins that act as catalysts in biochemical reactions. Studying
how enzymes work and how they can be regulated is crucial for understanding metabolic
pathways.

Page 1 of 208
Genetics: Understanding how genetic information is encoded in DNA and how it is expressed
and regulated in the form of RNA and proteins. This includes the study of gene structure,
function, and inheritance.

Cell Signaling: The study of how cells communicate with each other through chemical signals
and how these signals regulate cellular activities.

Structural Biology: The study of the physical structure of biological macromolecules, often
using techniques like X-ray crystallography, NMR spectroscopy, and cryo-electron
microscopy.

Importance and Applications

Biochemistry is fundamental to many areas of science and medicine. Its applications include:

Medical Research: Understanding diseases at a molecular level, leading to the development


of new treatments and diagnostic tools.

Pharmacology: Designing and developing new drugs.

Agriculture: Enhancing crop yields and resistance to pests.

Environmental Science: Developing biofuels and studying the impact of pollutants on


biological systems.

Historical Background

Biochemistry emerged as a distinct discipline in the early 20th century, although its roots can
be traced back to ancient times when humans first began to understand fermentation and
digestion. Key milestones include the discovery of enzymes, the elucidation of metabolic
pathways, and the development of techniques for studying biomolecules.

Page 2 of 208
Key Figures

Emil Fischer: Known for his work on the chemistry of proteins and enzymes.

Linus Pauling: Made significant contributions to understanding the nature of the chemical
bond and the structure of proteins.

James Watson and Francis Crick: Discovered the double-helix structure of DNA, which laid
the foundation for molecular biology.

Current Trends and Future Directions

Biochemistry continues to evolve with advancements in technology and interdisciplinary


approaches. Current trends include:

Genomics and Proteomics: Large-scale studies of genes and proteins.

Bioinformatics: Using computational tools to analyze biological data.

Synthetic Biology: Engineering new biological parts and systems.

In summary, biochemistry is a dynamic and rapidly advancing field that is crucial for
understanding the molecular basis of life. Its principles and techniques are foundational to
many scientific and medical disciplines.

Page 3 of 208
2. An Overview of the Cellular Foundation of Life

Cells are the fundamental building blocks of all living organisms, serving as the smallest unit
of life capable of performing all vital physiological functions. Understanding the cellular
foundation of life is essential for comprehending how organisms grow, reproduce, and interact
with their environment. Here, we will explore the basic structure and functions of cells, the

distinctions between different cell types, and the importance of cellular processes in the
context of life.

Basic Structure of Cells

Cells can be broadly categorized into two types: prokaryotic and eukaryotic. Despite their
differences, all cells share certain common features:

1. Cell Membrane: The cell membrane, or plasma membrane, is a phospholipid bilayer


that encloses the cell, providing a barrier that regulates the movement of substances
in and out of the cell. It plays a crucial role in maintaining homeostasis.
2. Cytoplasm: The cytoplasm is the jelly-like substance within the cell membrane,
containing cytosol (a fluid) and various organelles. It is the site of numerous metabolic
activities.
3. Genetic Material: All cells contain genetic material (DNA) that holds the instructions
for cellular processes. In prokaryotes, DNA is typically a single circular chromosome
located in the nucleoid region, while in eukaryotes, DNA is organized into linear
chromosomes within a membrane-bound nucleus.
4. Ribosomes: Ribosomes are molecular machines responsible for protein synthesis.
They are found in both prokaryotic and eukaryotic cells, either floating freely in the
cytoplasm or attached to the endoplasmic reticulum in eukaryotes.

Page 4 of 208
Prokaryotic vs. Eukaryotic Cells

Prokaryotic Cells:

 Simpler and smaller (typically 1-10 micrometers).


 Lack a membrane-bound nucleus and organelles.
 Include bacteria and archaea.
 Reproduce primarily through binary fission.

Eukaryotic Cells:

 Larger and more complex (typically 10-100 micrometers).


 Possess a membrane-bound nucleus and various organelles (e.g., mitochondria,
endoplasmic reticulum, Golgi apparatus).
 Include plants, animals, fungi, and protists.
 Reproduce through mitosis (for somatic cells) and meiosis (for gametes).

Cellular Organelles and Their Functions

Eukaryotic cells contain specialized structures called organelles, each with specific functions:

1. Nucleus: Contains genetic material and controls cellular activities by regulating gene
expression.
2. Mitochondria: Known as the powerhouses of the cell, mitochondria generate ATP
through cellular respiration.
3. Endoplasmic Reticulum (ER): The rough ER is studded with ribosomes and involved
in protein synthesis, while the smooth ER synthesizes lipids and detoxifies certain
chemicals.
4. Golgi Apparatus: Modifies, sorts, and packages proteins and lipids for transport
within the cell or secretion outside the cell.
Page 2 of 208
5. Lysosomes: Contain digestive enzymes to break down waste materials and cellular
debris.
6. Chloroplasts: Found in plant cells, chloroplasts are the sites of photosynthesis,
converting light energy into chemical energy stored in glucose.
7. Cytoskeleton: A network of protein filaments (microtubules, actin filaments,
intermediate filaments) that provides structural support, facilitates cell movement, and
organizes organelles.

Cellular Processes

Key cellular processes include:

1. Cell Division: Essential for growth, development, and repair. Involves mitosis for
somatic cells and meiosis for gametes.
2. Protein Synthesis: Transcription of DNA into mRNA in the nucleus, followed by
translation of mRNA into proteins at the ribosomes.
3. Metabolism: All chemical reactions within the cell, including catabolic pathways
(breaking down molecules for energy) and anabolic pathways (synthesizing complex
molecules).
4. Cell Signaling: Communication between cells through signaling molecules, receptors,
and signal transduction pathways, regulating various cellular activities.

Importance of Cellular Foundation

The cellular foundation of life is pivotal for understanding all biological processes. Cells are
the basic units of structure and function in organisms, and their coordinated activities enable
the complex behaviors and interactions observed in living beings. Advances in cellular biology
have led to significant breakthroughs in medicine, biotechnology, and environmental science,
highlighting the cell's central role in life.

Page 3 of 208
3. The Molecules of Life

Life at the molecular level is composed of a variety of organic molecules that interact in
complex ways to sustain cellular functions. These molecules are broadly categorized into four
main types: carbohydrates, lipids, proteins, and nucleic acids. Each type of molecule plays a
unique and essential role in the structure and function of cells.

1. Carbohydrates

Structure: Carbohydrates consist of carbon, hydrogen, and oxygen atoms, typically in a ratio
of 1:2:1 (C:H). They can be classified into three main categories based on their size and
complexity: monosaccharides, disaccharides, and polysaccharides.

 Monosaccharides: Simple sugars like glucose, fructose, and galactose. They serve
as the building blocks for more complex carbohydrates.

 Disaccharides: Formed by the linkage of two monosaccharides. Examples include


sucrose (glucose + fructose) and lactose (glucose + galactose).

 Polysaccharides: Long chains of monosaccharide units. Examples include starch


and glycogen (energy storage in plants and animals, respectively) and cellulose
(structural component in plant cell walls).

Functions:

 Energy Source: Glucose is a primary energy source for cells.

 Energy Storage: Glycogen in animals and starch in plants store energy for later use.

 Structural Role: Cellulose provides structural support in plant cell walls.

Page 4 of 208
2. Lipids

Structure: Lipids are a diverse group of hydrophobic molecules that include fats, oils,
phospholipids, and steroids. They are characterized by their insolubility in water and solubility
in nonpolar solvents.

 Fats and Oils: Composed of glycerol and fatty acids. Fats (solid at room temperature)
and oils (liquid at room temperature) are important energy storage molecules.

 Phospholipids: Contain two fatty acids, a glycerol backbone, and a phosphate group.
They are the main component of cell membranes.

 Steroids: Have a structure composed of four fused rings. Cholesterol and hormones
like testosterone and estrogen are examples of steroids.

Functions:

 Energy Storage: Fats store more energy per gram than carbohydrates.

 Cell Membranes: Phospholipids form the bilayer structure of cell membranes,


providing a barrier and matrix for membrane proteins.

 Signaling Molecules: Steroid hormones act as signaling molecules in various


physiological processes.

3. Proteins

Structure: Proteins are composed of amino acids linked by peptide bonds, forming
polypeptide chains that fold into specific three-dimensional structures. There are 20 different
amino acids, each with distinct side chains that influence protein structure and function.

Page 5 of 208
Levels of Protein Structure:

 Primary Structure: The linear sequence of amino acids.

 Secondary Structure: Local folding patterns such as alpha-helices and beta-sheets.

 Tertiary Structure: The overall three-dimensional shape of a single polypeptide chain.

 Quaternary Structure: The arrangement of multiple polypeptide chains in a multi-


subunit protein.

Functions:

 Enzymes: Catalyze biochemical reactions, increasing their rates.

 Structural Proteins: Provide support and shape to cells and tissues (e.g., collagen in
connective tissues).

 Transport Proteins: Carry molecules across cell membranes or through the


bloodstream (e.g., hemoglobin transports oxygen).

 Signaling Proteins: Hormones and receptors involved in communication within and


between cells.

4. Nucleic Acids

Structure: Nucleic acids are polymers of nucleotides, which consist of a sugar, a phosphate
group, and a nitrogenous base. The two main types of nucleic acids are DNA
(deoxyribonucleic acid) and RNA (ribonucleic acid).

 DNA: Consists of two strands forming a double helix. It contains the genetic blueprint
for the synthesis of proteins and is responsible for heredity.

 RNA: Single-stranded and involved in various roles in gene expression, including


messenger RNA (mRNA), transfer RNA (tRNA), and ribosomal RNA (rRNA).

Page 6 of 208
Functions:

 Genetic Information Storage: DNA stores genetic information in the sequence of its
bases (adenine, thymine, cytosine, and guanine).

 Protein Synthesis: RNA plays a central role in translating genetic information from
DNA into proteins.

 Regulation and Catalysis: Some RNA molecules have catalytic activity (ribozymes)
and are involved in regulating gene expression.

Interactions and Integration

The molecules of life do not function in isolation. They interact in intricate networks to support
the dynamic processes required for life. For example:

 Metabolic Pathways: Enzymes (proteins) catalyze reactions that convert


carbohydrates, lipids, and proteins into energy and building blocks for the cell.

 Gene Expression: DNA stores genetic information, which is transcribed into RNA and
translated into proteins that perform cellular functions.

 Cell Membrane Dynamics: Phospholipids form the cell membrane, while proteins
embedded in the membrane regulate transport and signaling.

Understanding these molecules and their interactions provides a fundamental basis for
exploring biological systems and developing applications in medicine, biotechnology, and
other fields. The study of these molecules continues to be a central focus of biochemistry and
molecular biology, driving discoveries that enhance our knowledge of life and its processes.

Page 7 of 208
3.1.1. Amino acids, Peptides and Proteins

Amino Acids

Amino acids are the building blocks of proteins, essential for numerous biological functions.
There are 20 standard amino acids commonly found in proteins, each with unique
characteristics that influence the structure and function of the proteins they form.

Structure of Amino Acids

Each amino acid has a central carbon atom (α-carbon) bonded to four different groups:

1. Amino Group (-NH2): A basic group that can accept a proton.

2. Carboxyl Group (-COOH): An acidic group that can donate a proton.

3. Hydrogen Atom (H): A single hydrogen atom.

4. R Group (Side Chain): A variable group that determines the specific properties of the
amino acid. The nature of the R group classifies amino acids into different categories.

Categories of Amino Acids

The R groups (side chains) of amino acids can be classified based on their properties:

1. Nonpolar (Hydrophobic) Amino Acids:

o Characteristics: These amino acids have side chains that are not attracted to
water.

o Examples: Alanine (Ala), Valine (Val), Leucine (Leu), Isoleucine (Ile),


Methionine (Met), Phenylalanine (Phe), Tryptophan (Trp), Proline (Pro).

2. Polar (Hydrophilic) Amino Acids:

o Characteristics: These amino acids have side chains that can form hydrogen
bonds with water, making them soluble in water.
Page 8 of 208
o Examples: Serine (Ser), Threonine (Thr), Asparagine (Asn), Glutamine (Gln),
Tyrosine (Tyr), Cysteine (Cys).

3. Charged Amino Acids:

o Acidic:

 Characteristics: These amino acids have side chains that are


negatively charged at physiological pH.

 Examples: Aspartate (Asp), Glutamate (Glu).

o Basic:

 Characteristics: These amino acids have side chains that are positively
charged at physiological pH.

 Examples: Lysine (Lys), Arginine (Arg), Histidine (His).

4. Aromatic Amino Acids:

o Characteristics: These amino acids have side chains with aromatic rings,
contributing to their unique properties.

o Examples: Phenylalanine (Phe), Tyrosine (Tyr), Tryptophan (Trp).

Functions of Amino Acids

Amino acids play a variety of critical roles in biological systems:

1. Protein Synthesis: Amino acids are polymerized into proteins, which perform a wide
array of structural and functional roles in cells.

2. Metabolic Functions:

o Energy Production: Amino acids can be metabolized to produce energy,


especially during periods of fasting or intense exercise.

Page 9 of 208
o Precursors to Biomolecules: Some amino acids serve as precursors for the
synthesis of other important biomolecules, such as nucleotides, hormones, and
neurotransmitters.

3. Signaling Molecules: Certain amino acids and their derivatives function as signaling
molecules. For example:

o Neurotransmitters: Glutamate and GABA (γ-aminobutyric acid) are


neurotransmitters derived from amino acids.

o Hormones: Thyroxine, a hormone produced by the thyroid gland, is derived


from the amino acid tyrosine.

4. Regulation of Metabolism: Amino acids can regulate metabolic pathways and gene
expression. For instance, leucine plays a role in the regulation of protein synthesis
through the mTOR pathway.

5. Detoxification: Some amino acids, like glutamine, help in the detoxification of


ammonia in the body by forming non-toxic compounds excreted in urine.

Essential and Non-Essential Amino Acids

Amino acids are also categorized based on their necessity in the human diet:

1. Essential Amino Acids: These cannot be synthesized by the human body and must
be obtained from the diet. They include:

o Histidine (His)

o Isoleucine (Ile)

o Leucine (Leu)

o Lysine (Lys)

Page 10 of 208
o Methionine (Met)

o Phenylalanine (Phe)

o Threonine (Thr)

o Tryptophan (Trp)

o Valine (Val)

2. Non-Essential Amino Acids: These can be synthesized by the body and are not
required to be obtained through the diet. They include:

o Alanine (Ala)

o Arginine (Arg) (conditionally essential)

o Asparagine (Asn)

o Aspartate (Asp)

o Cysteine (Cys) (conditionally essential)

o Glutamate (Glu)

o Glutamine (Gln) (conditionally essential)

o Glycine (Gly)

o Proline (Pro)

o Serine (Ser)

o Tyrosine (Tyr) (conditionally essential)

Amino acids are fundamental to the structure and function of proteins, which are crucial for
the myriad biochemical processes that sustain life. Understanding their properties and roles
provides insight into the molecular basis of health and disease.

Page 11 of 208
Proteins are complex biomolecules made up of one or more long chains of amino acids. They
play crucial roles in nearly all biological processes. Here is a detailed overview of proteins,
including their structure, functions, and importance:

Structure of Proteins

Proteins are composed of 20 different amino acids, which are linked together by peptide
bonds. The sequence and number of these amino acids determine the protein's structure and
function. Protein structure is organized into four levels:

Proteins can be classified into different types based on their molecular shape and structure.
Here's a detailed overview of their classification and the various levels of protein structure:

Classification of Proteins Based on Molecular Shape

1. Fibrous Proteins:

o Structure: Polypeptide chains run parallel and are held together by hydrogen
and disulfide bonds, forming a long, fiber-like structure.

o Solubility: Generally insoluble in water.

o Examples:

 Keratin: Found in hair, wool, and silk.

 Myosin: Found in muscles.

2. Globular Proteins:

o Structure: Polypeptide chains coil around to form a spherical shape.

o Solubility: Usually soluble in water.

o Examples:

 Insulin: Regulates glucose levels in the blood.

Page 12 of 208
 Albumins: Found in egg whites and blood plasma.

Levels of Protein Structure

1. Primary Structure:

o Definition: The exact sequence of amino acids in the polypeptide chain.

o Importance: The sequence determines the protein’s final fold and function.
Any change in the sequence can alter the protein’s function.

o Example: A sequence of amino acids like Methionine (Met), Glycine (Gly),


Serine (Ser), Threonine (Thr), Histidine (His), Leucine (Leu).

2. Secondary Structure:

o Definition: Local folded structures that form within a polypeptide due to


interactions between atoms of the backbone.

o Types:

 α-Helix: A right-handed coil where each amino acid forms a hydrogen


bond with the amino acid four residues ahead.

 Example: Keratin in hair.

 β-Pleated Sheet: Polypeptide chains lie side by side, held together by


hydrogen bonds, forming a sheet-like structure.

 Example: Silk fibroin.

3. Tertiary Structure:

o Definition: The overall three-dimensional shape of a single polypeptide chain,


further folding of the secondary structure.

Page 13 of 208
o Stabilization: Hydrogen bonds, electrostatic forces, disulfide linkages, and
Van der Waals forces.

o Shapes: Can form fibrous or globular shapes.

4. Quaternary Structure:

o Definition: The spatial arrangement of multiple polypeptide chains (subunits)


into a single functional protein complex.

o Examples:

 Hemoglobin: Composed of four subunits, each with its own polypeptide


chain.

Summary of Key Points

 Primary Structure: Sequence of amino acids held together by covalent peptide


bonds.

 Secondary Structure: Local folding into α-helices and β-pleated sheets stabilized by
hydrogen bonds.

 Tertiary Structure: Three-dimensional folding due to interactions among side chains


(R groups).

 Quaternary Structure: Arrangement of multiple polypeptide chains into a functional


protein complex.

Understanding these levels of protein structure is crucial for studying protein function and how
alterations in structure can lead to diseases. Each level of structure contributes to the protein’s
overall shape and stability, determining its role in biological processes.

Page 14 of 208
Figure 1: Protein structures

Page 15 of 208
Peptides are short chains of amino acids that are linked together by peptide bonds. They are
simpler than proteins but share similar building blocks and play various roles in biological
systems. Here is a detailed overview of peptides:

Structure of Peptides

1. Amino Acids:

o The basic building blocks of peptides.

o Each amino acid consists of an amino group (-NH2), a carboxyl group (-


COOH), a hydrogen atom, and a distinctive side chain (R group) attached to a
central carbon atom (α-carbon).

2. Peptide Bonds:

o Formed through a dehydration synthesis reaction between the carboxyl group


of one amino acid and the amino group of another, releasing a molecule of
water.

o The resulting bond is called a peptide bond (-CONH-).

3. Length:

o Peptides typically range from 2 to 50 amino acids in length.

o Dipeptides: Consist of two amino acids.

o Tripeptides: Consist of three amino acids.

o Oligopeptides: Short peptides, generally consisting of up to 20 amino acids.

o Polypeptides: Longer chains, consisting of more than 20 amino acids but


fewer than those in proteins.

Page 16 of 208
Types of Peptides

1. Signaling Peptides:

o Act as hormones and neurotransmitters, facilitating communication between


cells.

o Example: Insulin, which regulates blood sugar levels.

2. Antimicrobial Peptides:

o Serve as part of the immune response, providing defense against pathogens.

o Example: Defensins, which are found in the immune system and epithelial
cells.

3. Therapeutic Peptides:

o Used in medicine for their specific biological activities.

o Example: Peptide-based drugs like vasopressin, which regulates water


retention in the body.

Functions of Peptides

1. Biological Signaling:

o Peptides can function as signaling molecules, transmitting information between


cells and tissues.

o Example: Glucagon, which helps to raise blood glucose levels.

2. Immune Response:

Page 17 of 208
o Some peptides play a crucial role in the immune system by attacking pathogens
or modulating immune responses.

o Example: LL-37, a peptide with antimicrobial properties.

3. Structural Components:

o Certain peptides contribute to the structural integrity of cells and tissues.

o Example: Collagen peptides, which support the structure of skin, bones, and
connective tissues.

4. Regulation:

o Peptides can regulate various physiological processes, including metabolism,


growth, and repair.

o Example: Growth hormone-releasing peptides, which stimulate the release of


growth hormone.

Importance of Peptides

 Research and Medicine: Peptides are used extensively in biomedical research and
therapeutic applications due to their specific and potent biological activities.

 Biotechnology: Peptides are utilized in various biotechnological applications,


including the development of new drugs, vaccines, and diagnostic tools.

 Nutritional Supplements: Collagen peptides and other bioactive peptides are used
in dietary supplements to promote health and wellness.

Page 18 of 208
3.1.2. Protein Sequence and Evolution

Protein sequence refers to the specific order of amino acids in a polypeptide chain. This
sequence is crucial because it determines the protein's structure and function. Over time,
protein sequences can evolve, leading to variations that can impact an organism's adaptability
and survival.

1. Protein Sequence

 Amino Acids: Proteins are composed of 20 different amino acids. The sequence of
these amino acids in a polypeptide chain is determined by the genetic code in an
organism's DNA.

 Peptide Bonds: Amino acids are linked by peptide bonds, forming a linear chain.

 Primary Structure: The primary structure of a protein is its unique sequence of amino
acids. This sequence dictates how the protein will fold into its secondary, tertiary, and
quaternary structures.

Example: Consider a short segment of a protein sequence:

2. Protein Evolution

Protein evolution refers to the changes in protein sequences over time. These changes can
result from mutations, natural selection, and genetic drift.

 Mutations: Changes in the DNA sequence can lead to changes in the amino acid
sequence of proteins. These mutations can be:

o Silent: No change in the amino acid sequence.

o Missense: Change in one amino acid, which can affect protein function.

o Nonsense: Creates a stop codon, potentially truncating the protein.

Page 19 of 208
o Frameshift: Insertion or deletion of nucleotides that alter the reading frame.

 Natural Selection: Proteins that provide a survival advantage are more likely to be
passed on to future generations. Beneficial mutations are selected for, while harmful
ones are selected against.

 Genetic Drift: Random changes in protein sequences due to chance events. This can
lead to variations within populations, especially in small populations.

3. Mechanisms of Protein Evolution

 Gene Duplication: A gene can be duplicated, allowing one copy to maintain its
original function while the other copy is free to accumulate mutations and potentially
acquire a new function.

 Recombination: Exchange of genetic material between different DNA molecules can


create new combinations of amino acids in proteins.

 Exon Shuffling: Exons, the coding regions of genes, can be mixed and matched to
create new proteins with novel functions.

4. Evolutionary Relationships

By comparing protein sequences across different species, scientists can infer evolutionary
relationships. Proteins that are conserved across species suggest a common ancestry and
essential function.

 Homologous Proteins: Proteins that share a common ancestor. They can be:

o Orthologs: Homologous proteins in different species that perform the same


function.

o Paralogs: Homologous proteins within the same species that have evolved
new functions.

Page 20 of 208
 Phylogenetic Trees: Diagrams that represent the evolutionary relationships between
species based on protein or DNA sequence similarities and differences.

5. Examples of Protein Evolution

 Hemoglobin: Variations in hemoglobin sequences among different species reflect


adaptations to different oxygen environments.

 Cytochrome c: A highly conserved protein used in the study of evolutionary


relationships among diverse organisms.

Conclusion

Protein sequences are fundamental to the structure and function of proteins. The evolution of
these sequences is driven by mutations, natural selection, genetic drift, and recombination.
By studying protein sequences and their variations, scientists can gain insights into the
evolutionary history and relationships of organisms. Understanding protein evolution is crucial
for fields such as molecular biology, genetics, and evolutionary biology.

Page 21 of 208
3.1.3. Protein Sequence and Evolution: Structural and Functional Insights

The recent accumulation of thousands of protein three-dimensional (3D) structures, along


with the development of advanced tools for sequence similarity searches, has significantly
enhanced our understanding of the evolution of protein structures and functions. Studies have
revealed numerous examples of how protein structures evolve, leading to homologs with
different structural folds. This evolution has important implications for protein design and
engineering while posing challenges for homology modeling.

Evolution of Protein Structures

Protein structures evolve through various mechanisms, including gene duplication followed
by mutation and selection. This process can lead to the emergence of new protein structures
and functions.

 Gene Duplication and Mutation: Gene duplication creates copies of genes, which
can then accumulate mutations. Some of these mutations may lead to new protein
functions or structures.

 Pseudo-Twofold Symmetry: Many protein structural domains exhibit pseudo-twofold


symmetry, suggesting they evolved from the fusion of two primitive half-domains.
These symmetric molecules may retain sequence identity between the two halves or
diverge significantly.

Localized Structural Motifs and Fold Evolution

Motifs exhibiting similar sequences and localized structures can exist within seemingly
nonhomologous protein folds. These motifs, detected using sequence- and structure-based
searching methods, include:

 Heme Attachment Motifs

Page 22 of 208
 P-Loops (Walker A)

 FAD/NAD-Binding Motifs

 Zn Fingers

 Fe-S-Binding Motifs

 RNA-Binding Motifs

 Asp-Box Motifs

The existence of these motifs in different overall folds raises questions about their
evolutionary origins and whether they represent cases of convergent or divergent evolution.

Evolution of Protein Functions

The classification of emerging sequence and structural data into evolutionary families (such
as the SCOP and CATH databases) helps us understand the relationships between protein
sequence and structure. However, to fully comprehend protein evolution, we must also
consider the role of protein function.

 Conservation of Structure Over Sequence and Function: Protein 3D structure


tends to be more conserved than either sequence or function. Different protein folds
can perform similar functions, and similar sequences can result in different structures.

Challenges in Homology Modeling

The existence of proteins with similar sequences but different structures complicates
homology modeling. Accurate detection of evolutionary relatedness requires more than just
sequence or structural similarity.

Page 23 of 208
 Sequence Profile-Based Algorithms:

o PSI-BLAST (Position-Specific Iterated Basic Local Alignment Search


Tool): Identifies distant homologs by iteratively searching for sequence
profiles.

o HMMER (Hidden Markov Model): Uses hidden Markov models to detect


homologous sequences based on sequence profiles.

 Additional Considerations for Evolutionary Relatedness:

o Molecular Function Similarity: Evaluating the similarity of molecular


functions can provide additional evidence for common ancestry.

o Unusual Structural Features: Retention of unusual structural features can


support evolutionary relatedness.

o Domain Organization: Common domain organization among proteins


indicates shared evolutionary origins.

o Combined Features: A combination of sequence, structural, and functional


similarities offers the strongest evidence for homology.

