Sunteți pe pagina 1din 203

Life Sciences

Fundamentals and Practice I

Sixth edition

Pranav Kumar | Usha Mina


Life Sciences
Fundamentals and Practice I

Sixth edition

Pranav Kumar
Former faculty,
Department of Biotechnology
Jamia Millia Islamia (JMI),
New Delhi, India

Usha Mina
Associate Professor,
School of Environmental Sciences,
Jawaharlal Nehru University (JNU),
New Delhi, India

Pathfinder Publication
New Delhi, India
Pranav Kumar
Former faculty,
Department of Biotechnology,
Jamia Millia Islamia (JMI),
New Delhi, India

Usha Mina
Associate Professor,
School of Environmental Sciences,
Jawaharlal Nehru University (JNU),
New Delhi, India

Life Sciences : Fundamentals and Practice

Sixth edition

ISBN: 978-81-906427-0-5 (paperback)

Copyright © 2017 by Pathfinder Publication, all rights reserved.

This book contains information obtained from authentic and highly


regarded sources. Reasonable efforts have been made to publish reliable data
and information, but the author and the publisher cannot assume responsibility
for the validity of all materials or for the consequences of their use.
No part of this book may be reproduced by any mechanical, photographic, or
electronic process, or in the form of a phonographic recording, nor it may be
stored in a retrieval system, transmitted, or otherwise copied for public or
private use, without written permission from the publisher.

Publisher : Pathfinder Publication


Production editor : Ajay Kumar
Copy editor : Jomesh Joseph
Illustration and layout : Pradeep Verma
Cover design : Monu
Marketing director : Arun Kumar
Production coordinator : Murari Kumar Singh

Pathfinder Publication
A unit of Pathfinder Academy Private Limited, New Delhi, India.
pathfinderpublication.in
iii

Preface
Life Sciences have always been a fundamental area of science. The exponential increase in
the quantity of scientific information and the rate, at which new discoveries are made, require
very elaborate, interdisciplinary and up-to-date information and their understanding. This
sixth edition of Life sciences, Fundamentals and practice includes extensive revisions of the
previous edition. We have attempted to provide an extraordinarily large amount of information
from the enormous and ever-growing field in an easily retrievable form. It is written in clear
and concise language to enhance self-motivation and strategic learning skill of the students
and empowering them with a mechanism to measure and analyze their abilities and the
confidence of winning. We have given equal importance to text and illustrations. The sixth edition
has a number of new figures to enhance understanding. At the same time, we avoid excess detail,
which can obscure the main point of the figure. We have retained the design elements that have
evolved through the previous editions to make the book easier to read. Sincere efforts have been
made to support textual clarifications and explanations with the help of flow charts, figures and
tables to make learning easy and convincing. The chapters have been supplemented with self-tests
and questions so as to check one’s own level of understanding. Although the chapters of this book
can be read independently of one another, they are arranged in a logical sequence. Each page is
carefully laid out to place related text, figures and tables near one another, minimizing the need
for page turning while reading a topic. We have given equal importance to text and illustrations
as well. We hope you will find this book interesting, relevant and challenging.
Acknowledgements
Our students were the original inspiration for the first edition of this book, and we remain continually
grateful to them, because we learn from them how to think about the life sciences, and how to
communicate knowledge in most meaningful way. We thank, Dr. Diwakar Kumar Singh and Mr. Ajay
Kumar, reviewers of this book, whose comments and suggestions were invaluable in improving
the text. Any book of this kind requires meticulous and painstaking efforts by all its contributors.
Several diligent and hardworking minds have come together to bring out this book in this complete
form. We are much beholden to each of them and especially to Dr. Neeraj Tiwari. This book is a
team effort, and producing it would be impossible without the outstanding people of Pathfinder
Publication. It was a pleasure to work with many other dedicated and creative people of Pathfinder
Publication during the production of this book, especially Pradeep Verma.

Pranav Kumar
Usha Mina
v

Contents
Chapter 1
Biomolecules and Catalysis
1.1 Amino acids and Proteins 1

1.1.1 Optical properties 3

1.1.2 Absolute configuration 4

1.1.3 Standard and non-standard amino acids 5

1.1.4 Titration of amino acids 7

1.1.5 Peptide and polypeptide 12

1.1.6 Peptide bond 13

1.1.7 Protein structure 16

1.1.8 Denaturation of proteins 20

1.1.9 Solubilities of proteins 21

1.1.10 Simple and conjugated proteins 22

1.2 Fibrous and globular proteins 22

1.2.1 Collagen 23

1.2.2 Elastin 24

1.2.3 Keratins 25

1.2.4 Myoglobin 25

1.2.5 Hemoglobin, Hb 27

Oxygen transport 28

Functional differences between Mb and Hb 30

Factors affecting the affinity of Hb for oxygen 30

1.2.6 Models for the behavior of allosteric proteins 31

1.3 Protein folding 33

1.3.1 Molecular chaperones 34

1.3.2 Amyloid 35

1.3.3 Ubiquitin mediated protein degradation 36

1.3.4 N-end rule 38

1.4 Protein sequencing and assays 39

1.5 Nucleic acids 47

1.5.1 Nucleotides 47

1.5.2 Chargaff’s rules 51

1.6 Structure of dsDNA 53

1.6.1 B-DNA 53

1.6.2 Z-DNA 54
vi

1.6.3 Triplex DNA 55

1.6.4 G-quadruplex 56

1.6.5 Stability of the dsDNA helix 57

1.6.6 DNA denaturation 57

1.6.7 Quantification of nucleic acids 59

1.6.8 Supercoiled forms of DNA 59

Linking number 60

1.6.9 DNA: A genetic material 61

1.7 RNA 63

1.7.1 Alkali-catalyzed cleavage of RNA 64

1.7.2 RNA World hypothesis 65

1.7.3 RNA as genetic material 65

1.8 Carbohydrates 66

1.8.1 Monosaccharide 66

1.8.2 Epimers 68

1.8.3 Cyclic forms 68

1.8.4 Derivatives of monosaccharide 70

1.8.5 Disaccharides and glycosidic bond 71

1.8.6 Polysaccharides 73

1.8.7 Glycoproteins 75

1.8.8 Reducing and non-reducing sugar 76

1.9 Lipids 76

1.9.1 Fatty acids 77

1.9.2 Triacylglycerol and Wax 78

1.9.3 Phospholipids 79

1.9.4 Glycolipids 81

1.9.5 Steroid 82

1.9.6 Eicosanoid 82

1.9.7 Plasma lipoproteins 84

1.10 Vitamins 84

1.10.1 Water-soluble vitamins 85

Thiamine (Vitamin B1) 85

Riboflavin (Vitamin B2) 85

Niacin 86

Biotin 86

Pantothenic acid 87

Folic acid 87

Cobalamin (Vitamin B12) 87

Pyridoxine (Vitamin B6) 88

Ascorbic acid (Vitamin C) 88

1.10.2 Fat-soluble vitamins 89

Vitamin A (Retinol) 89
vii

Vitamin D 89

Vitamin K 90

Vitamin E 90

1.11 Reactive oxygen species and antioxidant 91

1.12 Enzymes 92

1.12.1 Naming and classification of enzyme 93

1.12.2 How enzymes operate? 94

1.12.3 Catalytic strategies 96

1.12.4 Enzyme kinetics 97

1.12.5 Enzyme inhibition 104

1.12.6 Regulatory enzymes 107

1.12.7 Isozymes 110

1.12.8 Zymogen 110

1.12.9 Ribozyme 111

1.12.10 Examples of enzymatic reactions 111

Chapter 2
Bioenergetics and Metabolism
2.1 Bioenergetics 119

2.2 Metabolism 124

2.3 Respiration 125

2.3.1 Aerobic respiration 125

2.3.2 Glycolysis 126

2.3.3 Pyruvate oxidation 131

2.3.4 Krebs cycle 133

2.3.5 Anaplerotic reaction 136

2.3.6 Oxidative phosphorylation 136

2.3.7 Inhibitors of electron transport 141

2.3.8 Electrochemical proton gradient 142

2.3.9 Chemiosmotic theory 143

2.3.10 ATP synthase 144

2.3.11 Uncoupling agents and ionophores 146

2.3.12 ATP-ADP exchange across the inner mitochondrial membrane 146

2.3.13 Shuttle systems 147

2.3.14 P/O ratio 149

2.3.15 Fermentation 150

2.3.16 Pasteur effect 152

2.3.17 Warburg effect 152

2.3.18 Respiratory quotient 152

2.4 Glyoxylate cycle 153

2.5 Pentose phosphate pathway 154


viii

2.6 Entner-Doudoroff pathway 156

2.7 Photosynthesis 157

2.7.1 Photosynthetic pigment 157

2.7.2 Absorption and action spectra 161

2.7.3 Fate of light energy absorbed by photosynthetic pigments 162

2.7.4 Concept of photosynthetic unit 164

2.7.5 Hill reaction 164

2.7.6 Oxygenic and anoxygenic photosynthesis 164

2.7.7 Concept of pigment system 165

2.7.8 Stages of photosynthesis 167

2.7.9 Light reactions 168

2.7.10 Prokaryotic photosynthesis 174

2.7.11 Non-chlorophyll based photosynthesis 176

2.7.12 Dark reaction: Carbon reduction and fixation cycle 176

2.7.13 Starch and sucrose synthesis 180

2.8 Photorespiration 181

2.8.1 C4 cycle 182

2.8.2 CAM pathway 184

2.9 Carbohydrate metabolism 187

2.9.1 Gluconeogenesis 187

2.9.2 Glycogen metabolism 192

2.10 Lipid metabolism 197

2.10.1 Synthesis and storage of triacylglycerols 197

2.10.2 Biosynthesis of fatty acid 199

2.10.3 Fatty acid oxidation 202

2.10.4 Biosynthesis of cholesterol 210

2.10.5 Steroid hormones and Bile acids 211

2.11 Amino acid metabolism 213

2.11.1 Amino acid synthesis 213

2.11.2 Amino acid catabolism 216

2.11.3 Molecules derived from amino acids 221

2.12 Nucleotide metabolism 222

2.12.1 Nucleotide synthesis 222

2.12.2 Nucleotide degradation 229

Chapter 3
Cell Structure and Functions
3.1 What is a Cell? 235

3.2 Structure of eukaryotic cells 236

3.2.1 Plasma membrane 236

3.2.2 ABO blood group 244

3.2.3 Transport across plasma membrane 246


ix

3.3 Membrane potential 253

3.4 Transport of macromolecules across plasma membrane 263

3.4.1 Endocytosis 263

3.4.2 Fate of receptor 268

3.4.3 Exocytosis 268

3.5 Ribosome 269

3.5.1 Protein targeting and translocation 271

3.6 Endoplasmic reticulum 272

3.6.1 Endomembrane system 276

3.6.2 Transport of proteins across the ER membrane 277

3.6.3 Transport of proteins from ER to cis Golgi 281

3.7 Golgi complex 283

3.7.1 Transport of proteins through cisternae 284

3.7.2 Transport of proteins from the TGN to lysosomes 285

3.8 Vesicle fusion 286

3.9 Lysosome 287

3.10 Vacuoles 289

3.11 Mitochondria 290

3.12 Plastids 293

3.13 Peroxisome 294

3.14 Nucleus 295

3.15 Cytoskeleton 299

3.15.1 Microtubules 299

3.15.2 Kinesins and Dyneins 302

3.15.3 Cilia and Flagella 303

3.15.4 Centriole 305

3.15.5 Actin filament 306

3.15.6 Myosin 307

3.15.7 Muscle contraction 309

3.15.8 Intermediate filaments 312

3.16 Cell junctions 314

3.17 Cell adhesion molecules 317

3.18 Extracellular matrix of animals 318

3.19 Plant cell wall 320

3.20 Cell signaling 322

3.20.1 Signal molecules 323

3.20.2 Receptors 323

3.20.3 GPCR and G-proteins 325

3.20.4 Ion channel-linked receptors 334

3.20.5 Enzyme-linked receptors 334

3.20.6 Nitric oxide 341

3.20.7 Two-component signaling systems 342


x

3.20.8 Chemotaxis in bacteria 343

3.20.9 Quorum sensing 344

3.20.10 Scatchard plot 345

3.21 Cell Cycle 347

3.21.1 Role of Rb protein in cell cycle regulation 356

3.21.2 Role of p53 protein in cell cycle regulation 358

3.21.3 Replicative senescence 360

3.22 Mechanics of cell division 360

3.22.1 Mitosis 360

3.22.2 Meiosis 367

3.22.3 Nondisjunction and aneuploidy 371

3.23 Apoptosis 374

3.24 Cancer 377

Molecular basis of cancer 379

Proto-oncogenes 380

Tumor suppressor genes 382

Carcinogen 383

Retinoblastoma 384

Oncovirus or tumor virus 385

Retroviral oncogenes 385

Chapter 4
Prokaryotes and Viruses
4.1 General features of Prokaryotes 391

4.2 Phylogenetic overview 392

4.3 Structure of bacterial cell 393

Staining 393

Cell Wall 395

Outer membrane 397

Glycocalyx 398

Plasma membrane 399

Cytoplasm 399

Surface appendages 400

Endospores 402

4.4 Bacterial genome: Bacterial chromosome and plasmid 403

4.5 Bacterial nutrition 408

4.5.1 Culture media 409

4.5.2 Bacterial growth 410

4.6 Horizontal gene transfer and genetic recombination 413

4.6.1 Transformation 414

4.6.2 Transduction 416


xi

4.6.3 Conjugation 420

4.7 Bacterial taxonomy 425

4.8 General features of important bacterial groups 426

4.9 Archaebacteria 429

4.10 Bacterial toxins 430

4.11 Control of microbial growth 432

4.12 Virus 436

4.12.1 Bacteriophage (Bacterial virus) 438

4.12.2 Life cycle of bacteriophage 439

4.12.3 Plaque assay 442

4.12.4 Genetic analysis of phage 445

4.12.5 Animal viruses 448

4.12.6 Plant viruses 458

4.13 Prions and Viroid 458

4.13.1 Bacterial and viral disease 460

Chapter 5
Immunology
5.1 Innate immunity 463

5.2 Adaptive immunity 466

5.3 Cells of the immune system 468

5.3.1 Lymphoid progenitor 468

5.3.2 Myeloid progenitor 470

5.4 Organs involved in the adaptive immune response 471

5.4.1 Primary lymphoid organs 471

5.4.2 Secondary lymphoid organs/tissues 472

5.5 Antigens 473

5.6 Major-histocompatibility complex 477

5.6.1 MHC molecules and antigen presentation 479

5.6.2 Antigen processing and presentation 480

5.6.3 Laboratory mice 482

5.7 Immunoglobulins: Structure and function 483

5.7.1 Basic structure of antibody molecule 483

5.7.2 Different classes of immunoglobulin 486

5.7.3 Action of antibody 488

5.7.4 Antigenic determinants on immunoglobulins 488

5.8 B-cell maturation and activation 490

Clonal selection theory 494

5.9 Kinetics of the antibody response 495

5.10 Monoclonal antibodies and Hybridoma technology 497

5.10.1 Engineered monoclonal antibodies 498


xii

5.11 Organization and expression of Ig genes 499

Mechanism of DNA rearrangements 502

Allelic exclusion 504

Class switching 504

5.12 Generation of antibody diversity 505

5.13 T-cells and CMI 507

T-cell maturation 510

Thymic selection 512

5.13.1 Superantigens 517

5.14 Cytokines 518

5.15 The complement system 521

5.16 Hypersensitivity 525

5.17 Autoimmunity 527

5.18 Transplantation 528

5.19 Immunodeficiency diseases 528

5.20 Failures of host defense mechanisms 529

5.21 Vaccines 531

Chapter 6
Diversity of Life
6.1 Taxonomy 537

6.1.1 Nomenclature 537

6.1.2 Classification 538

6.1.3 Biological species concept 538

6.1.4 Phenetic and phylogenetic principles of classification 539

6.2 The five-kingdom system 545

6.3 Protists 547

6.3.1 Protozoan protists 547

6.3.2 Photosynthetic protists 548

6.3.3 Slime mold 549

6.3.4 Oomycetes 550

6.4 Fungi 550

6.4.1 Mycorrhiza 552

6.4.2 Lichens 552

6.5 Plantae 553

6.5.1 Plant life cycle 553

6.5.2 Algae 555

6.5.3 Life cycle of land plants 557

6.5.4 Bryophytes 558

6.5.5 Pteridophytes 560

6.5.6 Gymnosperm 561

6.5.7 Angiosperms 562


xiii

6.6 Animalia 566

6.7 Animal’s classification 574

6.7.1 Phylum Porifera (Pore bearing animals) 574

6.7.2 Phylum Cnidaria (Coelenterata) 574

6.7.3 Phylum Platyhelminthes (Flatworms) 575

6.7.4 Phylum Aschelminthes (Roundworms) 575

6.7.5 Phylum Annelida 577

6.7.6 Phylum Mollusca 577

6.7.7 Phylum Arthropoda 578

6.7.8 Phylum Echinodermata 578

6.7.9 Phylum Hemichordata 579

6.7.10 Phylum Chordata 579

Answers of self test 587

Index 588
Chapter 01

Biomolecules and Catalysis

A biomolecule is a carbon-based organic compound that is produced by a living organism. More than 25 naturally
occurring chemical elements are found in biomolecules, but these biomolecules consist primarily of carbon, hydrogen,
nitrogen, oxygen, phosphorus and sulfur. In terms of the percentage of the total number of atoms, four elements
such as hydrogen, oxygen, nitrogen and carbon together make up over 99% of the mass of most cells.
Biomolecules include both small as well as large molecules. The small biomolecules are low molecular weight
(less than 1000) compound which include sugars, fatty acids, amino acids, nucleotides, vitamins, hormones,
neurotransmitters, primary and secondary metabolites. Sugars, fatty acids, amino acids and nucleotides constitute
the four major families of small biomolecules in cells. Large biomolecules which have high molecular weight are
called macromolecules and mostly are polymers of small biomolecules. These macromolecules are proteins,
carbohydrates and nucleic acids.
Small biomolecules Macromolecules
Sugars Polysaccharides
Amino acids Polypeptides (proteins)
Nucleotides Nucleic acids
Fatty acids

Nucleic acids and proteins are informational macromolecules. Proteins are polymers of amino acids and constitute
the largest fraction (besides water) of cells. The nucleic acids, DNA and RNA, are polymers of nucleotides. They
store, transmit, and translate genetic information. The polysaccharides, polymers of simple sugars, have two major
functions. They serve as energy-yielding fuel stores and as extracellular structural elements.

1.1 Amino acids and Proteins


Amino acids are compounds containing carbon, hydrogen, oxygen and nitrogen. They serve as monomers (building
blocks) of proteins and are composed of an amino group, a carboxyl group, a hydrogen atom, and a distinctive
side chain, all bonded to a carbon atom, the α-carbon. In an α-amino acid, the amino and carboxylate groups are
attached to the same carbon atom, which is called the α-carbon. The various α-amino acids differ with respect to
the side chain (R group) attached to their α-carbon. The general structure of an amino acid is:

a-carboxyl group

COO

+
a-amino group H3N Ca H

R
Side chain

Figure 1.1 General structure of an amino acid.


This page intentionally left blank.
Biomolecules and Catalysis 3

1.1.1 Optical properties


All amino acids except glycine are optically active i.e. they rotate the plane of plane polarized light. Optically active
molecules contain chiral carbon. A tetrahedral carbon atom with four different constituents are said to be chiral.
All amino acids except glycine have chiral carbon and hence they are optically active. An optically active compound
can rotate the plane of polarized light either clockwise (to the right) or counterclockwise (to the left). Optically
active compounds that rotate the plane of polarized light clockwise are said to be dextrorotatory. By convention,
this direction is designated by a plus sign (+). Optically active compounds that rotate the plane of polarized light
counterclockwise are said to be levorotatory. This is designated by a minus sign (–). The + and – forms have also
been termed d- and l-, respectively.
— —
COO COO
+ +
H3N Ca H H3N Ca H

Achiral H Chiral CH3


carbon carbon

Glycine Alanine

Figure 1.4 Amino acids showing achiral and chiral carbon.

Optical activity is measured by polarimeter. Optical activity is the ability of an optically active compound to rotate
the plane of linearly polarized light. Optical rotation is a quantitative measure of the rotation of light caused by
the compound. The magnitude of optical rotation indicates the extent to which plane of linearly polarized light is
rotated and sign represents the direction of rotation. Optical rotation of an optically active compound depends on
the concentration of the compound, temperature, wavelength of light used, solvent used to dissolve the sample
and light pathlength. The optical rotation of a solution at a given temperature and wavelength is given by

Å = [α]Tλ × C × l

where, Å = observed rotation in degrees


C = concentration of the solution in g/ml
l = light path length in decimeters (dm)
[α]Tλ = the specific rotation of compound at temperature, T (in degrees Celsius) and wavelength, λ (in nm).
If the wavelength of the light used is 589 nm, the symbol ‘D’ is used, [α]DT.

Specific rotation is the reference value of optical rotation for a given concentration of compound at a given
temperature and fixed wavelength. At a given temperature and for a given wavelength of light, the specific rotation
is defined as the observed value of optical rotation when plane polarized light is passed through a sample with a
path length of 1 decimeter and a sample concentration of 1g per milliliter.

Sample tube
containing a
chiral compound

Normal light Polarizer Plane-polarized Rotation of


light plane-polarized light

Figure 1.5 When plane polarized light is passed through a solution that contains an optically active compound, there
is net rotation of the plane polarized light. The light is rotated either clockwise (dextrorotatory) or counterclockwise
(levorotatory) by an angle that depends on the molecular structure and concentration of the compound, the path
length and the wavelength of the light.
This page intentionally left blank.
Biomolecules and Catalysis 7

Nonstandard amino acids


Although hundreds of different amino acids present in cells but only 22 different amino acids participate in protein
synthesis and are incorporated ribosomically into proteins. Such amino acids are called proteinogenic or standard
amino acids. Apart from the 22 standard amino acids, all other amino acids are not ribosomically incorporated
into proteins are called non-standard. In addition to the standard amino acids, some proteins may contain non-
standard amino acid residues formed by post-translational modification of standard amino acid residues already
incorporated into a polypeptide. These modifications are often essential for the function or regulation of a protein.
Examples of some of these amino acids are 4-Hydroxyproline (derivative of proline), 5-Hydroxylysine (derivative
of lysine), desmosine (derivative of lysine), N-acetylserine, N-formylmethionine and γ-carboxyglutamate (found in
the blood clotting protein prothrombin).
Besides their role in proteins, amino acids and their derivatives have many other biologically important functions.
Many nonstandard amino acids are not found in proteins. These amino acids often occur as intermediates in the
metabolic pathways for standard amino acids. For example, ornithine and citrulline are key intermediates in the
biosynthesis of arginine and in the urea cycle. Similarly, azaserine, a nonstandard amino acid, acts as an antibiotic.
It was originally thought that all unconventional amino acids were made by modifying one of the standard amino
acids after it was incorporated into protein, a process called a post-translational modification. But amino acids like
selenocysteine, pyrrolysine are inserted into proteins by the translational machinery.
Selenocysteine (Sec or U) is the 21st standard amino acid. It has a structure similar to that of cysteine, but it contains
selenium rather than sulphur. It is incorporated into polypeptides during translation. However, it is specified by a
triplet codon, UGA (a stop codon). Selenocysteine has its own tRNA containing the anticodon UCA and it is formed
by modifying a serine that has been attached to the selenocysteine tRNA. Enzymes like glutathione peroxidase and
formate dehydrogenase contain selenocysteine in their catalytic center. Pyrrolysine (Pyl or O) is the 22nd standard
amino acid. It is similar to lysine and is present in some bacterial proteins. It is coded by UAG codon.