Evolution of Protein Structures and Functions: Examples

 Divergent Evolution: Proteins with different functions but similar structures suggest
that they evolved from a common ancestor but adapted to different roles.

 Convergent Evolution: Proteins with similar functions but different structures


demonstrate that different evolutionary paths can lead to similar functional outcomes.

Page 24 of 208
3.1.4. The Three-Dimensional Structure of Proteins

Proteins are complex molecules that perform a wide array of functions in biological systems.
Their function is directly related to their three-dimensional (3D) structure, which is determined
by the sequence of amino acids in their primary structure. The 3D structure of proteins can
be divided into four levels: primary, secondary, tertiary, and quaternary structures.
Understanding these levels is crucial for grasping how proteins function and how they are
involved in various biological processes.

1. Primary Structure

 Definition: The primary structure is the linear sequence of amino acids in a


polypeptide chain, linked by peptide bonds.

 Significance: The sequence of amino acids determines the protein's final 3D shape
and function. Any change in this sequence can alter the protein's structure and,
consequently, its function.

2. Secondary Structure

 Definition: The secondary structure refers to local folded structures that form within a
polypeptide due to interactions between atoms of the backbone. These structures are
stabilized by hydrogen bonds.

 Types:

o Alpha Helix (α-helix): A right-handed coil where each amino acid forms a
hydrogen bond with the fourth amino acid ahead in the chain. This structure is
common in fibrous proteins and many enzymes.

Page 25 of 208
o Beta Sheet (β-sheet): Consists of beta strands connected laterally by at least
two or three backbone hydrogen bonds, forming a sheet-like arrangement.
Beta sheets can be parallel or antiparallel.

3. Tertiary Structure

 Definition: The tertiary structure is the overall 3D shape of a single polypeptide chain,
formed by the folding of the secondary structures. It is stabilized by various
interactions, including hydrogen bonds, disulfide bridges, hydrophobic interactions,
and ionic bonds.

 Significance: The tertiary structure determines the protein's functionality. The unique
3D shape allows the protein to interact specifically with other molecules.

4. Quaternary Structure

 Definition: The quaternary structure is the arrangement of multiple polypeptide chains


(subunits) into a single functional complex. The same types of interactions stabilize
this structure as the tertiary structure.

 Examples: Hemoglobin, which consists of four polypeptide subunits, and DNA


polymerase, which has multiple subunits that work together to replicate DNA.

Determinants of Protein Structure

 Amino Acid Properties: The chemical properties of the amino acid side chains (R-
groups) influence how a protein folds.

o Hydrophobic vs. Hydrophilic: Hydrophobic side chains tend to cluster in the


interior of the protein, while hydrophilic side chains are usually found on the
surface.

Page 26 of 208
o Charged Side Chains: Positively and negatively charged side chains often
form ionic bonds, stabilizing the structure.

 Environmental Factors: pH, temperature, and the presence of salts and other
chemicals can affect protein folding and stability.

 Chaperones: These are proteins that assist in the folding of other proteins, ensuring
they achieve the correct conformation.

Protein Misfolding

 Consequences: Incorrectly folded proteins can lead to loss of function or gain of toxic
function, contributing to diseases such as Alzheimer's, Parkinson's, and prion
diseases.

 Mechanisms: Cells have quality control mechanisms, including molecular


chaperones and proteolytic systems, to deal with misfolded proteins.

Techniques for Determining Protein Structure

 X-Ray Crystallography: Provides high-resolution details of protein structures by


analyzing the diffraction patterns of X-rays passing through a protein crystal.

 Nuclear Magnetic Resonance (NMR) Spectroscopy: Useful for studying proteins in


solution, providing information about the structure and dynamics of proteins.

 Cryo-Electron Microscopy (Cryo-EM): Allows the visualization of proteins in their


native state without the need for crystallization, useful for large complexes.

Page 27 of 208
Figure 2: De Novo Predicted 3D Models of Membrane Proteins with No Known Structure

Page 28 of 208
3.1.5. Protein Folding and Denaturation

Protein folding is a complex process by which a polypeptide chain assumes its functional
three-dimensional structure, crucial for its biological activity. This process involves the
formation of secondary and tertiary structures, driven by various interactions between amino
acids and the surrounding environment. On the other hand, denaturation refers to the loss of
a protein's native structure, disrupting its function. Understanding these processes is vital for
various fields, including biochemistry, medicine, and biotechnology.

Protein Folding

 Definition: Protein folding is the process by which a linear sequence of amino acids
(primary structure) folds into a specific three-dimensional shape (tertiary structure).

 Key Points:

o Primary Structure: The sequence of amino acids determines the folding


pathway and the final structure of the protein.

o Secondary Structure Formation: Local folding into alpha helices, beta


sheets, and turns stabilized by hydrogen bonds.

o Tertiary Structure Formation: Overall folding of the polypeptide chain,


stabilized by interactions such as hydrogen bonds, disulfide bonds,
hydrophobic interactions, and electrostatic attractions.

o Quaternary Structure (if applicable): Assembly of multiple polypeptide


chains into a functional protein complex.

Page 29 of 208
Factors Influencing Protein Folding

 Physicochemical Environment:

o pH and Temperature: Optimal conditions are required for proper folding;


deviations can lead to denaturation.

o Ionic Strength: Salt concentration affects protein stability and folding.

 Chaperones: Specialized proteins that assist in the folding of other proteins, ensuring
they attain their correct structure.

 Post-Translational Modifications: Chemical modifications of amino acids can


influence folding and function.

Protein Denaturation

 Definition: Denaturation is the process by which a protein loses its native structure
and consequently its biological activity.

 Causes:

o Heat: High temperatures disrupt non-covalent bonds and interactions


stabilizing the protein structure.

o pH Changes: Extremes of pH can alter ionization states of amino acids,


affecting folding.

o Organic Solvents: Disrupt hydrogen bonding and hydrophobic interactions.

o Chaotropic Agents: Such as urea and guanidine hydrochloride, which disrupt


hydrogen bonds and hydrophobic interactions.

 Consequences: Denatured proteins often lose their biological function and may
aggregate or precipitate out of solution.

Page 30 of 208
Techniques for Studying Protein Folding and Denaturation

 Circular Dichroism (CD) Spectroscopy: Measures changes in protein secondary


structure.

 Fluorescence Spectroscopy: Monitors changes in protein tertiary structure by


detecting tryptophan or tyrosine fluorescence.

 Differential Scanning Calorimetry (DSC): Measures heat absorption or release


during protein unfolding.

 Size Exclusion Chromatography (SEC): Analyzes protein aggregation states.

 X-Ray Crystallography, NMR Spectroscopy, and Cryo-Electron Microscopy:


Techniques for visualizing protein structures and understanding their folding
dynamics.

Applications and Implications

 Biotechnology: Understanding protein folding allows for the design of proteins with
specific functions or improved stability.

 Medicine: Misfolded proteins are associated with various diseases (e.g., Alzheimer's,
prion diseases), making protein folding a target for therapeutic interventions.

 Industrial Processes: Enzymes used in industry require stable folding for optimal
activity under different conditions.

Page 31 of 208
3.1.6. Methods to Study Proteins

Studying proteins involves a range of techniques aimed at understanding their structure,


function, interactions, and roles in biological processes. These methods are essential across
various scientific disciplines, including biochemistry, molecular biology, medicine, and
biotechnology. Here is an overview of key methods used to study proteins:

1. Protein Purification

Objective: Isolate a specific protein from a complex mixture for further study.

Techniques:

 Salting Out: Precipitation of proteins using high salt concentrations.

 Chromatography:

o Affinity Chromatography: Uses a specific ligand for the protein of interest.

o Ion Exchange Chromatography: Separates proteins based on charge.

o Size Exclusion Chromatography: Separates proteins based on size.

 Electrophoresis: Separates proteins based on charge and size using an electric field.

2. Protein Sequencing

Objective: Determine the amino acid sequence of a protein.

Techniques:

 Edman Degradation: Sequential removal of amino acids from the N-terminus.

 Mass Spectrometry: Measures mass-to-charge ratio of peptides for sequencing.

 Next-Generation Sequencing (NGS): High-throughput methods for large-scale


sequencing.

Page 32 of 208
3. Protein Structure Determination

Objective: Understand the three-dimensional structure of proteins.

Techniques:

 X-Ray Crystallography: Provides high-resolution structure from protein crystals.

 Nuclear Magnetic Resonance (NMR) Spectroscopy: Provides structure in solution.

 Cryo-Electron Microscopy (Cryo-EM): Visualizes protein structures without


crystallization.

4. Protein Detection and Quantification

Objective: Detect and measure proteins in samples.

Techniques:

 Western Blotting: Detects specific proteins using antibodies.

 Enzyme-Linked Immunosorbent Assay (ELISA): Quantifies proteins based on


antigen-antibody binding.

 Mass Spectrometry: Quantifies proteins based on peptide mass.

5. Protein-Protein Interactions

Objective: Identify and characterize interactions between proteins.

Techniques:

 Co-immunoprecipitation: Detects protein complexes using antibodies.

 Yeast Two-Hybrid System: Screens for protein-protein interactions in yeast.

 Surface Plasmon Resonance (SPR): Measures binding affinities of protein


interactions.

Page 33 of 208
6. Structural and Functional Studies

Objective: Investigate how protein structure relates to function.

Techniques:

 Site-Directed Mutagenesis: Introduces specific mutations to study their effects.

 Protein Engineering: Modifies proteins to improve stability or function.

 Functional Assays: Tests biochemical or biological activities of proteins.

7. Proteomics

Objective: Study all proteins in a system or organism (the proteome).

Techniques:

 Mass Spectrometry: Identifies and quantifies proteins in complex mixtures.

 2D Gel Electrophoresis: Separates proteins based on charge and size for proteome
analysis.

 Shotgun Proteomics: High-throughput sequencing and identification of peptides.

Applications and Implications

 Biomedical Research: Understanding disease mechanisms and drug targets.

 Biotechnology: Engineering proteins for industrial and medical applications.

 Clinical Diagnostics: Biomarker discovery and disease diagnosis.

 Environmental Science: Studying protein function in ecological systems.

Page 34 of 208
3.1.7. Determining protein structure

Determining protein structure is crucial for understanding its function and interactions within
biological systems. Here are some key methods used in protein structure determination:

1. X-ray Crystallography:

 Principle: Crystallizes proteins into a crystal lattice and exposes them to X-rays. The
diffraction pattern generated when X-rays pass through the crystal is used to
determine the electron density and thus the 3D structure of the protein.

 Strengths: Provides high-resolution structures (up to atomic level), suitable for small
to medium-sized proteins.

 Limitations: Requires protein crystallization, which can be challenging for some


proteins. In addition, the protein must be static in the crystal lattice, limiting its
application for dynamic proteins.

2. Nuclear Magnetic Resonance (NMR) Spectroscopy:

 Principle: Analyzes the interaction of atomic nuclei with magnetic fields in solution.
Provides information on atomic structure, dynamics, and interactions.

 Strengths: Can study proteins in near-native conditions (solution state), providing


insights into dynamics and flexibility. Suitable for smaller proteins (<30 kDa).

 Limitations: Limited to proteins of a certain size (<50 kDa) and requires significant
expertise in data interpretation and sample preparation.

3. Cryo-Electron Microscopy (Cryo-EM):

 Principle: Uses electron microscopy on samples frozen at cryogenic temperatures.


Provides high-resolution images of large protein complexes and membrane proteins.

Page 35 of 208
 Strengths: Can study proteins in their native environment, such as within cell
membranes or large complexes. Has advanced rapidly, allowing resolution
improvements.

 Limitations: Requires large quantities of pure protein and significant computational


resources for data processing. Resolution can be lower compared to X-ray
crystallography for smaller proteins.

4. Mass Spectrometry (MS):

 Principle: Measures mass-to-charge ratios of peptides and proteins, providing


information on their composition and structure.

 Strengths: Useful for analyzing protein modifications (e.g., phosphorylation) and


interactions (e.g., protein-protein interactions). Can be coupled with other techniques
for comprehensive structural analysis.

 Limitations: Provides structural information indirectly and may require extensive


fragmentation and computational analysis for full structural determination.

5. Computational Modeling:

 Principle: Uses computational methods to predict and refine protein structures based
on known templates, physical principles (e.g., energy minimization), and experimental
data.

 Methods: Includes homology modeling (using known structures as templates),


molecular docking (predicting binding interactions), and molecular dynamics
simulations (modeling protein dynamics over time).

 Strengths: Provides structural insights when experimental methods are challenging.


Can predict structures of novel proteins or protein complexes.

Page 36 of 208
 Limitations: Accuracy depends on the quality of input data and algorithms used.
Requires validation against experimental data for reliability.

4. Enzymes
4.1. Enzymes classification

The classification and nomenclature of enzymes are governed by specific principles to ensure
clarity and accuracy in scientific communication. Here are the key points based on the
principles outlined:

1. Single Enzyme Names: Enzyme names, especially those ending in "-ase," should
refer to single catalytic entities. Enzyme systems involving multiple enzymes should
be named as systems (e.g., succinate oxidase system).
2. Classification by Reaction Type: Enzymes are primarily classified and named based
on the specific chemical reaction they catalyze. This ensures that enzymes with similar
functions are grouped together.
3. Chemical Nature and Substrate Specificity: While less commonly used, enzymes
can also be classified based on their prosthetic groups or substrate specificity.
However, the primary basis remains the reaction catalyzed.
4. Grouping and Naming: Enzymes with similar catalytic properties are grouped
together, even if they come from different organisms (e.g., bacterial, plant, or animal
sources). Exceptions exist for enzymes with significantly different mechanisms or
substrate specificities.
5. Complex Reactions: Enzymes catalyzing complex transformations are classified
based on the essential first catalytic step. Intermediate reactions, whether catalyzed
or spontaneous, may influence naming conventions.

Page 37 of 208
6. Direction of Reaction: The direction of the reaction used for classification should be
consistent within enzyme classes. Systematic names are derived from a specific
reaction direction, while common names may reflect physiological relevance.
7. Examples: Enzyme classifications and naming examples include EC 1.1.1.9 (xylitol +
2-oxidoreductase) and its common name, D-xylulose reductase, illustrating the
principles applied to enzyme nomenclature.

The Enzyme Commission's approach to enzyme nomenclature reflects a balance between


systematic and practical considerations. Initially, there was a push for systematic names that
precisely describe enzyme actions but were criticized for being cumbersome. Over time, more
emphasis was placed on common or trivial names, which are shorter and more familiar,
though less systematic. Systematic names, however, remain important for classification and
clarity, especially since they are self-explanatory and can be systematically generated for new
enzymes.

In scientific papers, common names are generally used for enzymes that are not the primary
focus, while systematic names or reaction equations are preferred for enzymes under detailed
study. Additionally, citing the source of the enzyme is crucial for identification, especially when
multiple forms exist. The Enzyme List provides references supporting the existence and
specificity of each enzyme, focusing on key studies rather than comprehensive bibliographies.

This approach ensures clarity and consistency in enzyme naming, aiding communication and
research in the field of biochemistry and enzymology.

Page 38 of 208
The Enzyme Commission's classification system, devised in 1961 and still widely used,
categorizes enzymes into six main classes, each with subclasses and sub-subclasses
denoted by a four-element EC number. Here is a summary of the classification scheme:

1. Class 1: Oxidoreductases

o Catalyze oxidation-reduction reactions.

o Systematic name: donor

oxidoreductase.

o Common names often end in "-dehydrogenase" or "-reductase".

o Specificities such as the nature of the hydrogen donor and acceptor determine
subclassification.

2. Class 2: Transferases

o Transfer functional groups between molecules.

o Systematic name: donor grouptransferase.

o Common names often reflect the acceptor or donor.

o Includes special cases like transaminases (EC 2.6.1).

3. Class 3: Hydrolases

o Catalyze hydrolysis reactions, breaking bonds with the addition of water.

o Systematic name includes "hydrolase".

o Common names typically end in "-ase" derived from the substrate name.

4. Class 4: Lyases

Page 39 of 208
o Catalyze the removal of groups to form double bonds or rings, or add groups
to double bonds.

o Systematic name: substrate group-lyase.

o Common names include terms like "-decarboxylase" or "-aldolase".

5. Class 5: Isomerases

o Catalyze geometric or structural changes within one molecule.

o Includes various types like racemases, mutases, and tautomerases.

o Subclasses and sub-subclasses are based on the type of isomerism and


substrates involved.

6. Class 6: Ligases

o Catalyze the joining of two molecules using ATP or similar compounds.

o Systematic name: Xligase.

o Common names have historically used "synthetase" but now generally use
"ligase" for clarity.

Each enzyme is assigned an EC number reflecting its class, subclass, sub-subclass, and
specific enzyme within that category. This systematic approach facilitates clarity and
consistency in enzyme classification and nomenclature, aiding in the communication and
understanding of enzyme function across scientific research and applications.

Page 40 of 208
4.2. How enzymes work

Enzymes are biological catalysts that speed up chemical reactions within cells. They work by
lowering the activation energy required for a reaction to proceed, thereby increasing the rate
of reaction without being consumed in the process. Here is a breakdown of how enzymes
achieve this:

1. Substrate Binding: Enzymes have specific binding sites where their substrate(s)
bind. The substrate(s) are the molecule(s) upon which the enzyme acts.

2. Formation of Enzyme-Substrate Complex: When the substrate binds to the


enzyme's active site, it forms an enzyme-substrate complex. This complex is hel

3. d together by various interactions, such as hydrogen bonds, ionic bonds, and


hydrophobic interactions.

4. Catalysis of Reaction: Once the substrate is bound, the enzyme catalyzes the
conversion of the substrate(s) into product(s). This catalysis involves several
mechanisms:

o Induced Fit: The enzyme may undergo a conformational change upon


substrate binding, which brings catalytic groups into position to facilitate the
reaction.

o Active Site Environment: The microenvironment of the active site can


stabilize transition states or facilitate bond breaking or formation.

o Lowering Activation Energy: Enzymes lower the activation energy barrier by


providing an alternative reaction pathway that requires less energy to convert
substrates into products.

o Transition State Stabilization: Enzymes stabilize the transition state of the


reaction, making it easier for the reaction to proceed.

Page 41 of 208
5. Product Release: After catalyzing the reaction, the enzyme releases the product(s),
which are then free to diffuse away from the active site.

6. Enzyme Recycling: Once the product is released, the enzyme is free to catalyze
another reaction cycle with another substrate molecule.

This process allows enzymes to perform their specific functions with high efficiency and
specificity under physiological conditions, making them essential for life processes such as
metabolism, signal transduction, and cellular maintenance. If you need further details or have
specific aspects of enzyme function you would like to explore, feel free to ask!

Figure 3: Enzymes classifications

Page 42 of 208
 Binding of the substrate(s) to the enzyme at their active site takes place, thereby
forming an enzyme-substrate complex.

 Either enzymes catalyze the chemical reaction to take place, which can be a synthesis
reaction (favors bond formation) or a decomposition reaction (favors bond breakage).

 As a result, the formation of one or more products takes place, and the enzymes are
released for their reuse in the next reaction.

Figure 4: Enzyme key and lock model

Page 43 of 208
4.3. Enzyme kinetics
Enzyme kinetics is the study of the rates at which enzymes catalyze reactions and the factors
that influence these rates. Here are the key aspects of enzyme kinetics:

1. Michaelis-Menten Kinetics:

o Michaelis-Menten Equation: Describes the rate of enzymatic reactions where


an enzyme binds to a substrate to form an enzyme-substrate complex, which
then forms product PPP and releases the enzyme back into its free form.

Figure 5: Enzyme kinetics

Maximum velocity of the reaction, reached when all enzyme active sites are saturated with
substrate. Km, Michaelis constant, which represents the substrate concentration at which the
reaction rate is half of VmaxV. Turnover number, representing the maximum number of
substrate molecules converted to product per enzyme molecule per unit time when the
enzyme is fully saturated with substrate.

2. Lineweaver-Burk Plot: A graphical representation of the Michaelis-Menten equation,


which linearizes the data to determine Vmax and KmK

Page 44 of 208
Figure 6: Enzyme Inhibition

3. Enzyme Inhibition:

 Competitive Inhibition: Inhibitor competes with the substrate for binding to the
enzyme's active site.
 Non-competitive Inhibition: Inhibitor binds to the enzyme at a site other than the
active site, altering enzyme conformation and reducing catalytic activity.
 Mixed Inhibition: Inhibitor can bind to both the enzyme-substrate complex and the
free enzyme, affecting both KmK and Vmax.
 Uncompetitive Inhibition: Inhibitor binds only to the enzyme-substrate complex,
reducing VmaxV.

4. Enzyme Regulation:

o Allosteric Regulation: Regulation of enzyme activity by molecules binding to


allosteric sites, causing a conformational change.

o Covalent Modification: Addition or removal of functional groups (e.g.,


phosphorylation) that change enzyme activity.

o Enzyme Induction and Repression: Regulation of enzyme synthesis in


response to environmental conditions or substrate availability.

Page 45 of 208
5. Enzyme Assays: Experimental methods to measure enzyme activity often based on
changes in substrate or product concentration, or changes in absorbance,
fluorescence, or other properties.

Understanding enzyme kinetics is crucial in fields such as biochemistry, pharmacology, and


medicine, as it helps elucidate how enzymes function and how they can be targeted for
therapeutic purposes or industrial applications. If you have specific questions or want to delve
deeper into any of these topics, feel free to ask!

4.4. Factors affecting rate of enzyme catalyzed reaction

Factors affecting the rate of enzyme-catalyzed reactions can be diverse and depend on the
specific enzyme and reaction involved. Here are the key factors commonly influencing
enzyme kinetics:

1. Substrate Concentration:
o Effect: Generally, an increase in substrate concentration initially increases the
reaction rate because more substrate molecules are available to bind with
enzymes.
o Saturation: At high substrate concentrations, enzyme active sites may become
saturated (all enzymes are occupied), and the reaction rate levels off
(approaches Vmax).
2. Enzyme Concentration:
o Effect: Increasing enzyme concentration typically increases the reaction rate,
assuming substrate concentration is not limiting.
o Linear Relationship: Reaction rate is directly proportional to enzyme
concentration up to a point where substrate availability becomes limiting.
3. Temperature:
Page 46 of 208
o Effect: Enzyme-catalyzed reactions generally increase in rate with temperature
due to greater kinetic energy of molecules.
o Optimal Temperature: Each enzyme has an optimal temperature at which it
catalyzes reactions most effectively. Above this temperature, enzyme
denaturation can occur, decreasing activity.
4. pH:
o Effect: pH affects the ionization state of amino acid side chains in the enzyme
active site, influencing enzyme activity.
o Optimal pH: Enzymes have an optimal pH at which they function best.
Deviations from this pH can reduce enzyme activity due to altered enzyme
structure or charge interactions.
5. Co-factors and Co-enzymes:
o Effect: Many enzymes require non-protein molecules (cofactors or
coenzymes) to function properly.
o Essential Roles: Cofactors may assist in substrate binding, catalytic activity,
or serve as carriers of functional groups during reactions.
6. Inhibitors:
o Effect: Inhibitors can decrease enzyme activity by binding to the enzyme and
preventing substrate binding or catalytic activity.
o Types: Inhibitors can be competitive (compete with substrate for active site),
non-competitive (bind to enzyme at a site other than the active site), or
uncompetitive (bind only to enzyme-substrate complex).
7. Activators:
o Effect: Activators can increase enzyme activity by binding to the enzyme and
promoting substrate binding or catalytic activity.
o Regulation: Often involved in metabolic regulation, activators can enhance
enzyme function in response to cellular needs.
Page 47 of 208
8. Enzyme Isoforms and Variants:
o Effect: Different isoforms or variants of enzymes may have altered kinetic
properties (e.g., substrate specificity, pH optimum) depending on cellular or
environmental conditions.

Figure 7: Factors affecting enzyme activities

Page 48 of 208
4.5. Regulation of enzyme catalyzed reactions

Regulation of enzyme-catalyzed reactions is essential for maintaining metabolic balance and


responding to changing cellular conditions. Enzyme activity can be regulated in several ways,
allowing organisms to control when and how metabolic pathways operate. Here are the key
mechanisms of enzyme regulation:

1. Allosteric Regulation:
o Definition: Allosteric regulation occurs when a molecule binds to an enzyme
at a site other than the active site (allosteric site), altering the enzyme's activity.
o Effect: Allosteric regulators can be either activators or inhibitors, changing
enzyme conformation to enhance or reduce substrate binding and catalytic
efficiency.
o Examples: Hemoglobin (allosteric enzyme) binds oxygen in the lungs
(activator) and releases it in tissues (inhibitor), regulating oxygen transport.
2. Covalent Modification:
o Definition: Enzyme activity can be modified through covalent attachment or
removal of functional groups, such as phosphorylation or dephosphorylation.
o Effect: Phosphorylation by kinases often activates enzymes, while
dephosphorylation by phosphatases deactivates them, influencing metabolic
pathways.
o Examples: Glycogen phosphorylase is activated by phosphorylation for
glycogen breakdown in response to low glucose levels in cells.
3. Substrate Availability:
o Definition: Enzyme activity can be regulated by substrate availability, which is
influenced by cellular concentrations of substrates and products.

Page 49 of 208
o Effect: High substrate concentrations typically increase enzyme activity until
saturation is reached, while product accumulation may inhibit enzyme activity
(feedback inhibition).
o Examples: ATP is a key regulator of many metabolic pathways, inhibiting
enzymes involved in its biosynthesis when ATP levels are sufficient.
4. Gene Expression and Synthesis:
o Definition: Enzyme activity can be regulated by controlling the synthesis or
degradation of enzymes through gene expression.
o Effect: Cells can adjust enzyme levels in response to environmental stimuli or
metabolic demands, ensuring enzymes are present when needed.
o Examples: Induction or repression of enzyme synthesis in response to nutrient
availability or hormonal signals.
5. Competitive Inhibition:
o Definition: Competitive inhibitors resemble the substrate and compete for
binding at the enzyme's active site.
o Effect: They reduce enzyme activity by preventing substrate binding, often
reversible by increasing substrate concentration.
o Examples: Drugs that mimic natural substrates can competitively inhibit
enzymes, such as statins inhibiting HMG-CoA reductase in cholesterol
biosynthesis.
6. Non-competitive Inhibition:
o Definition: Non-competitive inhibitors bind to the enzyme at a site other than
the active site (allosteric site), altering enzyme conformation and reducing
activity.
o Effect: Unlike competitive inhibitors, non-competitive inhibitors do not compete
with substrates and cannot be overcome by increasing substrate concentration.

Page 50 of 208
o Examples: Heavy metals like mercury can non-competitively inhibit enzymes
by binding to sulfhydryl groups, altering enzyme structure and function.
7. Feedback Inhibition:
o Definition: Feedback inhibition occurs when the end product of a metabolic
pathway inhibits an enzyme earlier in the pathway.
o Effect: It prevents excess accumulation of products by shutting down the
pathway when the end product is abundant.
o Examples: ATP inhibits phosphofructokinase in glycolysis, slowing glucose
breakdown when ATP levels are high.

Understanding these regulatory mechanisms is crucial for maintaining metabolic homeostasis


and responding to environmental changes in living organisms. Enzyme regulation ensures
that metabolic pathways operate efficiently and adaptively to support cellular functions and
survival.

Page 51 of 208
5. Carbohydrates

Carbohydrates are essential biomolecules that serve multiple roles in living organisms,
primarily as a source of energy and structural components. Here is an overview covering their
structure, functions, and significance:

Structure of Carbohydrates

1. Monosaccharides:

o Definition: Single sugar units that cannot be further hydrolyzed into simpler
sugars.

o Examples: Glucose, fructose, and galactose.

o Structure: Typically have a backbone of carbon atoms, with hydroxyl groups


(-OH) attached to most carbons and either an aldehyde group (-CHO) or a keto
group (>C=O).

2. Disaccharides:

o Definition: Formed by the condensation of two monosaccharides with the


elimination of a water molecule.

o Examples: Sucrose (glucose + fructose), lactose (glucose + galactose), and


maltose (glucose + glucose).

3. Oligosaccharides:

o Definition: Short chains of monosaccharides (usually 3-10 units) linked by


glycosidic bonds.

o Function: Can serve as recognition signals on cell surfaces or participate in


cell-cell interactions.

Page 52 of 208
4. Polysaccharides:

o Definition: Complex carbohydrates composed of many monosaccharide units


linked together.

o Examples: Starch (energy storage in plants), glycogen (energy storage in


animals), and cellulose (structural component in plants).

Functions of Carbohydrates

1. Energy Source:

o Primary Role: Glucose and other sugars are broken down via cellular
respiration to generate ATP, the primary energy currency of cells.

2. Energy Storage:

o Starch: Plants store excess glucose as starch in roots, seeds, and tubers.

o Glycogen: Animals store glucose as glycogen in liver and muscle cells for rapid
energy release.

3. Structural Support:

o Cellulose: Forms the structural component of plant cell walls, providing rigidity
and support.

o Chitin: Found in the exoskeleton of arthropods and the cell walls of fungi,
providing structural support.

4. Cellular Communication:

o Glycoproteins and Glycolipids: Carbohydrates attached to proteins


(glycoproteins) or lipids (glycolipids) on cell membranes serve as recognition
sites and play roles in cell signaling and immune response.

Page 53 of 208
5. Dietary Fiber:

o Function: Indigestible polysaccharides (e.g., cellulose, pectins) in plant cell


walls that promote digestive health and regular bowel movements.

Significance of Carbohydrates

1. Nutritional Importance:

o Energy: Carbohydrates provide the main source of energy for human


metabolism.

o Fiber: Essential for maintaining digestive health and reducing the risk of
chronic diseases like heart disease and diabetes.

2. Biotechnological Applications:

o Biofuels: Sugars derived from carbohydrates can be fermented into biofuels


like ethanol.

o Food Production: Used as sweeteners, thickeners, and stabilizers in food


processing.

3. Medicinal and Pharmaceutical Uses:

o Vaccines: Carbohydrate antigens are used in vaccines to stimulate immune


responses.

o Drug Delivery: Glycosylated drugs and drug carriers are employed in targeted
drug delivery systems.

In summary, carbohydrates are vital molecules with diverse roles in biology, serving as energy
sources, structural components, and mediators of cellular interactions. Their study is
fundamental to understanding metabolism, nutrition, and various applications in
biotechnology and medicine.

Page 54 of 208
5.1. Monosaccharides

Monosaccharides are the simplest form of carbohydrates, consisting of single sugar units that
cannot be hydrolyzed into smaller sugars. They are classified based on the number of carbon
atoms they contain (triose, tetrose, pentose, hexose, etc.) and the functional groups they
carry.

Structure

1. General Formula: (CH2O)n, where n ranges from 3 to 7.

o Each carbon atom, except one, is attached to a hydroxyl group (-OH).

o One carbon is part of either an aldehyde group (-CHO), making it an aldose, or


a ketone group (>C=O), making it a ketose.

2. Examples:

o Trioses: Simplest monosaccharides with three carbon atoms.

 Glyceraldehyde: An aldose.

 Dihydroxyacetone: A ketose.

o Pentoses: Monosaccharides with five carbon atoms.

 Ribose: Found in RNA.

 Deoxyribose: Found in DNA.

o Hexoses: Monosaccharides with six carbon atoms.

 Glucose: A vital energy source in cellular respiration.

 Fructose: Found in fruits and honey.

o Heptoses: Monosaccharides with seven carbon atoms.

Page 55 of 208
 Sedoheptulose: Involved in the pentose phosphate pathway.

Functions

1. Energy Source: Monosaccharides like glucose are essential for cellular respiration,
providing ATP, the main energy currency of cells.

2. Structural Role: Monosaccharides contribute to the structure of more complex


carbohydrates like disaccharides, polysaccharides, and glycoproteins.

3. Metabolic Intermediates: Some monosaccharides, such as glucose, participate in


metabolic pathways beyond energy production, including the synthesis of amino acids
and nucleotides.

Biological Importance

1. Cellular Energy: Glucose is a primary energy source for cellular functions, from basic
metabolism to complex processes like muscle contraction and nerve impulse
transmission.

2. Metabolism Regulation: Blood sugar levels, regulated by monosaccharides, are


critical for maintaining homeostasis and overall health.

3. Health Impact: Dietary sugars, including monosaccharides, affect metabolic health,


insulin sensitivity, and obesity risk.

Applications

1. Food Industry: Monosaccharides like glucose and fructose are used as sweeteners
and preservatives in food products.

2. Medicine: Glucose is commonly used in intravenous solutions to replenish energy in


patients.

Page 56 of 208
3. Research: Understanding monosaccharide metabolism is crucial for studying
diseases related to carbohydrate metabolism disorders, such as diabetes mellitus.

Monosaccharides play essential roles in both biological processes and industrial applications,
making them fundamental to the study of biochemistry and nutrition.

5.2. Oligosaccharides

Oligosaccharides

Oligosaccharides are carbohydrates composed of a few (typically 3 to 10) monosaccharide


units linked together by glycosidic bonds. They are intermediate in size between
monosaccharides and polysaccharides.

Structure

1. Composition: Composed of 3 to 10 monosaccharide units joined by glycosidic bonds.

2. Types of Linkages: Glycosidic bonds can be of various types:

o Alpha (α) or Beta (β) configurations depending on the anomeric carbon


linkage.

o 1-4, 1-6, etc., indicating the positions of glycosidic bonds between


monosaccharide units.

3. Examples:

o Disaccharides: Simplest form of oligosaccharides consisting of two


monosaccharide units.

 Sucrose: Glucose + Fructose (α-1,2-glycosidic bond).

 Lactose: Galactose + Glucose (β-1,4-glycosidic bond).

Page 57 of 208
o Trisaccharides: Consist of three monosaccharide units.

 Raffinose: Galactose + Glucose + Fructose (α-1,6-glycosidic bond).

o Tetrasaccharides and Pentasaccharides: Four and five monosaccharide


units, respectively.

Functions

1. Prebiotic Effects: Oligosaccharides serve as prebiotics, promoting the growth of


beneficial gut bacteria.

2. Digestive Health: They aid in digestion and absorption of nutrients.

3. Cell Recognition: Oligosaccharides attached to proteins and lipids (glycoproteins,


glycolipids) play crucial roles in cell-cell recognition and signaling.

Biological Importance

1. Nutritional Value: Oligosaccharides like inulin and fructooligosaccharides (FOS) are


dietary fibers with potential health benefits, including improved digestion and gut
health.

2. Medical Applications: Oligosaccharides are used in infant formulas to mimic human


milk oligosaccharides, which support the development of a healthy gut microbiome in
infants.

3. Industrial Uses: Oligosaccharides are utilized in food products as sweeteners,


thickeners, and stabilizers.

Applications

1. Food Industry: Oligosaccharides are added to food products as dietary fibers,


sweeteners (e.g., maltodextrins), and health supplements.

Page 58 of 208
2. Pharmaceuticals: They are used in pharmaceutical formulations for their role in drug
delivery systems and as excipients.

3. Research: Studying oligosaccharides helps in understanding biological processes like


cell signaling, immune response modulation, and pathogen recognition.

Oligosaccharides play diverse roles in nutrition, health, and industry, making them significant
components in both biological systems and commercial applications.

5.2. Polysaccharides

Oligosaccharides

Oligosaccharides are carbohydrates composed of a few (typically 3 to 10) monosaccharide


units linked together by glycosidic bonds. They are intermediate in size between
monosaccharides and polysaccharides.

Structure

1. Composition: Composed of 3 to 10 monosaccharide units joined by glycosidic bonds.


2. Types of Linkages: Glycosidic bonds can be of various types:
o Alpha (α) or Beta (β) configurations depending on the anomeric carbon
linkage.
o 1-4, 1-6, etc., indicating the positions of glycosidic bonds between
monosaccharide units.
3. Examples:
o Disaccharides: Simplest form of oligosaccharides consisting of two
monosaccharide units.
 Sucrose: Glucose + Fructose (α-1,2-glycosidic bond).

Page 59 of 208
 Lactose: Galactose + Glucose (β-1,4-glycosidic bond).
o Trisaccharides: Consist of three monosaccharide units.
 Raffinose: Galactose + Glucose + Fructose (α-1,6-glycosidic bond).
o Tetrasaccharides and Pentasaccharides: Four and five monosaccharide
units, respectively.

Functions

1. Prebiotic Effects: Oligosaccharides serve as prebiotics, promoting the growth of


beneficial gut bacteria.
2. Digestive Health: They aid in digestion and absorption of nutrients.
3. Cell Recognition: Oligosaccharides attached to proteins and lipids (glycoproteins,
glycolipids) play crucial roles in cell-cell recognition and signaling.

Biological Importance

1. Nutritional Value: Oligosaccharides like inulin and fructooligosaccharides (FOS) are


dietary fibers with potential health benefits, including improved digestion and gut
health.
2. Medical Applications: Oligosaccharides are used in infant formulas to mimic human
milk oligosaccharides, which support the development of a healthy gut microbiome in
infants.
3. Industrial Uses: Oligosaccharides are utilized in food products as sweeteners,
thickeners, and stabilizers.

Page 60 of 208
Applications

1. Food Industry: Oligosaccharides are added to food products as dietary fibers,


sweeteners (e.g., maltodextrins), and health supplements.
2. Pharmaceuticals: They are used in pharmaceutical formulations for their role in drug
delivery systems and as excipients.
3. Research: Studying oligosaccharides helps in understanding biological processes like
cell signaling, immune response modulation, and pathogen recognition.

Oligosaccharides play diverse roles in nutrition, health, and industry, making them significant
components in both biological systems and commercial applications.

5.3. Polysaccharides

Polysaccharides

Polysaccharides are complex carbohydrates composed of many monosaccharide units linked


together by glycosidic bonds. They can be linear or branched and serve various structural
and storage functions in organisms.

Structure

1. Composition: Composed of hundreds to thousands of monosaccharide units linked


by glycosidic bonds.
2. Types of Linkages: Glycosidic bonds can vary:
o Alpha (α) or Beta (β) configurations.
o 1-4, 1-6, etc., indicating the positions of glycosidic bonds between
monosaccharide units.
3. Examples:
o Starch: A polysaccharide composed of glucose units.
Page 61 of 208
 Amylose: Linear chain of glucose units linked by α-1,4-glycosidic
bonds.
 Amylopectin: Branched chain of glucose units with α-1,4-glycosidic
bonds and occasional α-1,6-glycosidic bonds at branch points.
o Glycogen: Similar to starch but more highly branched, serving as an energy
storage molecule in animals and fungi.
o Cellulose: Structural polysaccharide in plant cell walls, composed of β-glucose
units linked by β-1,4-glycosidic bonds.
o Chitin: Found in the exoskeletons of arthropods and cell walls of fungi,
composed of N-acetylglucosamine units linked by β-1,4-glycosidic bonds.

Functions

1. Energy Storage: Polysaccharides like starch and glycogen serve as energy storage
molecules in plants and animals, respectively.
2. Structural Support: Polysaccharides like cellulose and chitin provide structural
support to plant cell walls and arthropod exoskeletons, respectively.
3. Cellular Recognition: Polysaccharides attached to proteins and lipids (glycoproteins,
glycolipids) play roles in cell-cell recognition and immune response.

Biological Importance

1. Nutritional Value: Polysaccharides like starch are major dietary sources of


carbohydrates, providing energy.
2. Industrial Applications: Polysaccharides are used in various industries for their
thickening, stabilizing, and gelling properties. For example, agar from seaweed is used
in microbiological culture media.

Page 62 of 208
3. Medical Uses: Polysaccharides are used in pharmaceutical formulations, such as in
drug delivery systems and wound dressings.

Applications

1. Food Industry: Polysaccharides are used as thickeners, stabilizers, and texture


modifiers in food products.
2. Biotechnology: Polysaccharides are studied for their potential in biomaterials,
biodegradable plastics, and as components of biofuels.
3. Research: Understanding polysaccharide structure and function is crucial for fields
like biochemistry, microbiology, and biotechnology.

Polysaccharides are essential components of biological systems, contributing to energy


storage, structural integrity, and cellular interactions across a wide range of organisms and
industrial applications.

5.4. Glycoconjugates

Glycoconjugates are molecules composed of a carbohydrate (glycan) covalently linked to a


non-carbohydrate entity, such as a protein, lipid, or another carbohydrate. These molecules
are crucial in various biological processes, including cellular recognition, signaling, and
immunity. Here is an overview:

Glycoconjugates

Types of Glycoconjugates

1. Glycoproteins:

o Definition: Proteins covalently attached to carbohydrates (glycans).

Page 63 of 208
o Structure: Carbohydrate chains are attached to the protein via N-linked or O-
linked glycosylation.

o Function: Play roles in protein folding, stability, cell-cell recognition, and


immune response.

2. Glycolipids:

o Definition: Lipids (such as glycerolipids or sphingolipids) covalently linked to


carbohydrates.

o Structure: Carbohydrates are typically attached to the lipid portion of the


molecule.

o Function: Found in cell membranes, involved in cell signaling, adhesion, and


recognition.

3. Proteoglycans:

o Definition: Proteins with one or more covalently attached glycosaminoglycan


(GAG) chains.

o Structure: Core protein with GAG chains (e.g., chondroitin sulfate, heparan
sulfate) attached via a linker region.

o Function: Provide structural support in tissues (e.g., cartilage), regulate cellular


processes, and interact with growth factors.

4. Glycosylphosphatidylinositols (GPIs):

o Definition: Glycolipids anchored to the cell membrane via a phosphatidylinositol


(PI) moiety.

o Structure: Carbohydrate chain attached to the inositol ring of the GPI anchor.

Page 64 of 208
o Function: Anchors proteins to the cell membrane, involved in signal
transduction and immune response.

Functions of Glycoconjugates

1. Cellular Recognition:

o Glycoconjugates on cell surfaces serve as recognition markers for immune


cells, pathogens, and other cells.

2. Signaling:

o Glycoproteins and glycolipids participate in cell signaling pathways, influencing


processes like growth, differentiation, and apoptosis.

3. Structural Support:

o Proteoglycans provide structural integrity to tissues like cartilage and


extracellular matrix through their interactions with collagen and other proteins.

4. Immune Response:

o Glycoconjugates on cell surfaces and secreted glycoproteins (e.g., antibodies)


play crucial roles in immune recognition and response.

5. Cell Adhesion:

o Glycoconjugates facilitate cell-cell and cell-extracellular matrix adhesion,


essential for tissue organization and wound healing.

Importance in Biology

 Biological Recognition: Glycoconjugates mediate interactions between cells and their


environment, influencing development, immunity, and disease progression.

Page 65 of 208
 Therapeutic Applications: Understanding glycoconjugate structures is crucial for
developing vaccines, diagnostic tools, and therapies targeting carbohydrate-based
interactions.

 Biotechnological Uses: Glycoengineering techniques are employed to modify


glycoconjugates for therapeutic and industrial applications, such as in drug delivery
systems.

In summary, glycoconjugates represent a diverse group of molecules essential for cellular


function, structural integrity, and intercellular communication. Their complex structures and
functions make them vital components in biological systems and targets for biomedical
research and applications.

Page 66 of 208
6. Lipids

Lipids are a diverse group of biomolecules that include fats, oils, waxes, phospholipids, and
steroids. They play essential roles in cellular structure, energy storage, signaling, and
insulation. Here is an overview of lipids:

Lipids

Classification of Lipids

1. Fatty Acids and Glycerolipids:

o Fatty Acids: Long hydrocarbon chains with a carboxyl group (COOH) at one
end. They can be saturated (no double bonds) or unsaturated (one or more
double bonds).

o Glycerolipids: Esters of fatty acids with glycerol. They include triglycerides


(storage lipids) and phospholipids (membrane lipids).

2. Phospholipids:

o Structure: Consist of a glycerol backbone linked to two fatty acids and a


phosphate group. They have hydrophobic tails (fatty acids) and a hydrophilic
head (phosphate group), making them amphipathic.

o Function: Main components of cell membranes, forming lipid bilayers with


hydrophilic heads facing outward and hydrophobic tails facing inward.

3. Steroids:

o Examples: Cholesterol, hormones like testosterone and estrogen.

o Structure: Four fused carbon rings with various functional groups attached.

Page 67 of 208
o Function: Structural components of cell membranes (cholesterol), and
regulatory molecules (hormones).

4. Waxes:

o Structure: Esters of long-chain fatty acids with long-chain alcohols.

o Function: Protective coatings on plant surfaces (cuticle) and animal fur,


providing water resistance and preventing dehydration.

Functions of Lipids

1. Energy Storage: Triglycerides store energy in adipose tissue, providing a concentrated


source of metabolic fuel.

2. Structural Components: Phospholipids and cholesterol maintain cell membrane


integrity and fluidity.

3. Cell Signaling: Lipid-derived signaling molecules (e.g., prostaglandins) regulate


physiological processes such as inflammation and blood clotting.

4. Insulation: Lipids in adipose tissue serve as thermal insulation and cushioning for
organs.

5. Hormone Production: Steroid hormones are synthesized from cholesterol and regulate
various physiological functions.

Importance in Biology

 Cell Membrane Function: Lipids form the basic structure of cell membranes, regulating
the passage of molecules and ions into and out of cells.

 Metabolic Regulation: Lipids play roles in metabolic pathways, including energy


production (through beta-oxidation of fatty acids) and storage.

Page 68 of 208
 Health Implications: Imbalances in lipid metabolism can lead to metabolic disorders
(e.g., obesity, diabetes) and cardiovascular diseases.

 Industrial Applications: Lipids are used in various industries; including food (cooking
oils, margarine), cosmetics (emollients), and pharmaceuticals (drug delivery systems).

6.1. Storage lipids

Storage lipids, primarily in the form of triglycerides, serve as a vital energy reserve in
organisms. Here is an overview of storage lipids:

Storage Lipids

Structure

 Composition: Storage lipids are predominantly triglycerides (triacylglycerols or


TAGs), consisting of three fatty acids esterified to a glycerol molecule.

 Fatty Acids: The fatty acids in triglycerides can vary in chain length and degree of
saturation, influencing their physical properties and energy density.

 Hydrophobic Nature: Triglycerides are hydrophobic molecules, making them highly


efficient for energy storage because they are compact and insoluble in water.

Function

1. Energy Storage: Triglycerides serve as the main energy reservoir in organisms. They
store approximately twice as much energy per gram as carbohydrates due to their
higher carbon-to-oxygen ratio.

2. Metabolic Fuel: During periods of energy demand (e.g., fasting, exercise),


triglycerides are broken down into fatty acids and glycerol through lipolysis. Fatty acids
are then oxidized in mitochondria to generate ATP, providing sustained energy for
cellular processes.

Page 69 of 208
3. Insulation and Protection: Adipose tissue, where triglycerides are stored, acts as a
thermal insulator and cushion around organs, protecting them from physical impact
and temperature fluctuations.

4. Regulation of Metabolism: Storage lipids play roles in hormone regulation and


signaling pathways related to energy balance. For example, adipose tissue secretes
hormones like leptin and adiponectin, which influence appetite and metabolism.

Importance in Biology

 Survival Adaptations: Triglycerides allow organisms to survive prolonged periods


without food by providing a concentrated source of metabolic fuel.

 Health Implications: Imbalances in storage lipid metabolism contribute to obesity,


diabetes, and cardiovascular diseases. Understanding lipid metabolism is crucial for
managing these health conditions.

 Industrial Applications: Triglycerides from plant and animal sources are used in food
production (cooking oils, margarine), biofuels, and pharmaceutical formulations (drug
carriers).

Research and Development

 Nutritional Sciences: Studying triglyceride metabolism informs dietary


recommendations and nutritional interventions for managing weight and metabolic
disorders.

 Medical Research: Investigating lipid metabolism provides insights into obesity-


related diseases and potential therapeutic targets.

In summary, storage lipids, particularly triglycerides, are essential for energy storage,
metabolic regulation, and physiological protection in organisms. Their efficient storage and

Page 70 of 208
release of energy play critical roles in maintaining cellular function and supporting survival
during times of nutrient scarcity.

6.2. Lipids as signal, cofactors and pigments

Lipids play diverse roles beyond energy storage in biological systems, serving as signals,
cofactors, and pigments. Here is an overview of these functions:

Lipids as Signals, Cofactors, and Pigments

1. Lipids as Signaling Molecules

 Steroid Hormones: Lipids such as cholesterol and its derivatives (e.g., cortisol,
estrogen, and testosterone) act as steroid hormones. These hormones regulate
various physiological processes including metabolism, growth, reproduction, and
stress responses.

 Prostaglandins and Eicosanoids: Derived from arachidonic acid, these lipids


function as local hormones (autocrine and paracrine signals) involved in inflammation,
blood clotting, and immune responses.

 Lipid Mediators: Lipids like sphingosine-1-phosphate (S1P) and ceramides act as


signaling molecules in cell proliferation, apoptosis (programmed cell death), and
immune responses.

2. Lipids as Cofactors

 Coenzyme Q (Ubiquinone): A lipid-derived molecule involved in electron transport in


mitochondria, crucial for ATP production in cellular respiration.

 Lipid-Derived Vitamins: Vitamins A, D, E, and K are fat-soluble vitamins derived from


lipids. They serve as cofactors in various enzymatic reactions and play roles in vision

Page 71 of 208
(vitamin A), bone health (vitamin D), antioxidant defense (vitamin E), and blood clotting
(vitamin K).

3. Lipids as Pigments

 Carotenoids: Lipid-soluble pigments found in plants, algae, and some bacteria. They
play roles in photosynthesis (as accessory pigments), provide coloration in fruits and
vegetables (e.g., β-carotene in carrots), and act as antioxidants.

 Chlorophylls: Lipid-containing pigments essential for photosynthesis in plants and


algae. Chlorophyll molecules absorb light energy during photosynthesis, converting it
into chemical energy.

Importance in Biology

 Cell Signaling: Lipid signaling molecules regulate cellular responses to environmental


cues, contributing to processes like growth, differentiation, and immune function.

 Metabolic Regulation: Lipid cofactors participate in enzymatic reactions essential for


energy metabolism, antioxidant defense, and blood clotting.

 Ecological Roles: Lipid pigments (e.g., carotenoids and chlorophylls) influence


organismal coloration, photosynthetic efficiency, and interactions in ecosystems.

Research and Applications

 Biomedical Research: Understanding lipid signaling pathways informs research on


diseases like cancer, inflammation, and metabolic disorders.

 Nutritional Sciences: Studying lipid-derived vitamins helps optimize dietary


recommendations and supplementation strategies.

 Environmental Sciences: Lipid pigments serve as biomarkers for environmental


studies, indicating ecosystem health and responses to climate change.

Page 72 of 208
In summary, lipids function as essential signaling molecules, cofactors in enzymatic reactions,
and pigments crucial for biological processes and ecological interactions. Their roles extend
beyond energy storage, highlighting their diverse contributions to health, metabolism, and
environmental dynamics.

6.3. Working with lipds

Signal transduction pathways involve complex mechanisms that transmit signals from the
extracellular environment to the cell's interior, influencing cellular responses. One crucial
component of these pathways is gated ion channels. Here’s an overview of their role:

Gated Ion Channels in Signal Transduction Pathways

1. Definition and Structure

 Gated Ion Channels: These are transmembrane proteins that span the lipid bilayer of
cells. They allow the selective passage of ions (such as Na+, K+, Ca2+, and Cl-)
across the membrane in response to specific stimuli.

 Types of Gating: Gated ion channels can be gated by various stimuli, including
voltage changes (voltage-gated channels), ligand binding (ligand-gated channels),
mechanical deformation (mechanically gated channels), or changes in temperature
(thermo-gated channels).

2. Role in Signal Transduction

 Neuronal Signaling: In neurons, ligand-gated ion channels (e.g., neurotransmitter


receptors) respond to neurotransmitters released from presynaptic neurons. Binding
of neurotransmitters to these receptors causes conformational changes that open or
close the ion channel, allowing ions to flow across the membrane and generating
electrical signals (action potentials).

Page 73 of 208
 Sensory Perception: Mechanically gated ion channels are critical in sensory cells
(e.g., hair cells in the ear, touch receptors in the skin). Physical stimuli such as sound
waves or touch deform the cell membrane, opening these channels to allow ion influx
or efflux, which triggers nerve impulses.

 Muscle Contraction: Calcium ion channels (Ca2+ channels) play a pivotal role in
muscle contraction. Upon receiving an action potential, voltage-gated Ca2+ channels
open in muscle cells, leading to an influx of calcium ions. Calcium ions then bind to
proteins involved in the contraction process, initiating muscle fiber contraction.