COO
+
H3N C H

CH2

CH2

CH2
— —
COO COO
CH2
+ +
H3N C H H3N C H
NH
CH2 CH2
C=O
SH Se H3C
N
H

Cysteine Selenocysteine Pyrrolysine

1.1.4 Titration of amino acids


Because amino acids contain ionizable groups, the predominant ionic form of these molecules in solution depends
on the pH. Titration of an amino acid illustrates the effect of pH on amino acid structure. Consider alanine, a simple
amino acid, which has two titrable groups (α-amino and α-carboxyl group). During titration with a strong base such
as NaOH, alanine loses two protons in a stepwise fashion. In a strongly acidic solution, alanine is present mainly in
the form in which the carboxyl group is uncharged. Under this condition the molecule’s net charge is +1, since the
ammonium group is protonated. However, an increase in the pH results in the deprotonation of α-carboxyl group.
8 Biomolecules and Catalysis
At this point, alanine has no net charge and is electrically neutral. The pH at which this occurs is called the isoelectric
point (pI).

+1 0 –1

— —
COOH COO COO
+ pk1 + pk2
H3N Ca H H3N Ca H H2N Ca H

CH3 CH3 CH3

Low pH (pH < pI) Intermediate pH High pH (pH > pI)


(pH = pI)

Because there is no net charge at the isoelectric point, amino acids are electrophoretically non-mobile and least
soluble at this pH. Further increase in pH i.e. lowering of the H+ concentration results in the deprotonation of the
charged amino group and an uncharged amino group forms. So at high pH, the net charge on the molecule is
–1, since the ammonium group is deprotonated and a net negative charge develops due to the presence of the
carboxylate group.

13
Alanine
pK2 = 9.7

7
pH pI = 6

pK1 = 2.34

0 0.5 1 1.5 2

OH (equivalents)

Figure 1.6 Titration curve of alanine (monoamino and monocarboxylic acid). A plot of the dependence of the pH on
the amount of OH– added is called a titration curve.

The isoelectric point for alanine may be calculated as follows:

pK1 + pK2
pI =
2

The pK1 and pK2 values for alanine are 2.34 and 9.7 respectively. The pI value for alanine is therefore,

2.34 + 9.7
pI = = 6.0
2

Amino acids with ionizable side chains have more complex titration curves. Glutamic acid, for example, has a carboxyl
side chain group. At low pH, glutamic acid has net charge +1. As the base is added (pH increases), the α-carboxyl
group loses a proton to become a carboxylate group (in a polyprotic acid, the protons are first lost from the group
with the lowest pKa). Glutamate now has no net charge. As still more base is added, the second carboxyl group
This page intentionally left blank.
Biomolecules and Catalysis 13

peptides are cyclic in nature. Two cyclic decapeptides (peptides containing 10 amino acid residues) produced by
the bacterium Bacillus brevis are common examples. Both of these peptides, gramicidin S and tyrocidine A, are
antibiotics, and both contain D-amino acids as well as L-amino acids. In addition, both contain the amino acid
ornithine, which does not occur in proteins. Small peptides play many roles in organisms. Some, such as oxytocin
and vasopressin, are important hormones. Others, like glutathione, regulate oxidation–reduction reactions. Still
others, such as enkephalins, are naturally occurring painkillers. Aspartame is a commercially synthesized dipeptide,
L-aspartylphenylalanyl methylester, and is used as an artificial sweetener.
When many amino acid residues are joined, the product is called a polypeptide. Amino acids which have been
incorporated into a peptide or polypeptide are termed amino acid residues. By convention, in a polypeptide the left
end represented by the first amino acid while the right end represented by the last amino acid. The first amino acid
is also called as N-terminal amino acid residue. The last amino acid is called the C-terminal amino acid residue.

H O H O H O
N-terminal C-terminal

H2N C C N C C N C C OH

R H R H R

Amino acid Amino acid Amino acid


residue residue residue

Figure 1.10 A series of amino acids joined by peptide bonds form a polypeptide chain, and each amino acid unit in a
polypeptide is called a residue. A polypeptide chain has polarity because its ends are different, with an α-amino group
at one end and an α-carboxyl group at the other.

The peptide bonds in proteins are formed between the α-amino and the α-carboxyl groups. But peptides do occur
naturally where the peptide linkage involves a carboxyl or amino group which is attached to a carbon atom other
than the α-carbon. For example a dipeptide formed between the γ-carboxyl group of glutamic acid and the amino
group of alanine is called γ-glutamylalanine.

1.1.6 Peptide bond


Peptides and polypeptides are linear and unbranched polymers composed of amino acids linked together by peptide
bonds. Peptide bonds are amide linkages formed between α-amino group of one amino acid and the α-carboxyl group
of another. This reaction is a dehydration reaction, that is, a water molecule is removed and the linked amino acids
are referred to as amino acid residues. Peptide bond formation is an endergonic process, with ΔG ~ +21kJ/mol.

H O H O

H2N C C OH H N C C OH

R1 H R2

H2O

H O H O

H2N C C N C C OH

R1 H R2

Figure 1.11 The formation of a peptide bond (also called an amide bond) between the α-carboxyl group of one amino
acid to the α-amino group of another amino acid is accompanied by the loss of a water molecule.
This page intentionally left blank.
16 Biomolecules and Catalysis

1.1.7 Protein structure


Proteins are unbranched polymers constructed from 22 standard α-amino acids. They have four levels of the
structural organization. Primary structure, the amino acid sequence, is specified by genetic information. As the
polypeptide chain folds, it forms certain localized arrangements of adjacent amino acids that constitute secondary
structure. The overall three-dimensional shape that a polypeptide assumes is called the tertiary structure. Proteins
that consist of two or more polypeptide chains (or subunits) are said to have a quaternary structure.

Primary structure
The primary structure (1° structure) of a polypeptide is its amino acid sequence. The amino acids are connected
by peptide bonds. Primary structure of polypeptide determines the higher levels of structural organization.

Secondary structure
The most common types of secondary structure (2° structure) are the α-helix and the β-pleated sheet. Both
α-helix and β-pleated sheet patterns are stabilized by hydrogen bonds between the carbonyl and N—H groups in
the polypeptide’s backbone.

α-helix
The α-helix is a rigid, rod like structure that forms when a polypeptide chain twists into a helical conformation. The
screw sense of α-helix can be right-handed (clockwise) or left-handed (counterclockwise). However, right-handed
helices are energetically more favorable. In almost all proteins, the helical twist of the α-helix is right-handed.
There are 3.6 amino acid residues per turn of the helix and the pitch (the distance between corresponding points
per turn) is 0.54 nm. Each residue is related to the next one by a rise of 1.5 Å (0.15 nm) along the helix axis. A
single turn of α-helix involves 13 atoms from O to the H of the H bond. For this reason, the α-helix is referred to as
the 3.613-helix. Length of α-helix is usually 10–15 amino acid residues. Intrachain hydrogen bonds form between
the N—H group of each amino acid and the carbonyl group of the amino acid four residues away.

O1 H O H 13 O
n+1 n+3 n+5

C 3
Ca 5 N 7 C 9
Ca 11 N C Ca
2 4 6 8 10 12
Ca N C Ca N C Ca N
n n+2 n+4

H O H O H

Figure 1.16 The hydrogen bonding arrangement in the α-helix. The α-helix is known as the 3.613-helix, where 3.6
is the number of residues per turn and 13 is the number of atoms in the hydrogen-bonded loop.

Except for amino acids near the ends of an α-helix, all the main-chain CO and NH groups are hydrogen bonded.
The side chains of amino acids extend outward from the helix. All H bonds lie parallel to the helix axis and point
in the same direction.
Helices can be formed from either D- or L-amino acids, but a given helix must be composed entirely of amino acids
of one configuration. The α-helix cannot be formed from a mixed copolymer of D- and L-amino acids. L-amino acids
can form either right or left handed α-helices.
Biomolecules and Catalysis 17

C
N
C
C
N
C
N C
C
C
N
C
C
N
C
C
N
C
C
N
0.15 nm 3.6 residues C
C
3.6 residues
N
C
C
N
C
C
N
N
C

(a) (b)

Figure 1.17 Describing the geometry of α-helix. The helix structure is defined by: the pitch (the distance along the
axis between successive turns) and the rise per residue. The number of residues per helical turn is 3.6. In the right
handed α-helix, a complete turn of the helix contains 3.6 amino acid residues, and the distance it rises per turn (its
pitch) is 0.54 nm. The R groups of each amino acid residues in an α-helix face outward (not shown in the figure). In
the α-helix, the hydrogen bonds are within a single helix and are almost parallel to the helix axis.

Amino acids have different propensities for forming α-helices. Amino acid residues such as alanine, glutamine,
glutamate, leucine, methionine, arginine show the higher tendency to form α-helices. Proline tends to disrupt
α-helices because it lacks an –NH group and because its ring structure restricts its φ value to near –60 degrees.

β-pleated sheets
β-pleated sheets form when two or more polypeptide chain segments line up side by side. Each individual segment
is referred to as a β-strand. Rather than being coiled, each β-strand is fully extended. The distance between
adjacent amino acids along a β-strand is approximately 3.5 Å, in contrast with a distance of 1.5 Å along an α-helix.
β-pleated sheets are stabilized by interchain hydrogen bonds that form between the polypeptide backbone N—H
and carbonyl groups of adjacent strands. Adjacent strand can be either parallel or antiparallel. In parallel β-pleated
sheet structures, the polypeptide chains are arranged in the same direction. However in antiparallel β-pleated
sheet chains run in opposite directions. Antiparallel β-sheets are more stable than parallel β-sheets because fully
collinear hydrogen bonds form.
18 Biomolecules and Catalysis
O H O H
Ca C N Ca C N Ca
C-terminus C N Ca C N Ca C N N-terminus

O H O H O H

O O O
H H H

N-terminus N C Ca N C Ca N C C-terminus
N N
Ca C Ca C Ca
H H
O O

O H O H
Ca C N Ca C N Ca
C-terminus C N Ca C N Ca C N N-terminus

O H O H O H

O H O H
Ca C N Ca C N Ca
C-terminus C N Ca C N Ca C N N-terminus

O H O H O H

Figure 1.18 In an antiparallel β-pleated sheet, adjacent strands run in opposite directions. Hydrogen bonds between
NH and CO groups connect each amino acid to a single amino acid on an adjacent strand. In parallel β-pleated sheet,
adjacent strands run in the same direction. Hydrogen bonds connect each amino acid on one strand with two different
amino acids on the adjacent strand. For each amino acid, the NH group is hydrogen bonded to the CO group of one
amino acid on the adjacent strand, whereas the CO group is hydrogen bonded to the NH group on the amino acid two
residues farther along the chain.

Turns
Most proteins have compact, globular shapes, requiring reversals in the direction of their polypeptide chains. Many
of these reversals are accomplished by a common structural element called the turn. Turns, composed of three to
five residues, are classified as a third type of secondary structure. These short, U-shaped secondary structures are
stabilized by a hydrogen bond between their end residues. Glycine and proline are commonly present in turns. The
lack of a large side chain in glycine and the presence of a built-in bend in proline allow the polypeptide backbone
to fold into a tight U shape. Turns allow large proteins to fold into highly compact structures. In contrast, loops are
longer than turns and do not have regular secondary structure.
Turns are classified according to the separation between the two end residues participating in hydrogen bonding:
α-turn, β-turn and γ-turn. In an α-turn, the donor and acceptor residues are separated by four peptide bonds
(involves five amino acid residues). H-bond forms between the carbonyl oxygen of residue (n) and the hydrogen
of the amide group of residue (n+4). β-turn (the most common form) is characterized by hydrogen bond(s) in
which the donor and acceptor residues are separated by three peptide bonds (n and n+3). Similarly, a γ-turn is
characterized by hydrogen bond(s) in which the donor and acceptor residues are separated by two peptide bonds (n
and n+2). Turns are located primarily on the protein surface and thus participate in interactions between proteins
and other molecules.

Supersecondary structures
Many globular proteins contain combinations of α-helix and β-pleated secondary structures. Specific geometric
arrangements of α-helices and β-strands connected through loops are called supersecondary structures (also
called motifs). These structures can be αα (two α-helices linked by a loop), ββ (two β-strands linked by a loop),
βαβ (two parallel β-strands are connected by an α-helix) or more complexes structures, like the Greek key motif
or the beta-barrel.
This page intentionally left blank.
Biomolecules and Catalysis 25

H H O

N C C

(CH2)2

H N N H
CH2
H C CH2 H2C CH2 CH2 C H

O C C O
+
N

CH2

(CH2)3

N C C

H H O

Figure 1.22 Intramolecular desmosine cross-links in elastin.

1.2.3 Keratins
Keratins are fibrous proteins present in eukaryotes. They form a large family, with about 30 members being
distinguished. Keratins have been classified as either α-keratins or β-keratins.
Proteins α-keratin β-keratin
Characteristics Tough, insoluble Soft, flexible
Conformation Helical Extended chain
Basic unit Protofibril Antiparallel β-pleated sheet

α-keratins are intermediate filament proteins present only in many metazoans, including vertebrates. In vertebrates,
α-keratins constitute almost the entire dry weight of hair, wool, feathers, nails, claws, scales, horns, hooves, and
much of the outer layer of skin. The α-keratin polypeptide chain which forms polymerized α-keratin structure, is a
right-handed α-helix and rich in hydrophobic amino acid residues Ala, Val, Leu, Ile, Met and Phe. Every α-keratin
polypeptide chain dimerizes to form heterodimer. The heterodimer is made up of type I (acidic) and the type II
(neutral/basic) α-keratin polypeptide chains. The two chains in heterodimer have a parallel arrangement. Two
heterodimers join in an antiparallel manner to form the fundamental tetrameric subunit (a protofilament). Two
protofilaments constitute a protofibril. Four protofibrils constitute a microfibril, which associates with other microfibrils
to form a macrofibril.

1.2.4 Myoglobin
Myoglobin (Mb), a globular protein, contains a single polypeptide chain of 153 amino acid residues (molecular
weight 17,800), and a single heme group. The inside of myoglobin consists almost exclusively of nonpolar residues,
whereas the outside contains both polar and nonpolar residues. About 75% of the polypeptide chain is α-helical.
There are eight helical segments. These eight helical segments are commonly labeled A–H, starting from the NH2-
terminal end. The interhelical regions are designated as AB, BC, CD,..., GH, respectively. The iron atom of the heme
is directly bonded to a nitrogen atom of a histidine side chain of globin.

Heme
Globin of Mb binds a single heme group by forming a co-ordinate bond. The heterocyclic ring system of heme is a
porphyrin derivative. The porphyrin in heme is known as protoporphyrin IX. It is made up of 4-pyrrole ring and
4-pyrroles are linked by methine (=CH–) bridges to form a tetrapyrrole ring. The Fe atom is present either in Fe2+
or Fe3+ oxidation state in the center of the protoporphyrin IX ring.
26 Biomolecules and Catalysis
Only myoglobin in Fe2+ state can bind O2. The iron atom has six co-ordination bond, 4 in the plane of flat porphyrin
ring and two perpendicular to it. The iron atom of the heme is directly bonded to one of the histidine called proximal
histidine (His93 or His F8) of globin protein. It is 93rd amino acid residue from the amino-terminal end of the
myoglobin polypeptide chain (i.e. His93) or the 8th residue in helix F (i.e. His F8). The O2-binding site is present on
the other side of the heme plane, at the sixth coordination position. A second histidine residue, termed as distal
histidine, His64 or His E7, (not bonded to the heme) makes H-bond with oxygen molecules. O2 binds directly to
the iron atom of the heme only.

CH2
Distal histidine
CH3 CH
M V
N

N N
N
M H M CH3 H
CH3
2+
N N N Fe N
O
P H V CH2 CH
N N O
CH2 CH2 N N

Fe Heme
COOH
P M N N
CH2 CH3 N
P : Propionic; V : Vinyl; M : Methyl
CH2
Protoporphyrin IX N
COOH
Proximal histidine
Heme

Figure 1.23 The pyrrole rings and methylene bridge carbons are coplanar, and the iron atom resides in almost the
same plane. The fifth and sixth coordination positions of iron atom are directed perpendicular to—and directly above
and below—the plane of the heme ring. The fifth coordination position of the iron is linked to a ring nitrogen of the
proximal histidine. The distal histidine lies on the side of the heme ring opposite to proximal histidine.

Binding of oxygen to Mb

Myoglobin, functions as an oxygen-storage protein, binds with one oxygen molecule. The dissociation of the
myoglobin-oxygen complex (MbO2) is described by

K
ˆˆˆˆ†
d
MbO2 ‡ˆˆˆˆ Mb + O2

Kd is the dissociation constant and used to express the affinity of a protein for a ligand. A lower value of Kd
corresponds to a higher affinity of ligand for the protein. Kd is given by:

[Mb][O2 ] [Mb][O2 ]
Kd = or [MbO2 ] =
[MbO2 ] Kd

The fractional saturation (Y) of myoglobin

Number of binding sites occupied [MbO2 ] [Mb][O2 ] / Kd [O2 ]


Y= = = =
Total number of binding sites [MbO2 ] + [Mb] [Mb][O2 ] / Kd + [Mb] [O2 ] + Kd

When all of the myoglobin molecules in solution are bound to oxygen, Y = 1.0. Myoglobin is saturated with oxygen
at this point. When Y = 0.5, myoglobin is half-saturated with oxygen. Kd is equal to the [O2] at which half of the
myoglobin molecules in solution are occupied or [O2]0.5.

[O2 ]
Y=
[O2 ] + [O2 ]0.5
This page intentionally left blank.
Biomolecules and Catalysis 31

Ascent to high altitude triggers a substantial rise in BPG concentration in red cells, with a consequent increase in
the availability of oxygen to tissues. The high altitude-induced increase in BPG causes decrease in hemoglobin’s
oxygen affinity. At sea level, the difference between arterial and venous pO2 is 70 torr and hemoglobin unloads 38%
of its bound O2. However, when the arterial pO2 drops to 55 torr, as it does at an altitude of 4500 m, hemoglobin
would be able to unload only 30% of its O2. High-altitude adaptation (which decreases the amount of O2 that
hemoglobin can bind in the lungs but, to a greater extent, increases the amount of O2 it releases at the tissues)
allows hemoglobin to deliver a near-normal 37% of its bound O2.
Similarly, the affinity of fetal hemoglobin (hemoglobin F) for O2, which is greater than that for adult hemoglobin
(hemoglobin A), facilitates the movement of O2 from the mother to the fetus. The cause of this greater affinity is
the poor binding of BPG by the γ-polypeptide chains that replace β-chains in fetal hemoglobin.

Effect of temperature: A rise in temperature shifts the curve to the right. Conversely, a fall in temperature shifts
the curve to the left, and a lower pO2 is required to bind a given amount of O2.

Left shift
(Higher affinity)
Decreased temperature
Decreased BPG concentration
Increased pH
1.0
Y (fractional saturation)

Figure 1.28 Effect of pH, BPG


Right shift concentration and temperature
(Lower affinity) on the oxygen affinity of
0.5
Increased temperature
hemoglobin. Change in these
Increased BPG concentration
Decreased pH factors shift the entire oxygen-
hemoglobin dissociation curve
either to the left (higher affinity)
0.0
0 50 100 or to the right (lower affinity).

pO2 (torr)

Carbon dioxide transport by Hb

Carbon dioxide binds to hemoglobin and forms carbaminohemoglobin. It combines with the amino groups of
N-terminal amino acids of α- and β-globin chains. Hemoglobin transports only about 23% of carbon dioxide. The
greatest percentage of carbon dioxide (about 70%) is transported through blood plasma as bicarbonate ions.

Sickle-cell hemoglobin (HbS)


HbS forms as a result of a single amino acid substitution in the β-chain of Hb. Replacement of the glutamate
residue at position 6 in the β-chain by a valine residue is the only chemical difference between HbA and sickle-cell
hemoglobin. This residue is present on the outer surface of the molecule. The change produces a sticky hydrophobic
spot on the surface that results in abnormal quaternary association of hemoglobin. This makes the deoxyHbS less
soluble than deoxyHbA. Insoluble deoxyHbS forms polymers that aggregate into tubular fibers. The formation of
insoluble deoxyHbS fibers distorts the RBC into the elongated sickle shape structure which is the characteristic of
the disease, sickle-cell anemia. Sickle-cell anemia and sickle cell trait are different. Sickle cell trait describes a
condition in which an individual has one abnormal allele of β-globin gene (heterozygous). A person with sickle cell
anemia has two copies of abnormal β-globin gene (homozygous).

1.2.6 Models for the behavior of allosteric proteins


Several models have been proposed to explain the behavior of allosteric proteins/enzymes. The concerted model
proposed by Monod, Wyman and Changeux (MWC) and the Sequential model suggested by Koshland, Nemethy
and Filmer (KNF) have been the most popular. Both these models explain the non-hyperbolic kinetics by assuming
This page intentionally left blank.
Biomolecules and Catalysis 33

1.3 Protein folding


Protein folding is the physical process by which a polypeptide folds into its characteristic and functional three-
dimensional conformation. The correct three-dimensional structure is essential to protein function. Failure to fold
into native structure produces inactive proteins. A protein molecule folds spontaneously during or after biosynthesis.
However, the process also depends on the nature of solvent, the concentration of salts, the temperature, and the
presence of molecular chaperones.
One of the most important experiment, which helped in understanding the process of protein folding was carried out
by Christian Anfinsen and colleagues in the early 1960s. C. Anfinsen studied the refolding of protein ribonuclease
A. Ribonuclease A isolated from bovine pancreas is an enzyme that has a molecular mass of 13,700 Da. It contains
124 amino acid residues and four disulfide linkages. In the presence of urea, a denaturant, and β-mercaptoethanol,
a reducing agent, ribonuclease is denatured and the disulfide bonds are broken. When the protein is allowed to
renature by removing the denaturant and the reductant, the protein regains its native conformation, including four
correctly paired disulfide bonds. This finding provided the first evidence that the amino acid sequence of a polypeptide
chain contains all the information required to fold the chain into its native three dimensional structure. However,
when the reductant is removed while the denaturant is still present, the disulfide bonds are formed again in protein
but most of the disulfide bonds are formed between incorrect partners. This indicates that weak interactions are
required for correct positioning of disulfide bonds and assumption of the native conformation.

Oxidation of the
sulfhydryl group in
the absence of urea
and b-mercaptoethanol

8 M urea Native ribonuclease

b-Mercaptoethanol

Native ribonuclease Denatured


ribonuclease Oxidation of the
sulfhydryl group in
the presence of
8 M urea Scrambled ribonuclease

Figure 1.31 Denaturation and renaturation of ribonuclease. Depending on the conditions for renaturation, we obtain
either native ribonuclease or scrambled ribonuclease.

The amino acid sequence of a protein determines its native conformation. But, if this is true, how do proteins find
the right conformation out of the simply endless number of potential three-dimensional forms that it could randomly
fold into? After all, the folding of a protein is not a chemical reaction. The folding pathway of a polypeptide is very
complicated, and not all the principles that guide the process have been worked out. However, there are several
models to explain folding. According to one model, folding is initiated by a spontaneous collapse of the unfolded
polypeptide chain into a partly organized globular state, mediated by hydrophobic interactions among nonpolar
residues (hydrophobic collapse). The collapsed state is referred to as a molten globule. This state is clearly
different from the native and the denatured state. The molten globule has most of the secondary structure of the
native state but it is less compact and the proper packing interactions in the interior of the protein have not been
formed. This event is very fast, usually completes within a few milliseconds. We therefore know almost nothing
about the process that leads to the molten globule. However we know some of the properties of this state. As
mentioned above the molten globule has most of the secondary structure of the native state. It is less compact
34 Biomolecules and Catalysis
than the native structure and lacks the proper packing interactions in the interior of the protein. The interior side
chains remain mobile, more closely resembling a liquid than the solid-like interior of the native state.

Fast Slow

Molten globule Folded

Unfolded

Figure 1.32 The molten globule state is an intermediate state in the folding pathway when a polypeptide chain
converts from an unfolded to a folded native state.