3. Regulation and Modulation

 Second Messengers: second messenger molecules can also regulate gated ion
channels. For instance, G protein-coupled receptors (GPCRs) activate intracellular
signaling cascades that can modulate the activity of ion channels indirectly through
the production of second messengers like cyclic AMP (cAMP) or inositol trisphosphate
(IP3).

 Disease and Drug Targets: Dysregulation of ion channels is implicated in various


diseases, including neurological disorders (e.g., epilepsy, Alzheimer's disease) and
cardiac arrhythmias. Ion channels are significant targets for therapeutic drugs that aim
to modulate ion flow and cellular responses.

Research and Applications

 Biomedical Research: Understanding the structure, function, and regulation of gated


ion channels is crucial for advancing treatments for neurological disorders, sensory
perception disorders, and cardiac diseases.

Page 74 of 208
 Drug Development: Ion channels represent valuable targets for developing
pharmacological agents that can either activate or inhibit specific channels to treat
diseases.

In conclusion, gated ion channels are integral components of signal transduction pathways,
allowing cells to respond dynamically to changes in their environment. Their precise
regulation and modulation are essential for normal physiological functions and are critical
targets for therapeutic interventions in various diseases.

Certainly! Receptors and secondary messengers are key components in signal transduction
pathways, facilitating the transmission of extracellular signals into intracellular responses.
Here’s an overview:

Receptors and Secondary Messengers in Signal Transduction Pathways

1. Receptors

 Definition: Receptors are proteins located on the cell surface or within the cell that
bind specific ligands (e.g., hormones, neurotransmitters, growth factors) from the
extracellular environment.

 Types of Receptors:

o Ligand-Gated Ion Channels: Receptors that, upon binding a ligand, open or


close ion channels in the cell membrane, allowing ion flux (e.g., acetylcholine
receptors).

o G Protein-Coupled Receptors (GPCRs): Receptors that activate intracellular


signal transduction pathways via interaction with G proteins upon ligand
binding (e.g., adrenaline receptors).

Page 75 of 208
o Enzyme-Linked Receptors: Receptors with intrinsic enzymatic activity or
associated with intracellular kinases that become activated upon ligand
binding, initiating phosphorylation cascades (e.g., receptor tyrosine kinases).

2. Secondary Messengers

 Definition: Secondary messengers are small molecules or ions that relay signals from
receptors to intracellular targets, amplifying and diversifying the initial signal.

 Types of Secondary Messengers:

o cAMP (Cyclic Adenosine Monophosphate): Produced from ATP by adenylyl


cyclase upon activation of GPCRs. cAMP activates protein kinase A (PKA),
leading to phosphorylation of downstream targets.

o cGMP (Cyclic Guanosine Monophosphate): Similar to cAMP, produced by


guanylyl cyclase upon receptor activation. It regulates ion channels and protein
kinases, influencing cellular responses.

o IP3 (Inositol Trisphosphate) and DAG (Diacylglycerol): Produced from


phosphatidylinositol bisphosphate (PIP2) by phospholipase C (PLC) upon
activation of certain receptors. IP3 triggers calcium release from intracellular
stores, while DAG activates protein kinase C (PKC).

o Calcium Ions (Ca2+): Regulates various cellular processes, including muscle


contraction, neurotransmitter release, and gene expression. Ca2+ levels are
controlled by channels in the endoplasmic reticulum and plasma membrane.

3. Signal Transduction Pathways

 Activation: Ligand binding to receptors induces conformational changes that activate


intracellular signaling molecules (e.g., G proteins, kinases) or directly alter ion channel
activity.

Page 76 of 208
 Propagation: Secondary messengers amplify and propagate the signal within the cell,
often leading to phosphorylation cascades, changes in gene expression, or alterations
in cellular metabolism.

 Integration: Signals from multiple receptors and secondary messengers are


integrated to coordinate complex cellular responses such as growth, differentiation,
metabolism, and apoptosis.

4. Clinical Relevance

 Drug Targets: Receptors and secondary messengers are targets for pharmaceutical
interventions. Drugs can modulate receptor activity (agonists, antagonists) or interfere
with secondary messenger production or action to treat diseases (e.g., hypertension,
cancer, and diabetes).

 Disease Mechanisms: Dysregulation of receptor signaling and secondary messenger


pathways underlies various diseases, making them critical areas of research and
therapeutic development.

In summary, receptors and secondary messengers play crucial roles in transmitting


extracellular signals into intracellular responses, regulating diverse physiological processes
and serving as key targets for therapeutic interventions in human health and disease.

Page 77 of 208
Certainly! Protein phosphorylation and dephosphorylating are fundamental regulatory
mechanisms in cellular signaling, influencing a wide range of cellular processes. Here’s an
overview:

Protein Phosphorylation and Dephosphorylation

1. Phosphorylation

 Definition: Phosphorylation is the addition of a phosphate group (PO₄³⁻) to a protein


molecule, typically on specific amino acid residues such as serine, threonine, or
tyrosine.

 Catalysis: Catalyzed by enzymes called kinases, which transfer the phosphate group
from ATP (adenosine triphosphate) to the protein substrate.

 Roles:

o Signal Transduction: Phosphorylation acts as a molecular switch, altering


protein conformation, activity, or interaction partners in response to
extracellular signals (e.g., hormones, growth factors).

o Enzyme Regulation: Many enzymes, including kinases themselves, are


regulated by phosphorylation, affecting their activity and substrate specificity.

o Cellular Processes: Regulates processes such as cell growth, differentiation,


metabolism, apoptosis, and response to stress.

 Types:

o Serine/Threonine Kinases: Phosphorylate serine and threonine residues,


regulating various cellular processes including cell cycle progression and
signaling pathways like MAPK (Mitogen-Activated Protein Kinase).

Page 78 of 208
o Tyrosine Kinases: Phosphorylate tyrosine residues on receptor tyrosine
kinases (RTKs) and non-receptor tyrosine kinases, controlling growth factor
signaling, cell adhesion, and differentiation.

2. Dephosphorylation

 Definition: Dephosphorylation is the removal of a phosphate group from a protein,


catalyzed by enzymes called phosphatases.

 Catalysis: Phosphatases hydrolyze the phosphate ester bond, restoring the protein
to its unphosphorylated state.

 Roles:

o Signal Termination: Dephosphorylation reverses the effects of


phosphorylation, deactivating proteins and terminating signal transduction
pathways.

o Enzyme Regulation: Phosphatases regulate the activity of kinases and other


phosphoproteins, maintaining cellular homeostasis.

o Cell Cycle Control: Dephosphorylation events are critical in regulating cell


cycle checkpoints and progression.

 Types:

o Protein Tyrosine Phosphatases (PTPs): Dephosphorylate tyrosine residues,


counteracting the action of tyrosine kinases and regulating RTK signaling.

o Serine/Threonine Phosphatases: Dephosphorylate serine and threonine


residues, playing roles in cell cycle regulation, metabolism, and stress
responses.

Page 79 of 208
3. Regulation and Dynamics

 Dynamic Equilibrium: Phosphorylation and dephosphorylation maintain a dynamic


equilibrium, fine-tuning protein function in response to changing cellular conditions and
signals.

 Spatial and Temporal Regulation: Spatially and temporally regulated by localization


of kinases and phosphatases within specific cellular compartments (e.g., plasma
membrane, nucleus, cytoplasm).

 Disease Implications: Dysregulation of phosphorylation signaling is implicated in


various diseases, including cancer, neurodegenerative disorders, and metabolic
diseases. Targeting kinases and phosphatases is a strategy for therapeutic
intervention.

In summary, protein phosphorylation and dephosphorylation are essential mechanisms for


cellular regulation, controlling a wide array of biological processes and serving as critical
points of intervention in disease treatment and management.

Defects in signaling pathways play a significant role in the development and progression of
cancer. Here’s an overview of how abnormalities in signaling pathways contribute to cancer:

Defects in Signaling Pathways and Cancer

1. Overview of Signaling Pathways in Cancer

 Cell Signaling: Cells communicate and respond to their environment through complex
signaling networks that regulate processes like growth, proliferation, differentiation,
and apoptosis.

Page 80 of 208
 Abnormalities: Genetic mutations, epigenetic changes, or alterations in protein
expression can disrupt signaling pathways, leading to uncontrolled cell growth and
cancer development.

2. Common Signaling Pathways Implicated in Cancer

 RTK/RAS/RAF/MEK/ERK Pathway:

o Function: Regulates cell proliferation, survival, and differentiation in response


to growth factors.

o Abnormalities: Mutations in RTKs (e.g., EGFR), RAS oncogenes (e.g.,


KRAS), or downstream effectors (e.g., BRAF) lead to constitutive activation,
promoting tumor growth (e.g., in colorectal cancer, lung cancer).

 PI3K/AKT/mTOR Pathway:

o Function: Controls cell growth, metabolism, and survival in response to growth


factors and nutrients.

o Abnormalities: Mutations in PI3KCA, loss of PTEN (a phosphatase that


opposes PI3K), or activation of AKT/mTOR pathway promote cell proliferation
and survival (e.g., in breast cancer, prostate cancer).

 Wnt/β-Catenin Pathway:

o Function: Regulates cell fate determination, proliferation, and stem cell


renewal.

o Abnormalities: Mutations in APC, β-catenin (CTNNB1), or Wnt ligands lead to


aberrant activation, contributing to colorectal cancer and other cancers.

Page 81 of 208
 Notch Pathway:

o Function: Mediates cell fate decisions during development and tissue


homeostasis.

o Abnormalities: Dysregulation of Notch signaling due to mutations or altered


expression promotes cancer development (e.g., leukemia, breast cancer).

 Hedgehog Pathway:

o Function: Controls cell proliferation and differentiation during embryonic


development and tissue regeneration.

o Abnormalities: Mutations in components like PTCH1 or SMO result in


constitutive activation, implicated in basal cell carcinoma, medulloblastoma,
and other cancers.

3. Consequences of Signaling Pathway Dysregulation

 Uncontrolled Cell Growth: Constitutive activation of oncogenic pathways promotes


cell cycle progression and inhibits apoptosis, leading to tumor formation and
progression.

 Metastasis: Dysregulated signaling pathways can enhance cell motility, invasiveness,


and resistance to anoikis (detachment-induced cell death), facilitating metastatic
spread.

 Therapeutic Implications: Targeting key signaling nodes (e.g., receptor tyrosine


kinases, downstream effectors) with small molecule inhibitors or monoclonal
antibodies is a strategy for cancer treatment (e.g., EGFR inhibitors in lung cancer,
BRAF inhibitors in melanoma).

Page 82 of 208
4. Clinical Relevance and Targeted Therapies

 Precision Medicine: Understanding specific mutations or alterations in signaling


pathways allows for targeted therapies that selectively inhibit aberrant pathways while
sparing normal cells.

 Resistance Mechanisms: Tumor cells can develop resistance to targeted therapies


through secondary mutations, pathway crosstalk, or activation of compensatory
pathways, necessitating combination therapies or development of new agents.

 Prognostic Markers: Biomarkers related to signaling pathway activation (e.g.,


phosphorylation status of key proteins) serve as prognostic indicators and guide
treatment decisions in clinical practice.

In conclusion, defects in signaling pathways contribute significantly to cancer by promoting


abnormal cell proliferation, survival, and metastasis. Understanding these mechanisms
provides insights into cancer biology and informs the development of targeted therapies
aimed at disrupting oncogenic signaling while minimizing off-target effects.

Page 83 of 208
Signal transduction in microorganisms involves processes by which cells sense and respond
to external stimuli, enabling them to adapt to changing environments. Here is an overview of
signal transduction in microorganisms:

Signal Transduction in Microorganisms

1. Types of Signals

 Environmental Signals: Microorganisms sense and respond to various


environmental cues such as nutrient availability, pH changes, temperature
fluctuations, and presence of toxins or other microorganisms.

 Cell-Cell Communication: Some microorganisms utilize signaling molecules (e.g.,


quorum sensing molecules) to communicate with nearby cells and coordinate group
behaviors like biofilm formation or virulence factor production.

2. Signal Reception

 Receptors: Microorganisms possess receptors on their cell surfaces or within their


cytoplasm that recognize specific signal molecules. These receptors can be:

o Transmembrane Proteins: Embedded in the cell membrane and capable of


sensing extracellular signals.

o Intracellular Sensors: Found in the cytoplasm and involved in detecting


internal signals such as changes in metabolite levels.

3. Signal Transduction Pathways

 Transduction Mechanisms: Once a signal is received, it is transmitted through a


series of biochemical reactions that amplify and relay the signal to effector molecules,
resulting in cellular responses.

 Common Pathways:

Page 84 of 208
o Two-Component Systems: Found in bacteria and some fungi, consisting of a
sensor histidine kinase that autophosphorylates in response to a specific
signal, and a response regulator protein that modulates gene expression or
enzyme activity.

o cAMP Signaling: In bacteria and lower eukaryotes like yeast, cyclic AMP
(cAMP) acts as a second messenger to regulate metabolic pathways and
responses to nutrient availability.

o Quorum Sensing: Used by bacteria to coordinate gene expression in


response to population density, involving the production and detection of small
signaling molecules (autoinducers).

4. Cellular Responses

 Gene Regulation: Signal transduction pathways often lead to changes in gene


expression, altering the synthesis of proteins and enzymes that allow microorganisms
to adapt to their environment.

 Metabolic Pathways: Responses may include adjustments in metabolic pathways to


optimize energy production or utilization of available nutrients.

 Virulence and Biofilm Formation: Some signal transduction pathways regulate


virulence factor production or biofilm formation, key for microbial pathogenesis and
survival in host environments.

5. Adaptation and Evolutionary Significance

 Adaptive Advantage: Efficient signal transduction systems confer a selective


advantage by allowing microorganisms to rapidly respond and adapt to changing
environmental conditions, enhancing survival and competitive fitness.

Page 85 of 208
 Evolutionary Conservation: Many signaling pathways are conserved across different
microbial species, reflecting their essential roles in microbial physiology and
adaptation strategies.

6. Application in Biotechnology and Medicine

 Biotechnological Applications: Understanding microbial signal transduction


pathways enables manipulation of microorganisms for biotechnological processes
such as industrial fermentation, bioremediation, and production of bioactive
compounds.

 Medical Relevance: Targeting microbial signaling pathways offers potential strategies


for developing novel antimicrobial agents or therapies that disrupt pathogenicity and
virulence mechanisms in infectious diseases.

In summary, signal transduction pathways in microorganisms are essential for sensing and
responding to environmental cues, facilitating adaptation, survival, and interactions within
complex microbial communities. Understanding these pathways provides insights into
microbial physiology, evolution, and potential applications in various fields.

Page 86 of 208
7. Central metabolic pathways and energy transduction

Central metabolic pathways and energy transduction refer to the fundamental biochemical
processes that organisms use to generate energy from nutrients and to synthesize essential
biomolecules. These pathways are crucial for sustaining life and maintaining cellular
functions. Here is an overview of these important processes:

Central Metabolic Pathways

1. Glycolysis

 Description: Glycolysis is the metabolic pathway that converts glucose into pyruvate,
generating ATP and NADH in the process.
 Steps: It involves a series of enzymatic reactions, starting with glucose and ending
with the production of two molecules of pyruvate, two ATP molecules (net gain), and
two NADH molecules.
 Importance: Glycolysis is a central pathway in both aerobic and anaerobic
metabolism, providing precursor molecules for other pathways and energy in the form
of ATP.

2. Citric Acid Cycle (Krebs Cycle)

 Description: The citric acid cycle is a series of chemical reactions used by all aerobic
organisms to generate energy through the oxidation of acetyl-CoA derived from
carbohydrates, fats, and proteins.
 Steps: Acetyl-CoA enters the cycle, where it undergoes a series of redox reactions
that release CO2 and generate NADH, FADH2, and GTP (which can be converted to
ATP).

Page 87 of 208
 Importance: The cycle not only produces ATP but also supplies intermediates for
biosynthesis and serves as a hub for integrating metabolic pathways.

3. Oxidative Phosphorylation

 Description: Oxidative phosphorylation is the process in which ATP is formed as a


result of the transfer of electrons from NADH or FADH2 to oxygen by a series of
electron carriers in the inner mitochondrial membrane (or plasma membrane in
prokaryotes).
 Steps: Electron transport chain (ETC) complexes pump protons across the
membrane, creating a proton gradient. ATP synthase then uses the energy from the
proton gradient to synthesize ATP from ADP and Pi.
 Importance: This is the primary mechanism for ATP synthesis in aerobic organisms,
coupling electron transport with ATP production.

Energy Transduction

1. ATP

 Role: Adenosine triphosphate (ATP) is the universal energy carrier in cells, providing
energy for metabolic reactions, biosynthesis, movement, and cellular processes.
 Formation: ATP is synthesized primarily through oxidative phosphorylation in
mitochondria (or through substrate-level phosphorylation in glycolysis and the citric
acid cycle).

2. Energy Currency

 Function: ATP hydrolysis releases energy that is used to drive endergonic reactions
(energy consuming) in the cell.

Page 88 of 208
 Regulation: Cellular ATP levels are tightly regulated to meet the energy demands of
the cell, maintaining homeostasis and responding to changes in metabolic needs.

3. Intermediary Metabolites

 Role: Besides ATP, central metabolic pathways produce intermediates (e.g., NADH,
FADH2, acetyl-CoA, oxaloacetate) that participate in other metabolic processes like
amino acid biosynthesis, lipid metabolism, and nucleotide synthesis.
 Integration: These intermediates serve as building blocks for cellular components and
play critical roles in coordinating metabolic fluxes and responding to cellular demands.

Conclusion

Central metabolic pathways and energy transduction are essential for converting nutrients
into usable energy (in the form of ATP) and metabolic intermediates necessary for cell growth,
maintenance, and function. These processes are highly conserved across all forms of life and
are crucial for understanding cellular physiology, disease mechanisms, and biotechnological
applications.

Page 89 of 208
7.1. Bioenergetics

Bioenergetics is the study of how living organisms acquire, convert, and utilize energy to
sustain life processes. It encompasses the biochemical processes and mechanisms involved
in energy capture, storage, and transformation within cells. Here's an overview of key
concepts in bioenergetics:

Key Concepts in Bioenergetics

1. Energy Acquisition

 Photosynthesis: In plants, algae, and some bacteria, photosynthesis captures light


energy and converts it into chemical energy in the form of ATP and NADPH, which are
used to synthesize glucose and other organic molecules.

 Chemotrophic Energy Acquisition: Organisms, including humans, acquire energy


by breaking down organic molecules (carbohydrates, fats, proteins) through processes
like glycolysis, the citric acid cycle, and oxidative phosphorylation.

2. Energy Storage and Transformation

 ATP: Adenosine triphosphate (ATP) is the primary energy carrier molecule in cells. It
stores energy in its phosphate bonds, which can be hydrolyzed to release energy for
cellular processes.

 Redox Reactions: Cellular respiration involves redox reactions where electrons are
transferred from donor molecules (e.g., NADH, FADH2) to acceptor molecules (e.g.,
oxygen), releasing energy that is used to synthesize ATP.

Page 90 of 208
3. Metabolic Pathways

 Glycolysis: The breakdown of glucose into pyruvate, producing ATP and NADH.

 Citric Acid Cycle: Also known as the Krebs cycle, it oxidizes acetyl-CoA to produce
NADH, FADH2, and ATP precursors.

 Oxidative Phosphorylation: Electron transport chain (ETC) complexes use the


energy from redox reactions to pump protons across membranes, generating a proton
gradient that drives ATP synthesis.

4. Energy Balance and Homeostasis

 Regulation: Cells tightly regulate ATP levels to meet energy demands, balancing
energy production with consumption.

 Coupling Reactions: ATP hydrolysis is coupled with energy-requiring reactions


(endergonic reactions) to drive cellular processes like biosynthesis, muscle
contraction, and nerve impulse transmission.

5. Thermodynamics of Bioenergetics

 First Law of Thermodynamics: Energy cannot be created or destroyed, only


converted from one form to another. In cells, chemical energy from nutrient molecules
is converted to ATP and heat.

 Second Law of Thermodynamics: Energy transformations increase the entropy


(disorder) of the universe. Cells manage entropy by maintaining ordered structures
through energy input.

Page 91 of 208
Significance of Bioenergetics

Understanding bioenergetics is crucial for various fields, including:

 Cell Biology: Exploring how cells produce and utilize energy to maintain cellular
functions.

 Physiology: Studying energy metabolism in tissues and organs to understand health


and disease.

 Ecology: Analyzing energy flow through ecosystems and nutrient cycling.

 Biotechnology: Harnessing bioenergetic processes for applications like biofuel


production and metabolic engineering.

7.2. Phosphoryl group transfer and ATP

Phosphoryl group transfer and ATP (adenosine triphosphate) play fundamental roles in
cellular energy metabolism and biochemical reactions. Here's an overview of these concepts:

Phosphoryl Group Transfer

Phosphoryl group transfer involves the transfer of a phosphate group (PO₄³⁻) from a donor
molecule, often ATP, to another molecule (acceptor). This process is crucial for regulating
cellular activities and driving energy-requiring reactions. Key points include:

 ATP as a Phosphoryl Group Donor: ATP donates its terminal phosphate group in
reactions, converting into ADP (adenosine diphosphate) or AMP (adenosine
monophosphate), releasing energy that can be used in cellular processes.

 Enzymatic Catalysis: enzymes known as kinases often catalyze Phosphoryl group


transfers. These enzymes facilitate the transfer of a phosphate group from ATP to
specific substrates, thereby activating or inactivating them.

Page 92 of 208
 Role in Cellular Signaling: Phosphorylation of proteins by kinases is a common
mechanism in cellular signal transduction pathways, regulating protein activity, gene
expression, and other cellular responses.

ATP (Adenosine Triphosphate)

ATP is often referred to as the "energy currency" of cells due to its role in storing and
transferring energy for biochemical reactions. Key aspects of ATP include:

 Structure: ATP consists of adenine (a nitrogenous base), ribose (a sugar), and three
phosphate groups (tri-phosphate).

 Energy Storage: Energy is stored in the high-energy bonds between the phosphate
groups. Hydrolysis of ATP to ADP and inorganic phosphate (Pi) releases
approximately 7.3 kcal/mol of energy, which is used to drive endergonic (energy-
requiring) reactions in cells.

 Cellular Functions: ATP is involved in various cellular processes, including muscle


contraction, nerve impulse transmission, biosynthesis of macromolecules (e.g.,
proteins, nucleic acids), and active transport of ions and molecules across cell
membranes.

 Regeneration: ATP is continuously regenerated from ADP and Pi through cellular


respiration (glycolysis, citric acid cycle, oxidative phosphorylation) or photosynthesis
in plants, ensuring a constant supply of energy for cellular activities.

Significance in Bioenergetics

Understanding phosphoryl group transfer and ATP is critical in bioenergetics for several
reasons:

 Energy Coupling: ATP hydrolysis provides the energy needed to drive coupled
reactions, such as biosynthesis and muscle contraction.

Page 93 of 208
 Metabolic Regulation: Phosphorylation and dephosphorylation of proteins regulate
enzyme activity, metabolic pathways, and cellular responses to environmental cues.

 Thermodynamic Considerations: ATP serves as a mediator of energy transfer and


a buffer against fluctuations in energy demand, maintaining cellular homeostasis.

In summary, phosphoryl group transfer and ATP are essential mechanisms in cellular energy
metabolism and signaling. They enable cells to efficiently capture, store, and utilize energy
for various biochemical processes necessary for life and adaptation to changing
environmental conditions.

7.3. Biological oxidation -reduction reactions

Biological oxidation-reduction (redox) reactions are crucial for energy production, metabolism,
and maintaining cellular functions. Here is an overview of their significance and mechanisms:

Significance of Redox Reactions

1. Energy Production: Redox reactions are central to energy metabolism, such as in


cellular respiration and photosynthesis, where they generate ATP (adenosine
triphosphate) for cellular energy.

2. Electron Transfer: These reactions involve the transfer of electrons (and often
protons) between molecules. This electron transfer is essential for transferring energy
and altering molecular structures.

3. Metabolic Pathways: Redox reactions are integral to various metabolic pathways,


including glycolysis, the citric acid cycle (Krebs cycle), and oxidative phosphorylation,
facilitating the breakdown of nutrients and the synthesis of cellular components.

Page 94 of 208
4. Oxidative Stress and Defense: Redox balance is critical for cellular homeostasis.
Imbalances can lead to oxidative stress, where reactive oxygen species (ROS)
damage cells. Antioxidant systems, such as glutathione and superoxide dismutase,
mitigate oxidative damage by neutralizing ROS.

Mechanisms of Biological Redox Reactions

1. Oxidation and Reduction:

o Oxidation: Involves the loss of electrons (and often hydrogen atoms) from a
molecule, increasing its oxidation state (e.g., from C₂H₆ to CO₂ in cellular
respiration).

o Reduction: Involves the gain of electrons (and often hydrogen atoms),


decreasing the oxidation state of a molecule (e.g., from NAD⁺ to NADH in
glycolysis).

2. Role of Coenzymes:

o NAD⁺/NADH and FAD/FADH₂ are key coenzymes involved in accepting and


donating electrons in redox reactions.

o NAD⁺ accepts two electrons and one proton to form NADH, while FAD accepts
two electrons and two protons to form FADH₂.

3. Electron Transport Chain (ETC):

o In cellular respiration, electrons extracted from nutrients (e.g., glucose) are


passed through a series of electron carriers (e.g., cytochromes) embedded in
mitochondrial membranes.

o This transfer generates a proton gradient across the membrane, driving ATP
synthesis through oxidative phosphorylation.

Page 95 of 208
4. Photosynthesis:

o In plants and algae, photosynthetic organisms use light energy to drive redox
reactions that convert carbon dioxide (CO₂) into glucose.

o Chlorophyll absorbs light energy, initiating a series of redox reactions that


produce ATP and NADPH, which are used to fix CO₂ into organic molecules.

Biological Redox Balance

1. Regulation and Homeostasis: Cells maintain redox balance through tightly regulated
pathways that control the production and neutralization of ROS.

2. Antioxidant Systems: Enzymes like catalase, superoxide dismutase, and


peroxidases neutralize ROS and prevent oxidative damage to cellular components like
DNA, proteins, and lipids.

3. Disease Implications: Dysregulation of redox signaling and oxidative stress are


implicated in various diseases, including neurodegenerative disorders, cancer, and
cardiovascular diseases.

In conclusion, biological oxidation-reduction reactions are fundamental to cellular energy


metabolism, nutrient breakdown, and maintaining cellular redox balance. Understanding
these processes is essential for comprehending how cells generate and utilize energy and for
exploring their implications in health and disease.

Page 96 of 208
7.4. Glycolysis, gluconeogenesis, and the pentose phosphate pathway

Glycolysis: an overview

Glycolysis is a central metabolic pathway that occurs in the cytoplasm of cells and is crucial
for both energy production and the synthesis of precursor molecules for other metabolic
pathways. Here’s an overview of glycolysis:

Overview of Glycolysis:

1. Location: Glycolysis takes place in the cytoplasm of cells, making it accessible to both
aerobic and anaerobic organisms.

2. Purpose: The primary goal of glycolysis is to convert glucose (a 6-carbon molecule)


into two molecules of pyruvate (a 3-carbon molecule). This process generates ATP
and NADH, which are used for cellular energy and redox balance.

3. Steps:

o Step 1: Phosphorylation of Glucose: Glucose is phosphorylated to glucose-


6-phosphate using one molecule of ATP. This step is catalyzed by the enzyme
hexokinase, and it traps glucose inside the cell as glucose-6-phosphate cannot
easily cross the cell membrane.

o Step 2: Isomerization: Glucose-6-phosphate is converted into its isomer,


fructose-6-phosphate, by the enzyme glucose phosphate isomerase.

o Step 3: Phosphorylation of Fructose-6-phosphate: Fructose-6-phosphate is


phosphorylated by ATP to form fructose-1,6-bisphosphate. This reaction is
catalyzed by the enzyme phosphofructokinase-1 (PFK-1) and is a key
regulatory step in glycolysis.

Page 97 of 208
o Step 4: Cleavage into Triose Phosphates: Fructose-1,6-bisphosphate is
cleaved into two triose phosphates: dihydroxyacetone phosphate (DHAP) and
glyceraldehyde-3-phosphate (G3P).

o Step 5: Interconversion of Triose Phosphates: DHAP is converted into


another molecule of G3P by the enzyme triose phosphate isomerase. This step
ensures that glycolysis continues with two molecules of G3P.

o Step 6: Oxidation and ATP Generation: Each G3P molecule is oxidized by


NAD⁺ to form 1,3-bisphosphoglycerate (1,3-BPG) while reducing NAD⁺ to
NADH. In the process, ATP is generated by substrate-level phosphorylation,
producing 2 molecules of ATP per molecule of glucose.

o Step 7: ATP Production: 1,3-BPG donates a phosphate group to ADP,


forming ATP and 3-phosphoglycerate (3-PG). This reaction is catalyzed by
phosphoglycerate kinase.

o Step 8: ATP Production and Substrate-level Phosphorylation: 3-PG is


converted into 2-phosphoglycerate (2-PG), and then into phosphoenolpyruvate
(PEP) by the enzyme enolase. PEP donates a phosphate group to ADP,
forming ATP and pyruvate.

o Step 9: Pyruvate Formation: Each PEP molecule is converted into pyruvate


by pyruvate kinase, generating another molecule of ATP.

Net Yield of Glycolysis:

 Energy Yield: Glycolysis produces a net of 2 molecules of ATP per glucose molecule
through substrate-level phosphorylation.

Page 98 of 208
 Redox Balance: It also generates 2 molecules of NADH, which carry high-energy
electrons to the electron transport chain (in aerobic conditions) or contribute to
fermentation (in anaerobic conditions).

Regulation of Glycolysis:

 Key Enzymes: Phosphofructokinase-1 (PFK-1) and pyruvate kinase are key


regulatory enzymes in glycolysis, controlling the rate of the pathway based on cellular
energy needs.

 Feedback Inhibition: ATP and citrate inhibit PFK-1, while AMP (indicative of low
energy levels) activates PFK-1, regulating the rate of glycolysis.

Importance of Glycolysis:

 Glycolysis is fundamental in both aerobic and anaerobic metabolism, providing ATP


and precursor molecules for various cellular processes.

 It serves as a starting point for other metabolic pathways, such as the citric acid cycle
and the pentose phosphate pathway.

Understanding glycolysis is essential for comprehending cellular energy metabolism and the
interconnected nature of metabolic pathways within cells.

Page 99 of 208
Figure 8: Glycolysis.

Page 100 of 208


The fate of pyruvate under anaerobic conditions (fermentation)

Under anaerobic conditions, when oxygen is not available as an electron acceptor for the
electron transport chain, pyruvate undergoes fermentation to regenerate NAD⁺ from NADH,
which is necessary for glycolysis to continue producing ATP. There are several types of
fermentation pathways, each characteristic of different organisms and yielding distinct
fermentation products. Here is an overview of the fate of pyruvate under anaerobic conditions
through fermentation:

Anaerobic Fermentation Pathways:

1. Lactic Acid Fermentation:

o Organisms: Lactic acid bacteria (e.g., lactobacilli) and some muscle cells in
animals.

o Process: NADH to form lactate directly reduces Pyruvate, regenerating NAD⁺.

o Equation:
Pyruvate + NADH + H⁺ ⟶ Lactate + NAD⁺

2. Alcoholic Fermentation:

o Organisms: Yeasts and some plants.

o Process: Pyruvate is decarboxylated to acetaldehyde, which is then reduced


by NADH to ethanol, regenerating NAD⁺.

o Equation:
Pyruvate ⟶ Acetaldehyde + CO₂
Acetaldehyde + NADH + H⁺ ⟶ Ethanol + NAD⁺

3. Other Fermentation Pathways:

Page 101 of 208


o Mixed Acid Fermentation: Found in some bacteria (e.g., Escherichia coli),
producing a mixture of fermentation products including lactate, ethanol,
acetate, formate, and succinate.

o Butyric Acid Fermentation: Produces butyrate and acetate, commonly


observed in certain bacteria.

Importance of Fermentation:

 Regeneration of NAD⁺: The primary role of fermentation is to regenerate NAD⁺ from


NADH produced during glycolysis. NAD⁺ is essential for the continued functioning of
glycolysis, which generates ATP through substrate-level phosphorylation.

 ATP Production: While fermentation itself does not directly produce ATP (as in
oxidative phosphorylation), it allows glycolysis to continue, yielding a net gain of 2 ATP
molecules per glucose molecule.

Differences from Aerobic Respiration:

 ATP Yield: Fermentation yields much less ATP compared to aerobic respiration,
which is more efficient due to the complete oxidation of glucose in the presence of
oxygen.

 Final Electron Acceptor: Fermentation uses organic molecules within the cell as the
final electron acceptors, unlike aerobic respiration which uses oxygen as the final
electron acceptor.

Industrial and Biological Significance:

 Food Production: Fermentation is crucial in the production of fermented foods and


beverages such as yogurt, cheese, beer, and wine.

Page 102 of 208


 Biotechnology: It is used in industrial processes for the production of biofuels and
various biochemicals.

 Cellular Regulation: Understanding fermentation pathways helps in understanding


metabolic regulation and adaptation to different environmental conditions.

In summary, under anaerobic conditions, pyruvate is metabolized through fermentation


pathways to regenerate NAD⁺, allowing glycolysis to continue producing ATP. The type of
fermentation pathway used depends on the organism and environmental conditions,
ultimately influencing the products formed.

Regulation of glycolysis

Regulation of glycolysis involves several mechanisms that control the activity of enzymes in
the pathway, ensuring that glucose metabolism is balanced and responsive to cellular needs.
Here's an overview of the key regulatory mechanisms involved in glycolysis:

1. Regulation by Enzyme Activity and Availability:

 Hexokinase (or Glucokinase):


o Regulation: Feedback inhibition by glucose-6-phosphate (product inhibition).
o Function: Catalyzes the phosphorylation of glucose to glucose-6-phosphate,
which initiates glycolysis.
 Phosphofructokinase-1 (PFK-1):
o Regulation: Allosteric regulation by ATP, ADP, and AMP.
 ATP: Inhibits PFK-1 activity.
 ADP: Activates PFK-1 activity.
 AMP: Strongly activates PFK-1 activity.
o Function: Catalyzes the conversion of fructose-6-phosphate to fructose-1,6-
bisphosphate, a key regulatory step in glycolysis.

Page 103 of 208


 Pyruvate Kinase:
o Regulation: Allosteric regulation by fructose-1,6-bisphosphate and ATP.
 Fructose-1,6-bisphosphate: Activates pyruvate kinase.
 ATP: Inhibits pyruvate kinase.
o Function: Catalyzes the conversion of phosphoenolpyruvate (PEP) to
pyruvate, producing ATP.

2. Regulation by Hormones and Signaling Pathways:

 Insulin (in Liver and Muscle Cells):


o Effect: Stimulates glycolysis by activating phosphofructokinase-1 and pyruvate
kinase.
o Mechanism: Increases glucose uptake and glycolytic enzyme activity in
response to high blood glucose levels.
 Glucagon and Epinephrine (in Liver Cells):
o Effect: Inhibit glycolysis to maintain blood glucose levels during fasting or
stress.
o Mechanism: Activate glycogen breakdown and gluconeogenesis pathways,
indirectly reducing glycolytic enzyme activity.

3. Regulation by Cellular Energy Status:

 AMP-Activated Protein Kinase (AMPK):


o Role: Monitors cellular AMP/ATP ratio, activated under low energy conditions
(high AMP).
o Effect: Inhibits anabolic pathways (including glycolysis) and activates catabolic
pathways to restore ATP levels.
 Citrate (in Liver Cells):

Page 104 of 208


o Effect: Inhibits phosphofructokinase-1 allosterically.
o Mechanism: Reflects high-energy status (plentiful ATP and citrate), signaling
reduced need for glycolysis.

4. Regulation by Substrate Availability and Metabolic Intermediates:

 Glucose Availability:
o Effect: Controls initial steps of glycolysis (glucose uptake and
phosphorylation).
o Mechanism: High glucose levels favor glycolysis, while low levels activate
gluconeogenesis and glycogen breakdown.
 Fructose-2,6-bisphosphate (in Liver and Muscle Cells):
o Effect: Potent allosteric activator of phosphofructokinase-1.
o Mechanism: Produced in response to insulin signaling, stimulates glycolysis
during high blood glucose levels.

5. Regulation by Transcriptional and Post-Translational Mechanisms:

 Gene Expression of Glycolytic Enzymes:


o Effect: Long-term regulation of glycolysis.
o Mechanism: Altered expression levels in response to hormonal signals and
cellular metabolic needs.
 Post-Translational Modifications:
o Effect: Rapid adjustments in enzyme activity.
o Mechanism: Phosphorylation, acetylation, and other modifications alter
enzyme activity in response to cellular signals.

Page 105 of 208


7.5. Gluconegenesis

Gluconeogenesis is the process by which glucose is synthesized from non-carbohydrate


precursors, primarily in the liver and to a lesser extent in the kidneys. It is essentially the
reverse of glycolysis, with some distinct enzymatic steps to bypass irreversible reactions of
glycolysis. Here is an overview of gluconeogenesis and its regulation:

Overview of Gluconeogenesis:

1. Substrates for Gluconeogenesis:


o Lactate: Produced by anaerobic glycolysis in muscles and red blood cells.
o Glycerol: Derived from hydrolysis of triglycerides in adipose tissue.
o Glucogenic Amino Acids: Amino acids that can be converted into
intermediates of the citric acid cycle (e.g., alanine).
o Propionate: A precursor derived from odd-chain fatty acids metabolism.
2. Location:
o Liver: Main site of gluconeogenesis.
o Kidneys: Contribute to gluconeogenesis during prolonged fasting or
starvation.
3. Key Enzymes and Steps:
o Pyruvate Carboxylase: Converts pyruvate to oxaloacetate, a citric acid cycle
intermediate.
o Phosphoenolpyruvate Carboxykinase (PEPCK): Converts oxaloacetate to
phosphoenolpyruvate (PEP), bypassing the irreversible step of pyruvate kinase
in glycolysis.
o Fructose-1,6-bisphosphatase: Converts fructose-1,6-bisphosphate to
fructose-6-phosphate.

Page 106 of 208


o Glucose-6-phosphatase: Converts glucose-6-phosphate to glucose, which
can be released into the bloodstream.

Regulation of Gluconeogenesis:

1. Reciprocal Regulation with Glycolysis:


o Mutual Inhibition: Glycolysis and gluconeogenesis are reciprocally regulated
to prevent futile cycling.
 Regulation of PFK-1 and FBPase-1: PFK-1 is inhibited by ATP and
activated by AMP, while FBPase-1 is inhibited by AMP and activated by
ATP.
2. Substrate Availability:
o Lactate: High lactate levels (e.g., during intense exercise) can stimulate
gluconeogenesis.
o Glycerol: Derived from triglycerides breakdown in adipose tissue during
fasting or starvation.
3. Hormonal Regulation:
o Glucagon: Released during low blood glucose levels, stimulates
gluconeogenesis by activating cAMP-dependent pathways.
o Insulin: Inhibits gluconeogenesis by promoting glycolysis and glycogen
synthesis.
o Cortisol: Stimulates gluconeogenesis during stress or fasting by increasing
gene expression of gluconeogenic enzymes.
4. Allosteric Regulation:
o Fructose-2,6-bisphosphate: Inhibits gluconeogenesis by activating PFK-1
and inhibiting FBPase-1.
o Acetyl-CoA and Citrate: Reflect cellular energy status and inhibit pyruvate
carboxylase, thereby regulating gluconeogenesis indirectly.
Page 107 of 208
5. Transcriptional Regulation:
o Glucagon and Cortisol: Increase transcription of genes encoding
gluconeogenic enzymes (e.g., PEPCK, glucose-6-phosphatase).

Physiological Role:

Gluconeogenesis ensures that glucose is synthesized and released into the bloodstream
during fasting, prolonged exercise, or periods of low carbohydrate intake. It helps maintain
blood glucose levels and provides energy to tissues (especially the brain) that rely heavily on
glucose for metabolism. The regulation of gluconeogenesis is tightly coordinated with
glycolysis and other metabolic pathways to maintain overall energy balance and metabolic
homeostasis in the body.

7.6. The pentose phosphate pathway

The pentose phosphate pathway (PPP), also known as the hexose monophosphate shunt, is
an important metabolic pathway parallel to glycolysis. Here is an overview of the pentose
phosphate pathway:

Overview of the Pentose Phosphate Pathway:

1. Purpose:
o Generation of NADPH: Provides reducing equivalents (in the form of NADPH)
needed for biosynthetic reactions (e.g., fatty acid synthesis, cholesterol
synthesis, and reduction of oxidized glutathione).
o Production of Ribose-5-Phosphate: Essential for nucleotide synthesis (e.g.,
for DNA and RNA).
2. Location:

Page 108 of 208


o Cytoplasm: Occurs in the cytoplasm of most cells, particularly in tissues
actively engaged in biosynthesis (e.g., liver, adipose tissue, and mammary
glands).
3. Phases of the Pentose Phosphate Pathway:
o Oxidative Phase:
 Glucose-6-Phosphate Dehydrogenase (G6PD): Catalyzes the first
and rate-limiting step, converting glucose-6-phosphate into 6-
phosphogluconolactone, producing NADPH.
 6-Phosphogluconate Dehydrogenase: Converts 6-
phosphogluconate into ribulose-5-phosphate, producing a second
molecule of NADPH.
o Non-Oxidative Phase (Interconversion of sugars):
 Transketolase and Transaldolase: Catalyze the rearrangement of
sugars in the pathway to produce ribose-5-phosphate and other
intermediates needed for nucleotide synthesis.
4. Regulation of the Pentose Phosphate Pathway:
o Regulated by NADP+: High levels of NADP+ stimulate G6PD activity,
increasing flux through the pathway when NADPH is required for biosynthesis.
o Feedback Inhibition: NADPH inhibits G6PD, reducing flux through the
pathway when NADPH levels are adequate.
o Substrate Availability: Levels of glucose-6-phosphate and NADP+ influence
pathway activity.
5. Physiological Roles:
o Antioxidant Defense: Provides NADPH for the regeneration of reduced
glutathione, which protects cells from oxidative stress.
o Biosynthetic Functions: Supplies ribose-5-phosphate for nucleotide
synthesis, important for DNA and RNA production.
Page 109 of 208
o Maintenance of Redox Balance: NADPH is essential for maintaining the
reduced state of cells, necessary for numerous biosynthetic reactions.
6. Clinical Relevance:
o G6PD Deficiency: Deficiency in glucose-6-phosphate dehydrogenase can
lead to hemolytic anemia due to reduced ability to protect red blood cells from
oxidative stress.
o Cancer Metabolism: Altered pentose phosphate pathway activity is observed
in cancer cells, supporting rapid proliferation and adaptation to oxidative stress.

Page 110 of 208


Figure 9: Pentose phosphate pathway.

Page 111 of 208


7.7. Metabolic regulation (glucose and glycogen as examples)

Metabolic Regulation: Glucose and Glycogen

Metabolic regulation refers to the control mechanisms that cells use to maintain energy
homeostasis, respond to changing energy demands, and ensure proper utilization of
metabolic fuels like glucose. Here, we will explore how glucose and glycogen metabolism are
regulated.

1. Glucose Metabolism Regulation:

 Glucose Uptake:

o Insulin: Secreted by the pancreas in response to high blood glucose levels,


insulin promotes glucose uptake into cells by increasing the translocation of
GLUT4 transporters to the cell membrane.

o Glucagon: Secreted by the pancreas in response to low blood glucose levels,


glucagon stimulates glycogen breakdown (glycogenolysis) in the liver to
release glucose into the bloodstream.

 Glycolysis Regulation:

o Enzyme Regulation: Enzymes involved in glycolysis, such as


phosphofructokinase-1 (PFK-1), are regulated allosterically and hormonally to
control the rate of glycolysis.

o Feedback Inhibition: High levels of ATP and citrate inhibit PFK-1, slowing
down glycolysis when energy levels are sufficient.

Page 112 of 208


 Gluconeogenesis Regulation:

o Reciprocal Regulation: Gluconeogenesis is reciprocally regulated with


glycolysis to maintain glucose homeostasis. It is mainly activated by glucagon
and cortisol, and inhibited by insulin.

2. Glycogen Metabolism Regulation:

 Glycogen Synthesis (Glycogenesis):

o Insulin: Promotes glycogen synthesis by activating glycogen synthase and


inhibiting glycogen phosphorylase, thus storing excess glucose as glycogen in
liver and muscle cells.

o Glucagon and Epinephrine: Inhibit glycogen synthesis and stimulate


glycogen breakdown (glycogenolysis) to release glucose.

 Glycogen Breakdown (Glycogenolysis):

o Glucagon and Epinephrine: Stimulate glycogen breakdown in the liver (and


muscles for epinephrine) to release glucose into the bloodstream during times
of fasting or stress.

 Glycogen Phosphorylase: This enzyme is the key regulatory enzyme in


glycogenolysis and is activated by phosphorylation (via protein kinase A under
glucagon/epinephrine signaling) to break down glycogen into glucose-1-phosphate.

3. Integrated Regulation:

 Energy Status Regulation: Hormonal signals (insulin, glucagon, epinephrine)


integrate with cellular energy status (ATP/AMP ratio) to regulate both glucose
uptake/utilization and glycogen storage/breakdown.

Page 113 of 208


 Tissue-Specific Regulation: Liver and muscle tissues have different roles in glucose
metabolism regulation. The liver regulates blood glucose levels via gluconeogenesis
and glycogenolysis, while muscles primarily store and use glycogen for energy.

Clinical Relevance:

 Diabetes Mellitus: Type 1 diabetes involves insufficient insulin production, leading to


uncontrolled blood glucose levels. Type 2 diabetes involves insulin resistance, where
cells do not respond effectively to insulin, disrupting glucose metabolism regulation.

 Glycogen Storage Diseases: Genetic disorders affecting enzymes involved in


glycogen metabolism lead to abnormal glycogen storage in tissues, affecting energy
metabolism.

Understanding these regulatory mechanisms is essential for managing metabolic disorders


and optimizing energy metabolism in health and disease.

Page 114 of 208


7.8. The citric acid cycle

7.8.1. Krebs cycle (TCA Cycle) Steps:

1. Condensation (Citrate Synthesis):


o Acetyl CoA (2C) combines with oxaloacetate (4C) to form citrate (6C).
o Enzyme: Citrate synthase
2. Isomerization:
o Citrate is converted to isocitrate (6C).
o Enzyme: Aconitase
3. Dehydrogenation and Decarboxylation:
o Isocitrate undergoes oxidative decarboxylation to form α-ketoglutarate (5C)
and CO2.
o NAD+ is reduced to NADH.
o Enzyme: Isocitrate dehydrogenase
4. Oxidative Decarboxylation:
o α-Ketoglutarate is oxidatively decarboxylated to form succinyl-CoA (4C), CO2,
and NADH.
o Enzyme: α-Ketoglutarate dehydrogenase complex
5. Substrate-level Phosphorylation:
o Succinyl-CoA undergoes substrate-level phosphorylation to form succinate
(4C), GTP (which converts to ATP), and CoA-SH.
o Enzyme: Succinyl-CoA synthetase
6. Oxidation of Succinate:
o Succinate is oxidized to fumarate, reducing FAD to FADH2.
o Enzyme: Succinate dehydrogenase (part of the electron transport chain)
7. Hydration of Fumarate:
o Fumarate is hydrated to form malate.

Page 115 of 208


o Enzyme: Fumarase
8. Regeneration of Oxaloacetate:
o Malate is oxidized to oxaloacetate, reducing NAD+ to NADH.
o Oxaloacetate can then combine with another molecule of acetyl CoA to begin
the cycle again.
o Enzyme: Malate dehydrogenase

7.8.2. Regulation of Citric Acid cycles

The regulation of the citric acid cycle (TCA cycle) is tightly controlled to match cellular energy
demands and metabolic conditions. Here are some key regulatory mechanisms:

1. Feedback Inhibition:

o High concentrations of ATP, NADH, and succinyl-CoA act as feedback


inhibitors of key enzymes in the TCA cycle.

o ATP inhibits citrate synthase and isocitrate dehydrogenase.

o NADH inhibits isocitrate dehydrogenase and α-ketoglutarate dehydrogenase.

o Succinyl-CoA inhibits α-ketoglutarate dehydrogenase.

2. Substrate Availability:

o The availability of substrates such as acetyl-CoA, oxaloacetate, and NAD+


influences the rate of the cycle.

o Acetyl-CoA availability is determined by the breakdown of fatty acids and


carbohydrates.

o Oxaloacetate availability affects the initial step with citrate synthase.

3. Allosteric Regulation:

Page 116 of 208


o Enzymes in the TCA cycle are regulated by allosteric modulators.

o For example, citrate synthase is inhibited by ATP and NADH, while it is


activated by ADP and calcium ions.

4. Activation by Calcium Ions:

o Calcium ions can activate enzymes like pyruvate dehydrogenase and isocitrate
dehydrogenase, thereby enhancing TCA cycle activity during times of
increased cellular activity.

5. Hormonal Regulation:

o Hormones such as insulin can influence TCA cycle activity indirectly by


regulating glucose uptake and metabolism, thereby affecting the availability of
substrates like pyruvate and acetyl-CoA.

6. Transcriptional Regulation:

o Long-term regulation of enzymes in the TCA cycle can occur through changes
in gene expression influenced by transcription factors and cellular signaling
pathways.

These regulatory mechanisms ensure that the TCA cycle operates efficiently under varying
metabolic conditions, balancing energy production with the metabolic needs of the cell.

Page 117 of 208


7.8.3. TCA intermediates as precursors for biosynthesis

The regulation of the citric acid cycle (TCA cycle) is tightly controlled to match cellular energy
demands and metabolic conditions. Here are some key regulatory mechanisms:

1. Feedback Inhibition:

o High concentrations of ATP, NADH, and succinyl-CoA act as feedback


inhibitors of key enzymes in the TCA cycle.

o ATP inhibits citrate synthase and isocitrate dehydrogenase.

o NADH inhibits isocitrate dehydrogenase and α-ketoglutarate dehydrogenase.

o Succinyl-CoA inhibits α-ketoglutarate dehydrogenase.

2. Substrate Availability:

o The availability of substrates such as acetyl-CoA, oxaloacetate, and NAD+


influences the rate of the cycle.

o Acetyl-CoA availability is determined by the breakdown of fatty acids and


carbohydrates.

o Oxaloacetate availability affects the initial step with citrate synthase.

3. Allosteric Regulation:

o Enzymes in the TCA cycle are regulated by allosteric modulators.

o For example, citrate synthase is inhibited by ATP and NADH, while it is


activated by ADP and calcium ions.

Page 118 of 208


4. Activation by Calcium Ions:

o Calcium ions can activate enzymes like pyruvate dehydrogenase and isocitrate
dehydrogenase, thereby enhancing TCA cycle activity during times of
increased cellular activity.

5. Hormonal Regulation:

o Hormones such as insulin can influence TCA cycle activity indirectly by


regulating glucose uptake and metabolism, thereby affecting the availability of
substrates like pyruvate and acetyl-CoA.

6. Transcriptional Regulation:

o Long-term regulation of enzymes in the TCA cycle can occur through changes
in gene expression influenced by transcription factors and cellular signaling
pathways.

These regulatory mechanisms ensure that the TCA cycle operates efficiently under varying
metabolic conditions, balancing energy production with the metabolic needs of the cell.

7.8.4. Glyoxylate cycle

The glyoxylate cycle is a variant of the citric acid cycle (TCA cycle) found in certain
microorganisms and plants, particularly those that utilize acetate or fatty acids as their primary
carbon source. Here's an overview of the glyoxylate cycle:

Overview:

1. Purpose: The glyoxylate cycle allows organisms to synthesize carbohydrates from


fats or lipids (such as acetate or fatty acids) when glucose or other sugars are not
available. It bypasses the two decarboxylation steps of the TCA cycle, which
conserves carbon for gluconeogenesis.

Page 119 of 208


2. Key Enzymes: It shares several enzymes with the TCA cycle but includes additional
enzymes necessary for its unique functions:

o Isocitrate lyase: This enzyme cleaves isocitrate into succinate and glyoxylate,
bypassing the decarboxylation step that occurs in the TCA cycle.

o Malate synthase: This enzyme then condenses acetyl-CoA with glyoxylate to


form malate.