1.3.1 Molecular chaperones


Not all proteins fold spontaneously after or during synthesis in the cell. Folding of many proteins requires molecular
chaperones. Molecular chaperones are a class of proteins which bind to incompletely folded or unfolded proteins in
order to assist their folding or prevent them from aggregating. Chaperones function mainly by preventing formation
of incorrect structures rather than by promoting formation of correct structures. Chaperones may also be required
to assist the refolding of stress-denatured proteins, formation of oligomeric structures, protein trafficking through
membranes and assistance in proteolytic degradation.
Molecular chaperones were first identified in bacteria E. coli but are present in both prokaryotes and eukaryotes
(ubiquitous). Several molecular chaperones are included among the heat-shock proteins (hence their designation as
Hsp), because they are synthesized in increased amounts after a brief exposure of cells to an elevated temperature
(for example, 42°C for cells that normally live at 37°C). Chaperones are usually classified according to their
molecular weight (Hsp40, Hsp60, Hsp70, Hsp90, Hsp100 and the small Hsps). There are two major families of
molecular chaperones known as the Hsp60 and Hsp70 families. The members of these two chaperone families
function differently.
The members of Hsp70 family (Hsp70, Hsc70, Hsp40 and GrpE) act early in the life of many proteins, binding to
a string of about seven hydrophobic amino acids before the protein leaves the ribosome. The Hsp70 polypeptide
chain is divided into two functional regions, one that binds and hydrolyses ATP and a second that binds hydrophobic
segments of unfolded polypeptide chains. The polypeptide binding domain is an antiparallel C-terminal region. Hsp70
is induced by stress (e.g. heat shock) whereas Hsc70 is constitutively expressed in cells. Cytosolic Hsp70s prevent
misfolding and maintain the polypeptide chain in unfolded condition. Cytosolic Hsp70s are also necessary for normal
translocation of protein from cytosol into either ER or mitochondria. Hsp70 (DnaK in E. coli) works in tandem with
Hsp40 (DnaJ in E. coli). The ATP-dependent reaction cycle of Hsp70 is regulated by the Hsp40. Hydrolysis of ATP
to ADP is strongly accelerated by Hsp40.
The Hsp60 family of molecular chaperones (also called chaperonins) forms a large barrel-shaped structure that acts
later in a protein’s life, after it has been fully synthesized. Chaperonins bind unfolded, partly folded and incorrectly
folded protein molecules but not protein in their native state. This type of chaperone forms an isolation chamber
into which misfolded proteins are fed, preventing their aggregation and providing them a favorable environment
to refold. The typical structure is a ring of many subunits, forming a cylinder. Hsp60 itself in eukaryotes (GroEL
in E. coli) forms a structure consisting of 14 subunits that are arranged in two heptameric rings stacked on top of
each other in an inverted orientation. This structure associates with a ring shaped heptamer formed of subunits of
Hsp10 (GroES in E. coli), also described as co-chaperonin.
This page intentionally left blank.
Biomolecules and Catalysis 47

1.5 Nucleic acids


Nucleic acid was first discovered by Friedrich Miescher from the nuclei of the pus cells (Leukocytes) from discarded
surgical bandages and called it nuclein. Nuclein was later shown to be a mixture of a basic protein and a phosphorus-
containing organic acid, now called nucleic acid. There are two types of nucleic acids (polynucleotides): ribonucleic
acid (RNA) and deoxyribonucleic acid (DNA).

1.5.1 Nucleotides
The monomeric units of nucleic acids are called nucleotides. Nucleic acids therefore are also called polynucleotides.
Nucleotides are phosphate esters of nucleosides and made up of three components:
1. A base that has a nitrogen atom (nitrogenous base)
2. A five carbon sugar
3. An ion of phosphoric acid

Nitrogenous bases
Nitrogenous bases are heterocyclic, planar and relatively water insoluble aromatic molecules. There are two general
types of nitrogenous bases in both DNA and RNA, pyrimidines and purines.

H H
7
C6 5 N C4 5
1N C 3N CH
8
2 CH 2
HC C HC CH
4 N9 6
N H N
3 1

Purine Pyrimidine

Purines

Two different nitrogenous bases with a purine ring (composed of carbon and nitrogen) are found in DNA. The two
common purine bases found in DNA and RNA are adenine (6-aminopurine) and guanine (6-oxy-2-aminopurine).
Adenine has an amino group (–NH2) on the C6 position of the ring (carbon at position 6 of the ring). Guanine has
an amino group at the C2 position and a carbonyl group at the C6 position.

Pyrimidines

The two major pyrimidine bases found in DNA are thymine (5-methyl-2,4-dioxypyrimidine) and cytosine (2-oxy-4-
aminopyrimidine) and in RNA they are uracil (2,4-dioxypyrimidine) and cytosine. Thymine contains a methyl group
at the C5 position with carbonyl groups at the C4 and C2 positions. Cytosine contains a hydrogen atom at the C5
position and an amino group at C4. Uracil is similar to thymine but lacks the methyl group at the C5 position. Uracil
is not usually found in DNA. It is a component of RNA.

NH2 O NH2 O O
C C C C C
N N
N C HN C N CH HN CH HN C CH3
CH CH
HC C C C C CH C CH C CH
N H2N N O N O N O N
N H N H H H H

Adenine Guanine Cytosine Uracil Thymine

Sugars

Naturally occurring nucleic acids have two types of pentose sugars: ribose and deoxyribose sugar.
Ribose sugar is found in RNA. It is a five carbon monosaccharide with a hydroxyl group (–OH) on each carbon.
48 Biomolecules and Catalysis
Deoxyribose sugar is found in DNA. It is a five carbon monosaccharide, lacking one oxygen atom at 2’ position.
The hydroxyl group (–OH) at 2’ position of ribose sugar is replaced by a hydrogen (–H).

5’ 5’
HOCH2 OH HOCH2 OH
O O
4’ 1’ 4’ 1’

H 3’ 2’ H H 3’ 2’ H
HO OH HO H

b-D-Ribose b-D-2-Deoxyribose

The carbon atoms of the ribose and deoxyribose present in nucleoside or nucleotides are designated with a prime
(’) mark to distinguish them from the backbone numbering in the nitrogenous bases. Unprimed numbers refer to
the atoms of the nitrogenous base.

Sugar pucker
Pentose sugar is non-planar. This non-planarity is termed puckering. Pentose ring can be puckered in two basic
conformations: envelope and twisted. In the envelope form, the four carbons of the pentose sugar are nearly
coplanar and the fifth is away from the plane. In twisted form three atoms are coplanar and the other two lie away
on opposite sides of this plane. Twisting the C2’ and C3’ carbons relative to the other atoms results in twisted
forms of the sugar ring.
Sugar pucker can be endo or exo. C2’ or C3’ endo pucker means that C2’ or C3’ are on the same side as the base
and C4’-C5’ bond. Exo-pucker describes a shift in the opposite direction. Purines show a preference for the C2’-
endo pucker conformational type whereas pyrimidines favour C3’-endo. In RNA we find predominantly the C3’-endo
conformation.

5’ C N 5’ C
3’ N
3’

O 1’
4’ 4’ O 1’
2’

2’

Envelope form, C3’ endo Twisted form, C3’ endo and C2’ exo

5’ C 5’ C
N N
2’ 2’

O 1’ O
4’ 4’ 1’
3’

3’
Envelope form, C2’ endo Twisted form, C2’ endo and C3’ exo

Figure 1.42 Sugar puckers.

Nucleoside
Sugar and nitrogenous base join to form nucleoside. The bond between the sugar and the base is called the
N-glycosidic bond. The nitrogenous base lies above the plane of the sugar when the structure is written in the
standard orientation; that is, the configuration of the N-glycosidic linkage is β.
This page intentionally left blank.
Biomolecules and Catalysis 53

1.6 Structure of dsDNA


Watson and Crick first described the structure of the DNA double helix in 1953 using X-ray diffraction data of DNA
fibers obtained by R. Franklin and M. Wilkins. Watson, Crick, and Wilkins were awarded the 1962 Nobel Prize for
Medicine for discovering the molecular structure of DNA. R. Franklin died of cancer before the Nobel prize was
awarded. Nobel prizes are not awarded posthumously.
The Watson-Crick double helix model describes the features of the B form of DNA. However, there are many other
forms or conformations of DNA (such as A-, C-, Z-froms) which are very distinct from the B form. The form that
DNA would adopt depends on several factors: the hydration level, DNA sequence, chemical modifications of the
bases, the type and concentration of metal ions in solution.

1.6.1 B-DNA
B-form of DNA has following major features:
• Two long polynucleotide strands coiled around a central axis.
• Strands are wrapped plectonemically in a right handed helix.
• Strands are antiparallel i.e. one strand is oriented in the 5’→3’ direction and the other in the 3’→5’ direction.
• Strands interact by hydrogen bonds between complementary base pairs.
• G forms three hydrogen bonds with C.
• A forms two hydrogen bonds with T.

Major groove Major groove

H
HN–H CH3 O
O H–N
N N
5 4 7 5 7
4
6 8 8
5 6 5
6 C 3 N 6 T
H–N 1
9 3 N–H N
9
1
G 4 N 1 A 4 N
2 1 2
N 2 3 N 2 3
N Sugar N Sugar
Sugar O Sugar O
H– NH

Minor groove Minor groove

Figure 1.47 Standard base pairing between guanine and cytosine and between adenine and thymine via hydrogen
bonds.

• Angle of interaction between base pairs result in major and minor grooves. The angle between the C1’ atoms
is larger on one side than the other, generating two dissimilar grooves in the B-DNA. The side containing N7
of purines is termed the major groove, while the other side, containing N3 of purines is the minor groove.
• Helix diameter is 20Å.
• Helix rise per base pair is 3.32Å.
• Helix pitch (distance along the axis per 360 degree turn) is 33.2Å.
• 10.4 base pairs per helical turn.
• Base pairs are in the inside of the molecule stacked close to each other.
54 Biomolecules and Catalysis
Diameter
~20 Å

Minor groove

Helix pitch
Major groove 33.2 Å

3.32 Å
Axial rise

Figure 1.48 The Watson-Crick double helix is composed of about 10.4 bps per helical turn. Since 360° constitutes
one helical turn, there would be a 34.3° twist angle or rotation per residue between adjacent base pairs.

The position of the base pairs relative to the helix axis are described by another three parameters:

Base pair tilts: It is a shift of the base pairs short axis relative to the vertical helix axis. The tilt angle is
measured by considering the angle made by a line drawn through the two hydrogen bonded bases relative to
a line drawn perpendicular to the helix axis. The tilt angle opens in the direction of the phosphate backbone.

Base pair roll: It is a shift of the base pairs long axis relative to the vertical helix axis.

Propeller twist: It is the twist of the bases in a base pair against each other. A base pair is rarely a perfect
flat plane with each base in the same plane. Rather, each base has a slightly different roll angle with respect
to the other base. This makes the two bases look like an airplane propeller.

Tilt Roll Propeller Twist

1.6.2 Z-DNA
Z-DNA is a left-handed double helical structure with two anti-parallel strands that are held together by Watson-
Crick base pairing. The transition from B- to Z-DNA conformation occurs most readily in DNA segments containing
alternating purines and pyrimidines, especially alternations of C and G on one strand (and also in DNA segments
containing alternations of T G on one strand and C A on the other). The existence of Z-DNA was first suggested
by optical studies demonstrating that a polymer of alternating C and G in one strand in a 4 M NaCl solution. The
This page intentionally left blank.
Biomolecules and Catalysis 65

1.7.2 RNA World hypothesis


The concept of an RNA World is a way of answering the basic problem of what was the self-replicating molecule
present at the beginning of life. This hypothesis proposes that RNA was actually the first life-form on earth, later
developing a cell membrane around it and becoming the first prokaryotic cell (the phrase RNA World was first used
by Walter Gilbert in 1986). This hypothesis is supported by the RNA’s ability to store, transmit, and duplicate genetic
information, just like DNA does and to catalyze chemical reactions, just like protein does. Because RNA can perform
the tasks of both genetic materials and enzymes, RNA is believed to have once been capable of independent life.

1.7.3 RNA as genetic material


Some viruses contain an RNA as genetic material. One of the first experiments that established RNA as the genetic
material in RNA viruses was the reconstitution experiment of H.Fraenkel-Conrat and B.Singer. They took two
different strains of Tobacco Mosaic Virus (TMV), separated the RNAs from their protein coats, and reconstituted
hybrid viruses by mixing the proteins of one strain with the RNA of the second strain, and vice versa. When the
hybrid virus was spread on tobacco leaves, the lesions that developed corresponded to the TMV from which the
RNA had been obtained. Thus, it was concluded that RNA serves as the genetic material in TMV.

TMV type A

Infection of
tobacco leaf

RNA from TMV type A TMV type A


and Protein (capsid)
from TMV type B

TMV type B

Figure 1.59 In vivo reconstitution of a hybrid TMV virus. There are two strains of virus (TMV type A and type B)
which were separated into protein and RNA. The protein of one strain (type B) was allowed to recombine with the RNA
of the other (type A). The in vivo progeny of this hybrid had the protein originally associated with its RNA. This proves
that the genetic material of TMV is RNA, not protein.

Problem

What is the approximate molecular weight of duplex DNA required to code for glyceraldehyde phosphate dehydrogenase
(MW 40,000)?

Solution

The average molecular weight of an amino acid residue in a protein is 110. Thus, a protein whose molecular weight is
40,000 contains 40,000/110 = ~364 amino acids and requires a minimum DNA duplex of 3 × 364 = ~1090, nucleotide
pairs. Since each nucleotide pair has an average molecular weight of about 650, the molecular weight of this gene would
be about 1090 × 650 = 708,500. On the average, the molecular weight of coding DNA is about 18 times that of the cor-
responding protein.

Problem

The molecular weight of bacteriophage T4 dsDNA is 1.3 × 108.


1. How many amino acids can be coded for by T4 DNA?
2. How many different proteins of MW 55000 could be coded for by T4 DNA?
66 Biomolecules and Catalysis
Solution

1. The genetic code is a triplet code. It will take a sequence of three nucleotides on the coding strand of DNA to specify
one amino acid. The DNA of T4 contains:
1.3 ´ 108
= 2 ´ 105 nucleotide pairs = 2 ´ 105 nucleotides in the coding strand.
650

2 ´ 105
= ~ 6.7 ´ 104 codons.
3

2. The average MW of an amino acid residue is 110. A protein of MW 55000 contains:

55000
= 500 amino acids.
110

6.7 ´ 104
6.7 ´ 104 codons can yield: = 134.
500

Nucleic acid conversion factors


Average MW of a DNA base pair = 650 Da
1 A260 unit = ~50 microgram/ml of double strand DNA
1 A260 unit = ~40 microgram/ml of single strand RNA
1 A260 unit = ~33 microgram/ml of single strand DNA
1000 bp DNA open reading frame = 333 amino acids = 37,000 Da protein
To calculate the concentration of plasmid DNA in solution using absorbance at 260 nm:
(Observed A260) × (dilution factor) × (0.050) = DNA concentration in μg/μl

1.8 Carbohydrates
Carbohydrates are polyhydroxy aldehydes or polyhydroxy ketones, or compounds that can be hydrolyzed to them.
In the majority of carbohydrates, H and O are present in the same ratio as in water, hence also called as hydrates
of carbon. Carbohydrates are the most abundant biomolecules on Earth. Carbohydrates are classified into following
classes depending upon whether these undergo hydrolysis and if so on the number of products form:

Monosaccharides are simple carbohydrates that consist of a single polyhydroxy aldehyde or ketone unit.

Oligosaccharides are polymers made up of two to ten monosaccharide units joined together by glycosidic linkages.
Oligosaccharides can be classified as di-, tri-, tetra- depending upon the number of monosaccharides present.
Amongst these the most abundant are the disaccharides, with two monosaccharide units.
Polysaccharides are polymers with hundreds or thousands of monosaccharide units. Polysaccharides are not sweet
in taste hence they are also called non-sugars.

1.8.1 Monosaccharide
Monosaccharides consist of a single polyhydroxy aldehyde or ketone unit. Monosaccharides are the simple sugars
and they have a general formula CnH2nOn. Monosaccharides are colorless, crystalline solids that are freely soluble
in water but insoluble in nonpolar solvents. The most abundant monosaccharide in nature is the D-glucose.
Monosaccharides can be further sub classified on the basis of:

Number of the carbon atoms

Monosaccharides can be named by a system that is based on the number of carbons with the suffix-ose added.
Monosaccharides with four, five, six and seven carbon atoms are called tetroses, pentoses, hexoses and heptoses,
respectively.
This page intentionally left blank.
76 Biomolecules and Catalysis
O-linked glycosidic bond N-linked glycosidic bond

CH2OH CH2OH
C O O C O
O O CH2 CH Ser O NH C CH2 CH Asn
1 1
OH NH OH NH
O H O H

Monosaccharide Monosaccharide
Core protein Core protein

Figure 1.67 Carbohydrates are covalently attached to many different proteins to form glycoproteins. Carbohydrates
are attached either to the amide nitrogen atom in the side chain of asparagine (termed an N-linkage) or to the oxygen
atom in the side chain of serine or threonine (termed an O-linkage).

1.8.8 Reducing and non-reducing sugar


Sugars capable of reducing ferric or cupric ion are called reducing sugar. A reducing sugar is any sugar that either
has an aldehyde group or is capable of forming one in solution through isomerization. This functional group allows
the sugar to act as a reducing agent.
All monosaccharides whether aldoses and ketoses, in their hemiacetal and hemiketal form are reducing sugars.
All disaccharides formed from head to tail condensation are also reducing sugar i.e. disaccharides except sucrose,
trehalose are reducing sugars. All reducing sugars undergo mutarotation in aqueous solution.
Disaccharides like sucrose, trehalose not capable of reducing ferric or cupric ion are called non-reducing sugar.
In sucrose and trehalose, anomeric carbons of both monosaccharides participate in glycosidic bond formation. So,
they do not contain free anomeric carbon atoms. Sucrose and trehalose are therefore non-reducing sugar, and have
no reducing end. So it cannot be oxidized by cupric or ferric ions. In describing disaccharides or polysaccharides,
the end of a chain that has a free anomeric carbon (i.e. is not involved in a glycosidic bond) is called the reducing
end of the chain.

1.9 Lipids
Biological lipids are a chemically diverse group of organic compounds which are insoluble or only poorly soluble
in water. They are readily soluble in nonpolar solvents such as ether, chloroform, or benzene. The hydrophobic
nature of lipids is due to the predominance of hydrocarbon chains (—CH2—CH2—CH2—) in their structures. Unlike
the proteins, nucleic acids, and polysaccharides, lipids are not polymers.

Functions
Biological lipids have diverse functions. The four general functions of biological lipids have been identified.
• They serve as a storage form of metabolic fuel.
• They serve as a transport form of metabolic fuel.
• They provide the structural components of membranes.
• They have protective functions in bacteria, plants, insects, and vertebrates, serving as a part of the outer
coating between the body of the organism and the environment.

Apart from the general functions biological lipids serve as pigments (carotene), hormones (vitamin D derivatives,
sex hormones), signaling molecules (eicosanoids, phosphatidylinositol derivatives), cofactors (vitamin K), detergents
(bile salt) and many other specialized functions.
Biomolecules and Catalysis 77

1.9.1 Fatty acids


Fatty acids are the simplest form of lipids and serve as constituents in a large number of complex forms of lipids.
Fatty acids are long-chain hydrocarbons (4 to 36 carbons long) with one carboxyl group. Fatty acids in biological
systems usually contain an even number of carbon atoms. The 16- and 18-carbon fatty acids are most common.
The alkyl chain may be saturated or unsaturated. Unsaturated fatty acids may contain one or more double bonds.
Fatty acids are amphipathic by nature; that is, they have both nonpolar and polar ends.
O
b
w C OH
H3C a
Hydrocarbon chain

Fatty acyl chain

Figure 1.68 Structure of fatty acid.

By an older system, in a fatty acid second carbon is referred to as the α-carbon, third carbon as the β-carbon and
the end methyl carbon as the ω-carbon.

Table 1.16 Predominant naturally occurring fatty acids

Common name Systematic name Carbon atoms : Double bonds

Saturated fatty acid

Lauric acid Dodecanoic acid 12 : 0

Myristic acid Tetradecanoic acid 14 : 0

Palmitic acid Hexadecanoic acid 16 : 0

Stearic acid Octadecanoic acid 18 : 0

Arachidic acid Eicosanoic acid 20 : 0

Unsaturated fatty acid

Palmitoleic acid cis-Δ9-Hexadecenoic acid 16 : 1

Oleic acid cis-Δ9-Octadecenoic acid 18 : 1

Linoleic acid all cis-Δ9, 12-Octadecadienoic acid 18 : 2


9, 12, 15
Linolenic acid all cis-Δ -Octadecatrienoic acid 18 : 3

Arachidonic acid all cis-Δ5, 8, 11,14-Eicosatetraenoic acid 20 : 4

Saturated and unsaturated fatty acids


Saturated fatty acids have no double bonds in the chain. Their general formula is CH3—(CH2)n—COOH where n
specifies the number of methylene groups between the methyl and carboxyl carbons. Examples of predominant
saturated fatty acids are lauric, myristic, palmitic and others.
Unsaturated fatty acids have one or more double bonds, and called monounsaturated or polyunsaturated respectively.
The double bonds in naturally occurring fatty acids are generally in a cis as opposed to a trans configuration. The
double bonds of polyunsaturated fatty acids are almost never conjugated (alternating single and double bonds).
H H H

CH2 C C CH2 CH2 C C CH2

H
Cis Trans
78 Biomolecules and Catalysis
The systematic name includes the number of carbons, the number of double bonds, and the positions of the double
bonds. For example, stearic acid (a saturated fatty acid) has 18 carbons and has the systematic name octadecanoic
acid (18:0). The notation 18:0 denotes an 18 carbons fatty acid with no double bonds. Similarly, oleic acid is an
18 carbons fatty acid with one double bond and has the systematic name octadecenoic acid (18:1). An 18 carbons
fatty acid with two double bonds is octadecadienoic acid (18:2). The notation 18:1 denotes an 18 carbons fatty
acid with one double bond, whereas 18:2 signifies that there are two double bonds.
Two systems are used for designating the position of double bonds in an unsaturated fatty acid. In carboxyl-
reference system, fatty acid carbon atoms are numbered starting from the carboxyl terminus. The positions of the
double bonds are described by counting from the carboxyl carbon. The position of a double bond is represented by
the symbol Δ followed by a superscript number. For example, cis-Δ9 means that there is a cis double bond between
carbon atoms 9 and 10; trans-Δ2 means that there is a trans double bond between carbon atoms 2 and 3. In this
nomenclature the carboxyl carbon is designated carbon 1. For example, palmitoleic acid has 16 carbons and has
a double bond between carbons 9 and 10. It is designated as 16:1:Δ9.
In omega-reference system, the position of the double bond are indicated relative to the omega carbon (i.e.
number 1 is assigned to the omega carbon). For example, ω6 indicates a double bond on the sixth carbon counting
from the ω-carbon.

Essential fatty acids

Essential fatty acids are those fatty acids which are not synthesized by animals and must be obtained from diet.
Linoleate and linolenate are the two essential fatty acids for humans and other animals. Humans lack the enzymes
to introduce double bonds at carbon atoms beyond C-9 in the fatty acid chain. Hence, humans cannot synthesize
linoleate and linolenate. Fatty acids that can be endogenously synthesized are termed as nonessential fatty acids.
They are nonessential in the sense that they do not have to be obligatorily obtained from diet.

Melting point of fatty acids

The melting point of fatty acids depends on chain length, presence or absence of double bond and number of double
bonds (i.e. degree of unsaturation). The longer the chain length, the higher the melting point, and the greater
the number of double bonds, the lower the melting point. The presence of double bonds makes unsaturated chain
more rigid. As a result, unsaturated chains cannot pack themselves in crystals efficiently and densely as saturated
chain, so, they have a lower melting point as compared to saturated fatty acids. Similarly, the unsaturated fatty
acids with cis configuration have lower melting points than the unsaturated fatty acids with trans configuration.

Problem

Why unsaturated fatty acids have low melting points?

Solution

The presence of double bonds makes unsaturated chain more rigid. As a result, unsaturated chains cannot pack them-
selves in crystals efficiently and densely as saturated chain, so, they have lower melting point as compared to saturated
fatty acids.