3. Steps:

o Step 1: Isocitrate is cleaved by isocitrate lyase into succinate and glyoxylate.

o Step 2: Acetyl-CoA combines with glyoxylate to form malate via malate


synthase.

o Malate can then be converted back to oxaloacetate, which replenishes the pool
of TCA cycle intermediates and supports gluconeogenesis.

4. Importance:

o The glyoxylate cycle enables organisms to utilize acetate or fatty acids


efficiently for the synthesis of carbohydrates, particularly in seeds of plants and
certain bacteria like Mycobacterium tuberculosis, which use fatty acids as a
primary carbon source during infection.

5. Regulation:

o Similar regulatory mechanisms as the TCA cycle apply, such as substrate


availability and allosteric regulation of enzymes involved in the glyoxylate cycle.

Page 120 of 208


Significance:

 Plants: In plants, the glyoxylate cycle plays a crucial role in seed germination, allowing
the conversion of stored lipids into carbohydrates for growth until photosynthesis
begins.

 Microorganisms: In bacteria, particularly pathogens like Mycobacterium tuberculosis,


the glyoxylate cycle supports their ability to survive and replicate within host cells by
utilizing fatty acids as a carbon source.

Page 121 of 208


7.9. Fatty Acid catabolism
7.9.1. Digestion, mobilization and transport of fats

To discuss the digestion, mobilization, and transport of fats, we delve into the physiological
processes involved in lipid metabolism:

Digestion of Fats:

1. Digestion in the Gut:

o Mouth and Stomach: Limited fat digestion occurs due to lingual lipase in the
mouth and gastric lipase in the stomach.

o Small Intestine: The main site for fat digestion. Upon entry, fats are emulsified
by bile salts from the liver, forming small droplets. Pancreatic lipase then breaks
down triglycerides into monoglycerides and free fatty acids.

2. Absorption:

o Monoglycerides and fatty acids are absorbed by intestinal epithelial cells


(enterocytes).

o Inside enterocytes, they are re-esterified into triglycerides and packaged into
chylomicrons (lipoprotein particles).

Mobilization of Fats:

1. Adipose Tissue:

o Stored triglycerides in adipocytes are mobilized through lipolysis, mediated by


hormone-sensitive lipase (HSL) and adipose triglyceride lipase (ATGL).

o Hormones like adrenaline (epinephrine) and glucagon stimulate lipolysis during


fasting or stress.

Page 122 of 208


2. Transport in Blood:

o Triglycerides, along with cholesterol and other lipids, are transported in the
bloodstream within lipoprotein particles such as chylomicrons, VLDL (very-low-
density lipoproteins), LDL (low-density lipoproteins), and HDL (high-density
lipoproteins).

Regulation and Transport:

1. Lipoproteins:

o Chylomicrons: Transport dietary lipids from the intestine to tissues.

o VLDL: Transport endogenous triglycerides synthesized in the liver to peripheral


tissues.

o LDL: Delivers cholesterol to cells.

o HDL: Involved in reverse cholesterol transport, bringing cholesterol from


tissues back to the liver for excretion or recycling.

2. Metabolic Regulation:

o Insulin: Promotes fat storage by enhancing triglyceride synthesis in adipose


tissue.

o Glucagon and adrenaline: Stimulate lipolysis during fasting or stress.

o Leptin: Hormone that regulates appetite and energy balance, influencing fat
storage and utilization.

Page 123 of 208


Clinical Relevance:

Understanding fat digestion, mobilization, and transport is crucial for managing lipid
metabolism disorders like hyperlipidemia (high blood lipids) and understanding the role of
lipids in cardiovascular health, metabolic syndrome, and obesity-related conditions.

This overview provides a glimpse into the intricate processes that enable the body to digest,
mobilize, and transport fats for energy production and storage.

7.9.2. Oxidation of fatty acids

To discuss the oxidation of fatty acids, we explore the processes involved in breaking down
fats for energy production:

Oxidation of Fatty Acids:

1. Fatty Acid Activation:

o Cytoplasm: Fatty acids are activated by attaching Coenzyme A (CoA) to form


fatty acyl-CoA. This step requires ATP hydrolysis.

o Outer Mitochondrial Membrane: Long-chain fatty acyl-CoA is transported into


the mitochondria via carnitine shuttle system.

2. Beta-Oxidation:

o Mitochondrial Matrix: The process of beta-oxidation breaks down fatty acyl-


CoA molecules into acetyl-CoA molecules:

 Step 1: Dehydrogenation: Fatty acyl-CoA is oxidized by FAD-linked


acyl-CoA dehydrogenase, producing FADH2 and trans-2-enoyl-CoA.

 Step 2: Hydration: Water is added across the double bond by enoyl-


CoA hydratase, forming L-3-hydroxyacyl-CoA.

Page 124 of 208


 Step 3: Dehydrogenation: L-3-hydroxyacyl-CoA is oxidized by NAD+-
linked L-3-hydroxyacyl-CoA dehydrogenase, producing NADH and 3-
ketoacyl-CoA.

 Step 4: Thiolytic cleavage: Thiolysis of 3-ketoacyl-CoA by thiolase


yields acetyl-CoA and a fatty acyl-CoA molecule shortened by two
carbons.

3. Repeat Cycle:

o Each cycle shortens the fatty acid chain by two carbons, generating additional
acetyl-CoA, NADH, and FADH2 until the entire fatty acid is oxidized.

4. Energy Production:

o Acetyl-CoA: Enters the citric acid cycle (TCA cycle) to generate ATP through
oxidative phosphorylation.

o NADH and FADH2: Produced during beta-oxidation and used in the electron
transport chain (ETC) to generate more ATP.

Regulation of Fatty Acid Oxidation:

 Enzyme Regulation: The rate-limiting step is often the transport of fatty acyl-CoA into
the mitochondria via the carnitine shuttle system.

 Hormonal Regulation: Hormones such as insulin and glucagon regulate fatty acid
oxidation in response to energy needs:

o Insulin: Promotes fatty acid synthesis and storage.

o Glucagon and adrenaline: Stimulate lipolysis and fatty acid oxidation during
fasting or stress.

Page 125 of 208


7.9.3. Ketone bodies

Ketone bodies are water-soluble molecules produced primarily in the liver during periods of
fasting, carbohydrate restriction, or prolonged exercise. They serve as an alternative energy
source, especially for tissues like the brain, heart, and skeletal muscles when glucose
availability is limited. Here's an overview of ketone bodies and their metabolic significance:

Types of Ketone Bodies:

1. Acetoacetate:

o Acetoacetate is the first ketone body produced in the liver from acetyl-CoA
during conditions of increased fatty acid oxidation.

o It can be further converted into two other forms:

 β-hydroxybutyrate (β-HB): A more stable form of ketone body.

 Acetone: A minor byproduct that is spontaneously produced from


acetoacetate.

2. Formation and Regulation:

o Fatty Acid Oxidation: When glucose availability is low (e.g., during fasting),
fatty acids are oxidized to produce acetyl-CoA.

o Acetyl-CoA Availability: Excess acetyl-CoA, due to limited oxaloacetate


availability (used in the TCA cycle), leads to ketogenesis.

o Regulation: Hormones like insulin (low levels) and glucagon stimulate


ketogenesis, while insulin inhibits it.

Page 126 of 208


Metabolic Roles and Significance:

1. Energy Source: Ketone bodies serve as an alternative fuel source to glucose during
fasting or starvation, reducing the reliance on glucose and sparing muscle protein
breakdown.

2. Brain Fuel: Although the brain predominantly uses glucose, during prolonged fasting
or starvation, ketone bodies can cross the blood-brain barrier and serve as an energy
substrate for neurons.

3. Transport and Utilization: Ketone bodies are transported in the bloodstream to


peripheral tissues, where they are converted back into acetyl-CoA for energy
production in mitochondria via the TCA cycle and oxidative phosphorylation.

Clinical Implications:

1. Ketosis and Ketogenesis: Ketosis refers to elevated levels of ketone bodies in the
blood, which can occur in conditions like fasting, low-carbohydrate diets (e.g.,
ketogenic diets), or untreated diabetes mellitus (diabetic ketoacidosis).

2. Therapeutic Applications: Ketogenic diets, which induce ketosis, are used


therapeutically in conditions like epilepsy, where ketone bodies may provide
neuroprotective effects.

3. Disorders: Disorders of ketone metabolism, such as certain inherited metabolic


disorders (e.g., ketolysis defects), can lead to abnormal accumulation of ketone bodies
and metabolic disturbances.

Understanding ketone body metabolism is crucial for comprehending metabolic adaptation to


fasting, dietary interventions, and pathological conditions associated with altered
carbohydrate and lipid metabolism.

Page 127 of 208


7.10. Amino acid oxidation
7.10.1. Metabolic fates of amino acids

The metabolic fate of amino acids involves several pathways depending on the body's
metabolic state and the specific amino acid involved. Here is an overview of how amino acids
are metabolized:

1. Protein Turnover:

 Protein Degradation: Amino acids are released from dietary proteins or from the
breakdown of body proteins (e.g., muscle proteins during fasting or starvation).

 Transport: Amino acids are transported to the liver and other tissues via the
bloodstream for utilization or storage.

2. Deamination:

 Amino Acid Catabolism: Amino acids undergo deamination (removal of the amino
group) primarily in the liver and kidneys.

 Ammonia Formation: Ammonia (NH₃) is released as a byproduct, which is toxic and


must be converted to urea in the liver (urea cycle) or excreted as ammonia or urea via
the kidneys.

3. Glucogenic and Ketogenic Amino Acids:

 Glucogenic Amino Acids: Most amino acids (except leucine and lysine) can be
converted into intermediates of the TCA cycle or glycolysis, leading to glucose
synthesis (gluconeogenesis).

 Ketogenic Amino Acids: Some amino acids (e.g., leucine and lysine) can be
converted into acetyl-CoA or acetoacetyl-CoA, which can be used for ketone body
synthesis.

Page 128 of 208


4. Fate of Carbon Skeletons:

 TCA Cycle Intermediates: Carbon skeletons from amino acids can enter the TCA
cycle as intermediates such as α-ketoglutarate, succinyl-CoA, and oxaloacetate.

 Energy Production: These intermediates can be oxidized to produce ATP via


oxidative phosphorylation.

5. Special Cases:

 Glutamine and Alanine: Glutamine is a major carrier of nitrogen in the bloodstream,


transporting ammonia to the kidneys. Alanine is transported from muscles to the liver
where it undergoes gluconeogenesis.

 Branch-Chain Amino Acids (BCAAs): Leucine, isoleucine, and valine are essential
amino acids that can be oxidized directly by muscle tissue for energy.

Clinical Relevance:

 Nitrogen Balance: The balance between amino acid intake and excretion (as urea) is
crucial for maintaining nitrogen balance in the body.

 Inborn Errors of Metabolism: Disorders affecting amino acid metabolism (e.g.,


phenylketonuria, maple syrup urine disease) can lead to toxic accumulation of amino
acid byproducts and metabolic disturbances.

Understanding the metabolic fate of amino acids is essential for comprehending overall
protein metabolism, energy production, and metabolic adaptations during various
physiological states such as fasting, exercise, and dietary changes.

Page 129 of 208


7.10.2. Nitrogen excretion and the urea cycle

The urea cycle, also known as the ornithine cycle, plays a crucial role in nitrogen excretion in
mammals, including humans. Here is a brief overview:

Urea Cycle Overview:

Purpose: The urea cycle converts toxic ammonia, which is generated during amino acid
metabolism, into urea, a less toxic compound that can be safely excreted by the kidneys in
urine.

Location: The urea cycle primarily occurs in the liver and to a lesser extent in the kidneys.

Steps:

Step 1: Ammonia (NH₃) combines with carbon dioxide (CO₂) to form carbamoyl phosphate
in a reaction catalyzed by carbamoyl phosphate synthetase I (CPS I).

Step 2: Ornithine reacts with carbamoyl phosphate to form citrulline, catalyzed by ornithine
transcarbamylase (OTC).

Step 3: Citrulline is transported out of the mitochondria into the cytosol.

Step 4: Citrulline reacts with aspartate to form argininosuccinate, catalyzed by


argininosuccinate synthetase.

Step 5: Argininosuccinate is cleaved into arginine and fumarate by argininosuccinate lyase.

Step 6: Arginine undergoes hydrolysis to form urea and regenerate ornithine, catalyzed by
arginase.

Regulation: The urea cycle is regulated by the availability of substrates, particularly ammonia
and ornithine, as well as by the activity of the enzymes involved in each step.

Page 130 of 208


Clinical Relevance: Defects in enzymes of the urea cycle can lead to urea cycle disorders,
which are characterized by the accumulation of ammonia in the blood (hyperammonemia),
causing neurological symptoms and potentially leading to severe health complications. The
urea cycle is essential for maintaining nitrogen balance in the body and for eliminating excess
nitrogen generated from protein metabolism.

Figure 10: Uric acid

Page 131 of 208


7.10.3. Pathways of amino acid degradation

Pathways of Amino Acid Degradation:

Amino acids can be degraded through different pathways depending on their specific
structures and the need for energy or substrates. The primary pathways include:

1. Transamination:
o Overview: Amino acids undergo transamination to transfer their amino group
(-NH₂) to α-ketoglutarate, forming glutamate and α-keto acid. This process is
reversible and occurs in most tissues.
o Enzyme: Catalyzed by aminotransferases (transaminases).
2. Oxidative Deamination:
o Overview: Glutamate, generated from transamination, undergoes oxidative
deamination by glutamate dehydrogenase, releasing ammonia (NH₃) and
forming α-ketoglutarate.
o Regulation: This step is crucial for ammonia detoxification in the liver.
3. Urea Cycle:
o Overview: Ammonia produced from oxidative deamination of amino acids is
incorporated into the urea cycle to form urea, which is excreted in urine.
o Location: Mainly occurs in the liver.
4. Decarboxylation:
o Overview: Some amino acids undergo decarboxylation, where the amino
group is removed as ammonia (NH₃), leaving behind an α-keto acid.
o Examples: Histidine, serine, threonine.
5. Ketogenesis:
o Overview: Some amino acids can be converted to ketone bodies, particularly
during prolonged fasting or starvation when glucose levels are low.

Page 132 of 208


o Examples: Leucine and lysine contribute to ketone body formation.
6. Glycogenesis and Gluconeogenesis:
o Overview: Amino acids can also contribute to glucose production through
gluconeogenesis in the liver during fasting or starvation.
o Examples: Alanine is converted to pyruvate, which can be used for
gluconeogenesis.
7. Creatine Synthesis:
o Overview: Arginine and glycine can be utilized for creatine synthesis, important
for energy metabolism in muscles.
8. Uric Acid Formation:
o Overview: Purine nucleotide degradation leads to the formation of uric acid,
which is excreted primarily by the kidneys.

Clinical Significance:

 Urea Cycle Disorders: Defects in enzymes of the urea cycle lead to


hyperammonemia, which can cause neurological symptoms and metabolic crises.
 Amino Acid Disorders: Inborn errors of metabolism can result from deficiencies in
enzymes involved in amino acid catabolism, leading to accumulation of toxic
intermediates or deficiency of essential molecules.

Understanding these pathways is crucial for comprehending how amino acids are
metabolized under normal physiological conditions and how disruptions can lead to various
metabolic disorders.

Page 133 of 208


Figure 11: Degradation of amino acids

Page 134 of 208


7.11. Oxidative Phosphorylation

Oxidative phosphorylation is the process by which ATP (adenosine triphosphate), the main
energy currency of cells, is synthesized using energy derived from the oxidation of nutrients.
This process occurs in the inner mitochondrial membrane and involves a series of enzyme
complexes and electron carriers.

1. Location: Occurs in the inner mitochondrial membrane of eukaryotic cells and the
plasma membrane of prokaryotes.
2. Key Components:
o Electron Transport Chain (ETC):
 Series of protein complexes (Complex I to IV) and electron carriers
(ubiquinone and cytochrome c) embedded in the inner mitochondrial
membrane.
 Electrons from NADH and FADH₂ are transferred through the
complexes, releasing energy in the form of protons (H⁺) pumped across
the membrane.
o Chemiosmosis:
 Proton gradient formation across the inner membrane due to proton
pumping by ETC complexes.
 Protons flow back through ATP synthase (Complex V) down their
electrochemical gradient, driving ATP synthesis from ADP and
phosphate (Pi).
3. Steps:
o NADH Dehydrogenase (Complex I): Accepts electrons from NADH, pumps
protons across the membrane.

Page 135 of 208


o Succinate Dehydrogenase (Complex II): Accepts electrons from FADH₂
(from succinate in the TCA cycle), feeds electrons into ubiquinone.
o Cytochrome c Reductase (Complex III): Transfers electrons from ubiquinone
to cytochrome c, pumps protons.
o Cytochrome c Oxidase (Complex IV): Transfers electrons to oxygen (forming
water), pumps protons.
4. ATP Synthase (Complex V):
o Uses the proton gradient generated by the ETC to drive ATP synthesis.
o Protons flow through ATP synthase, causing conformational changes that bind
ADP and phosphate (Pi) together to form ATP.
5. Chemical Reactions:
o Oxidation: Nutrients (such as glucose, fatty acids, and amino acids) are
oxidized to generate NADH and FADH₂.
o Reduction: Electrons from NADH and FADH₂ are passed through the ETC,
releasing energy.
o Phosphorylation: ATP synthesis occurs as protons flow back through ATP
synthase.
6. Energy Yield: The complete oxidation of one molecule of glucose can yield up to 36-
38 ATP molecules through oxidative phosphorylation, depending on the shuttle
systems used to transfer electrons into the mitochondria.
7. Regulation: Oxidative phosphorylation is tightly regulated by the availability of
substrates (NADH, FADH₂, oxygen) and by feedback mechanisms to maintain cellular
ATP levels and redox balance.

Page 136 of 208


7.11.1. The chemiosmotic theory and the mechanism of ATP synthesis

The chemiosmotic theory, proposed by Peter Mitchell in 1961, is a fundamental concept in


bioenergetics, explaining how ATP (adenosine triphosphate) is synthesized in cells. This
theory describes the process by which a proton gradient across a membrane drives the
synthesis of ATP, the primary energy currency of the cell.

Key Components of Chemiosmotic Theory:

1. Electron Transport Chain (ETC):


o Located in the inner mitochondrial membrane (in eukaryotes) or the plasma
membrane (in prokaryotes).
o Electrons are transferred through a series of protein complexes (Complex I-IV)
and other molecules like coenzyme Q and cytochrome c.
o The energy released from electron transfer is used to pump protons (H+) from
the mitochondrial matrix to the intermembrane space, creating a proton
gradient (proton motive force).
2. Proton Gradient:
o The proton gradient results in a higher concentration of protons in the
intermembrane space compared to the matrix.
o This gradient creates both a pH gradient (chemical potential) and a charge
difference (electrical potential) across the membrane.
3. ATP Synthase:
o ATP synthase is a multi-subunit enzyme complex embedded in the inner
mitochondrial membrane.
o It consists of two main parts: F0 (a proton channel) and F1 (catalytic site for
ATP synthesis).

Page 137 of 208


o Protons flow back into the matrix through the F0 channel, driven by the proton
gradient.

Mechanism of ATP Synthesis:

1. Proton Flow and ATP Synthase Activation:


o Protons move through the F0 subunit of ATP synthase, causing it to rotate.
o This rotational motion induces conformational changes in the F1 subunit, where
ADP and inorganic phosphate (Pi) bind.
2. Conformational Changes:
o The rotation of the F0 subunit causes the active sites in the F1 subunit to
undergo a series of conformational changes.
o These changes facilitate the binding of ADP and Pi, and their subsequent
condensation to form ATP.
3. ATP Release:
o After ATP is synthesized, the active site returns to its original conformation,
releasing the newly formed ATP molecule into the mitochondrial matrix.

Summary of Chemiosmotic Theory:

 The electron transport chain generates a proton gradient across the inner
mitochondrial membrane.
 The proton motive force drives protons back into the mitochondrial matrix through ATP
synthase.
 The energy from this proton flow is used to catalyze the synthesis of ATP from ADP
and Pi.

Importance of Chemiosmotic Theory:

Page 138 of 208


 The chemiosmotic theory revolutionized our understanding of how cells generate
energy.
 It explained the coupling of electron transport and ATP synthesis, a process essential
for cellular respiration and photosynthesis.
 Peter Mitchell was awarded the Nobel Prize in Chemistry in 1978 for his pioneering
work on chemiosmotic theory.

Understanding this process is crucial for fields like bioenergetics, cellular biology, and
biochemistry, providing insights into how energy is harnessed and utilized by living organisms.

7.11.2. The electron transport system

The Electron Transport System (ETS), also known as the Electron Transport Chain (ETC), is
a series of protein complexes and other molecules within the inner mitochondrial membrane
(in eukaryotes) or the plasma membrane (in prokaryotes). The primary role of the ETS is to
transfer electrons from electron donors to electron acceptors via redox reactions, coupled
with the translocation of protons (H+) across a membrane, which creates a proton gradient
that drives ATP synthesis.

Key Components of the Electron Transport System:

1. Complex I (NADH: Ubiquinone Oxidoreductase):


o Accepts electrons from NADH, oxidizing it to NAD+.
o Transfers electrons to ubiquinone (coenzyme Q).
o Pumps protons from the matrix into the intermembrane space.
2. Complex II (Succinate: Ubiquinone Oxidoreductase):
Page 139 of 208
o Accepts electrons from FADH2, generated from the Krebs cycle.
o Transfers electrons to ubiquinone.
o Does not pump protons, thus contributing less to the proton gradient than
Complex I.
3. Ubiquinone (Coenzyme Q):
o A lipid-soluble electron carrier that shuttles electrons between Complexes I/II
and Complex III.
o Transfers electrons while moving freely within the inner mitochondrial
membrane.
4. Complex III (Cytochrome bc1 Complex):
o Accepts electrons from reduced ubiquinone (ubiquinol).
o Transfers electrons to cytochrome c.
o Pumps protons from the matrix into the intermembrane space.
5. Cytochrome c:
o A small, soluble electron carrier protein located in the intermembrane space.
o Transfers electrons from Complex III to Complex IV.
6. Complex IV (Cytochrome c Oxidase):
o Accepts electrons from cytochrome c.
o Transfers electrons to molecular oxygen (O2), reducing it to water (H2O).
o Pumps protons from the matrix into the intermembrane space.

Mechanism of Electron Transport:

1. NADH and FADH2 Oxidation:


o NADH and FADH2, produced in the Krebs cycle, donate electrons to Complex
I and Complex II, respectively.
o These complexes then transfer the electrons to ubiquinone.
2. Electron Transfer and Proton Pumping:
Page 140 of 208
o As electrons are passed from one complex to the next, the energy released is
used to pump protons across the inner mitochondrial membrane.
o Complex I, III, and IV pump protons, creating a proton gradient across the
membrane.
3. Formation of Proton Gradient:
o The proton pumping creates a high concentration of protons in the
intermembrane space and a low concentration in the mitochondrial matrix.
o This proton gradient represents potential energy, known as the proton motive
force.
4. Reduction of Oxygen:
o At Complex IV, electrons are transferred to oxygen, the final electron acceptor,
reducing it to water.
o This step is crucial for maintaining the flow of electrons through the ETS.

Role in ATP Synthesis:

 The proton gradient created by the ETS drives protons back into the mitochondrial
matrix through ATP synthase, a process known as chemiosmosis.
 The flow of protons through ATP synthase provides the energy needed to synthesize
ATP from ADP and inorganic phosphate (Pi).

Summary of the Electron Transport System:

 The ETS consists of four main protein complexes and two mobile electron carriers.
 It functions to transfer electrons from NADH and FADH2 to oxygen, creating a proton
gradient in the process.
 The proton gradient generated by the ETS is essential for the production of ATP
through oxidative phosphorylation.

Page 141 of 208


Understanding the ETS is fundamental for grasping how cells produce energy, making it a
key concept in bioenergetics, cellular respiration, and overall cellular metabolism.

Page 142 of 208


7.11.3. Regulation of Oxidative Phosphorylation

Oxidative phosphorylation is a critical process in cellular respiration, involving the production


of ATP through the electron transport chain (ETC) and chemiosmosis. The regulation of
oxidative phosphorylation ensures that ATP production matches the energy demands of the
cell. Several factors and mechanisms contribute to this regulation:

1. Availability of Substrates:

ADP and Pi: The availability of ADP and inorganic phosphate (Pi) is crucial. ATP synthesis
only occurs when ADP and Pi are present, a concept known as respiratory control or acceptor
control.

NADH and FADH2: These electron donors, generated from the Krebs cycle and other
metabolic pathways, supply the electrons to the ETC. Their availability regulates the rate of
electron transport and subsequent ATP production.

2. Oxygen Concentration:

Oxygen is the final electron acceptor in the ETC. Adequate oxygen levels are necessary for
the proper functioning of Complex IV (cytochrome c oxidase). Low oxygen levels (hypoxia)
can limit the rate of oxidative phosphorylation.

3. Proton Gradient and Membrane Potential:

The proton gradient (proton motive force) across the inner mitochondrial membrane drives
ATP synthesis. If the gradient becomes too steep, it can slow down the electron transport
chain. Conversely, if the gradient dissipates, ATP synthesis will stop.

Page 143 of 208


4. ATP/ADP Ratio:

The ratio of ATP to ADP is a key regulatory factor. High ATP levels inhibit oxidative
phosphorylation, while high ADP levels stimulate it. This feedback mechanism ensures that
ATP is synthesized only when needed.

5. Allosteric Regulation of Enzymes:

Certain enzymes involved in oxidative phosphorylation can be regulated allosterically. For


example, ATP synthase activity can be influenced by ADP and Pi concentrations.

6. Calcium Levels:

Calcium ions can stimulate the activity of certain dehydrogenases in the Krebs cycle (e.g.,
isocitrate dehydrogenase and alpha-ketoglutarate dehydrogenase), increasing NADH
production and thereby enhancing oxidative phosphorylation.

7. Hormonal Regulation:

Hormones like insulin can influence the rate of oxidative phosphorylation by regulating the
availability of substrates and enzymes involved in the process.

8. Regulation by Reactive Oxygen Species (ROS):

The production of reactive oxygen species (ROS) is a byproduct of oxidative phosphorylation.