1.9.2 Triacylglycerol and Wax


Triacylglycerols (also called triglycerides) are triesters of fatty acids and glycerol. They are composed of three fatty
acids and a glycerol molecule. Triacylglycerols are of two types – simple and mixed type. Those containing a single
kind of fatty acids are called simple triacylglycerols and with two or more different kinds of fatty acids are called
mixed triacylglycerols. The general formula of triacylglycerol is given below:
This page intentionally left blank.
84 Biomolecules and Catalysis
H
O

COOH
H

CH3

Figure 1.76 Structure of leukotriene A.

Table 1.18 Biological effects of eicosanoids

Type Major functions

Prostaglandins Mediation of inflammatory response

Regulation of nerve transmission

Inhibition of gastric secretion

Sensitization to pain

Stimulation of smooth muscle contraction

Thromboxanes Platelet aggregation

Aorta constriction

Prostacyclins Thromboxane antagonists

Leukotrienes Bronchoconstriction

Leukotaxis

1.9.7 Plasma lipoproteins


Triacylglycerols, phospholipids, cholesterol and cholesterol esters are transported in human plasma in association
with proteins as lipoproteins. Blood plasma contains a number of soluble lipoproteins, which are classified, according
to their densities, into four major types. These lipid-protein complexes function as a lipid transport system because
isolated lipids are insoluble in blood. There are four basic types of lipoproteins in human blood: chylomicrons, very
low density lipoproteins (VLDL), low density lipoproteins (LDL), and high density lipoproteins (HDL). A lipoprotein
contains a core of neutral lipids, which includes triacylglyerols and cholesterol esters. This core is coated with a
monolayer of phospholipids in which proteins (called apolipoprotein) and cholesterol are embedded.

Table 1.19 Some properties of major classes of human plasma lipoproteins

Density Phospho- Free Cholesterol Triacyl-


Lipoprotein Protein Apolipoprotein
(g/mL) lipids cholesterol esters glycerols

Chylomicrons <1.006 1.5–2.5 7–9 1–3 3–5 85 A-I, C-I, B-48

VLDL 0.95–1.006 5–10 15–20 5–10 10–15 50 B-100, C-I, C-II

LDL 1.006–1.063 20–25 15–20 7–10 35–40 7–10 B-100

HDL 1.063–1.210 50–55 20–25 3–4 15 3–4 A-I, A-II, C-I

1.10 Vitamins
Vitamins are organic compounds required by the body in trace amounts to perform specific cellular functions. They
can be classified according to their solubility and their functions in metabolism. The requirement for any given
vitamin depends on the organisms. Not all vitamins are required by all organisms. Vitamins are not synthesized by
humans, and therefore must be supplied by the diet. Vitamins may be water soluble or fat soluble. Nine vitamins
(thiamines, riboflavin, niacin, biotin, pantothenic acid, folic acid, cobalamin, pyridoxine, and ascorbic acid) are
classified as water soluble, whereas four vitamins (vitamins A, D, E and K) are termed fat-soluble. Except for
vitamin C, the water soluble vitamins are all precursors of coenzymes.
Biomolecules and Catalysis 85

1.10.1 Water-soluble vitamins


Thiamine (Vitamin B ) 1

Thiamine pyrophosphate (TPP) is the biologically active form of the vitamin, formed by the transfer of a pyrophosphate
group from ATP to thiamine. Thiamine is composed of a substituted thiazole ring joined to a substituted pyrimidine
by a methylene bridge.

Thiazolium Aminopyrimidine
Reactive H NH2
H NH2 carbon +
S N N
+
S N N AMP
ATP
N CH3
CH3
N CH3
CH3 O
TPP synthetase
O —
O P O
H
Thiamine O

O P O

O

Thiamine pyrophosphate (TPP)

Figure 1.77 Structure of thiamine and thiamine pyrophosphate.

TPP serves as a coenzyme in the oxidative decarboxylation of α-keto acid, and in the formation or degradation of
α-ketols (hydroxy ketones) by transketolase.

Pyruvate (α-keto acid) Acetaldehyde + CO2 (Enzyme: Pyruvate decarboxylase)

Xylulose-5-P + Ribose-5-P Glyceraldehyde-3-P + Sedoheptulose-7-P (Enzyme: Transketolase)

Beri-Beri is a severe thiamine-deficiency syndrome found in areas where polished rice is the major component of
the diet.

Riboflavin (Vitamin B ) 2

Riboflavin is a constituent of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). FMN is synthesized
after the addition of phosphate in riboflavin and FAD formed by the transfer of an AMP moiety from ATP to FMN.
FMN and FAD are each capable of reversibly accepting two hydrogen atoms, forming FMNH2 or FADH2. The oxidized
form of the isoalloxazine structure absorbs light around 450 nm. The color is lost, when the ring is reduced.
H
O O
Isoalloxazine

H3C N H3C N N
NH NH 2H
+


N N O N N O 2e N
H3C H3C

CH2 CH2 H
H C OH ADP PPi H C OH FADH2 (Reduced)
ATP ATP
Ribitol H C OH FMN H C OH

H C OH H C OH

CH2OH H2C O P P Adenosine

Riboflavin FAD (Oxidized)

Figure 1.78 Structure and biosynthesis of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD).
86 Biomolecules and Catalysis

Niacin
Niacin, or nicotinic acid, is a substituted pyridine derivative. The biologically active coenzyme forms are nicotinamide
adenine dinucleotide (NAD+) and its phosphorylated derivative, nicotinamide adenine dinucleotide phosphate (NADP+).
Nicotinamide, is a derivative of nicotinic acid that contains an amide instead of a carboxyl group. NAD+ and NADP+
serve as coenzymes in oxidation-reduction reactions in which the coenzyme undergoes reduction of the pyridine
ring by accepting a hydride ion (H–). The reduced forms of NAD+ and NADP+ are NADH and NADPH, respectively.
Deficiency of niacin causes pellagra, a disease involving the skin and central nervous system. The symptoms of
pellagra progress through the three Ds: Dermatitis, Diarrhoea, Dementia, and, if untreated, death.

O O
H H H
NH2
C C
+
N NH2 H NH2
O O

5’ 5’ + 2e

:
N H2C O P O P O CH2 N N
N
O
O O— O—
H H NADH (Reduced)
1’
H H H
H
OH OH
OH OH

Adenosine

Figure 1.79 Structure of NAD+ (Oxidized).

Biotin
Biotin is a coenzyme in carboxylation reactions, in which it serves as a mobile carboxyl group carrier. Biotin is
covalently bound to the enzyme by an amide linkage between the carboxyl group of its valerate side chain and the
ε-amino group of an enzyme Lys residue to form a biocytin (alternatively, biotinyllysine) residues.

C
2'
HN 1' NH
3'

H C C H
4 3

5 2 H
H2C 1
C

S CH2 CH2 CH2 CH2 COO

Valerate side chain

Figure 1.80 Structure of Biotin.

Most biotin-dependent carboxylations use bicarbonate as the carboxylating agent and transfer the carboxyl group
to a substrate carbanion. Examples of some important biotin-dependent carboxylations are given below:

ATP + HCO—
3
+ Pyruvate Oxaloacetate + ADP (Enzyme: Pyruvate carboxylase)

ATP + HCO—
3
+ Acetyl-CoA Malonyl-CoA + ADP (Enzyme: Acetyl-CoA carboxylase)

ATP + HCO—
3
+ Propionyl-CoA Methylmalonyl-CoA + ADP (Enzyme: Propionyl-CoA carboxylase)

Biotin deficiency does not occur naturally because the vitamin is widely distributed in foods. Raw egg white contains
a glycoprotein, avidin, which tightly binds biotin and prevents its absorption from the intestine. The avidin homolog
streptavidin, which is secreted by the Streptomyces avidinii, also has high affinity for biotin.
This page intentionally left blank.
92 Biomolecules and Catalysis

1.12 Enzymes
An enzyme is a biocatalyst that increases the rate of chemical reaction without itself being changed in the overall
process. Virtually all cellular reactions or processes are mediated by enzymes. Enzymes have several properties
that make them unique.
• Most (but not all) enzymes are proteins. With the exception of a small group of catalytic RNA molecules, all
enzymes are proteins.
• Enzymes are highly specific. They are specialized proteins and have a high degree of specificity for their substrates.
• Enzymes exhibit enormous catalytic power. It increases the rate of a reaction by lowering the activation energy.
• Enzymes do not change the equilibrium state of a biochemical reaction. It changes only the rate at which
equilibrium is achieved.
Proteinaceous enzymes can be divided into two general classes: simple enzymes, consist entirely of amino acids
and conjugated enzymes, consist of proteins as well as non-protein components. The non-protein component is
called a cofactor, which is required for catalytic activity. Removal of cofactor from a conjugated enzyme leaves
only protein component, called an apoenzyme, which generally is biologically inactive. The complete, biologically
active conjugated enzyme (simple enzyme plus cofactor) is called a holoenzyme. A cofactor can be linked to the
protein portion of the enzyme either covalently or non-covalently. Some cofactors are simple metal ions and other
cofactors are complex organic compounds, which are also called coenzymes. Many coenzymes are vitamins or
contain vitamins as part of their structure. Some coenzymes are only transiently associated with a given enzyme
molecule, so that they function as cosubstrates. Coenzymes which are tightly associated with the protein covalently
or non-covalently are called prosthetic group.

Table 1.21 Vitamin B-complex and their coenzyme forms

Vitamin Coenzyme form Reaction or process promoted

Thiamine (B1) Thiamine pyrophosphate Decarboxylation, aldehyde group transfer

Riboflavin (B2) FAD and FMN Redox reaction

Pyridoxine (B6) Pyridoxal phosphate Amino group transfer


+ +
Nicotinic acid (niacin) NAD and NADP Redox reaction

Pantothenic acid (B5) Coenzyme A Acyl group transfer

Biotin Biocytin Carboxylation

Folic acid Tetrahydrofolic acid One-carbon group transfer

Vitamin B12 Deoxyadenosylcobalamin Intramolecular rearrangements

The vitamins in the human diet that are coenzyme precursors are all water soluble vitamins.

Table 1.22 Example of some enzymes and their cofactors

Fe2+ or Fe3+ Cytochrome oxidase, Catalase, Peroxidase, Xanthine oxidase

Cu2+ Cytochrome oxidase, Lysyl oxidase, Superoxide dismutase


2+
Zn Carbonic anhydrase, Alcohol dehydrogenase, Carboxypeptidase
2+
Mg Hexokinase, Enolase, Glucose-6-phosphatase

Mn2+ Arginase, Enolase, Pyruvate carboxylase


+
K Pyruvate kinase

Ni2+ Urease

Mo Dinitrogenase, Xanthine oxidase

Se Glutathione peroxidase
Biomolecules and Catalysis 93

1.12.1 Naming and classification of enzyme


Many enzymes have common names. For example, trypsin, a proteolytic enzyme, is secreted by the pancreas.
Common names provide little information about the reactions that enzymes catalyze. Many enzymes are named
for their substrates and for the reactions that they catalyze, with the suffix-ase added. As for example, ATPase is
an enzyme that helps in breaking down ATP, whereas ATP synthase is an enzyme that helps in synthesis of ATP.
Because of the confusion that arose from these common names, an International Commission on enzymes was
established to create a systematic basis for enzyme nomenclature.
The enzyme commission has developed a rule for naming enzymes. According to this rule, each enzyme is classified
and named according to the type of chemical reaction it catalyzes. The Enzyme Commission (EC) has given each
enzyme a number with four parts, like EC 2.7.1.2 (Hexokinase). The first three numbers define major class,
subclass, and sub-subclass, respectively. The last number is a serial number in the sub-subclass, indicating the
order in which each enzyme is added to the list.

Common name and EC numbers of some enzyme


Alcohol dehydrogenase EC 1.1.1.1
Phosphofructokinase EC 2.7.1.11
Glutamine synthetase EC 6.3.1.2
Acetylcholinesterase EC 3.1.1.7

Systematic classification

The first integer in the EC number designates the class of enzymes. There are six classes to which different enzymes
belong. These classes are:

EC 1 Oxidoreductase
Oxidoreductase catalyzes oxidation-reduction reactions.

Ared + Box ⎯→ Aox + Bred

Example
Oxidases Use oxygen as an electron acceptor but do not incorporate it into the substrate.
Dehydrogenases Use molecules other than oxygen (e.g. NAD+) as an electron acceptor.
Oxygenases Directly incorporate oxygen into the substrate.
Peroxidases Use H2O2 as an electron acceptor.

EC 2 Transferases
Transferases catalyze reactions that involve the transfer of groups from one molecule to another. Examples of such
groups include amino, carboxyl, carbonyl, methyl, phosphoryl and acyl (RC=O). Common trivial names for the
transferases often include the prefix trans.

A–B + C ⎯→ A + B–C

Examples
Transcarboxylases Transfers a carboxylate group to a substrate.
Transaminases Transfer amino group from amino acids to keto acids.
Kinases Transfer phosphate from ATP to a substrate.
Phosphorylases Transfer inorganic phosphate to a substrate.

EC 3 Hydrolases
Hydrolases catalyze reactions in which the cleavage of bonds is accomplished by adding water.

A–B + H2O ⎯→ A–H + B–OH


This page intentionally left blank.
102 Biomolecules and Catalysis
Hanes-Woolf plot

The Lineweaver-Burk equation may be rearranged to yield the linear equation for the Hanes-Woolf plot.

[S] 1 Km
= [S] +
V Vmax Vmax

Thus, a plot of [S]/V versus [S] is linear with a slope of 1/Vmax. The intercept on the [S]/V axis gives Km / Vmax.

Eadie-Hofstee plot

Another widely used linear form of the Michaelis-Menten equation is the Eadie-Hofstee plot, which is described by

V V V
=- + max
[S] Km Km

The plot of V/[S] versus V is linear with slope of –1/Km. The intercept on the V axis gives Vmax.

Vmax
Km

[S] V 1
1 Slope = – K
V Slope = [S]
Vmax m

Km
Vmax Vmax

0 [S] 0 V

Hanes plot Eadie-Hofstee plot

Effect of temperature and pH on enzyme’s catalytic activity


Enzymes generally function in a narrow range of temperature and pH. Each enzyme shows its highest activity at
the optimum temperature and optimum pH. Activity declines both below and above the optimum value. Hence,
when the activity is plotted against temperature or pH, a bell-shaped curve is usually obtained.
Temperature: The rate of most enzyme catalyzed reactions increases with increase in temperature. As a rule of
thumb, a typical reaction rate approximately doubles for every 10°C rise in temperature. But enzyme catalyzed
reactions show a more complex temperature dependence because raising the temperature above certain value may
cause denaturation of proteins so, an enzyme may lose its catalytic activity. Thus, both high and low temperature
influence the catalytic activity of an enzyme. The optimum temperature varies greatly from enzyme to enzyme and
also from organism to organism. For example, enzyme like Taq DNA Polymerase present in thermophilic bacteria
Thermus aquaticus has optimal temperature 72°C.
Percent maximum activity

100

Figure 1.92
50 The effect of temperature on enzyme
activity. The relative activity of an
enzymatic reaction as a function of
temperature. The decrease in the
20 40 60 80 activity above 50°C is due to thermal
denaturation.
Temperature (°C)

pH: Enzymes are sensitive to pH. Most enzymes are active within only a narrow pH range, typically 5 to 9. This pH
dependence is due to the presence of charged amino acids at the active site. Variations in pH cause changes in overall
This page intentionally left blank.
110 Biomolecules and Catalysis

1.12.7 Isozymes
Isozymes (also known as isoenzymes) are enzymes that differ in amino acid sequence but catalyze the same chemical
reaction. They are encoded by different genes located at different loci. These enzymes usually display different
kinetic parameters or different regulatory properties. Isozymes differ from allozymes, which are enzymes that arise
from alternative forms of the same gene i.e. alleles. A common example of isozyme is lactate dehydrogenase (LDH)
which catalyzes the reversible conversion of pyruvate to lactate. Human beings have two isozymic polypeptide
chains for this enzyme: H isozyme and M isozyme. The functional enzyme is tetrameric (homo– or heterotetramer).
Two different subunits combine randomly with each other forming five isozymes.
HHHH (H4) LDH1 In the heart and RBCs
HHHM (H3M) LDH2 In the reticuloendothelial system
HHMM (H2M2) LDH3 In the lungs
HMMM (HM3) LDH4 In the kidneys, placenta
MMMM (M4) LDH5 In the liver and striated muscle

Similarly, there are four important mammalian hexokinase isozymes that vary in subcellular locations and kinetics
with respect to different substrates and conditions. They are designated hexokinases I, II, III and IV or hexokinases
A, B, C and D. Hexokinases catalyze the phosphorylation of hexose (such as glucose). While hexokinases I, II and
III are capable of phosphorylating several hexoses, hexokinase IV (also referred to as glucokinase) acts only on
glucose as hexose. Glucokinase is mainly expressed in liver and pancreatic β-cells and is characterized by a low
affinity to its substrate glucose. Hexokinases I, II and III are referred to as ‘low-Km’ isozymes because of a high
affinity for glucose even at low concentrations (below 1 mM) and all three are strongly inhibited by their product,
glucose-6-phosphate. Glucokinase can only phosphorylate glucose if the concentration of this substrate is high
enough (its Km for glucose is 100 times higher than that of hexokinases I, II and III) and is not allosterically inhibited
by its product, glucose-6-phosphate.

1.12.8 Zymogen
An inactive precursor of an enzyme is called zymogen. Zymogen is cleaved to form the active enzyme. Many
proteolytic enzymes like chymotrypsin, trypsin are initially synthesized as inactive precursor chymotrypsinogen
and trypsinogen. Specific cleavage causes conformational changes in the inactive precursor that expose the
enzyme active site. In the duodenum, the pancreatic zymogens, trypsinogen, chymotrypsinogen, proelastase and
procarboxypeptidase are converted into active enzymes by enteropeptidase and trypsin, as shown in figure 1.99.

Trypsinogen

Enteropeptidase

Trypsin
Proelastase

Chymotrypsinogen Elastase

Procarboxypeptidase
Chymotrypsin

Carboxypeptidase

Figure 1.99 Activation of pancreatic zymogens.

Chymotrypsinogen, a single polypeptide chain of 245 amino acid residues, is converted to α-chymotrypsin,
which has three polypeptide chains linked by two of the five disulfide bonds present in the primary structure of
chymotrypsinogen.
Biomolecules and Catalysis 111

1 245
Chymotrypsinogen
(inactive)

Arg Ile Trypsin

p-chymotrypsin 1 15 16 245
(active)
p-chymotrypsin

14 15 147 148
Ser — Arg + Thr — Asn

Leu Ile Tyr Ala

a-chymotrypsin 1 13 16 146 149 245


(active)

Figure 1.100 Formation of chymotrypsin (active) from chymotrypsinogen (inactive). The numbering of amino acid
residues represents their positions in the primary sequence of the zymogen, chymotrypsinogen. The three polypeptide
chains (A, B and C) of chymotrypsin are linked by disulfide bonds.

1.12.9 Ribozyme
A ribozyme (term introduced by Kelly Kruger, also called RNA enzyme) is an RNA molecule, that catalyzes a chemical
reaction. Before the discovery of ribozymes, proteins were the only known biological catalysts. In 1967, Carl Woese,
Francis Crick, and Leslie Orgel were the first to suggest that RNA could act as a catalyst based upon findings that
it can form complex secondary structures. The first ribozyme was discovered in the 1980s by Thomas R. Cech,
who was studying RNA splicing in the ciliated protozoan Tetrahymena thermophila and Sidney Altman, who was
working on ribonucleoprotein, RNaseP. These ribozymes were found in the intron of an RNA transcript and RNA
component of RNaseP, which is involved in processing of pre-tRNAs. In 1989, Thomas R.Cech and Sydney Altman
won the Nobel prize in chemistry for their discovery of catalytic properties of RNA. Ribozymes often require divalent
metal ions such as Mg2+ as cofactors.
Although ribozymes are quite rare in the cell, their roles are sometimes essential for life. For example, the functional
part of the ribosome, the molecular machine that translates RNA into proteins, is fundamentally a ribozyme. Some
known ribozymes include RNase P, Group I and Group II introns, hairpin ribozyme, hammerhead ribozyme, hepatitis
delta virus ribozyme, and tetrahymena ribozyme.

Biochemical reactions catalyzed by ribozymes:


Many natural ribozymes catalyze either their own cleavage or the cleavage of other RNAs, but they have also been
found to catalyze the aminotransferase activity of the ribosome. Ribozymes that are known today catalyze two
types of biochemical reactions:
1. Cleavage of phosphodiester bonds (RNaseP, group I intron and group II intron)
2. Synthesis of peptide bonds (Peptidyl transferase)
However in the test tube, synthetic RNA molecules have been shown to carry out other biochemical reactions such
as synthesis of ribonucleotides and RNA molecules.

1.12.10 Examples of enzymatic reactions


Lysozyme
Lysozyme is an enzyme (EC 3.2.1.17), present in number of secretions such as tears, sweat and also found in egg
white. Alexander Fleming, who discovered penicillin, described lysozyme in 1922. Its structure was described by
David Chilton Phillips in 1965.
Lysozyme (present in Hen Egg White) is a monomer with 129 amino acid residues. It cleaves the β(1–4) glycosidic
bond that connects N-acetyl muramic acid (NAM) with the fourth carbon atom of N-acetylglucosamine (NAG) of
This page intentionally left blank.
Chapter 02

Bioenergetics and Metabolism

2.1 Bioenergetics
Bioenergetics is the quantitative study of the energy transductions that occur in living cells and of the nature and
functions of the chemical processes underlying these transductions.

Thermodynamic principles
The First law of thermodynamics states that the energy is neither created nor destroyed, although it can be
transformed from one form to another i.e. the total energy of a system, including surroundings, remains constant.

Mathematically, it can be expressed as:


U = q – w
U is the change in internal energy,
q is the heat exchanged from the surroundings,
w is the work done by the system.

If q is positive, heat has been transferred to the system, giving an increase in internal energy. When q is negative,
heat has been transferred to the surroundings, giving a decrease in internal energy. When w is positive, work has
been done by the system, giving a decrease in internal energy. When w is negative, work has been done by the
surroundings, giving an increase in internal energy.

The Second law of thermodynamics states that the total entropy of a system must increase if a process is to occur
spontaneously. Mathematically, it can be expressed as:

Dq
DS ³ where, S is the change in entropy of the system
T

Entropy is unavailable form of energy and it is very difficult to determine it, so a new thermodynamic term called
free energy is defined.

Free energy
Free energy or Gibb’s free energy indicates the portion of the total energy of a system that is available for useful
work (also known as chemical potential). The change in free energy is denoted as G.
Under constant temperature and pressure, the relationship between free energy change (G) of a reacting system
and the change in entropy (S) is expressed by following equation:

G = H – TS

Where, H is the change in enthalpy and T is absolute temperature. H is the measure of change in heat content
of reactants and products. The change in the free energy, G, can be used to predict the direction of a reaction at
constant temperature and pressure.
120 Bioenergetics and Metabolism
If G is negative, the reaction proceeds spontaneously with the loss of free energy (exergonic),
G is positive, the reaction proceeds only when free energy can be gained (endergonic),
G is 0, the system is at equilibrium; both forward and reverse reactions occur at equal rates,
G of the reaction A  B depends on the concentration of reactant and product. At constant temperature and
pressure, the following relation can be derived:

[B]
DG = DG0 + RT ln
[A]

Where, G0 is the standard free energy change;


R is the gas constant;
T is the absolute temperature;
[A] and [B] are the actual concentrations of reactant and product.

Standard free energy change


The actual change in free energy (G) during a reaction is influenced by temperature, pressure and the initial
concentrations of reactants and products, and usually differs from standard free energy change, G0.
The chemical reaction has a characteristic standard free energy change and it is constant for a given reaction. It can
be calculated from the equilibrium constant of the reaction under standard conditions i.e. at a solute concentration of
1.0M, at temperature of 25°C and at 1.0 atm pressure. The free energy change which corresponds to this standard
state is known as standard free energy change, G0 .

Relationship between G0 and Keq


In a reaction A  B, a point of equilibrium is reached at which no further net chemical change takes place–that is,
when A is being converted to B, B is also being converted to A, as fast as A into B. In this state, the ratio of [B] to
[A] is constant, regardless of the actual concentrations of the two compounds:

[B]eq
Keq =
[A]eq

where Keq is the equilibrium constant, and [A]eq and [B]eq are the concentrations of A and B at equilibrium. The
concentration of reactants and products at equilibrium define the equilibrium constant, Keq. The equilibrium constant
Keq depends on the nature of reactants and products, the temperature and the pressure. Under standard physical
conditions (25°C and 1 atm pressure, for biological systems), the Keq is always the same for a given reaction,
whether or not a catalyst is present.

If the reaction A B is allowed to go to equilibrium at constant temperature and pressure, then at equilibrium
the overall free energy change (G) is zero. Therefore,

[B]eq
DG0 = –RT ln
[A]eq

So, G0 = –RT ln Keq

This equation allows some simple predictions:


Keq G0 Reaction
> 1.0 Negative proceeds forward
1.0 Zero is at equilibrium
< 1.0 Positive proceeds in reverse
As we know, the ionic composition of an acid or base varies with pH. So, the standard free energy calculated
according to the biochemistry convention is valid only at pH=7. Hence, under biochemistry convention, G0 is
symbolized by G0’ and likewise, the biochemical equilibrium constant is represented by K’eq.
This page intentionally left blank.
124 Bioenergetics and Metabolism

2.2 Metabolism
All cells function as biochemical factories. Within living cell, biomolecules are constantly being synthesized and
transformed into some other biomolecules. These synthesis and transformation constantly occur through enzyme
catalyzed chemical reactions. Together all the interconnected chemical reactions occurring within a cell are called
metabolism (derived from the Greek word for change). Each of the chemical reactions results in the transformation
of chemical compounds and the chemical compounds involved in this process are known as metabolites. Majority of
metabolic reactions do not occur in isolation but are always linked to some other reactions. It occurs in a series of
linked chemical reactions called metabolic pathways in which one chemical is transformed through a series of steps
into another chemical. Metabolic pathways proceed in a stepwise manner, transforming substrates into end products
through many specific chemical intermediates. Each step of metabolic pathways is catalyzed by a specific enzyme.

A Starting molecule

Reaction 1 Enzyme 1

Reaction 2 Enzyme 2

Reaction 3 Enzyme 3

D Product

Metabolic pathways can be linear (such as glycolysis), cyclic (such as the citric acid cycle) or spiral (such as the
biosynthesis of fatty acids). Flow of metabolites through metabolic pathway has a definite rate and direction.
Because the biomolecules within the cell are in a continual state of degradation and resynthesis. This is called as
dynamic state of body constituents.

Metabolism serves two fundamentally different purposes:


1. Generation of energy to drive vital functions and
2. The synthesis of biological molecules.

To achieve these metabolic pathways fall into two categories: Anabolic and catabolic pathways. Anabolic pathways
are involved in the synthesis of compounds and consume energy (i.e. endergonic in nature). Synthesis of amino
acids or synthesis of polypeptide from amino acids is an example of an anabolic process. Catabolic pathways are
involved in the oxidative breakdown of larger complex molecules and release of energy (i.e. exergonic in nature).
For example, when glucose is degraded to lactic acid in our skeletal muscle, energy is liberated. Living organisms
have learnt to trap this energy liberated during degradation and store it in the form of chemical bonds. As and when
needed, this bond energy is utilized for biosynthetic and mechanical works. The most important form of energy
currency in living systems is the ATP.
Some pathways can be either anabolic or catabolic, depending on the energy conditions in the cell. They are referred
to as amphibolic pathways. Amphibolic pathways occur at the ‘crossroads’ of metabolism, acting as links between
the anabolic and catabolic pathways, e.g. the citric acid cycle.
Bioenergetics and Metabolism 125

2.3 Respiration
Living cells require an input of free energy. Energy is required for the maintenance of highly organized structures,
synthesis of cellular components, movement, generation of electrical currents and for many other processes. Cells
acquire free energy from the oxidation of organic compounds that are rich in potential energy.
Respiration is an oxidative process, in which free energy released from organic compounds is used in the formation
of ATP. The compounds that are oxidized during the process of respiration are known as respiratory substrates,
which may be carbohydrates, fats, proteins or organic acids. Carbohydrates are most commonly used as respiratory
substrates.
During oxidation within a cell, all the energy contained in respiratory substrates is not released free in a single step.
Free energy is released in multiple steps in a controlled manner and used to synthesise ATP, which is broken down
whenever (and wherever) energy is needed. Hence, ATP acts as the energy currency of the cell.
During cellular respiration, respiratory substrates such as glucose may undergo complete or incomplete oxidation.
The complete oxidation of substrates occurs in the presence of oxygen, which releases CO2, water and a large
amount of energy present in the substrate. A complete oxidation of respiratory substrates in the presence of oxygen
is termed as aerobic respiration.
Although carbohydrates, fats and proteins can all be oxidized as fuel, but here processes have been described
by taking glucose as a respiratory substrate. Oxidation of glucose is an exergonic process. An exergonic reaction
proceeds with a net release of free energy. When one mole of glucose (180 g) is completely oxidized into CO2 and
water, approximately 2870 kJ or 686 kcal energy is liberated. Part of this energy is used for synthesis of ATP. For
each molecule of glucose degraded to carbon dioxide and water by respiration, the cell makes up to about 30 or
32 ATP molecules, each with 7.3 kcal/mol of free energy.

C6H12O6 + 6O2  6CO2 + 6H2O + Energy (ATP + Heat)

The incomplete oxidation of respiratory substrates occurs under anaerobic conditions i.e. in the absence of oxygen.
As the substrate is never totally oxidized, the energy generated through this type of respiration is lesser than that
during aerobic respiration.

2.3.1 Aerobic respiration


Enzyme catalyzed reactions during aerobic respiration can be grouped into three major processes: glycolysis, citric
acid cycle and oxidative phosphorylation. Glycolysis takes place in the cytosol of cells in all living organisms. The
citric acid cycle takes place within the mitochondrial matrix of eukaryotic cells and in the cytosol of prokaryotic
cells. The oxidative phosphorylation takes place in the inner mitochondrial membrane. However, in prokaryotes,
oxidative phosphorylation takes place in the plasma membrane.

Table 2.3 Intracellular location of major processes of aerobic respiration

In eukaryotes,

Glycolysis Cytosol

Citric acid cycle Mitochondrial matrix

Oxidative phosphorylation Inner mitochondrial membrane

In prokaryotes,

Glycolysis Cytosol

Citric acid cycle Cytosol

Oxidative phosphorylation Plasma membrane


This page intentionally left blank.
Bioenergetics and Metabolism 133

CoA CH3 C S CoA

O 3

H3C C
SH
S

SH
HS
O E2
CH3 C COO— TPP FAD
NADH
Pyruvate
2 4
1 E1 E3 5

+
NAD
CO2 CH3 FADH2
S
CH OH S

TPP
Hydroxyethyl-TPP

Figure 2.6 Structure of pyruvate dehydrogenase and its catalytic activities. Catalytic activities occur in four steps:
Step 1. Decarboxylation of pyruvate occurs with formation of hydroxy ethyl – TPP. Step 2. Transfer of the two carbon
unit to lipoic acid. Step 3. Formation of acetyl-CoA. Step 4. Lipoic acid is re-oxidized.

2.3.4 Krebs cycle


Krebs cycle (also known as the citric acid cycle or tricarboxylic acid cycle) was discovered by H. A. Kreb, a German
born British Biochemist, who received the Nobel prize in 1953. This cycle occurs in the matrix of mitochondria
(cytosol in prokaryotes). The whole cycle is explained in the following figure. The net result of Kreb cycle is that
for each acetyl group entering the cycle as acetyl-CoA, two molecules of CO2 are produced.

Step 1: The Krebs cycle begins with the condensation of an oxaloacetate (four carbon unit), and the acetyl group
of acetyl-CoA (two-carbon unit). Oxaloacetate reacts with acetyl-CoA and H2O to yield citrate and coenzyme A. This
reaction, which is an aldol condensation followed by a hydrolysis, is catalyzed by citrate synthase. Citrate has no
chiral center but has the potential to react asymmetrically if an enzyme with which it interacts has an active site
that is asymmetric. Such molecule is called prochiral molecule.

Step 2a and 2b: An isomerization reaction, in which water is first removed and then added back, moves the hydroxyl
group from one carbon atom to its neighbour. The enzyme catalyzing this step, aconitase (nonheme iron protein),
is the target site for the toxic compound fluoroacetate (used as a pesticide). Fluoroacetate blocks the citric acid
cycle by its metabolic conversion of fluorocitrate, which is a potent inhibitor of aconitase.

Step 3: Isocitrate is oxidized and decarboxylated to -ketoglutarate (also called oxoglutarate). In the first of four
oxidation steps in the cycle, the carbon carrying the hydroxyl group is converted to a carbonyl group. The immediate
product is unstable, losing CO2 while still bound to the enzyme. The oxidative decarboxylation of isocitrate is
catalyzed by isocitrate dehydrogenase.

Step 4: A second oxidative decarboxylation reaction results in the formation of succinyl-CoA from -ketoglutarate.
-ketoglutarate dehydrogenase catalyzes this oxidative step and produces NADH, CO2, and a high-energy thioester
bond to coenzyme A.
Step 5: The cleavage of the thioester bond of succinyl CoA is coupled with the phosphorylation of an ADP or a GDP
(substrate level phosphorylation). This step is catalyzed by succinyl CoA synthetase (succinate thiokinase). ATP and
GTP are energetically equivalent. This is the only step in the citric acid cycle that directly yields a compound with
high phosphoryl transfer potential through a substrate-level phosphorylation. Animal cells have two isozymes of
succinyl-CoA synthetase, one specific for ADP and the other for GDP. The GTP formed by succinyl-CoA synthetase
This page intentionally left blank.
Bioenergetics and Metabolism 139

2QH2
2e 2e 2e The path of electrons through complex III.
Fe-S Cyt c1 Cyt c
Two molecules of QH2 are oxidized to Q and
2e
bL release a total of four electrons. Two of the
2Q 2e
electrons are transported via iron-sulfur
bH center protein and cytochrome c1, directly
Q
to cytochrome c. The other two electrons
2e
move through cytochrome bL and bH and
QH2 reduce an oxidized Q to QH2.

Q-cycle
The mechanism of the participation of ubiquinone in the electron transport process was proposed by Peter Mitchell
and termed as a proton motive Q-cycle. Ubiquinones are hydrophobic and uncharged, and hence can migrate along
the hydrophobic core of the membrane. Diffusion of one ubiquinol takes place to the QP binding site adjacent to
the iron-sulfur protein at the P face of the mitochondrial membrane. One electron is transferred to Fe-S protein
and the second electron is transferred to the heme bL and two protons are released to the P face. The Fe-S protein
transfers the electron along the chain to Cyt c1 and cytochrome oxidase. The electron moves from heme bL to heme
bH. Ubiquinone then binds to bH at the Qn site and electron from the reduced bH forms ubisemiquinone at this site.
Now, a second ubiquinol molecule is oxidized at the QP site, the process follows as described above and the second
electron formed completes the reduction of ubisemiquinone to ubiquinol. Two protons are taken from the matrix for
this purpose and released to the P face. The ubiquinol, then, goes back to the pool and the Q-cycle is completed.
+ +
2H 2H
Cytoplasm (P-face) Cytoplasm (P-face)

QH2 QH2 QH2 QH2 —



e Fe–S Cyt c1 e Fe–S Cyt c1
QP or Qo site — QP site —
e bL e bL
Q Q
Q
Q pool Q e

Q pool e

QH2
.—
Q —
Q —
e bH e bH
Qn or Qc site Q
.— Qn site QH2

Matrix (N-face) First half of Q cycle Matrix (N-face) Second half of Q cycle
+
2H

Figure 2.11 A simplified outline of Q-cycle in mitochondria. In the first half of the cycle, two electrons of a bound
QH2 are transferred, one to cytochrome c1 and the other to a bound Q in a second binding site to form the ubisemiqui-
none. The newly formed Q dissociates and enters the Q pool. In the second half of the cycle, a second QH2 also gives
up its electrons to complex III, one to a molecule of cytochrome c1 and the other to reduce Q•— to QH2. The second
electron transfer results in the uptake of two protons from the matrix. Ubiquinol – QH2, Ubisemiquinone – Q•— and
Ubiquinone – Q.

Cytochromes are heme proteins having distinctive visible-light spectra. The major respiratory cytochromes are classified
as b, c or a, depending on the wavelengths of the spectral absorption peaks. Within each class, the cytochromes are
distinguished by smaller spectral differences. In the respiratory electron carriers, there are two b-type cytochromes
(b566 and b562), two c-type cytochromes (c and c1), and two a-type cytochromes (a and a3).
The heme prosthetic groups of a and b cytochromes are tightly, but not covalently, bound to their associated proteins;
whereas heme groups of c-type cytochromes are covalently attached through Cys residues. Cytochrome c is present
in all aerobic organisms. It is a peripheral protein of the inner mitochondrial membrane and binds via electrostatic
interactions to acidic phospholipids in intermembrane space. It is composed of a single polypeptide chain of 104 amino
acid residues covalently bound to the heme group. The degree of sequence homology in cytochrome c among species
has been used as a measure of the evolutionary distances that separate species.
140 Bioenergetics and Metabolism
Complex IV

Complex IV or cytochrome c oxidase catalyzes the transfer of electrons from the reduced form of cytochrome c
to molecular oxygen. It consists of 13 subunits and contains two heme groups and three copper ions, arranged
as two copper centers. The two heme groups termed heme a and heme a3, have distinct properties because they
are located in different environments within cytochrome c oxidase. The two copper centers are designated as a
and b. One center, Cua, contains two copper ions linked by two bridging cysteine residues. The second center, Cub,
is coordinated by three histidine residues. Cytochrome c transports electrons, one at a time, to the complex IV.
Within this complex, electrons are transferred, first to a Cua center, then to Cyt a, next to Cub center and Cyt a3,
and finally to O2, the ultimate electron acceptor, yielding H2O. Together, heme a3 and Cub form the active center
at which O2 is reduced to H2O.
+
2H

2e
Cua
Cyt c P-face

Cyt a

Cyt a3 N-face
Cub
+
1 + 2H
2 O2+2H H2O

Figure 2.12 The electron transfer pathway for cytochrome oxidase (Cyt c Cua Cyt a Cub. Cyt a3 O2).
Cytochrome c (a peripheral protein) binds on the P face of inner mitochondrial membrane, transferring electrons
through the copper and heme centers to reduce O2 on the matrix side of the membrane.

Two electrons, sequentially released from two molecules of reduced cytochrome c together with two protons from
the matrix, combine with one O atom to form one water molecule. Additionally, for each electron transferred from
cytochrome c to oxygen, one proton is transported from the matrix to the intermembrane space, or a total of four
electrons are transferred for each O2 molecule reduced to two H2O molecules.
+ + +
4H 4H 2H
Intermembrane
space

2e– 2e– 2e– Inner mitochondrial


Q Cyt c
membrane
Complex I Complex III Complex IV
Mitochondrial
2e– matrix
+
NAD 1
O2 + 2H
+
H2O
NADH 2

+ +
4H 2H
Intermembrane
space

2e– 2e– 2e– Inner mitochondrial


Q Cyt c
membrane
Complex II Complex III Complex IV
Mitochondrial
2e– matrix
1 +
FADH2 FAD O2 + 2H H2O
2

Figure 2.13 Flow of electrons through the respiratory chain complexes, showing the entry points for reducing equiv-
alents. Q and cyt c are mobile components of the system.
This page intentionally left blank.
Bioenergetics and Metabolism 147

ATP-ADP translocase function is inhibited by a toxic glycoside atractyloside. ATP-ADP exchange is energetically
expensive; transmembrane proton gradient across the inner mitochondrial membrane powers the exchange. A
second membrane transport system is the phosphate translocase, which promotes symport of one H2PO4– and one
H+ into the matrix. At pH 7, Pi is present as both HPO42– and H2PO4– ; the phosphate translocase is specific for H2PO4–.
This transport process is also powered by the transmembrane proton gradient. Thus, the proton-motive force is
responsible for both ATP synthesis and for transporting substrates (ADP and Pi) into and product (ATP) out of the
mitochondria matrix. A complex of the ATP synthase and both translocases (ATP-ADP translocase and phosphate
translocase) is called ATP synthasome.

Phosphate
translocase
+ – +
H
+ –
+ – Matrix –
H2PO4
3–
ADP

ATP-ADP
translocase
4–
ATP

Figure 2.19 The phosphate and ATP/ADP translocase system in the inner mitochondrial membrane.

2.3.13 Shuttle systems


The glycolytic pathway is a primary source of NADH formation. NADH synthesized during the glycolytic process finally
transfers the electrons to electron transport chain. But, NADH cannot cross the inner mitochondrial membrane. So,
two different shuttle systems help in the transfer of electrons from NADH to the electron transport chain.
The malate-aspartate shuttle is the principal mechanism for the movement of NADH from the cytoplasm into the
mitochondrial matrix. The electrons are carried into the mitochondrial matrix in the form of malate. Cytoplasmic
malate dehydrogenase reduces oxaloacetate to malate while oxidizing NADH to NAD+. Malate then enters the
mitochondrial matrix, where the reverse reaction is carried out by mitochondria malate dehydrogenase and the
regeneration of NADH occurs.

NADH
Oxaloacetate
+
NAD

Malate Asp
Matrix

Malate Cytosol Asp

+
NAD
Oxaloacetate
NADH

Figure 2.20 The malate-aspartate shuttle for transporting NADH from cytosol into the mitochondrial matrix. NADH
in the cytosol transfers electrons to oxaloacetate, producing malate. Malate is transported across the inner membrane
by the help of transporter. In the matrix, malate passes electrons to NAD+; the resulting matrix NADH is finally oxi-
dized by the mitochondrial respiratory chain.
148 Bioenergetics and Metabolism
In glycerol 3-phosphate shuttle, electrons from NADH can enter the mitochondrial electron transport chain by
being used to reduce dihydroxyacetone phosphate to glycerol 3-phosphate. Glycerol 3-phosphate is reoxidized by
electron transfer to an FAD prosthetic group in a membrane-bound glycerol 3-phosphate dehydrogenase. FADH2
subsequently transfers electrons to Q to form QH2, which allows these electrons to enter the electron-transport chain.

Glycerol Mitochondrial
3-phosphate glycerol 3-phosphate
dehydrogenase

+
NAD FAD
Cytosolic
glycerol 3-phosphate
dehydrogenase
NADH
FADH2

Dihydroxyacetone
phosphate Cytosol Matrix

Figure 2.21 Glycerol 3-phosphate shuttle. Electrons from NADH can enter the mitochondrial electron transport
chain by being used to reduce dihydroxyacetone phosphate to glycerol 3-phosphate. Glycerol 3-phosphate is reox-
idized by electron transfer to a FAD prosthetic group in a membrane-bound glycerol 3-phosphate dehydrogenase.

Summary of ATP synthesis from the oxidation of one molecule of glucose

Glycolysis (cytoplasm) NADH FADH2 ATP

Glucose  Glucose-6-phosphate –1

Fructose-6-phosphate  Fructose-1,6-bisphosphate –1

2 Glyceraldehyde-3-phosphate  2 Glycerate-1,3-bisphosphate +2

2 Glycerate-1,3-bisphosphate  2 Glycerate-3-phosphate +2

2 Phosphoenolpyruvate  2 Pyruvate +2

Mitochondrial reactions

2 Pyruvate  2 Acetyl-CoA +2

Citric acid cycle

2 Isocitrate  2 -ketoglutarate +2

2 -ketoglutarate  2 Succinyl-CoA +2

2 Succinyl-CoA  2 Succinate +2

2 Succinate  2 Fumarate +2

2 Malate  2 Oxaloacetate +2

10 NADH 2 FADH2 4ATP

Total yield of ATP from the complete oxidation of one molecule of glucose (via glycolysis, pyruvate dehydrogenase
complex reaction, Krebs cycle and oxidative phosphorylation) = 32 (or 30). This is calculated as 2.5 ATP per NADH
and 1.5 ATP per FADH2. The total number is either 30 or 32 depending on the mechanisms used to shuttle NADH
equivalents from the cytosol to the mitochondrial matrix. In prokaryotic organisms, complete oxidation of one
molecule of glucose yields 32 ATP, because no shuttle systems are required for transport of NADH.
This page intentionally left blank.
150 Bioenergetics and Metabolism
Problem

When pure reduced cytochrome c is added to carefully prepared mitochondria along with ADP, Pi, antimycin A and oxygen,
the cytochrome c becomes oxidized and ATP is formed with a P/O ratio (the ratio of ATP synthesized to O atoms reduced)
approaching 1.0.
a. Indicate the probable flow of electrons in this system.
b. Why was antimycin A added?
c. What does this experiment tell you about the location of coupling sites for oxidative phosphorylation?

Solution

a. Cyt c  Cyt a  Cyt a3  O2


b. To block oxidation of endogenous substrates.
c. One site associated with cytochrome oxidase.

2.3.15 Fermentation
Pyruvate, the end product of glycolysis has two fates. In the absence of oxygen, it undergoes fermentation and
anaerobic respiration whereas in the presence of oxygen it enters the aerobic respiration. Anaerobic respiration
is different from fermentation. In anaerobic respiration, final electron acceptor in the electron transport chain is
an inorganic molecule other than O2 like nitrate, sulfate and carbonate whereas in fermentation, the final electron
acceptor is an organic molecule. Fermentation is a self contained process and no outside electron acceptor is
involved. It does not involve an electron transport system. In this section we have discussed only fermentation.

Cytosol

Glucose

Glycolysis

2 Pyruvate
O2 absent O2 present

2 Pyruvate
2 Ethanol or lactate

Fermentation
2 Acetyl-CoA

Krebs
cycle
4CO2

Aerobic respiration

Figure 2.23 Glycolysis is common to fermentation and aerobic respiration. In a facultative anaerobe, which is
capable of both aerobic respiration and fermentation, pyruvate is committed to one of those two pathways, usually
depending on whether or not oxygen is present.

During fermentation, when O2 is not present (or the cell cannot use it) NAD+ is regenerated from NADH by transferring
electrons to organic molecules. In this process, pyruvate is converted into organic molecules like lactate, ethanol.
Fermentations are classified in terms of either the substrate fermented or the fermentation products formed. Some
of the major types of fermentations on the basis of products formed are alcoholic, lactic acid, propionic acid and
butyric acid fermentation.
Bioenergetics and Metabolism 151

Lactic acid fermentation


Muscle cells and certain bacterial species (e.g. Lactobacillus) oxidize NADH by transforming pyruvate into lactate
and the process is known as lactic acid fermentation. Lactic acid fermentation is catalyzed by an enzyme, lactate
dehydrogenase (LDH).
Lactate
Glycolysis dehydrogenase
Glucose 2 Pyruvate 2 Lactate

2 NAD+ 2 NADH 2 NADH 2 NAD+

Figure 2.24 In lactic acid fermentation, the NADH formed in the oxidation of glyceraldehyde 3-phosphate is con-
sumed in the reduction of pyruvate. The regeneration of NAD+ in the reduction of pyruvate to lactate sustains the
continued operation of glycolysis under anaerobic conditions.

In animals, lactate formed in the muscles is recycled to glucose in the liver. Lactate produced in muscle is transported
from the muscle to the liver, where it is reoxidized by liver LDH to pyruvate. By the process of gluconeogenesis,
pyruvate is finally converted into glucose in the liver. Liver again exports glucose to muscle for glycolysis. This
cycle is referred to as the Cori cycle (also known as Lactic acid cycle). The Cori cycle is named for Carl and Gerty
Cori, who received the Nobel Prize in physiology or medicine in 1947 for their studies of glycogen metabolism and
blood glucose regulation.

Glucose Glucose

Glycolysis Gluconeo-
genesis

2 Pyruvate 2 Pyruvate

2 Lactate 2 Lactate

Skeletal muscle Blood Liver

Figure 2.25 The Cori cycle refers to the metabolic pathway in which lactate produced by fermentation in the muscles
moves to the liver and is converted to glucose, which then returns to the muscles and is converted back to lactate.