High levels of ROS can cause damage to the mitochondrial membrane and proteins, leading
to feedback inhibition of the ETC to prevent excessive ROS production.

9. Inhibitors and Uncouplers:

Natural Inhibitors: Certain metabolites can act as natural inhibitors of ETC complexes (e.g.,
high levels of ATP inhibiting cytochrome c oxidase).

Chemical Inhibitors: Compounds like cyanide and carbon monoxide inhibit cytochrome c
oxidase, halting electron transport and ATP synthesis.

Page 144 of 208


Uncouplers: Chemicals like dinitrophenol (DNP) dissipate the proton gradient by allowing
protons to re-enter the mitochondrial matrix without passing through ATP synthase, thereby
uncoupling electron transport from ATP synthesis.

10. Mitochondrial Dynamics:

Mitochondria constantly undergo fission and fusion, processes that can influence their
function and efficiency. Changes in mitochondrial morphology can affect oxidative
phosphorylation.

Summary of Regulation of Oxidative Phosphorylation:

Substrate Availability: ADP, Pi, NADH, and FADH2 levels regulate the process.

Oxygen Concentration: Essential for the terminal step of electron transport.

Proton Gradient: Drives ATP synthesis; its balance is crucial.

ATP/ADP Ratio: Feedback mechanism to match ATP production with demand.

Allosteric Enzyme Regulation: Influences key enzymes in the pathway.

Calcium Levels: Enhance dehydrogenase activity in the Krebs cycle.

Hormonal Influence: Modulates substrate and enzyme availability.

ROS Regulation: Prevents excessive ROS damage.

Inhibitors and Uncouplers: Affect the efficiency of oxidative phosphorylation.

Mitochondrial Dynamics: Structural changes affect functionality.

Understanding these regulatory mechanisms ensures a comprehensive grasp of how cells


maintain energy homeostasis and respond to varying metabolic demands.

Page 145 of 208


8. Biosynthesis
8.1. Carbohydrate Biosynthesis in Plants and Bacteria

Carbohydrate biosynthesis is a fundamental biological process in both plants and bacteria,


allowing these organisms to produce essential molecules like glucose, starch, and glycogen.
This process is critical for energy storage, structural integrity, and various metabolic functions.

Carbohydrate Biosynthesis in Plants

Photosynthesis:

Photosynthesis is the primary process by which plants synthesize carbohydrates. It occurs in


the chloroplasts and involves two main stages: the light-dependent reactions and the Calvin
cycle (light-independent reactions).

1. Light-Dependent Reactions:

o Occur in the thylakoid membranes of chloroplasts.

o Capture energy from sunlight to produce ATP and NADPH.

o Water is split to release oxygen.

2. Calvin Cycle (C3 Pathway):

o Occurs in the stroma of chloroplasts.

o Uses ATP and NADPH from the light-dependent reactions to convert carbon
dioxide (CO2) into glucose.

o Key steps include carbon fixation, reduction phase, carbohydrate formation,


and regeneration of ribulose-1,5-bisphosphate (RuBP).

Page 146 of 208


Calvin Cycle Details:

 Carbon Fixation: CO2 is fixed by the enzyme ribulose-1,5-bisphosphate


carboxylase/oxygenase (RuBisCO) to form 3-phosphoglycerate (3-PGA).

 Reduction Phase: 3-PGA is reduced to glyceraldehyde-3-phosphate (G3P) using


ATP and NADPH.

 Carbohydrate Formation: Two molecules of G3P are used to form glucose and other
carbohydrates.

 Regeneration of RuBP: Remaining G3P molecules are used to regenerate RuBP,


allowing the cycle to continue.

Sucrose and Starch Biosynthesis:

 Sucrose: Synthesized in the cytoplasm from G3P, transported to different parts of the
plant for energy and storage.

 Starch: Synthesized in the chloroplasts from ADP-glucose, stored in plastids as an


energy reserve.

Page 147 of 208


Figure 12: Calcin cycle in plants

Carbohydrate Biosynthesis in Bacteria

Glycolysis and Gluconeogenesis:

Bacteria use glycolysis and gluconeogenesis for carbohydrate metabolism, with


gluconeogenesis playing a key role in carbohydrate biosynthesis.

1. Glycolysis:

o Breaks down glucose to pyruvate, generating ATP and NADH.

Page 148 of 208


o Key pathway for energy production.

2. Gluconeogenesis:

o Synthesizes glucose from non-carbohydrate precursors like pyruvate, lactate,


and glycerol.

o Shares several enzymes with glycolysis but has distinct enzymes for bypassing
irreversible steps of glycolysis.

o Key enzymes include pyruvate carboxylase, phosphoenolpyruvate


carboxykinase (PEPCK), fructose-1,6-bisphosphatase, and glucose-6-
phosphatase.

Polysaccharide Biosynthesis:

Bacteria synthesize polysaccharides for structural and storage purposes.

1. Peptidoglycan:

o Essential component of bacterial cell walls.

o Synthesized from UDP-N-acetylglucosamine (UDP-GlcNAc) and UDP-N-


acetylmuramic acid (UDP-MurNAc).

o Cross-linking of peptide chains provides structural integrity.

2. Glycogen:

o Storage polysaccharide similar to starch in plants.

o Synthesized from ADP-glucose by glycogen synthase.

o Branching enzyme introduces α-1,6 linkages for a branched structure.

Page 149 of 208


3. Exopolysaccharides:

o Secreted by bacteria to form biofilms and protect against environmental stress.

o Synthesized from nucleotide sugars like UDP-glucose and GDP-mannose.

o Examples include alginate, xanthan, and cellulose.

Summary of Carbohydrate Biosynthesis:

In Plants:

 Photosynthesis (light-dependent reactions and Calvin cycle) synthesizes glucose.

 Sucrose and starch are the primary storage forms of carbohydrates.

In Bacteria:

 Glycolysis and gluconeogenesis manage carbohydrate metabolism.

 Polysaccharides like peptidoglycan, glycogen, and exopolysaccharides are


synthesized for structural and storage purposes.

Understanding these pathways provides insights into how organisms produce and utilize
carbohydrates for energy, growth, and survival.

8.2. Photosynthetic Carbohydrate Synthesis

Photosynthetic carbohydrate synthesis is the process by which plants, algae, and certain
bacteria convert carbon dioxide (CO2) and water into carbohydrates using the energy from
sunlight. This process primarily takes place in the chloroplasts of plant cells and involves two
main stages: the light-dependent reactions and the Calvin cycle.

Page 150 of 208


1. Light-Dependent Reactions

These reactions occur in the thylakoid membranes of chloroplasts and require light to produce
ATP and NADPH, which are then used in the Calvin cycle.

Key Steps:

 Photon Absorption: Chlorophyll and other pigments absorb light, exciting electrons
to a higher energy level.

 Water Splitting (Photolysis): Water molecules are split to release electrons, protons,
and oxygen (O2).

 Electron Transport Chain: Excited electrons move through a series of proteins in the
thylakoid membrane, creating a proton gradient.

 ATP Synthesis: Protons flow back into the thylakoid lumen through ATP synthase,
generating ATP.

 NADPH Formation: Electrons ultimately reduce NADP+ to NADPH.

2. Calvin Cycle (C3 Pathway)

The Calvin cycle takes place in the stroma of chloroplasts and does not require light directly.
It uses ATP and NADPH from the light-dependent reactions to fix CO2 into organic molecules,
ultimately producing glucose.

Key Steps:

1. Carbon Fixation:

o CO2 is fixed by the enzyme ribulose-1,5-bisphosphate carboxylase/oxygenase


(RuBisCO).

Page 151 of 208


o CO2 combines with ribulose-1,5-bisphosphate (RuBP) to form two molecules
of 3-phosphoglycerate (3-PGA).

2. Reduction Phase:

o 3-PGA is phosphorylated by ATP to form 1,3-bisphosphoglycerate (1,3-BPG).

o 1,3-BPG is reduced by NADPH to glyceraldehyde-3-phosphate (G3P).

3. Carbohydrate Formation:

o Some G3P molecules are used to synthesize glucose and other carbohydrates.

o Glucose can be further polymerized to form starch or sucrose.

4. Regeneration of RuBP:

o Remaining G3P molecules are used to regenerate RuBP, enabling the cycle to
continue.

o ATP is used in the regeneration process.

Summary of Photosynthetic Carbohydrate Synthesis:

1. Light-Dependent Reactions:

o Capture light energy to produce ATP and NADPH.

o Occur in the thylakoid membranes.

o Involve water splitting, electron transport, and proton gradient formation.

2. Calvin Cycle (C3 Pathway):

o Fix CO2 into organic molecules using ATP and NADPH.

o Occurs in the stroma.

o Produces glucose, which can be stored as starch or transported as sucrose.

Page 152 of 208


Key Points:

 Starch Synthesis: In the chloroplasts, glucose units are polymerized to form starch,
a storage polysaccharide.

 Sucrose Synthesis: In the cytoplasm, G3P is converted to sucrose, which is


transported to different parts of the plant for energy and growth.

C4 and CAM Pathways:

Some plants have adaptations to enhance photosynthetic efficiency under certain conditions.

 C4 Pathway:

o Found in plants like maize and sugarcane.

o CO2 is initially fixed into a four-carbon compound (oxaloacetate) in mesophyll


cells.

o Oxaloacetate is converted to malate, which is transported to bundle-sheath


cells where CO2 is released for the Calvin cycle.

 CAM Pathway:

o Found in plants like cacti and succulents.

o CO2 is fixed at night into a four-carbon compound (malate) and stored in


vacuoles.

o During the day, CO2 is released from malate for the Calvin cycle, allowing the
stomata to remain closed and reduce water loss.

Page 153 of 208


8.3. Photorespiration and C4 and CAM Pathways

Photorespiration is a process in plants where the enzyme ribulose-1,5-bisphosphate


carboxylase/oxygenase (RuBisCO) oxygenates RuBP, leading to a wasteful pathway that
reduces the efficiency of photosynthesis. C4 and CAM pathways are adaptations in some
plants to minimize photorespiration and optimize photosynthesis under specific environmental
conditions.

Photorespiration

Process:

 Occurs when RuBisCO reacts with O2 instead of CO2.

 Produces one molecule of 3-phosphoglycerate (3-PGA) and one molecule of 2-


phosphoglycolate.

 2-phosphoglycolate is recycled through a series of reactions involving the chloroplast,


peroxisome, and mitochondrion.

 Results in the release of CO2 and NH3, and consumes ATP and reducing power
without producing glucose.

Consequences:

 Reduces the efficiency of photosynthesis by diverting RuBP and energy away from
carbohydrate production.

 More prevalent under conditions of high light intensity, high temperatures, and low
CO2 concentrations.

Page 154 of 208


C4 Pathway

Adaptation to Minimize Photorespiration:

 Found in plants like maize, sugarcane, and sorghum.

 Separates initial CO2 fixation and the Calvin cycle into different cell types (mesophyll
and bundle-sheath cells).

Key Steps:

1. CO2 Fixation in Mesophyll Cells:

o CO2 is fixed into a four-carbon compound, oxaloacetate, by the enzyme


phosphoenolpyruvate carboxylase (PEP carboxylase), which has a higher
affinity for CO2 than RuBisCO and does not react with O2.

o Oxaloacetate is converted to malate.

2. Transport to Bundle-Sheath Cells:

o Malate is transported to bundle-sheath cells, which are tightly packed around


the leaf veins.

3. CO2 Release in Bundle-Sheath Cells:

o Malate is decarboxylated to release CO2, which is then fixed by RuBisCO in


the Calvin cycle.

o Pyruvate, the byproduct, is transported back to mesophyll cells and converted


to PEP, completing the cycle.

Page 155 of 208


Benefits:

 Concentrates CO2 around RuBisCO, reducing photorespiration.

 Enhances photosynthetic efficiency under high light intensity, high temperatures, and
drought conditions.

CAM Pathway

Adaptation to Arid Conditions:

 Found in succulent plants like cacti, agaves, and some orchids.

 Temporally separates CO2 fixation and the Calvin cycle to reduce water loss.

Key Steps:

1. Nighttime CO2 Fixation:

o Stomata open at night to minimize water loss.

o CO2 is fixed into oxaloacetate by PEP carboxylase and then converted to


malate.

o Malate is stored in vacuoles as malic acid.

2. Daytime CO2 Release:

o Stomata close during the day to conserve water.

o Malic acid is decarboxylated to release CO2 for the Calvin cycle.

o CO2 is fixed by RuBisCO in the Calvin cycle.

Benefits:

 Allows photosynthesis to occur with minimal water loss.

 Efficient under arid conditions with high temperatures and intense sunlight.

Page 156 of 208


Summary:

 Photorespiration is a wasteful process that occurs when RuBisCO reacts with O2


instead of CO2, reducing the efficiency of photosynthesis.

 C4 Pathway:

o Adaptation in certain plants to minimize photorespiration by spatially separating


CO2 fixation and the Calvin cycle.

o CO2 is initially fixed in mesophyll cells and then transported to bundle-sheath


cells where the Calvin cycle occurs.

o Found in plants like maize and sugarcane, effective under high light intensity
and high temperatures.

 CAM Pathway:

o Adaptation in succulent plants to minimize water loss by temporally separating


CO2 fixation and the Calvin cycle.

o CO2 is fixed at night and stored as malate, then used during the day when
stomata are closed.

o Found in plants like cacti and agaves, effective under arid conditions.

These adaptations illustrate how plants have evolved to optimize photosynthesis and reduce
photorespiration under different environmental conditions.

Biosynthesis of Starch and Sucrose

In plants, the biosynthesis of starch and sucrose involves converting simple sugars produced
during photosynthesis into these storage and transport forms of carbohydrates. Both starch
and sucrose play crucial roles in plant metabolism and energy management.

Page 157 of 208


Starch Biosynthesis

Starch is a storage polysaccharide in plants, composed of two types of molecules: amylose


and amylopectin. It is synthesized in the chloroplasts of leaves and the amyloplasts of storage
tissues (e.g., tubers and seeds).

Key Steps in Starch Biosynthesis:

1. Glucose-1-Phosphate Formation:

o Glucose-6-phosphate is converted to glucose-1-phosphate by the enzyme


phosphoglucomutase.

2. ADP-Glucose Synthesis:

o Glucose-1-phosphate reacts with ATP to form ADP-glucose, catalyzed by the


enzyme ADP-glucose pyrophosphorylase.

o This step is regulated and serves as a key control point in starch synthesis.

3. Polymerization:

o ADP-glucose acts as a glucose donor for the polymerization of glucose units.

o Starch Synthase: Catalyzes the addition of glucose units to the growing starch
chain, forming amylose.

o Branching Enzyme: Introduces α-1,6 linkages, creating branch points in the


amylopectin molecule.

4. Starch Granule Formation:

Page 158 of 208


o The newly formed amylose and amylopectin molecules aggregate to form semi-
crystalline starch granules.

Sucrose Biosynthesis

Sucrose is a disaccharide composed of glucose and fructose. It is synthesized in the


cytoplasm of plant cells and serves as the primary transport sugar, moving from source
tissues (e.g., leaves) to sink tissues (e.g., roots, fruits).

Key Steps in Sucrose Biosynthesis:

1. Formation of UDP-Glucose:

o Glucose-6-phosphate is converted to glucose-1-phosphate by


phosphoglucomutase.

o Glucose-1-phosphate reacts with UTP to form UDP-glucose, catalyzed by


UDP-glucose pyrophosphorylase.

2. Formation of Fructose-6-Phosphate:

o Fructose-6-phosphate is produced from glucose-6-phosphate via


isomerization, catalyzed by phosphoglucose isomerase.

3. Formation of Fructose-1,6-Bisphosphate:

o Fructose-6-phosphate is phosphorylated to fructose-1,6-bisphosphate by


phosphofructokinase.

4. Formation of Fructose-6-Phosphate:

o Fructose-1,6-bisphosphate is dephosphorylated to fructose-6-phosphate by


fructose-1,6-bisphosphatase.

Page 159 of 208


5. Sucrose-6-Phosphate Formation:

o UDP-glucose reacts with fructose-6-phosphate to form sucrose-6-phosphate,


catalyzed by sucrose-phosphate synthase.

6. Formation of Sucrose:

o Sucrose-6-phosphate is dephosphorylated to sucrose by sucrose-phosphate


phosphatase.

Summary of Biosynthesis:

Starch:

 Synthesized in chloroplasts (leaves) and amyloplasts (storage tissues).

 Involves conversion of glucose-1-phosphate to ADP-glucose, followed by


polymerization into amylose and amylopectin.

 Starch granules form from the aggregation of amylose and amylopectin.

Sucrose:

 Synthesized in the cytoplasm.

 Involves the conversion of glucose-6-phosphate to UDP-glucose and fructose-6-


phosphate.

 UDP-glucose and fructose-6-phosphate react to form sucrose-6-phosphate, which is


then dephosphorylated to sucrose.

These biosynthetic pathways highlight the intricate regulation and coordination required for
plants to produce and store energy efficiently.

Page 160 of 208


Figure 13: Biosynthesis of starch and sucrose

8.4. Synthesis of Plant and Bacterial Cell Wall Polysaccharides

Cell wall polysaccharides are crucial for maintaining structural integrity, protecting against
environmental stress, and regulating cell growth in both plants and bacteria. The synthesis
pathways for these polysaccharides are complex and involve multiple enzymatic steps.

Synthesis of Plant Cell Wall Polysaccharides

Plant cell walls are composed mainly of cellulose, hemicellulose, and pectin. Each component
is synthesized through distinct pathways and then integrated into the cell wall matrix.

Page 161 of 208


1. Cellulose Synthesis:

Cellulose is a linear polymer of β-1,4-linked glucose units and is the primary structural
component of the plant cell wall.

Key Steps:

 Cellulose Synthase Complex: Cellulose is synthesized at the plasma membrane by


a cellulose synthase complex (CSC). This complex consists of multiple cellulose
synthase (CESA) proteins.

 Glucose Polymerization: UDP-glucose serves as the substrate, and the CESA


enzymes polymerize glucose units into cellulose chains.

 Microfibril Formation: Multiple cellulose chains associate through hydrogen bonding


to form microfibrils, which are extruded into the extracellular space and integrated into
the cell wall.

2. Hemicellulose Synthesis:

Hemicelluloses are branched polysaccharides, such as xyloglucan, xylan, and mannans, that
cross-link cellulose microfibrils.

Key Steps:

 Nucleotide Sugars: Hemicellulose synthesis begins with nucleotide sugars like UDP-
glucose, UDP-xylose, and GDP-mannose.

 Glycosyltransferases: Enzymes called glycosyltransferases transfer sugar residues


from nucleotide sugars to form the backbone and side chains of hemicelluloses.

Page 162 of 208


 Golgi Apparatus: Synthesis primarily occurs in the Golgi apparatus, where
hemicelluloses are assembled and then transported to the cell wall in vesicles.

3. Pectin Synthesis:

Pectins are a group of complex polysaccharides rich in galacturonic acid, which contribute to
the porosity and charge properties of the cell wall.

Key Steps:

 Nucleotide Sugars: Precursors like UDP-galacturonic acid and UDP-rhamnose are


used in pectin synthesis.

 Glycosyltransferases: Various glycosyltransferases synthesize different pectin


domains, such as homogalacturonan, rhamnogalacturonan I, and
rhamnogalacturonan II.

 Golgi Apparatus: Similar to hemicelluloses, pectins are synthesized in the Golgi


apparatus and transported to the cell wall in vesicles.

Synthesis of Bacterial Cell Wall Polysaccharides

Bacterial cell walls are primarily composed of peptidoglycan, a polymer consisting of sugars
and amino acids, which provides structural strength and shape to the cell.

1. Peptidoglycan Synthesis:

Peptidoglycan is a mesh-like structure made of glycan chains cross-linked by peptides.

Key Steps:

 UDP-N-Acetylglucosamine (UDP-GlcNAc) Formation:

Page 163 of 208


o UDP-GlcNAc is synthesized from fructose-6-phosphate through a series of
enzymatic reactions.

 UDP-N-Acetylmuramic Acid (UDP-MurNAc) Formation:

o UDP-GlcNAc is converted to UDP-MurNAc through the addition of a lactyl


group.

 Peptide Addition:

o A peptide chain is added to UDP-MurNAc, forming UDP-MurNAc-pentapeptide.

 Lipid Carrier Involvement:

o The UDP-MurNAc-pentapeptide is transferred to a lipid carrier (bactoprenol) in


the membrane, forming lipid I.

o UDP-GlcNAc is then added to form lipid II.

 Polymerization:

o Lipid II is flipped to the outer side of the cytoplasmic membrane, where it is


polymerized into glycan chains by transglycosylase enzymes.

o Cross-linking of glycan chains by transpeptidase enzymes forms the


peptidoglycan mesh.

2. Teichoic Acids and Other Polysaccharides:

In Gram-positive bacteria, teichoic acids are also essential components of the cell wall.

Key Steps:

 Glycerol or Ribitol Phosphate Backbone:


Page 164 of 208
o Synthesized in the cytoplasm from precursors like glycerol-3-phosphate or
ribitol-5-phosphate.

 Polymerization and Modification:

o The backbone is polymerized and modified with D-alanine or sugars.

 Attachment to Peptidoglycan:

o Teichoic acids are covalently linked to the peptidoglycan layer or to membrane


lipids in the case of lipoteichoic acids.

Summary of Cell Wall Polysaccharide Synthesis:

In Plants:

 Cellulose: Synthesized by cellulose synthase complexes at the plasma membrane.

 Hemicellulose: Synthesized in the Golgi apparatus by glycosyltransferases, then


transported to the cell wall.

 Pectin: Synthesized in the Golgi apparatus, involving various glycosyltransferases,


and transported to the cell wall.

In Bacteria:

 Peptidoglycan: Synthesized from UDP-GlcNAc and UDP-MurNAc, involving lipid


carriers and polymerization by transglycosylase and transpeptidase enzymes.

 Teichoic Acids: Synthesized in the cytoplasm and linked to peptidoglycan or


membrane lipids.

These biosynthetic pathways highlight the complexity and precision required for constructing
robust cell walls, essential for the survival and functionality of both plants and bacteria.

Page 165 of 208


Figure 14: Membrane steps of the bacterial peptidoglycan synthesis pathway

Figure 15: Synthesis of plant cell wall polysaccharides

Page 166 of 208


8.5. Lipid biosynthesis
8.5.1. Biosynthesis of Fatty Acids and Triglycerides

Fatty acids and triglycerides are essential components of cellular structures and energy
storage in both plants and animals. The biosynthesis of these molecules involves distinct but
interconnected pathways.

Fatty Acid Biosynthesis

Fatty acid biosynthesis primarily occurs in the cytoplasm of cells, involving the stepwise
addition of two-carbon units to a growing fatty acid chain. The process is catalyzed by the
fatty acid synthase (FAS) complex.

Key Steps in Fatty Acid Biosynthesis:

1. Initiation:

o Acetyl-CoA Formation: Acetyl-CoA is produced from carbohydrates via


glycolysis and pyruvate dehydrogenase, or from amino acids and fatty acids.

o Transport to Cytoplasm: In animals, acetyl-CoA is transported from the


mitochondria to the cytoplasm as citrate, which is then cleaved back to acetyl-
CoA and oxaloacetate by ATP-citrate lyase.

2. Malonyl-CoA Formation:

o Acetyl-CoA Carboxylase (ACC): Acetyl-CoA is carboxylated to form malonyl-


CoA. This is the rate-limiting step and is tightly regulated.

o Biotin: This reaction requires biotin as a coenzyme.

Page 167 of 208


3. Elongation Cycle:

o Fatty Acid Synthase (FAS) Complex: A multifunctional enzyme complex in


animals (or a series of separate enzymes in plants and bacteria) carries out the
elongation process.

o Loading: Acetyl-CoA and malonyl-CoA are transferred to acyl carrier protein


(ACP) to form acetyl-ACP and malonyl-ACP.

o Condensation: Acetyl-ACP and malonyl-ACP undergo a condensation


reaction to form β-ketoacyl-ACP, releasing CO2.

o Reduction: β-Ketoacyl-ACP is reduced to β-hydroxyacyl-ACP by NADPH-


dependent β-ketoacyl-ACP reductase.

o Dehydration: β-Hydroxyacyl-ACP is dehydrated to form enoyl-ACP by β-


hydroxyacyl-ACP dehydratase.

o Reduction: Enoyl-ACP is reduced to acyl-ACP by NADPH-dependent enoyl-


ACP reductase.

o This cycle repeats, adding two-carbon units (from malonyl-CoA) to the growing
fatty acid chain until the desired chain length is achieved.

4. Termination:

o Thioesterase: The completed fatty acid is released from the FAS complex by
thioesterase, resulting in a free fatty acid, typically palmitate (C16:0).

Triglyceride Biosynthesis

Triglycerides (triacylglycerols) are composed of three fatty acids esterified to a glycerol


backbone. The biosynthesis occurs in the endoplasmic reticulum and cytoplasm.

Page 168 of 208


Key Steps in Triglyceride Biosynthesis:

1. Glycerol-3-Phosphate Formation:

o Glycolysis: Dihydroxyacetone phosphate (DHAP) from glycolysis is reduced


to glycerol-3-phosphate by glycerol-3-phosphate dehydrogenase.

o Glycerol Kinase: Alternatively, glycerol can be phosphorylated to glycerol-3-


phosphate by glycerol kinase (mainly in the liver and kidneys).

2. Acylation:

o Glycerol-3-Phosphate Acyltransferase (GPAT): Adds a fatty acyl-CoA to


glycerol-3-phosphate, forming lysophosphatidic acid (LPA).

o Lysophosphatidic Acid Acyltransferase (LPAAT): Adds a second fatty acyl-


CoA to LPA, forming phosphatidic acid (PA).

3. Phosphatidic Acid Conversion:

o Phosphatidic Acid Phosphatase (PAP): Dephosphorylates PA to form


diacylglycerol (DAG).

4. Final Acylation:

o Diacylglycerol Acyltransferase (DGAT): Adds a third fatty acyl-CoA to DAG,


forming triacylglycerol (TAG or triglyceride).

Regulation of Fatty Acid and Triglyceride Biosynthesis

The biosynthesis of fatty acids and triglycerides is tightly regulated at several levels:

1. Hormonal Regulation:

o Insulin: Stimulates fatty acid and triglyceride synthesis by activating acetyl-


CoA carboxylase and promoting glucose uptake and glycolysis.