Alcoholic fermentation
In alcoholic fermentation that occurs in yeast and several bacterial species, pyruvate is converted into ethanol
in a two-step pathway. In yeast, pyruvate is decarboxylated to form acetaldehyde, which is then reduced by
NADH to form ethanol. The non-oxidative decarboxylation of pyruvate to acetaldehyde is catalyzed by pyruvate
decarboxylase and NADH-dependent reduction of acetaldehyde to ethanol is catalyzed by alcohol dehydrogenase.
Pyruvate decarboxylase requires thiamine pyrophosphate as a coenzyme. This coenzyme, derived from vitamin B1,
participates in a number of group transfer reactions involving an activated aldehyde moiety.

Pyruvate Alcohol
Glycolysis decarboxylase dehydrogenase
Glucose 2 Pyruvate 2 Acetaldehyde 2 Ethanol
2 CO2
+ +
2 NAD 2 NADH 2 NADH 2 NAD

Figure 2.26 In alcoholic fermentation, conversion of glucose into ethanol occurs. The NADH generated by the oxi-
dation of glyceraldehyde 3-phosphate is consumed in the reduction of acetaldehyde to ethanol. Thus, there is no net
oxidation-reduction in the conversion of glucose into ethanol.
152 Bioenergetics and Metabolism
Table 2.6 Comparison of aerobic respiration, fermentation and anaerobic respiration

Aerobic respiration Oxygen dependent


Final electron acceptor: Molecular oxygen
Type of phosphorylation used to generate ATP: Substrate-level and oxidative

Fermentation Oxygen independent


Final electron acceptor: An organic substance
Type of phosphorylation used to generate ATP: Substrate-level

Anaerobic respiration Oxygen independent


Final electron acceptor: An inorganic substance such as nitrate, sulfate but not oxygen
Type of phosphorylation used to generate ATP: Substrate-level and oxidative

2.3.16 Pasteur effect


Louis Pasteur observed that when yeast is exposed to aerobic conditions (in the presence of oxygen), their glucose
consumption and ethanol production drop. Whereas in the absence of oxygen glucose consumption increases several
fold. Reason for the decrease in consumption of glucose is that fermentation results in the production of 2 ATPs
per glucose whereas aerobic respiration yields 32 ATPs per glucose. Hence, for the generation of same amount of
ATP to perform essential metabolic activities, more consumption of glucose is needed in anaerobic condition. This
accounts for Pasteur’s observation that yeast consumes more glucose when growing anaerobically than aerobically
(called Pasteur effect).

2.3.17 Warburg effect


Most cancer cells exhibit increased glycolysis and use this metabolic pathway for generation of ATP as a main source
of their energy supply. This phenomenon is known as the Warburg effect. However, the advantage it confers to
cancer cells has been unclear. Biochemical and molecular studies suggest several possible mechanisms by which this
metabolic alteration may evolve during cancer development. These mechanisms include mitochondrial defects and
malfunction, adaptation to hypoxic tumor microenvironment and abnormal expression of enzyme, pyruvate kinase.

Problem

When O2 is added to an anaerobic suspension of cells consuming glucose at a high rate, the rate of glucose consumption
declines greatly as the O2 is used up, and accumulation of lactate ceases. This effect is characteristic of most cells capable
of both aerobic and anaerobic glucose catabolism.
a. Why does the accumulation of lactate cease after O2 is added?
b. Why does the presence of O2 decrease the rate of glucose consumption?

Solution

a. Because NADH is reoxidized via electron transfer instead of lactic acid fermentation.
b. Oxidative phosphorylation is more efficient and release more energy from one mole of glucose.

2.3.18 Respiratory quotient


Respiration involves the oxidation of respiratory substrates such as glucose and fats. The oxidation involves the
release of carbon dioxide along with the release of energy. The organic substances, which are catabolised in the
living cells to release energy are called as respiratory substrates. Though carbohydrate, fat or protein may act as
a respiratory substrate, the common respiratory substrate is carbohydrate.
This page intentionally left blank.
Bioenergetics and Metabolism 157

2.7 Photosynthesis
Photosynthesis is a physiochemical process by which photosynthetic organisms convert light energy into chemical
energy in the form of reducing power (as NADPH) and ATP, and use these chemicals to drive carbon dioxide fixation.

Sun

Light reaction

ATP + NADPH

CO2 Calvin cycle Triose phosphate Glucose

Figure 2.32 Photosynthesis is a two stage process. The first process is a light dependent one (light reactions) that
requires the direct energy of light to make energy carrier molecules that are used in the second process. The Calvin cycle
(light independent process) occurs when the products of the light reaction are used in the formation of carbohydrate.

On the basis of generation of oxygen during photosynthesis, the photosynthetic organisms may be oxygenic or
anoxygenic. Oxygenic photosynthetic organisms include both eukaryotes as well as prokaryotes whereas anoxygenic
photosynthetic organisms include only prokaryotes.

Oxygenic photosynthetic organisms


Eukaryotes – Plants and Photosynthetic protists
Prokaryotes – Cyanobacteria

Anoxygenic photosynthetic organisms


Prokaryotes – Green and purple photosynthetic bacteria

In oxygenic photosynthetic organisms, photosynthetic oxygen generation occurs via the light-dependent oxidation
of water to molecular oxygen. This can be written as the following simplified chemical reaction:

Light
nCO2 + nH2O (CH2O)n + nH2O + nO2
Chl

2.7.1 Photosynthetic pigment


The solar energy required for photosynthesis is captured by photosynthetic pigment molecules. Different types of
pigments, described as photosynthetic pigment, participate in this process. The major photosynthetic pigment is
the chlorophyll.

Chlorophylls
Chlorophyll, a light-absorbing green pigment, contains a polycyclic, planar tetrapyrrole ring structure. Chlorophyll
is a lipid soluble pigment. It has the following important features:
1. The central metal ion in chlorophyll is Mg2+.
2. Chlorophyll has a cyclopentanone ring (ring V) fused to pyrrole ring III.
3. The propionyl group on a ring IV of chlorophyll is esterified to a long-chain tetraisoprenoid alcohol. In chlorophyll
a and b it is phytol.
158 Bioenergetics and Metabolism
R1 R2

3
H3C I II 4 R3
N N

Mg

N N
IV III CH3
H3C
R1 R2 R3
V
CH2 Chlorophyll a CH CH2 CH3 CH2CH3

CH2 O Chlorophyll b CH CH2 CHO CH2CH3


C O
O C O CH3
O

H2C

Phytyl side chain

Figure 2.33 Chlorophyll structures. Chlorophyll is composed of two parts; the first is a porphyrin ring with mag-
nesium at its center, the second is a hydrophobic phytol tail. The tail is a 20 carbon chain that is highly hydrophobic.

Table 2.8 Differences between Chlorophyll a and Chlorophyll b

Chlorophyll a Chlorophyll b

1. It is C55H72O5N4Mg. It is C55H70O6N4Mg.

2. In the pure state, chlorophyll a is blue-green. In the pure state, chlorophyll b is olive-green.

3. It is an essential photosynthetic pigment. It is accessory photosynthetic pigment.

4. Pyrrole ring II contains methyl (—CH3) group. Pyrrole ring II contains aldehyde (—CHO) group.

5. It absorbs more red wavelengths than violet-blue It absorbs more violet-blue wavelength than red
wavelength of light. wavelength of light.

Oxygenic photosynthetic organisms contain different types of chlorophyll molecules like Chl a, Chl b, Chl c and
Chl d. These chlorophyll molecules differ by having different substituent groups on the tetrapyrrole ring. Anoxygenic
photosynthetic organisms contain bacteriochlorophyll molecules. They are related to chlorophyll molecules. Different
groups of anoxygenic photosynthetic organisms contain different types of bacteriochlorophyll: BChl a, BChl b,
BChl c, BChl d and BChl e. Bacteriochlorophyll molecules absorb light at longer wavelengths as compared to
chlorophyll molecules.

Accessory pigments
Besides the major light-absorbing chlorophyll molecules, there are two groups of accessory pigments which absorb
light in the wavelength region, where chlorophylls do not absorb strongly. The two types of accessory pigments
are carotenoids and phycobilins.
Carotenoids are long-chain, conjugated hydrocarbons containing a string of isoprene residues and distinguished
from one another by their end groups. They are generally C40 terpenoid compounds formed by the condensation of
eight isoprene units. Carotenoids are lipid soluble pigments and can be subdivided into two classes, xanthophylls
(which contain oxygen) and carotenes (which are purely hydrocarbons, and contain no oxygen). The characteristic
absorption spectrum (400 to 500 nm) of carotenoids is responsible for the yellow color of leaves in autumn and
the orange color of carrots, for which this group of pigments is named. The upper wavelength limit of absorption
by carotenoids depends on the number of conjugated double bonds, the isomeric configuration and the nature of
the end groups.
Bioenergetics and Metabolism 159

b-carotene (Carotene)
OH

HO
Lutein (Xanthophyll)

Carotenoid acts as an accessory light-harvesting pigment. It captures light energy and feeds it to the photo-
chemical reaction center. The other role of carotenoids is to provide protection against photo-oxidative damage
(photoprotection). In providing photoprotection, it acts as quencher as well as scavenger.
High light intensity may damage many cellular components, especially lipids. The photoprotection mechanism
prevents the damage by removing excess energy. When the energy stored in chlorophylls in the excited state is
rapidly dissipated by excitation transfer or photochemical reaction, the excited state is said to be quenched. This is
called photochemical quenching. If the excited state of chlorophyll is not rapidly quenched by excitation transfer
or photochemical reaction, it can react with molecular oxygen to form an excited state of oxygen known as singlet
oxygen (1O2*).

Singlet excited state


1
Chl*

3
Chl* Triplet state

O2

Chl 1
O*2
Ground state

The extremely reactive singlet oxygen damages many cellular components. The photoproducts can damage the D1
protein of photosystem II. The damage leads to photoinhibition. Carotenoids exert their photoprotective action
by rapidly quenching the excited state of chlorophyll. The excited state of carotenoids does not have sufficient
energy to form singlet oxygen, so it decays back to its ground state while losing its energy as heat. It is called
non-photochemical quenching. Non-photochemical quenching is the quenching of excitation energy by process
other than photochemistry. In this case, chlorophylls in the excited state can return to the ground state by emitting
the energy as heat. Three xanthophylls– violaxanthin, antheraxanthin and zeaxanthin – participate in this process.
The molecular mechanism of non-photochemical quenching is not well understood. If this defense is not sufficient
and toxic photoproducts form, carotenoids eliminate the reactive photoproducts by acting as a scavenger.
Phycobilins are non-cyclic, linear tetrapyrroles structures, which are structurally similar to the bile pigment bilirubin.
Chlorophylls are also composed of four pyrroles, but the pyrroles are arranged in a ring and contain a Mg2+ ion in
the center. The three photosynthetic phycobilins are phycoerythrobilin, phycocyanobilin and allophycocyanobilin.
They are unique among the photosynthetic pigments in that they are covalently bound (thioether bond) to certain
water-soluble proteins to form phycobiliproteins. There are two major types of phycobiliproteins – phycoerythrins
and phycocyanins. Phycoerythrin contains protein and phycoerythrobilin whereas phycocyanin contains protein
and phycocyanobilin. All phycobiliproteins are water soluble and therefore cannot exist within the membrane like
carotenoids, but aggregate into clusters that adhere to the membrane called phycobilisomes. Phycobilins are
present abundantly in red algae and cyanobacteria. They act as accessory light-harvesting pigments, which capture
light energy and feed it to the chlorophyll molecules present at photochemical reaction center.
This page intentionally left blank.
Bioenergetics and Metabolism 197

Glycogen storage diseases


Glycogen storage diseases are caused by a genetic deficiency of one or another of the enzymes of glycogen
metabolism. Many diseases have been characterized that result from an inherited deficiency of the enzyme. These
defects are listed in the table.

Table 2.17 Glycogen storage diseases

Name Enzyme deficiency

Von Gierke’s disease Liver glucose-6-phosphatase

Pompe’s disease Lysosomal 1  4 and 1  6 glucosidase (acid maltase)

Hers’ disease Liver phosphorylase

Tarui’s disease Muscle and erythrocyte phosphofructokinase 1

McArdle’s disease Muscle glycogen phosphorylase

Andersen’s disease Amylo (1,4  1,6) transglycosylase (Branching enzyme)

2.10 Lipid metabolism


2.10.1 Synthesis and storage of triacylglycerols
All animals and plants have the ability to synthesize triacylglycerol (TAG). In animals, many cell types and organs
have the ability to synthesize triacylglycerols, but the liver and intestines are most active. Within all cell types, even
those of the brain, triacylglycerols are stored as cytoplasmic lipid droplets (also termed fat globules, oil bodies, lipid
particles, adiposomes, etc.) enclosed by a monolayer of phospholipids and hydrophobic proteins, such as the perilipins
in adipose tissue or oleosins in seeds. Two main biosynthetic pathways are known, the sn-glycerol-3-phosphate
pathway, which predominates in liver and adipose tissue, and a monoacylglycerol pathway in the intestines. The
most important route to triacylglycerol biosynthesis is the sn-glycerol-3-phosphate or Kennedy pathway.

O O O
CH2 OH CH2 O C R1 CH2 O C R1 CH2 O C R1
O O
1 2 3
CH OH CH OH CH O C R2 CH O C R2
Fatty Fatty
acyl-CoA acyl-CoA Pi
CH2 OP CH2 OP CH2 OP CH2 OH

Glycerol-3-phosphate Lysophosphatidic acid Phosphatidic acid Diacylglycerol

Fatty 4
acyl-CoA
Enzymes
1 Glycerol-3-phosphate acyltransferase O
2 Acylglycerophosphate acyltransferase CH2 O C R1
3 Phosphatidic acid phosphohydrolase O
4 Diacylglycerol acyltransferase CH O C R2
O
CH2 O C R3

Triacylglycerol

Figure 2.72 Triacylglycerol biosynthetic pathway.


198 Bioenergetics and Metabolism
Digestion and transport

Digestion and transport of lipids pose unique problems relating to the insolubility of lipids in water. Enzymes that act
on lipids are soluble proteins or membrane proteins at the aqueous interface. Lipids, and products of their digestion,
must be transported through aqueous compartments within the cell as well as in the blood and tissue spaces.

Release of fatty acids from stored triacylglycerols of adipose cells


Fats (triacylglycerols) are important energy reserve in the organisms. In animals, they are mostly stored in insoluble
form in the cells of adipose tissue—the adipocytes—where they are constantly being synthesized and broken down
again. The hydrolysis of triacylglycerols by lipases is referred to as lipolysis. Triacylglycerols in adipose tissue are
converted into free fatty acids and glycerol under the influence of hormones like epinephrine, norepinephrine,
glucagon. A hormone-sensitive lipase (TAG lipase) initiates the process. The free fatty acids are released into the
blood, where they bind to serum albumin. Albumin is a monomeric protein that comprises about half of the blood
serum proteins.

Glycerol Glycerol Glycerol


Fatty acid

Fatty acid

Fatty acid

Fatty acid

Fatty acid

Fatty acid
TAG lipase DAG lipase MAG lipase
Glycerol
Fatty acid

Fatty acid

Fatty acid
Figure 2.73 Liberation of fatty acids from triacylglycerols in adipose tissue.

Release of fatty acids from dietary triacylglycerols


Most dietary triacylglycerols are digested by pancreatic lipases in the intestinal lumen. Although the process actually
begins in the stomach with lingual lipase released by the serous gland and gastric lipase produced by the chief cells.
Pancreatic lipase cleaves fatty acids from C-1 and the C-3 position of triacylglycerol. Resulting monoacylglycerols
with fatty acids at C-2 are hydrolyzed by intestinal lipases. Before hydrolytic activity of the lipases, emulsification
of triacylglycerol occurs. In the intestinal lumen bile salts act on triacylglycerols. Bile salts are detergent substances
synthesized in the liver and stored in the gallbladder. A bile salt molecule is made up of a bile acid, such as cholic
acid, and an associated cation. The bile salt molecule has both hydrophobic and hydrophilic surfaces. It acts as
the detergent and emulsifies lipids and yield micelles. Water soluble digestive enzymes digest it and facilitates
the absorption of lipid through intestinal mucosal cells. Bile salts not only facilitate the lipid digestion but are also
essential for the absorption of lipid digestion products. The products of fat digestion comprise a mixture of glycerol,
free fatty acids, monoacylglycerols, and diacylglycerols. Less than 10% of the original triacylglycerol also remains
unhydrolyzed. During absorption through intestinal mucosal cells, resynthesis of triacylglycerols occurs from the
hydrolysis products. This resynthesis occurs in the endoplasmic reticulum and the Golgi complex of mucosal cells.
Triacylglycerol packaged with cholesterol (dietary) and apolipoproteins (ApoC-II) into lipoprotein aggregates called
chylomicrons. Lipoproteins are spherical particles composed of a central core of non-polar lipids (cholesterol
esters and triacylglycerol) and a surface monolayer made up of phospholipids, free cholesterol and apolipoproteins.
Lipoproteins are generally classified according to their density as chylomicron, VLDL (very-low density lipoprotein),
IDL (intermediate density lipoprotein), LDL (low density lipoprotein) and HDL (high density lipoprotein).
Chylomicrons move through the lymphatic and blood vascular system to the tissues. Lipoprotein lipase in capillary
converts triacylglycerols components of chylomicrons to fatty acids and glycerol. Finally, fatty acids enter cells
where they are oxidized as fuel or reesterified for storage.
This page intentionally left blank.
216 Bioenergetics and Metabolism
Serine family : The members of the serine family — serine, glycine, and cysteine—derive their carbon
skeletons from glycerate-3-phosphate.
Aspartate family : The aspartate family includes aspartate, asparagine, lysine, methionine, and threonine and
derive their carbon skeletons from oxaloacetate.
Pyruvate family : The pyruvate family consists of alanine, valine, leucine, and isoleucine and derive their carbon
skeletons from pyruvate.
Aromatic family : The members of the aromatic family — phenylalanine, tyrosine, and tryptophan — derive
their carbon skeletons from phosphoenol pyruvate and erythrose 4-phosphate.
Histidine : The member of histidine family–histidine–derive their carbon skeletons from ribose 5-phosphate.

2.11.2 Amino acid catabolism


The catabolism of the amino acids usually begins by removing the amino group. Amino groups can then be disposed
of in urea synthesis. The carbon skeletons produced from the standard amino acids are then degraded to TCA
intermediates or their precursors so that they can be metabolized to CO2 and H2O or used in gluconeogenesis. So,
catabolic pathway of amino acids involves three common stages:
1. Removal of -amino group from amino acids (amino acid deamination) and conversion of amino group to
ammonia.
2. Incorporation of ammonia into urea.
3. Conversion of amino acid’s carbon skeletons to common metabolic intermediate.

Protein

Free amino acids

Deamination

+
a-Keto acid NH4

Citric acid cycle

CO2 Biosynthesis
+ +
H2O Urea cycle & excretion
+
ATP

Figure 2.90 General pathway showing the stages of amino acid catabolism.

Amino acid deamination

The removal of the -amino group from amino acids involves two types of biochemical reactions: transamination
and oxidative deamination.

Transamination

The dominant reactions involved in removing amino acid nitrogen are known as transaminations. This class of reactions
funnels nitrogen from all free amino acids into a small number of compounds; then, either they are oxidatively
Bioenergetics and Metabolism 217

deaminated, producing ammonia, or their amine groups are converted to urea by the urea cycle. Transaminations
involve moving an -amino group from a donor -amino acid to the keto carbon of an acceptor -keto acid. As a
result of transfer, -keto derivatives of amino acid and corresponding amino acid forms.
All amino acids except lysine, threonine and proline participate in transamination during catabolism. Transamination
is readily reversible. This reaction is catalyzed by the enzyme called aminotransferases (also called transaminase).
Each aminotransferase is specific for one or at most a few amino group donors. Aminotransferases are named
after the specific amino group donor, because the acceptor of the amino group is almost always -ketoglutarate.
+
NH3 O
— —
R1 CH C O R1 C C O a-keto acid

O O

+
O NH3
— —
R2 C C O R2 CH C O a-amino acid

O O

Figure 2.91 Transamination involves the transfer of amino group to an -keto acid to yield the -keto acid of the
original amino acid and a new amino acid.

The most common compounds involved as a donor/acceptor pair in transamination reactions are glutamate and
-ketoglutarate, which participate in reactions with many different aminotransferases.
H O O H
— — — — — —
CH3 C COO + OOC CH2 CH2 C COO CH3 C COO + OOC CH2 CH2 C COO
+ +
NH3 NH3

Amino acid a-Ketoglutarate a-Keto acid Glutamate

All the amino nitrogen from amino acids that undergo transamination can be concentrated in glutamate. This is
important because L-glutamate is the only amino acid that undergoes oxidative deamination at an appreciable rate.

Oxidative deamination

Transamination does not result in any net deamination. During oxidative deamination, an amino acid is converted
into the corresponding keto acid by the removal of the amine functional group as ammonia and the amine functional
group is replaced by the ketone group. The ammonia eventually goes into the urea cycle.
Deamination occurs mainly through the oxidative deamination of glutamate by glutamate dehydrogenase. The
reaction requires an oxidizing agent NAD+ or NADP+. Glutamate dehydrogenase is allosterically inhibited by GTP
and NADH and activated by ADP and NAD+.

H +
O
NAD(P)
— — — — +
OOC CH2 CH2 C COO OOC CH2 CH2 C COO + NH4
+
NH3

Glutamate a-Ketoglutarate

Urea cycle
Living organisms excrete the excess nitrogen resulting from the metabolic breakdown of amino acids in one of
three ways. Many aquatic animals simply excrete ammonia. Where water is less plentiful, however, processes have
evolved that convert ammonia to less toxic waste products that therefore require less water for excretion. One
such product is urea and other is uric acid. Accordingly, living organisms are classified as being either ammonotelic
(ammonia excreting), ureotelic (urea excreting) or uricotelic (uric acid excreting).
This page intentionally left blank.
Chapter 03

Cell Structure and Functions

3.1 What is a Cell?


The basic structural and functional unit of cellular organisms is the cell. It is an aqueous compartment bound by cell
membrane, which is capable of independent existence and performing the essential functions of life. All organisms,
more complex than viruses, consist of cells. Viruses are noncellular organisms because they lack cell or cell-like
structure. In the year 1665, Robert Hooke first discovered cells in a piece of cork and also coined the word cell. The
word cell is derived from the Latin word cellula, which means small compartment. Hooke published his findings in
his famous work, Micrographia. Actually, Hooke only observed cell walls because cork cells are dead and without
cytoplasmic contents. Anton van Leeuwenhoek was the first person who observed living cells under a microscope
and named them animalcules, meaning little animals.
On the basis of the internal architecture, all cells can be subdivided into two major classes, prokaryotic cells and
eukaryotic cells. Cells that have unit membrane bound nuclei are called eukaryotic, whereas cells that lack a
membrane bound nucleus are prokaryotic. Eukaryotic cells have a much more complex intracellular organization
with internal membranes as compared to prokaryotic cells. Besides the nucleus, the eukaryotic cells have other
membrane bound organelles (little organs) like the endoplasmic reticulum, Golgi complex, lysosomes, mitochondria,
microbodies and vacuoles. The region of the cell lying between the plasma membrane and the nucleus is the
cytoplasm, comprising the cytosol (or cytoplasmic matrix) and the organelles. The prokaryotic cells lack such unit
membrane bound organelles.

Cell theory
In 1839, Schleiden, a German botanist, and Schwann, a British zoologist, led to the development of the cell theory
or cell doctrine. According to this theory all living things are made up of cells and cell is the basic structural and
functional unit of life. In 1855, Rudolf Virchow proposed an important extension of cell theory that all living cells
arise from pre-existing cells (omnis cellula e cellula). The cell theory holds true for all cellular organisms. Non-
cellular organisms such as virus do not obey cell theory. Over the time, the theory has continued to evolve. The
modern cell theory includes the following components:
• All cellular organisms are made up of one or more cells.
• The cell is the structural and functional unit of life.
• All cells arise from pre-existing cells by division.
• Energy flow occurs within cells.
• Cells contain hereditary information (DNA) which is passed from cell to cell.
• All cells have basically the same chemical composition.

Evolution of the cell


The earliest cells probably arose about 3.5 billion years ago in the rich mixture of organic compounds, the primordial
soup, of prebiotic times; they were almost certainly chemoheterotrophs. Primitive heterotrophs gradually acquired the
236 Cell Structure and Functions
capability to derive energy from certain compounds in their environment and to use that energy to synthesize more
and more of their own precursor molecules, thereby becoming less dependent on outside sources of these molecules-
less extremely heterotrophic. A very significant evolutionary event was the development of photosynthetic ability
to fix CO2 into more complex organic compounds. The original electron (hydrogen) donor for these photosynthetic
organisms was probably H2S, yielding elemental sulfur as the byproduct, but at some point, cells developed the
enzymatic capacity to use H2O as the electron donor in photosynthetic reactions, producing O2. The cyanobacteria
are the modern descendants of these early photosynthetic O2 producers.
One important landmark along this evolutionary road occurred when there was a transition from small cells with
relatively simple internal structures - the so-called prokaryotic cells, which include various types of bacteria - to a
flourishing of larger and radically more complex eukaryotic cells such as are found in higher animals and plants. The
fossil record shows that earliest eukaryotic cells evolved about 1.5 billion years ago. Details of the evolutionary path
from prokaryotes to eukaryotes cannot be deduced from the fossil record alone, but morphological and biochemical
comparison of modern organisms has suggested a reasonable sequence of events consistent with the fossil evidence.
Three major changes must have occurred as prokaryotes gave rise to eukaryotes. First, as cells acquired more DNA,
mechanisms evolved to fold it compactly into discrete complexes with specific proteins and to divide it equally between
daughter cells at cell division. These DNA-protein complexes called chromosomes become especially compact at the
time of cell division. Second, as cells became larger and intracellular membrane organelles developed. Eukaryotic
cells have a nucleus which contains most of the cell’s DNA, enclosed by a double layer of membrane. The DNA is,
thereby, kept in a compartment separate from the rest of the contents of the cell, the cytoplasm, where most of
the cell’s metabolic reactions occur.
Finally, primitive eukaryotic cells, which were incapable of photosynthesis or of aerobic metabolism, pooled their
assets with those of aerobic bacteria or photosynthetic bacteria to form symbiotic associations that became
permanent. Some aerobic bacteria evolved into the mitochondria of modern eukaryotes, and some photosynthetic
cyanobacteria became the chloroplasts of modern plant cells.

3.2 Structure of eukaryotic cells


3.2.1 Plasma membrane
Plasma membrane is a dynamic, fluid structure and forms the external boundary of cells. It acts as a selectively
permeable membrane and regulates the molecular traffic across the boundary. The plasma membrane exhibits
selective permeability; that is, it allows some solutes to cross it more easily than others. Different models were
proposed to explain the structure and composition of plasma membranes. In 1972, Jonathan Singer and Garth
Nicolson proposed fluid-mosaic model, which is now the most accepted model. In this model, membranes are viewed
as quasi-fluid structures in which proteins are inserted into lipid bilayers. It describes both the mosaic arrangement
of proteins embedded throughout the lipid bilayer as well as the fluid movement of lipids and proteins alike.

Peripheral protein

Phospholipid
bilayer

Integral
protein Peripheral
protein

Figure 3.1 Fluid mosaic model for membrane structure. The fatty acyl chains in the lipid bilayer form a fluid, hydro-
phobic region. Integral proteins float in this lipid bilayer. Both proteins and lipids are free to move laterally in the plane
of the bilayer, but movement of either from one face of the bilayer to the other is restricted.
This page intentionally left blank.
Cell Structure and Functions 241

Transmembrane proteins may be single-pass (monotopic) or multipass (polytopic). Glycophorin is a major single
pass transmembrane protein of RBC. It is a small glycoprotein of 131 amino acid residues. It was the first membrane
protein for which the complete amino acid sequence was determined. Despite their abundance in the plasma
membrane of RBC, their function remains unknown.

Peripheral protein

Lipid bilayer

Lipid
Transmembrane linked protein
protein Peripheral
protein

Figure 3.5 Schematic outline of typical transmembrane protein, peripheral protein and lipid linked protein in a lipid
bilayer.

Another prominent protein present on the plasma membrane of RBC is band 3 protein or chloride-bicarbonate
exchanger, a 95 kDa multipass membrane protein. It is responsible for mediating the electroneutral exchange of
chloride for bicarbonate. Transmembrane proteins are characterized by having transmembrane α-helices. They
contain a stretch of 21 to 26 hydrophobic amino acid residues coiled into an α-helix that is believed to facilitate
the spanning of a membrane bilayer. In a few membrane proteins, the transmembrane portion comprises β-barrel
made up of antiparallel β-strands. The number of β-strands varies widely (8 to 22 strands). One common example
is porin, a pore-forming protein, which allows hydrophilic solutes (up to 600 daltons) to diffuse. The porin β-barrel
is formed from a 16 stranded antiparallel β-sheet. The β-barrel is stabilized by extensive hydrogen bonding between
the β-strands. The interior of the barrel is hydrophilic.
Membrane proteins which are covalently bound to lipid molecules are called lipid-linked (or lipid anchored) membrane
proteins. They form covalent attachments with three classes of lipids: isoprenoid groups such as farnesyl and
geranylgeranyl residues; fatty acyl groups such as myristoyl and palmitoyl residues; and glycosylated phospholipid
(such as glycosylphosphatidylinositol, GPI). An isoprenoid group that contains 15 (farnesyl) or 20 (geranylgeranyl)
carbon atoms attaches to a cysteine residue at the C-terminus via a thioether linkage.
A myristic acid (14-carbon, saturated fatty acid) molecule attaches to a protein through an amide linkage to the
alpha-amino group of an N-terminal Gly residue. A palmitic acid (16-carbon, saturated fatty acid) molecule attaches
to a Cys residue close to the N- or C-terminus via a thioester linkage. A glycosylphosphatidylinositol molecule (GPI)
attaches to the C-terminal amino acid via an amide linkage. GPI-linked proteins occur on the exoplasmic face of
the plasma membrane.

Table 3.2 Characteristics of lipid-linked proteins

Lipid anchor Attachment site Subcellular location

Isoprenyl (Farnesyl/Geranylgeranyl) Cysteine residue at C-terminus Intracellular

Myristoyl Glycine residue at N-terminus Intracellular

Palmitoyl Cysteine residue near N- or C-terminus Intracellular

GPI-linked proteins Various residues at C-terminus Cell surface


242 Cell Structure and Functions

Prenylated
Fatty acylated
protein
protein
Fatty acylated
protein Cys

OOC Cys

ly CH2
G CH2

HN Amide S Thioester S Thioether


linkage linkage linkage
C O C O CH2
Cytosolic

Lipid bilayer

Noncytosolic
Core tetrasaccharide

Phosphoethanolamine
Amide linkage

GPI linked protein

Figure 3.6 Lipid-linked membrane proteins are attached to the bilayer either through a fatty acid chain or prenyl
group in the cytoplasmic monolayer or, less often, via an oligosaccharide, to a minor phospholipid, phosphatidyli-
nositol, in the noncytoplasmic monolayer. A fatty acid chain, myristic acid, is attached via an amide linkage to an
amino-terminal glycine. The palmitic acid is joined to a protein in thioester linkage to a specific Cys residue. A prenyl
group (either farnesyl or a longer geranylgeranyl group) is attached via a thioether linkage to a cysteine residue at
carboxyl terminus. GPI anchors are derivatives of phosphatidylinositol in which the inositol bears a short oligosaccha-
ride covalently joined to phosphoethanolamine.

Hydropathy plot
Transmembrane proteins, like glycophorin, contain membrane spanning region(s). An α-helical sequence of 20 to 25
residues span the thickness (30 Å) of the lipid bilayer. Typically most of the residues in membrane spanning region
are nonpolar and almost none of them are charged. This information can be used to identify putative membrane-
spanning regions in a transmembrane protein. Once the amino acid sequence of a transmembrane protein is known,
the number and positions of transmembrane segments can be inferred from a hydropathy (or hydrophobicity) plot.
It is a method to determine the hydrophobic and hydrophilic regions of a protein sequence.
The relative polarity of each amino acid has been determined experimentally by measuring the free-energy change
accompanying the movement of that amino acid side chain from a hydrophobic solvent into water. This free energy
change is expressed as a hydropathy index. A positive value indicates that free energy is required for transfer
to water (i.e. the segment is hydrophobic) and the value assigned is an index of the amount of energy needed.
The overall hydropathy index of a sequence of amino acids is estimated by summing the free energies of transfer
for the residues in the sequence. To scan a polypeptide sequence for potential membrane-spanning segments, an
investigator calculates the hydropathy index for successive segments of a given size, usually around 10-20 amino
acids. The span of 10 to 20 residues chosen for this calculation is called a window. We can take the amino acid
sequence of a protein and estimate the free-energy change that takes place when a hypothetical a-helix formed
of residues 1 through 20 is transferred from the membrane interior to water. The same calculation can be made
for residues 2 through 21, 3 through 22 and so forth, until we reach the end of the sequence. The free-energy
change for each window is plotted against the first amino acid residue of every window to create a hydropathy
plot. A region with more than 20 residues of high hydropathy index is presumed to be a transmembrane segment.
This page intentionally left blank.
244 Cell Structure and Functions
S S1 S2
S1

S2
Uniport Symport Antiport

Cotransport

Figure 3.8 Three types of carrier mediated transport involving carrier proteins functioning as uniporters, symporters
and antiporters.

Channel proteins transport solutes down their concentration gradients. Channels may be gated or nongated. Gated
channels open only in response to specific signals. The main types of signals that cause ion channels to open are
a change in the voltage across the membrane (voltage-gated channels), a mechanical stress (mechanically gated
channels), or the binding of a ligand (ligand-gated channels). The ligand can be either an extracellular mediator or
an intracellular mediator, such as an ion (ion-gated channels), or a nucleotide (nucleotide gated channels).
Carrier mediated transport may be active or passive, but channel mediated transport is always passive and transport
through channel proteins occurs at a much faster rate than mediated by carrier proteins.

Cell coat or glycocalyx


Carbohydrates are present on the surface of all eukaryotic cells. These carbohydrates as oligosaccharides occur
both as glycoproteins and glycolipids. Plasma membranes also contain integral proteoglycan molecules with surface-
exposed polysaccharide chains. The term cell coat, or glycocalyx is used to describe the carbohydrate coating on
the cell surface. Cell coat helps to protect the cell surface from mechanical and chemical damage and also mediate
specific, transient, cell-cell adhesion events.

3.2.2 ABO blood group


The plasma membrane of RBCs contain many antigens. Based on the presence or absence of various antigens,
blood is categorized into different blood groups. There are at least 24 blood groups. The ABO blood group (A,
B, AB and O) is based on presence of ABO antigens. The ABO antigens are complex oligosaccharides present in
form of glycoproteins and glycolipids. In the formation of A and B antigens, H molecule (coded by H gene) acts
as a precursor. H molecule (antigen) is found in every individual. In case of A antigen, H contains an additional
N-acetylgalactosamine (GalNAc). Similarly, in case of B antigen an additional galactose (Gal) is linked to H molecule.
The addition of the terminal GalNAc to the H molecule is catalyzed by GalNAc transferase, a product of A gene.
Similarly, the product of the B gene is the Gal transferase adding the Gal residue to the H molecule. Individuals
of type AB possess both enzymes and thus have two A and B antigens and individuals of type O contain only H
molecule. The RBCs of people with type A blood group contain A and H antigens, those with type B blood group
contain B and H antigens, those with type AB blood group have both A, B and H antigens and those with type O
blood groups have only H antigen (do not have any A or B antigens).

A antigen
Antigens present
RBC RBC RBC RBC B antigen
on RBC
H antigen
A A A B B B A B
Genotype I I |I i I I |I i I I ii
Blood group A B AB O

The IA and IB alleles are responsible for the production of A and B antigens. The allele IA for the A antigen is
codominant with the allele IB for the B-antigen. Both IA and IB are completely dominant to the allele i, which fails
to specify A or B antigens. The hierarchy of dominance relationship is symbolized as (IA=IB) > i.
This page intentionally left blank.
Cell Structure and Functions 247

through the lipid bilayer or through the transport proteins. The characteristics of lipid-phase diffusion and protein
mediated transport are basically different. Transport across the plasma membrane is of two types:

1. Passive transport

Passive transport occurs along the concentration gradient and without the use of metabolic energy. If a transported
substance carries a net charge, its movement is influenced by both its concentration gradient and the membrane
potential, the electric potential across the membrane. The combination of these two forces, called the electrochemical
gradient, determines the direction of transport of a charged molecule across a membrane. Thus, an ion diffuses
not simply down its concentration gradient but, more exactly, down its electrochemical gradient. Passive transport
may be-

a. Simple diffusion: During simple diffusion, a molecule simply dissolves in the phospholipid bilayer, diffuses
across it. No membrane proteins are involved and the direction of transport is determined simply by the rela-
tive concentrations of the molecule inside and outside of the cell. The relative diffusion rate of any substance
across a pure phospholipid bilayer is proportional to its concentration gradient across the layer and to its
hydrophobicity and size. The hydrophobicity of a substance is measured by its partition coefficient. It is the
equilibrium constant for partition of the molecule between oil and water. The higher the substance partition
coefficient, the more lipid soluble it is. Movement of solutes by diffusion is always from a higher to a lower
concentration, and the rate is described by Fick’s Law of Diffusion.

æ Dc ö
J = -D ç ÷
è Dx ø

Where J is the flux per unit area, D is the diffusion coefficient (usually expressed as cm2/sec), and Δc is the
difference in concentration between two regions separated by a distance Δx (membrane thickness in case of
membrane transport). The negative sign accounts for the fact that diffusion is towards the lower concentration.
Simple diffusion is a nonselective process by which any molecule able to dissolve in the phospholipid bilayer is
able to cross the plasma membrane and equilibrate between inside and outside of the cell. The rate of diffusion
of the molecule will be proportional to its hydrophobicity. Gases (such as O2 and CO2), hydrophobic molecules
(such as benzene), and small polar but uncharged molecules (such as H2O and ethanol) are able to diffuse
across the plasma membrane.

b. Facilitated diffusion: Like simple diffusion, facilitated diffusion involves the movement of solutes along the
concentrations gradient. However, the passage is mediated by transport protein (carriers and channels) and
is selective in nature. Facilitated diffusion may be carrier proteins or channel proteins mediated. The rate of
transport of the molecule across the membrane is far greater in facilitated diffusion as compared to simple
diffusion. Facilitated diffusion allows polar and charged molecules, such as carbohydrates, amino acids, nucle-
osides and ions to cross the plasma membrane.

Vmax

Approaches maximum
Rate of transport

rate when all facilitators


are occupied

Facilitated
diffusion
Simple diffusion

0
Concentration difference
across membrane

Figure 3.11 Facilitated and simple diffusion.


248 Cell Structure and Functions
Classes of proteins that mediate facilitated diffusion:

Carrier proteins mediated


Carrier proteins (called transporters or permeases) non-covalently bind specific molecules to be transported on
one side of the membrane. They then undergo conformational changes that allow the molecule to pass through the
membrane and be released on the other side. A classic example is the movement of glucose mediated by carrier
protein, glucose transporter (GLUT). There are various isoforms of the glucose transporter (GLUT) family. These
carriers are composed of ~500 amino acids and possess 12 transmembrane segments. Individual members differ
in their kinetic properties, tissue distribution and regulation. The GLUT-1 (erythrocyte), GLUT-2 (liver), GLUT-3
(brain) and GLUT-5 (small intestine) isoforms are constitutively expressed on the cell surface, whereas GLUT-4, the
predominant isoform of skeletal muscle and adipose tissue, is stored in intracellular vesicles, which upon insulin
stimulation, fuses with the plasma membrane, thereby increasing the rate of sugar uptake by several-fold.
The rate of transport (V) of glucose through GLUT can be well described by the following equation that is analogous
to Michaelis-Menten equation:

Vmax
V=
æ Km ö
ç1 + C ÷
è ø

Where Vmax is the maximal transport velocity, Km is the substrate (glucose) concentration at which the half-maximal
transport rate is attained and C is the concentration of the substrate. This relationship is formally equivalent with the
Michaelis-Menten equation, which relates the velocity of enzyme catalyzed reactions to the substrate concentration.
The Km values can be viewed as the affinity of the isoforms for glucose.
The chloride-bicarbonate exchanger (also called anion exchange protein) present on plasma membrane of RBC
mediates the transport of two anions simultaneously. HCO3– ion moves in one direction and Cl– moves in the opposite
direction. The exchange is electroneutral because of no net transfer of charge (electrical character of ion transport
may be electroneutral i.e. electrically silent either by symport of the oppositely charged ions or antiport of similarly
charged ions or electrogenic i.e. result in charge separation across the membrane).

H+
Carbonic
+
– –
CO2 + H2O HCO3 Cl
Anhydrase

Chloride-bicarbonate
exchange protein
– –
HCO3 Cl

Figure 3.12 The anion exchange protein (chloride-bicarbonate exchange protein) of the RBC membrane allows the
exit of HCO3–. For each HCO3– ion that moves in one direction, one Cl– ion moves in the opposite direction. The result
of this paired movement of two monovalent anions is no net change in the charge or electrical potential across the
RBC membrane.

Channel proteins mediated


Channel proteins form open pores through the membrane, allowing the free diffusion of any molecule of the
appropriate size and charge. Rate of diffusion mediated by channel protein is higher than those mediated by
carriers. Channels typically show less stereospecificity than carriers and are usually non-saturable. Channel proteins
concerned specifically with inorganic ions transport are called ion channels. Ion channels are highly selective. Most
of the ion channels are not permanently open. Some channels (called voltage gated channels) open in response
to change in electric potential; others (called ligand gated channels) open in response to the binding of ligands.
Cell Structure and Functions 249

Many animal and plant cells contain specialized water channel in their plasma membrane to facilitate the water
flow called aquaporins. These are a family of transport proteins that allow water and a few other small uncharged
molecules, such as glycerol, to cross membrane. Aquaporins (AQP) assemble as homotetramers in which each
monomer, consists of six membrane-spanning α-helical domains with cytoplasmically oriented amino and carboxy
termini. Each monomer functions as an independent pore. Two hydrophobic loops contain conserved, asparagine–
proline–alanine (NPA) motif for selectivity of the channel.
The water permeability of epithelial cells lining the renal collecting duct in the kidney is due to the presence of an
aquaporin (AQP-2). Antidiuretic hormone (ADH) regulates the retention of water by mobilizing AQP-2 molecules
stored in vesicle membranes within the epithelial cells, much as insulin mobilizes GLUT4 in muscle and adipose
tissue. When the vesicles fuse with the epithelial cell plasma membrane, water permeability greatly increases and
more water is reabsorbed from the collecting duct and returned to the blood. When the ADH level drops, AQP-2 is
resequestered within vesicles, reducing water retention.
The properties of ion channels can be studied by means of the patch clamp technique. In this technique, a glass
pipette with a very small opening is used to make tight contact with a tiny area or patch of membrane. After the
application of a small amount of suction to the back of the pipette, the seal between pipette and membrane becomes
so tight that no ions can flow between the pipette and the membrane. Thus, all the ions that flow when a single ion
channel opens must flow into the pipette. The resulting electrical current, though small, can be measured with an
ultrasensitive electronic amplifier connected to the pipette. This technique has provided lots of valuable information
about ion channels and its inventors, Erwin Neher and Bert Sakmann, were awarded the Nobel Prize in 1991.

Ionophores
Ionophores are small hydrophobic lipid-soluble molecules that dissolve in lipid bilayers and increase their permeability
to specific inorganic ions. Ionophores shield the charge of the ion to be transported, enabling it to penetrate the
hydrophobic interior of the lipid bilayer. There are two classes of ionophores – mobile ion carriers (which move
within in the bilayer) and channel formers (which span the lipid bilayer).
Valinomycin (isolated from Streptomyces fulvissimus) is an example of a mobile ion carrier. It transports K+ down
its electrochemical gradient. Similarly, ionophore momensin acts as a carrier for Na+ ions. Gramicidin A (produced
by the bacterium Bacillus brevis) is a 15-residue peptide with alternating D- and L-amino acids. In membranes it
forms a channel. The channel permits the passage of water and univalent cations, but not anions.

2. Active transport

Active transport occurs against the concentration gradient and is mediated by carrier proteins. Metabolic energy is
used to move ions or molecules against a concentration gradient. Active transport results in the accumulation of
solute on one side of membrane. Active transport is different from carrier proteins mediated facilitated diffusion.
Comparison of facilitated and active transport is given in the following table.

Table 3.3 Comparison of facilitated diffusion and active transport

Facilitated diffusion Active transport

Selective Selective

Passive Active

Occurs along the concentration gradient Occurs against the concentration gradient

Transport protein involved Transport protein involved

Saturable Saturable

Entropy increases Entropy decreases


This page intentionally left blank.
Cell Structure and Functions 263

Problem

When a neurotoxin is placed in the solution bathing an isolated neuron, it affects the action potential of the neuron as
shown in the figure below. What is the probable mechanism of action of this drug on this neuron?
80

Membrane potential (mV)


+Drug
40

0
Neuron
depolarized
–40 –Drug

–80 Time

Solution

One possibility is that the drug maintains the voltage-gated Na+ channels in an open position, although the drug is not
capable of opening voltage-gated channels by itself. Another possibility is that it prevents opening of the voltage-gated
K+ channels that are responsible for the quick return to the resting potential.

3.4 Transport of macromolecules across plasma membrane


The plasma membrane is a dynamic structure that functions to segregate the chemically distinct intracellular milieu
(the cytoplasm) from the extracellular environment by regulating and coordinating the entry and exit of small
and large molecules. Essential small molecules, such as amino acids, sugars and ions, can traverse the plasma
membrane through the action of integral membrane protein pumps or channels. Macromolecules must be carried
into the cell in membrane bound vesicles derived by the invagination and pinching-off of pieces of the plasma
membrane in a process termed endocytosis.

3.4.1 Endocytosis
The term endocytosis was coined by Christian de Duve in the year 1963. Endocytosis is a process whereby eukaryotic
cells internalize material from their surrounding environment. Internalization is achieved by the formation of
membrane-bound vesicles at the cell surface that arise by progressive invagination of the plasma membrane,
followed by pinching off and release of free vesicles into the cytoplasm.
Classically, endocytosis has been divided into phagocytosis (cellular eating) and pinocytosis (cellular drinking).
Phagocytosis or cell eating (first reported by Metchnikoff) describes the internalization of large particles following
particle binding to specific plasma membrane receptors and by the formation of large endocytic vesicles (generally
>250 nm in diameter) called phagosomes. The phagosomes fuse with lysosomes to form phagolysosomes. In
protozoa, phagocytosis is a form of feeding: large food particles taken up into phagosomes end up in lysosomes.
In multicellular eukaryotes, few specialized cells – so called professional phagocytes perform phagocytosis for non-
nutritive purposes. In mammals, two classes of white blood cells act as professional phagocytes—macrophage and
neutrophils. Phagocytosis is an active, actin mediated and highly regulated process involving specific cell-surface
receptors and signalling cascades mediated by Rho-family GTPases.
Pinocytosis or cell drinking (also termed as fluid-phase endocytosis) involves the ingestion of fluid by the formation
of small endocytic vesicles (termed pinocytic vesicles) of about 100 nm in diameter. Virtually all eukaryotic cells
perform pinocytosis. Uptake of soluble material dissolved in extracellular fluid during pinocytosis occurs both
selectively as well as non-selectively. Selective and efficient uptake occurs when solutes are captured by specific
high-affinity receptors (receptor mediated endocytosis). In receptor-mediated endocytosis, a specific receptor on the
cell surface binds tightly to the extracellular macromolecule (the ligand) that it recognizes. The plasma membrane
264 Cell Structure and Functions
region containing the receptor-ligand complex then undergoes endocytosis, becoming a transport vesicle. Receptor
ligand complexes are selectively incorporated into the intracellular transport vesicles. Pinocytosis occurs in all cells
by at least four basic mechanisms: macropinocytosis, clathrin-mediated endocytosis, caveolae-mediated endocytosis
and clathrin- and caveolae independent endocytosis.