Page 169 of 208


o Glucagon and Epinephrine: Inhibit fatty acid synthesis by phosphorylating
and inactivating acetyl-CoA carboxylase through AMP-activated protein kinase
(AMPK).

2. Nutritional Status:

o Fed State: High levels of glucose and insulin promote fatty acid and triglyceride
synthesis.

o Fasting State: Low insulin and high glucagon levels promote fatty acid
oxidation and inhibit synthesis.

3. Allosteric Regulation:

o Citrate: Activates acetyl-CoA carboxylase, indicating high availability of acetyl-


CoA.

o Palmitoyl-CoA: Inhibits acetyl-CoA carboxylase, indicating high levels of end


products.

Summary:

Fatty Acid Biosynthesis:

 Involves the carboxylation of acetyl-CoA to malonyl-CoA.

 Elongation occurs via the fatty acid synthase complex, adding two-carbon units from
malonyl-CoA.

 Produces palmitate as the primary product.

Triglyceride Biosynthesis:

 Glycerol-3-phosphate is acylated to form lysophosphatidic acid, then phosphatidic


acid, and finally diacylglycerol.

Page 170 of 208


 Diacylglycerol is acylated to form triglycerides.

These pathways provide the building blocks for cellular membranes and energy storage
molecules essential for cell function and survival.

Figure 16: Biosynthesis of fatty acids and triglycerides

Page 171 of 208


8.5.2. Biosynthesis of Membrane Phospholipids

Phospholipids are essential components of cellular membranes, contributing to membrane


structure, fluidity, and function. The biosynthesis of phospholipids involves the formation of
key intermediates and the addition of various head groups.

Key Types of Phospholipids

1. Phosphatidic Acid (PA): The precursor to most phospholipids.

2. Phosphatidylcholine (PC): Major component of cell membranes.

3. Phosphatidylethanolamine (PE): Important for membrane integrity and curvature.

4. Phosphatidylserine (PS): Involved in cell signaling and apoptosis.

5. Phosphatidylinositol (PI): Involved in signaling pathways.

6. Cardiolipin (CL): Specific to mitochondrial membranes.

Steps in Phospholipid Biosynthesis

1. Formation of Phosphatidic Acid (PA):

PA is the central intermediate in the synthesis of other phospholipids.

 Glycerol-3-Phosphate Acylation:

o Glycerol-3-phosphate (G3P): Derived from glycolysis or gluconeogenesis.

o GPAT (Glycerol-3-phosphate acyltransferase): Catalyzes the addition of a


fatty acyl-CoA to G3P, forming lysophosphatidic acid (LPA).

o AGPAT (1-acylglycerol-3-phosphate acyltransferase): Adds another fatty


acyl-CoA to LPA, forming PA.

Page 172 of 208


2. Conversion of PA to Diacylglycerol (DAG):

 Phosphatidic Acid Phosphatase (PAP): Dephosphorylates PA to produce DAG, a


key intermediate for the synthesis of triacylglycerols and phospholipids.

3. Synthesis of Phosphatidylcholine (PC) and Phosphatidylethanolamine (PE):

 CDP-Choline Pathway (Kennedy Pathway) for PC:

o Choline Phosphorylation: Choline is phosphorylated by choline kinase to


form phosphocholine.

o Activation: Phosphocholine reacts with CTP to form CDP-choline, catalyzed


by CTP

cytidylyltransferase.

o Final Step: CDP-choline reacts with DAG to form PC, catalyzed by CDP-
choline

Choline phosphotransferase.

 CDP-Ethanolamine Pathway for PE:

o Ethanolamine Phosphorylation: Ethanolamine is phosphorylated by


ethanolamine kinase to form phosphoethanolamine.

o Activation: Phosphoethanolamine reacts with CTP to form CDP-


ethanolamine.

o Final Step: CDP-ethanolamine reacts with DAG to form PE, catalyzed by CDP-
ethanolamine

Ethanolamine phosphotransferase.

Page 173 of 208


4. Synthesis of Phosphatidylserine (PS):

 Base Exchange Reaction:

o PS is synthesized by exchanging the head group of PC or PE with serine,


catalyzed by phosphatidylserine synthase (PSS).

5. Synthesis of Phosphatidylinositol (PI):

 CDP-Diacylglycerol Pathway:

o Formation of CDP-DAG: PA reacts with CTP to form CDP-diacylglycerol


(CDP-DAG).

o Inositol Addition: CDP-DAG reacts with inositol to form PI, catalyzed by


phosphatidylinositol synthase.

6. Synthesis of Cardiolipin (CL):

 Mitochondrial Specific Synthesis:

o Formation of CDP-DAG: Similar to PI synthesis, occurs in mitochondria.

o Reaction with Glycerol-3-Phosphate: CDP-DAG reacts with another


molecule of PA to form CL, catalyzed by cardiolipin synthase.

Regulation of Phospholipid Biosynthesis

Phospholipid biosynthesis is regulated at multiple levels to ensure membrane homeostasis


and adaptation to changing cellular needs:

Page 174 of 208


1. Enzyme Activation/Inactivation:

o Phosphorylation/Dephosphorylation: Key enzymes such as CTP

cytidylyltransferase are regulated by phosphorylation, which affects their activity and


localization.

2. Substrate Availability:

o Fatty Acyl-CoA and Glycerol-3-Phosphate: The availability of these


substrates can regulate the rate of PA and DAG formation.

3. Transcriptional Regulation:

o Gene Expression: The expression of genes encoding enzymes involved in


phospholipid biosynthesis can be regulated by transcription factors in response
to cellular signals and environmental conditions.

4. Feedback Inhibition:

o Product Inhibition: End products like PC can inhibit upstream enzymes to


prevent overproduction.

Summary:

Phosphatidic Acid (PA): Central intermediate, formed from glycerol-3-phosphate and fatty
acyl-CoA. Diacylglycerol (DAG): Intermediate for PC, PE, and PS synthesis.
Phosphatidylcholine (PC): Synthesized via CDP-choline pathway.
Phosphatidylethanolamine (PE): Synthesized via CDP-ethanolamine pathway.
Phosphatidylserine (PS): Formed by base exchange with PC or PE. Phosphatidylinositol
(PI): Synthesized from CDP-DAG and inositol. Cardiolipin (CL): Synthesized in mitochondria
from CDP-DAG and PA.

Page 175 of 208


These pathways underscore the complexity and regulation required to maintain membrane
structure and function across different cellular environments.

Figure 17: Biosynthesis of membrane phospholipids

8.5.3. Biosynthesis of Cholesterol, Steroids, and Isoprenoids

Cholesterol, steroids, and isoprenoids are diverse classes of molecules crucial for various
biological functions, including membrane structure, signaling, and hormone synthesis. Their
biosynthesis involves complex pathways that start from simple precursors and undergo
multiple enzymatic reactions.

Biosynthesis of Cholesterol

Cholesterol is a sterol molecule synthesized in the endoplasmic reticulum of cells, primarily


in the liver and intestines. It serves as a vital component of cell membranes and is a precursor
for steroid hormones and bile acids.

Page 176 of 208


Key Steps in Cholesterol Biosynthesis:

1. Acetyl-CoA to Mevalonate:

o Acetyl-CoA Formation: Acetyl-CoA is produced from citrate in the


mitochondria and transported to the cytoplasm.

o Formation of Mevalonate: Acetyl-CoA is converted to mevalonate by a series


of enzymatic reactions catalyzed by acetyl-CoA acetyltransferase, HMG-CoA
synthase, and HMG-CoA reductase (the rate-limiting step).

2. Mevalonate to Isoprene Units:

o Formation of Isopentenyl Pyrophosphate (IPP) and Dimethylallyl


Pyrophosphate (DMAPP): Mevalonate undergoes phosphorylation and
decarboxylation to form IPP and DMAPP, catalyzed by mevalonate kinase,
phosphomevalonate kinase, and mevalonate diphosphate decarboxylase.

3. Isoprenoid Chain Elongation:

o Formation of Geranyl Pyrophosphate (GPP) and Farnesyl Pyrophosphate


(FPP): IPP and DMAPP are condensed to form GPP (5-carbon) and
subsequently FPP (15-carbon), catalyzed by prenyltransferases.

4. Formation of Squalene:

o Condensation of FPP: Two molecules of FPP condense to form squalene, a


linear 30-carbon compound, catalyzed by squalene synthase.

5. Cholesterol Synthesis:

o Conversion of Squalene to Cholesterol: Squalene undergoes cyclization


and subsequent enzymatic modifications to form lanosterol, which is further

Page 177 of 208


processed to cholesterol through a series of steps involving several enzymes,
including lanosterol synthase and several cytochrome P450 enzymes.

Biosynthesis of Steroids

Steroids are a diverse group of molecules derived from cholesterol and play essential roles
as hormones (e.g., cortisol, testosterone, estrogen) and signaling molecules.

Key Steps in Steroid Biosynthesis:

1. Conversion of Cholesterol to Pregnenolone:

o Side Chain Cleavage: Cholesterol is converted to pregnenolone by


cholesterol side-chain cleavage enzyme (CYP11A1 or P450scc).

2. Formation of Steroid Hormones:

o Pregnenolone as a Precursor: Pregnenolone serves as a precursor for


various steroid hormones depending on the specific enzymes and tissues
involved.

o Examples: Cortisol, aldosterone, testosterone, estrogen, and progesterone


are synthesized in adrenal glands, gonads, and other steroidogenic tissues.

Biosynthesis of Isoprenoids

Isoprenoids are a large class of compounds derived from isopentenyl pyrophosphate (IPP)
and dimethylallyl pyrophosphate (DMAPP), which serve as building blocks for a wide range
of molecules including vitamins, hormones, and signaling molecules.

Page 178 of 208


Key Steps in Isoprenoid Biosynthesis:

1. Formation of IPP and DMAPP:

o Mevalonate Pathway: IPP and DMAPP are synthesized from acetyl-CoA via
the mevalonate pathway in the cytoplasm.

2. Formation of Geranyl Pyrophosphate (GPP) and Farnesyl Pyrophosphate (FPP):

o Condensation: IPP and DMAPP condense to form GPP (10-carbon) and


subsequently FPP (15-carbon), catalyzed by prenyltransferases.

3. Diverse Isoprenoids:

o Examples: Ubiquinone (Coenzyme Q), dolichol, heme A, heme B, and various


plant secondary metabolites (e.g., carotenoids, phytoene, phytofluene).

Regulation of Cholesterol and Isoprenoid Biosynthesis

The biosynthesis of cholesterol, steroids, and isoprenoids is tightly regulated at multiple


levels:

 Transcriptional Regulation: Enzymes involved in these pathways are regulated by


transcription factors that respond to cellular and hormonal signals.

 Post-Translational Modification: Enzyme activity can be modulated through


phosphorylation, proteolytic cleavage, or allosteric regulation.

 Feedback Inhibition: End products like cholesterol can inhibit key enzymes in earlier
steps, regulating their own synthesis.

Page 179 of 208


Summary

 Cholesterol: Synthesized from acetyl-CoA via the mevalonate pathway, crucial for
membrane structure and hormone synthesis.

 Steroids: Derived from cholesterol, serve as hormones and signaling molecules.

 Isoprenoids: Derived from IPP and DMAPP, involved in diverse functions including
electron transport, hormone synthesis, and antioxidant defense.

These pathways illustrate the intricate biochemical processes that cells employ to synthesize
essential molecules for cellular structure, function, and signaling.

Page 180 of 208


Figure 18: Biosynthesis of cholesterol,

Figure 19: Biosynthesis of isoprenoids in plants

Page 181 of 208


Page 182 of 208
8.6. Overview of Nitrogen Metabolism

Nitrogen metabolism in living organisms is crucial for the synthesis of biomolecules such as
amino acids, nucleotides, and various cofactors. It involves processes that assimilate,
incorporate, and recycle nitrogen-containing compounds. Here’s an overview of nitrogen
metabolism:

1. Nitrogen Fixation

Definition: The conversion of atmospheric nitrogen (N2) into ammonia (NH3) or ammonium
ions (NH4+) usable by living organisms.

 Biological Nitrogen Fixation: Carried out by nitrogen-fixing bacteria (e.g.,


Rhizobium, Azotobacter) and some archaea. These organisms possess nitrogenase
enzymes that reduce N2 to NH3.
 Industrial Nitrogen Fixation: Haber-Bosch process converts N2 and H2 into NH3
industrially, crucial for synthetic fertilizers.

2. Ammonium Assimilation

Definition: Incorporation of ammonium ions (NH4+) into organic molecules.

 Glutamine Synthesis: Glutamine synthetase catalyzes the ATP-dependent


condensation of glutamate and ammonia to form glutamine, a nitrogen donor in many
biosynthetic reactions.
 Glutamate Synthesis: Glutamate dehydrogenase and glutamate synthase convert α-
ketoglutarate and ammonia into glutamate, a central molecule in nitrogen metabolism.

Page 183 of 208


3. Nitrate Assimilation

Definition: Reduction of nitrate (NO3-) to ammonium ions (NH4+), then incorporation into
organic molecules.

 Nitrate Reduction: Nitrate reductase catalyzes the reduction of NO3- to nitrite (NO2-
), then nitrite reductase further reduces NO2- to NH4+.
 Ammonium Incorporation: NH4+ produced from nitrate reduction is assimilated into
glutamine and glutamate via similar pathways as for ammonium assimilation.

4. Amino Acid Biosynthesis

Definition: Synthesis of amino acids from intermediates of central metabolism (e.g.,


glycolysis, TCA cycle) and nitrogen donors.

 Transamination: Amino groups are transferred between amino acids and α-keto
acids, catalyzed by aminotransferases.
 Glutamate as a Central Molecule: Glutamate serves as a nitrogen donor in the
biosynthesis of many amino acids through the transamination process.

5. Nitrogen Recycling

Definition: Reutilization of nitrogen-containing compounds from cellular breakdown


processes.

 Urea Cycle: In animals, the urea cycle detoxifies ammonia generated from amino acid
catabolism into urea for excretion.
 Nitrogen-Containing Waste Products: Excreted in various forms depending on the
organism (e.g., ammonia, urea, uric acid).

Page 184 of 208


6. Regulation of Nitrogen Metabolism

Definition: Control mechanisms that maintain nitrogen homeostasis and respond to


environmental conditions.

 Feedback Inhibition: Key enzymes in nitrogen metabolism are regulated by end


products (e.g., glutamine, glutamate).
 Gene Expression Regulation: Transcription factors modulate the expression of
genes involved in nitrogen metabolism in response to nitrogen availability.
 Post-Translational Modification: Enzyme activity can be regulated by
phosphorylation, allosteric interactions, or proteolytic cleavage.

Significance of Nitrogen Metabolism

Nitrogen metabolism is essential for:

 Protein Synthesis: Amino acids are building blocks of proteins.


 Nucleotide Synthesis: Nitrogenous bases (purines and pyrimidines) are essential for
DNA and RNA.
 Energy Metabolism: Nitrogen-containing compounds participate in electron transport
and energy storage (e.g., ATP).

Summary

Nitrogen metabolism is a complex network of biochemical pathways that ensures organisms


have sufficient nitrogen for growth and maintenance. From nitrogen fixation to amino acid
biosynthesis and nitrogen recycling, these processes are tightly regulated to balance nitrogen
availability with metabolic demands in various environmental conditions.

Page 185 of 208


Figure 20: Biosynthesis of Amino Acids

8.7. Biosynthesis of Amino Acids

Amino acids are fundamental building blocks of proteins and play essential roles in numerous
biochemical processes within living organisms. They are synthesized through complex
pathways that vary depending on the specific amino acid and the metabolic state of the
organism. Here's an overview of the biosynthesis of amino acids:

1. General Pathways

a. Central Metabolic Intermediates

Amino acids are synthesized using intermediates from central metabolic pathways such as
glycolysis, the citric acid cycle (TCA cycle), and the pentose phosphate pathway.

b. Nitrogen Donors

Ammonia (NH3) or amino groups derived from glutamate or glutamine serve as nitrogen
donors in amino acid biosynthesis.

Page 186 of 208


2. Classification

Amino acids can be classified into different groups based on their biosynthetic pathways:

a. Non-Essential Amino Acids

 Synthesized de novo by most organisms.


 Examples include alanine, serine, glycine, and glutamine.

b. Essential Amino Acids

 Cannot be synthesized by the organism and must be obtained from the diet.
 Examples include lysine, tryptophan, phenylalanine, and leucine.

3. Biosynthetic Pathways

a. Glycine, Serine, and Cysteine

 Glycine: Derived from serine through removal of a methyl group.


 Serine: Derived from 3-phosphoglycerate via a series of enzymatic steps.
 Cysteine: Derived from serine through the addition of sulfur.

b. Glutamate and Glutamine

 Glutamate: Synthesized from α-ketoglutarate via glutamate dehydrogenase or


transamination.
 Glutamine: Synthesized from glutamate and ammonia via glutamine synthetase.

c. Aspartate and Asparagine

 Aspartate: Synthesized from oxaloacetate via transamination or reduction.


 Asparagine: Synthesized from aspartate and glutamine.

Page 187 of 208


d. Alanine

 Synthesized from pyruvate via transamination with glutamate.

e. Arginine

 Derived from glutamate and aspartate via several enzymatic steps.

f. Histidine

 Synthesized from phosphoribosyl pyrophosphate (PRPP) and ATP, incorporating


nitrogen from glutamine.

g. Proline

 Derived from glutamate via a series of enzymatic reactions.

h. Threonine

 Synthesized from aspartate-derived oxaloacetate and glycine.

i. Tyrosine

 Derived from phenylalanine via hydroxylation.

4. Regulation

Amino acid biosynthesis is tightly regulated to ensure balanced cellular metabolism and
respond to environmental cues:

 Feedback Inhibition: End products of amino acid biosynthesis pathways often inhibit
the enzymes catalyzing their own synthesis.

Page 188 of 208


 Transcriptional Regulation: Gene expression of enzymes involved in amino acid
biosynthesis is regulated by transcription factors in response to metabolic needs.
 Post-Translational Modification: Enzyme activity can be modulated by
phosphorylation, allosteric interactions, or proteolytic cleavage.

5. Significance

 Protein Synthesis: Amino acids are essential for protein structure and function.
 Metabolic Pathways: Amino acids serve as precursors for nucleotide synthesis,
neurotransmitters, and other important molecules.
 Health and Nutrition: Essential amino acids must be obtained from the diet,
highlighting their importance in human nutrition.

Page 189 of 208


Page 190 of 208
Figure 21: Amino acid biosynthesis overview.

Page 191 of 208


Biosynthesis and degradation of nucleotides are crucial processes in cells, providing the
building blocks for DNA and RNA synthesis, energy transfer (ATP), and signaling molecules
(e.g., cAMP). Here's a brief overview:

8.8. Biosynthesis of Nucleotides

1. Purine Biosynthesis:

o De Novo Synthesis: Begins with PRPP (5-Phosphoribosyl-1-pyrophosphate)


and involves multiple enzymatic steps to form IMP (Inosine monophosphate),
which is further converted to AMP (Adenosine monophosphate) and GMP
(Guanosine monophosphate).

2. Pyrimidine Biosynthesis:

o De Novo Synthesis: Starts with carbamoyl phosphate and aspartate to form


UMP (Uridine monophosphate), which is then phosphorylated to UDP (Uridine
diphosphate) and CTP (Cytidine triphosphate).

Degradation of Nucleotides:

1. Purine Degradation:

o Purines are broken down into uric acid through a series of enzymatic steps.
This process yields intermediate products like hypoxanthine, xanthine, and
ultimately uric acid, which is excreted in humans.

2. Pyrimidine Degradation:

o Pyrimidines are degraded to β-alanine or β-aminoisobutyrate, with bases like


uracil and thymine being catabolized through specific pathways in different
organisms.

Page 192 of 208


Regulation and Importance:

 Regulation: Enzyme activities in these pathways are tightly regulated to maintain


balanced nucleotide pools required for DNA and RNA synthesis.

 Importance: Nucleotides are essential for cellular functions, including replication,


transcription, energy transfer (ATP), and cell signaling (cAMP, cGMP).

Understanding these processes is fundamental in biochemistry and molecular biology,


influencing various aspects of cellular physiology and disease mechanisms.

Figure 22: The de novo purine nucleotide biosynthesis pathway.

Page 193 of 208


Figure 23: Synthesis of nucleotides

Page 194 of 208


Figure 24: Degradation of purines and biochemical basis of allopurinol treatment of gout.

Page 195 of 208


8.9. Regulation of nitrogen metabolism

Regulation of nitrogen metabolism is critical for organisms to maintain nitrogen homeostasis,


essential for synthesizing biomolecules like proteins and nucleic acids. Here's an overview of
how nitrogen metabolism is regulated:

Nitrogen Assimilation:

1. Ammonia Assimilation:

o Glutamine Synthetase-Glutamate Synthase (GS-GOGAT) Cycle: Ammonia


(NH3) is assimilated into glutamine and glutamate. Glutamine synthetase (GS)
catalyzes the ATP-dependent formation of glutamine from glutamate and NH3.
Glutamate is then converted to α-ketoglutarate via glutamate synthase
(GOGAT), producing two molecules of glutamine.

2. Nitrate Assimilation:

o Nitrate Reductase (NR) Pathway: Plants and some bacteria reduce nitrate
(NO3-) to nitrite (NO2-) and then to ammonia using nitrate reductase and nitrite
reductase enzymes.

Regulation Mechanisms:

1. Feedback Inhibition:

o End-product Inhibition: Enzymes involved in nitrogen metabolism are often


regulated by feedback inhibition. For instance, glutamine synthetase is
inhibited by high levels of glutamine, preventing unnecessary ammonia
assimilation.

Page 196 of 208


2. Transcriptional Regulation:

o Ntr (Nitrogen Regulatory) System: In bacteria, this system regulates genes


involved in nitrogen metabolism in response to nitrogen availability. When
nitrogen sources are limited, NtrC activates transcription of nitrogen
assimilation genes.

3. Post-translational Modifications:

o Nitrogen Sensing and Signaling: Various organisms use mechanisms like


protein phosphorylation and proteolysis to regulate nitrogen metabolism
enzymes based on intracellular nitrogen levels.

4. Alternative Pathway Regulation:

o Ammonium Transporters and Nitrogen Sources: Organisms regulate the


uptake of different nitrogen sources (ammonium, nitrate, urea) based on
availability and metabolic needs.

Importance:

 Cellular Function: Nitrogen is crucial for synthesizing amino acids, nucleotides, and
coenzymes required for growth and metabolism.

 Environmental Adaptation: Regulation of nitrogen metabolism allows organisms to


adapt to varying nitrogen concentrations in their environment, optimizing nutrient
utilization.

Understanding the regulation of nitrogen metabolism is essential in fields such as agriculture


(e.g., optimizing fertilizer use), microbiology, and biochemistry, impacting both cellular
physiology and environmental sustainability.

Page 197 of 208


References

1. Harper’s Illustrated Biochemistry, A Lange Medical Book, International Edition, 2006,


27th Edition.
2. Krishna Swamy K. Selenium in Human Health, ICMR Bulletin, 1990.
3. Murray. Harpers Biochemistry, Harper & Row, 1990.
4. Ramakrishnan S, Swamy J. Textbook of Clinical (Medical) Biochemistry and
Immunology, 1st edn, TR Publications, 1995.
5. Sittes DP. Basic and Clinical Immunology, 4th edn, Lange Medical Publication, 1982.
6. Harper’s Biochemistry: A Lange Medical Book, 25th edn, 1999.
7. Thompson RHS, Wotton IDP. Biochemical Disorders in Human Diseases, 3rd edn, J
and A Churchill Ltd, London, 1970.
8. Thorpe WB, Bray HG, James HP. Biochemistry for Medical Students, 9th edn,
Churchill, London, 1970.
9. Underwood EJ. Trace Elements in Human and Animal Nutrition, 4th edn, Academic
Press, New York, 1977.
10. Varley H. Practical Clinical Biochemistry, William Heinemann Medical Books Ltd,
London, 1969.
11. von Euler, Eliasson R. Prostaglandins, Academic Press, New York, 1967.
12. Wasserman RH (Ed). Calcium Binding Proteins and Calcium Function, Elsevier, 1977.
13. Weisberg HF. Water, Electrolytes and Acid-Base Balance, 2nd edn, Williams and
Wilkins, Baltimore, 1962.
14. West ES, Todd WR, Mason HS, Van Bruggen JT. Textbook of Biochemistry,
Macmillan, New York, 1966.
15. Wikipedia, the free encyclopedia.
16. Wilkinson R. Isoenzymes, 2nd edn, Chapman and Hall, London, 1970.

Page 198 of 208


17. Wilkinson JH (Ed). Principles and Practice of Diagnostic Enzymology, Edward Arnold,
London, 1976.
18. Williams RH. Textbook of Endocrinology, WB Saunders, Philadelphia, Indian Reprint,
1970.
19. Wootton IDP. Microanalysis in Medical Biochemistry, 6th edn, J and A. Churchill Ltd.,
London, 1982.
20. Wyllie AH. Apoptosis, Recent Advances in Histopathology.
21. Wyllie AH, Carder PJ, Clarke AR. Apoptosis in carcinogenesis; the role of p53, Gold
Spring Harbor Symposia on Quantitative Biology, 403-09, 1994.
22. Yudkin M, Offord K. Comprehensive Biochemistry, Longman (England), 1973.
23. Zubay Geoffrey. Biochemistry, 2nd edn, Maxwell Macmillan (International edn), 1989.
24. Fruton JS, Simmonds SS. General Biochemistry, 2nd edn, John Wiley & Sons, New
York, 1965.
25. Ganong WF. Review of Medical Physiology, 6th edn, Lange Medical Publications,
1973.
26. Goldberger, Emanuel. A Primer of Water, Electrolytes and Acid-base Syndromes, 4th
edn, Lea and Febiger, Philadelphia, 1971.
27. Goodhart RS, Maurice E Shils. Modern Nutrition in Health and Disease, 5th edn, Lea
and Febiger, 1973.
28. Gopalan C, Rao, Nara Singa BS. Dietary Allowances for Indians, Indian Council of
Medical Research, New Delhi, 1980.
29. Halkerston Ian DK. Biochemistry, 2nd edn, John Wiley and Sons, 1990.
30. Hoffman WS. The Biochemistry of Clinical Medicine, 4th edn, Year Book Medical
Publishers, Chicago, 1970.

Page 199 of 208

You might also like