Phagocytosis Macropinocytosis
(>1µm) Caveolin

Clathrin-mediated Caveolae-mediated Clathrin- and


endocytosis endocytosis caveolae-independent
(~120 nm) (~80 nm) endocytosis (~90 nm)

Figure 3.23 The endocytic pathways differ with regard to the size of the endocytic vesicle, the nature of the cargo
(ligands, receptors and lipids) and the mechanism of vesicle formation.

Macropinocytosis
Macropinocytosis involves the membrane ruffling that is induced in many cell types upon stimulation by growth
factors or other signals. Like phagocytosis, the signalling cascades that induce macropinocytosis involve Rho-family
GTPases, which trigger the actin-driven formation of membrane protrusions. However, unlike phagocytosis, these
protrusions do not ‘zipper up’ along a ligand coated particle, but they collapse onto and fuse with the plasma
membrane to generate large endocytic vesicles, called macropinosomes.

Clathrin mediated endocytosis


Clathrin mediated endocytosis was previously referred to as receptor mediated endocytosis, but it is now clear that
this is a misnomer, because most pinocytic pathways involve specific receptor and ligand interactions. In case of
clathrin mediated endocytosis, macromolecules bind to cell-surface receptors, accumulate in clathrin coated pits,
and enter the cell as a receptor and ligand complexes in clathrin-coated vesicles. Each clathrin consists of three
copies each of heavy chain and light chain, forming a three-legged structure called a triskelion. Clathrin triskelions
are the assembly units of the polygonal lattice and assemble into a basket like convex framework of hexagons
and pentagons to form coat. Clathrin assembles into coats on the cytoplasmic side of the plasma membrane by
interacting with its adaptor proteins. There are four structurally related adaptor protein complexes (AP1, AP2,
AP3 and AP4). Each of these four complexes are localized to different intracellular compartments and vary in their
receptor specificity.

Terminal globular
domain

Clathrin light chains

Vertex

Clathrin heavy chains

Figure 3.24 Structure of clathrin triskelion. Each triskelion consists of three clathrin heavy chains radiating from a
central vertex, with a terminal globular domain at the tip of each triskelion leg and a clathrin light chain bound to the
inner half of each leg.
Cell Structure and Functions 265

A common example of clathrin-mediated endocytosis is the uptake of cholesterol-containing particles called low-
density lipoprotein (LDL). LDL is a sphere of 20–25 nm in diameter. It has an outer phospholipid monolayer
containing a large protein called apo B100. It contains free cholesterol, cholesterol ester and triacylglycerol. Receptor
mediated endocytosis of LDL occurs via clatherin/AP2 coated pits and vesicles. AP2 is heterotetramer consists of
two large α and β2, one medium μ2, and one small σ2 subunit.
Michael Brown and Joseph Goldstein demonstrated that the uptake of LDL by mammalian cells requires the binding
of LDL to a specific cell surface receptor that is concentrated in clathrin-coated pits and internalized by endocytosis.
When a cell needs cholesterol for membrane synthesis, it makes transmembrane receptor proteins for LDL called
LDL receptor and inserts them into its plasma membrane. The LDL receptor, made up of 839 amino acid residues,
is a single-pass transmembrane glycoprotein. LDL receptors have four-residue sequence, Asn–Pro–X–Tyr (X = any
amino acid) in the cytosolic domain that is crucial for internalization. This sequence binds to the AP2 complex. In
most cell surface receptors such as transferrin receptor, the amino acid sequence present is Tyr-X-X-ø, where X
can be any amino acid and ø is a bulky hydrophobic amino acid, such as Phe, Leu, or Met residue.
Any LDL particles bound to LDL receptors in the clathrin/AP2 coated pits are rapidly internalized in clathrin/AP2 coated
vesicles. Clathrin-coated vesicles are ~120 nm in diameter. These coated vesicles from the plasma membrane move
to endosomes. Dynamin, a cytosolic protein, is essential for release of complete vesicles. Dynamin polymerizes
around the neck portion and then hydrolyzes GTP. The energy derived from GTP hydrolysis is essential for the final
pinching off of a completed clathrin-coated vesicle. The clathrin coat is rapidly lost shortly after the vesicle forms.
An Hsp70 chaperone function as an uncoating ATPase to peel off the clathrin coat. Auxillin is believed to activate
the ATPase.

Exoplasmic face

Clathrin coat Cytosolic face Plasma


Dynamin
membrane

Clathrin-coated vesicle

Figure 3.25 Dynamin-mediated pinching off of clathrin coated vesicles. When clathrin polymerizes on a donor mem-
brane, it does so in association with adaptor protein, which assembles between the clathrin lattice and the membrane.
After a vesicle bud forms, dynamin polymerizes over the neck. Dynamin-catalyzed hydrolysis of GTP leads to release
of the vesicle from the donor membrane. Adapted from K. Takel et al., 1995, Nature 374:186.

Endocytic vesicles derive from the plasma membrane are directed to fuse with and deliver their membrane and content
to the intracellular organelles of the endocytic pathway, the endosomes and the lysosomes. Endosomes are single
membrane bound heterogeneous structure. It is of three types – Early endosome (closer to plasma membrane),
late endosome (closer to the nucleus) and recycling endosome. The endocytic vesicles deliver their contents and
their membrane to early endosomes. Early endosomes mature into late endosomes that subsequently fuse with
lysosomes. The fusion of an endosome with a lysosome generates a transient hybrid organelle, the endolysosome,
in which active degradation takes place. Recycling endosomes are a sub-compartment of early endosomes that
return material to the plasma membrane. Late endosomes are more acidic than early endosomes. The interior
of the endosomal compartment is kept acidic (pH ~6) by ATP driven H+ pumps in the endosomal membrane that
pump H+ into the lumen from the cytosol.
A specialized late endosome that contains membrane-bound intraluminal vesicles is called multivesicular body (or
multivesicular endosome). These vesicles form by budding into the lumen of the multivesicular body. The content
of multivesicular bodies can be degraded, via fusion with lysosomes, or released into the extracellular space, via
fusion with the plasma membrane.
This page intentionally left blank.
Cell Structure and Functions 283

Clearly, the transport of newly synthesized proteins from the RER to the Golgi cisternae is a highly selective and
regulated process. The selective entry of proteins into membrane-bound transport vesicles is an important feature
of protein targeting as we will encounter them several times in our study of the subsequent stages in the maturation
of secretory and membrane proteins.

3.7 Golgi complex


The Golgi complex was first discovered in 1897 by Italian physician Camillo Golgi. The Golgi complex, also termed
as Golgi body or Golgi apparatus, is a single membrane bound organelle and part of endomembrane system. It
consists of five to eight flattened membrane-bound sacs called the cisternae. Each stack of cisternae is termed
as Golgi stack (or dictyosome). The cisternae in Golgi stack vary in number, shape and organization in different
cell types. The typical diagrammatic representation of three major cisternae (cis, medial and trans) as shown in
the figure 3.43 is actually a simplification. In some unicellular flagellates, however, as many as 60 cisternae may
combine to make up the Golgi stack. The number of Golgi complexes in a cell varies according to its function. A
mammalian cell typically contains 40 to 100 stacks. In mammalian cells, multiple Golgi stacks are linked together
at their edges.
Each Golgi stack has two distinct faces: a cis face (or entry face or forming face) and a trans face (or maturing
face). Both cis and trans faces are closely associated with special compartments: the cis Golgi network (CGN) and
the trans Golgi network (TGN), respectively. TGN was earlier known as GERL (Golgi apparatus - ER - lysosome).
Proteins and lipids enter the cis Golgi network in vesicular tubular clusters arriving from the ER and exit from the
trans Golgi network. Both networks are thought to be important for protein sorting. As we have seen, proteins
entering the CGN can either move onward in the Golgi apparatus or be returned to the ER. Similarly, proteins
exiting from the TGN can either move onward and be sorted according to whether they are destined for lysosomes,
secretory vesicles, or the cell surface, or be returned to an earlier compartment.

ER

Phosphorylation of sugar cis Golgi network

Removal of Man cis cisterna

Removal of Man, addition of GlcNAc Golgi stack


medial cisterna

Addition of Gal, addition of NANA trans cisterna

Sulfation of tyrosines and carbohydrates trans Golgi network

Plasma membrane

Figure 3.43 The functional compartmentalization of the Golgi apparatus.

The Golgi apparatus is especially prominent in cells that are specialized for secretion, such as the goblet cells of
the intestinal epithelium, which secrete large amounts of polysaccharide-rich mucus into the gut. In such cells,
unusually large secretory vesicles are found on the trans side of the Golgi apparatus. Secretory vesicles form from
284 Cell Structure and Functions
the trans Golgi network, and they release their contents to the cell exterior by exocytosis. The membrane proteins
and the lipids in these vesicles provide new components for the cell’s plasma membrane, while the soluble protein
and other compounds inside the vesicles are secreted to the extracellular space.
The Golgi apparatus is often considered the distribution and shipping department for the cell’s chemical products. It
modifies proteins and lipids that have been built in the endoplasmic reticulum and prepares them for export outside
of the cell or for transport to other locations in the cell. Proteins and lipids from the smooth and rough endoplasmic
reticulum bud off in tiny bubble-like vesicles that move through the cytoplasm until they reach the Golgi apparatus.
The vesicles fuse with the Golgi membranes and release their internally stored molecules into the organelle. Once
inside, the compounds are further processed by the Golgi apparatus. When completed, the product is extruded
from the Golgi apparatus in a vesicle and directed to its final destination inside or outside the cell.
The modifications to molecules that take place in the Golgi apparatus occur in an orderly fashion. Substances from
ER enter into the cis face of a Golgi stack for processing and exit from trans face. Consequently, the cis face is found
near the endoplasmic reticulum and the trans face is positioned near the plasma membrane of the cell. The chemical
make-up of each face is different and the enzymes contained in the cisternae between the faces are distinctive.

Glycosylation of proteins
N-linked oligosaccharide chains on proteins are altered as the proteins pass through the Golgi cisternae en route
from the ER. Further modifications of N-linked oligosaccharide in the Golgi apparatus gives two broad classes of
N-linked oligosaccharides, the complex oligosaccharides and the high-mannose oligosaccharides. High-mannose
oligosaccharides have no new sugars added to them in the Golgi apparatus. They contain just two N-acetylglucosamines
and many mannose residues. Complex oligosaccharides, by contrast, can contain more than the original two
N-acetylglucosamines as well as a variable number of galactose and sialic acid residues and, in some cases, fucose.
The complex oligosaccharides are generated by both removal of existing sugars and addition of new sugars.
Some proteins undergo O-linked glycosylation in the cisternae. O-linked oligosaccharides are linked to the hydroxyl
group of serine or threonine via N-acetylgalactosamine (in collagens to the hydroxyl group of hydroxylysine via
galactose). O-linked oligosaccharides are generally short, often containing only one to four sugar residues.
O-linked sugars are added one at a time, and each sugar transfer is catalyzed by a different glycosyltransferase
enzyme. Typical N-linked oligosaccharides, in contrast, always contain mannose as well as N-acetylglucosamine
and usually have several branches each terminating with a negatively charged sialic acid residue.

Lipid and polysaccharide metabolism


In addition to its activities in processing and sorting glycoproteins, the Golgi apparatus functions in lipid metabolism—in
particular, in the synthesis of glycolipids and sphingomyelin. Ceramide, which is synthesized in the ER, is converted
either to sphingomyelin or to glycolipids in the Golgi apparatus.
The Golgi apparatus also acts as a major site of carbohydrate metabolism. In plant cells, it acts as the site where
complex polysaccharides of the cell wall are synthesized. Complex cell wall polysaccharides such as hemicelluloses
and pectins are synthesized in the Golgi apparatus and then transported in vesicles to the cell surface. In animals,
most of the glycosaminoglycans of the extracellular matrix are synthesized in the Golgi apparatus.

3.7.1 Transport of proteins through cisternae


Several models have been proposed to explain the ordered transport of proteins through the cisternae. Two most
accepted models are cisternal maturation model and vesicular transport model.
According to cisternal maturation model, stacks of cisternae are dynamic. The cis-most cisterna is assembled by
the fusion of transport vesicles from the ER, carrying newly synthesized cargo. Each Golgi cisterna matures as
it migrates outward through a stack and at each stage, the Golgi resident proteins that are carried forward in a
cisterna are moved backward to an earlier compartment. In this way each cisterna progresses into the next one
and COPI-coated vesicles function to transport resident Golgi components in the retrograde direction.
This page intentionally left blank.
Cell Structure and Functions 287

as R-SNAREs (arginine containing SNAREs) or Q-SNAREs (glutamine-containing SNAREs), based on the identity
of a highly conserved residue.
Recognition step is controlled by members of a family of monomeric GTPases called Rab proteins, which check
that the interaction between a v-SNARE and a t-SNARE is correct. Cells contain many types of Rab proteins, each
associated with membrane of particular organelle and transport vesicle. There are over 60 Rab proteins. Rab proteins
cycle between a membrane and the cytosol. In their GDP-bound state, they are inactive and remain in cytosol while
in their GTP-bound state they are active and tightly associated with the membrane of an organelle or transport
vesicle. Membrane bound Rab-GEFs activate Rab proteins. Once in GTP-bound and membrane bound state, Rab
proteins bind to other proteins, called Rab effector, which act as tethering protein. Rab effector proteins interact
with active Rab proteins located on target membrane, vesicle membrane or both. After tethering, SNAREs mediate
docking and fusion. The binding of v-SNARE to t-SNARE causes the vesicle to remain bound for long enough to
allow the Rab protein to hydrolyze its bound GTP, which locks the vesicle onto the target membrane.

Docking

Formation of trans
Tethering Fusion SNARE
SNARE complex
Target
dissociation
membrane
Rab effector t-SNARE
v-SNARE
SNAP NSF
cis
v-SNARE SNARE
GTP GDP complex

Rab-GTP Soluble Rab-GDP


Vesicle

Figure 3.46 Complementary sets of vesicle-SNAREs (v-SNAREs) and target-membrane SNAREs (t-SNAREs) deter-
mine the selectivity of transport-vesicle docking. v-SNAREs of transport vesicles from the donor membrane, bind to
complementary t-SNAREs in the target membrane. The v- and t-SNARE proteins can be in trans (on opposing mem-
branes) or cis (on the same membrane) conformations. The Rab protein hydrolyzes its bound GTP, locking the vesicle
onto the target membrane and releasing Rab-GDP into the cytosol, from where it can be reused in a new round of
transport. The vesicle then fuses with the target membrane. NSF is an ATPase that hydrolyzes ATP to release the
complex once it has done its job.

Interacting SNAREs need to separate apart before they can function again. Most SNARE proteins in cells participate
in multiple rounds of vesicular transport. The complexes have to disassemble before the SNAREs can mediate new
rounds of transport. After the v-SNAREs and t-SNAREs have mediated the fusion of a vesicle on a target membrane,
the NSF (NEM Sensitive Factor) binds to the SNARE complex via adaptor proteins, SNAPs (Soluble NSF Attachment
Proteins) proteins. NSF, a hexamer of identical subunits, and SNAPs are not necessary for actual membrane fusion
but rather are required for regeneration of free SNARE proteins. NSF is a soluble ATPase and hydrolyzes ATP to
dissociate the SNAREs apart.

3.9 Lysosome
Lysosomes are single membrane-bound organelles present in animal cells. They are heterogeneous structure and
greatly vary in size and shape. Lysosomes have acidic internal pH (about 5) and are filled with hydrolytic enzymes.
They contain about 40 different types of hydrolytic enzymes (including proteases, nucleases, glycosidases, lipases,
phospholipases, phosphatases and sulfatases) which are responsible for the controlled intracellular digestion of
macromolecules. All are acid hydrolases because they require an acidic environment for optimal activity and the
lysosome provides this by maintaining a pH of about 5.0. A vacuolar H+ ATPase in the lysosomal membrane uses
the energy of ATP hydrolysis to pump H+ into the lysosome, thereby maintaining the internal acidic pH.
288 Cell Structure and Functions
0.2–0.5 µm

Lysosome Cytosol
pH ~5 pH ~7.2

+
H

ATP ADP

Figure 3.47 The interior of lysosomes has a pH of about 5.0. To create the low pH, V- type H+ ATPase located in
the lysosomal membrane pump protons into the lysosome using energy supplied from ATP. All the lysosomal enzymes
work most efficiently at acidic pH and collectively are termed acid hydrolases.

There are two types of lysosomes: primary lysosomes (do not contain materials for intracellular digestion) and
secondary lysosomes (contain materials that are undergoing digestion or that already have been digested).
Lysosomes are responsible for the digestion of both extracellular as well as intracellular materials. Lysosomal
digestion of materials can be classified into autophagy and heterophagy. The process by which substances are
taken into the cell from external environment and broken down by lysosome is called heterophagy. In contrast,
the degradation of cytoplasmic components within lysosomes is called autophagy.
In heterophagy, there are two different pathways that brings extracellular materials to lysosomes for degradation.
Phagocytic cells, such as macrophages and neutrophils in vertebrates, engulf large particles by the process of
phagocytosis. During phagocytosis, a single-membrane phagosome is generated, and this compartment fuses
directly with a lysosome to form a phagolysosome.
Virtually all eucaryotic cells continually internalize fluid substances in small pinocytic (endocytic) vesicles by the
process of pinocytosis. Most of endocytosed substances eventually end up in lysosomes, where they degraded. In
this process, the endocytosed substances first move from the endocytic vesicles to the endosomes. At the end of
this pathway, the late endosomes convert to endolysosomes and lysosomes as a result of both their fusion with
preexisting lysosomes and progressive acidification.

Cytosol
Phagocytosis
Phagosome Plasma
membrane
Early
Late endosome
endosome

(a) Figure 3.48


Pinocytosis
Lysosome (a) Schematic overview
of three pathways by which
materials are moved to
lysosomes: phagocytosis,
Phagophore Autophagosome
pinocytosis and autophagy.
Autophagy
(b) Process of autophagy.

Phagophore Autophagosome

Fusion
(b)
Engulfing
cytoplasmic
components
Autolysosome Degradation of
cytoplasmic components

Lysosome
Cell Structure and Functions 289

Autophagy is an intracellular degradation process of cytoplasmic constituents within lysosomes. During autophagy,
sequestration begins with the formation of a phagophore. Phagophores form de novo in the cytoplasm from a
cup-shaped membrane that expands into a double-membrane bound autophagosome surrounding a portion of the
cytoplasm. The autophagosome may fuse with an endosome. The product of the endosome-autophagosome fusion
is called an amphisome. The completed autophagosome or amphisome fuses with a lysosome, which supplies acid
hydrolases. The enzymes in the resulting compartment, an autolysosome, breakdown the inner membrane from the
autophagosome and degrade the materials. The resulting macromolecules are released and recycled in the cytosol.
Some lysosomes participate in exocytosis. This enables cells to eliminate undigested contents. For most cells, this
seems to be a minor pathway, used only when the cells are stressed. Some cell types, however, contain specialized
lysosomes that have acquired the necessary machinery for fusion with the plasma membrane. Melanocytes (melanin-
producing cells) in the skin, for example, produce and store melanin pigments in their lysosomes. These pigment
containing melanosomes release their pigment into the extracellular space of the epidermis by exocytosis.

Table 3.10 Example of some acid hydrolases present in lysosomes

Enzyme Natural substrate

Phosphatases

Acid phosphatase Most phosphomonoesters

Acid phosphodiesterase Oligonucleotides and other phosphodiesters

Nucleases

Acid ribonuclease RNA

Acid deoxyribonuclease DNA

Polysaccharide/mucopolysaccharide hydrolyzing enzymes

β-Galactosidase Galactosides

α-Glucosidase Glycogen

α-Mannosidase Mannosides, glycoproteins

β-Glucuronidase Polysaccharides and mucopolysaccharides

Hyaluronidase Hyaluronic acids; chondroitin sulfates

Proteases

Cathepsin(s) Proteins

Collagenase Collagen

Peptidases Peptides

Lipid-degrading enzymes

Esterase(s) Fatty acyl esters

Phospholipase(s) Phospholipids

3.10 Vacuoles
Most plants and fungal cells contain one or several very large, fluid-filled vesicles called vacuoles. They are
surrounded by single membrane called tonoplast and related to the lysosomes of animal cells, containing a variety
290 Cell Structure and Functions
of hydrolytic enzymes, but their functions are remarkably diverse. Like a lysosome, the lumen of a vacuole has an
acidic pH, which is maintained by similar transport proteins in the vacuolar membrane. The plant vacuole contains
water and dissolved inorganic ions, organic acids, sugars, enzymes and a variety of secondary metabolites. Solute
accumulation causes osmotic water uptake by the vacuole, which is required for plant cell enlargement. This water
uptake generates the turgor pressure.
The vacuole is different from contractile vacuole. A contractile vacuole is an organelle involved in osmoregulation.
It pumps excess water out of the cell. It is found predominantly in protists (such as Paramecium, Amoeba) and in
unicellular algae (Chlamydomonas). It was previously known as pulsatile or pulsating vacuole.

3.11 Mitochondria
Mitochondria (term coined by C. Benda) are energy-converting organelles, which are present in virtually all
eukaryotic cells. They are the sites of aerobic respiration. They produce cellular energy in the form of ATP, hence
they are called ‘power houses’ of the cell. Mitochondria are membrane-bound mobile as well as plastic organelle.
Each mitochondrion is a double membrane-bound structure with outer and inner membranes. The space between
the outer and inner membranes is called intermembrane space. The outer membrane is fairly smooth. But the
inner membrane is highly convoluted; forming folds called cristae. The inner membrane is also very impermeable
to many solutes due to very high content of a phospholipid called cardiolipin. The cristae greatly increase the
inner membrane’s surface area. The two faces of this membrane are referred to as the matrix side (N-side) and
the cytosolic side (P-side). Inner membrane contains enzyme complex called ATP synthase (or F0-F1 ATPase or
oxysome) that makes ATP. The outer membrane protects the organelle, and contains specialized transport proteins
such as porin which allows free passage for various molecules into the intermembrane space of the mitochondria.
Mitochondrial porins, or voltage-dependent anion-selective channels (VDAC) allow the passage of small molecules
across the mitochondrial outer membrane.

Inner membrane

Outer membrane
Matrix
Intermembrane space

ATP synthase (F0-F1 ATPase)

Figure 3.49 A mitochondrion has double-membraned organization and contains: the outer mitochondrial mem-
brane, the intermembrane space (the space between the outer and inner membranes), the inner mitochondrial mem-
brane, and the matrix (space within the inner membrane).

The matrix (large internal space) contains multiple copies of the dsDNA (as genetic material), mitochondrial
ribosomes (ranging from 55S-75S), tRNAs and various proteins. Mitochondrial dsDNA is mostly circular. The size
of mitochondrial DNA also varies greatly among different species.

Organisms Size (kb)


Human 16.6
Xenopus (frog) 18.4
Drosophila (fruit fly) 18.4
Saccharomyces (yeast) 75.0

Arabidopsis (mustard plant) 367.0

Kinetoplastids such as Trypanosoma brucei, Trypanosoma cruzi, possess a network of DNA, known as the kinetoplast
DNA (kDNA), within mitochondria. It consists of two types of circular DNA molecules, maxicircles and minicircles,
which are topologically interlocked into a single, massive network.
This page intentionally left blank.
Cell Structure and Functions 293

Targeting to inner-membrane
Proteins enter into inner mitochondrial membrane contain either N-terminal cleavable matrix targeting sequences or
internal signal sequences. Unlike targeting to the matrix, targeting of proteins to the inner mitochondrial membrane
requires different strategies. In most single-pass transmembrane proteins, N-terminal cleavable matrix targeting
sequences first direct the precursor proteins into the matrix; however, the precursor proteins additionally contain
a stop transfer signal that is located after the matrix-targeting presequence. This signal arrests translocation in the
mitochondrial inner membrane with the precursor proteins being released laterally into the lipid layer. During targeting,
the N-terminal cleavable matrix targeting sequences is recognized by Tom20 receptor of the TOM complex. The
protein is then translocated through the TIM23 complex so that it transiently spans both mitochondrial membranes.
An another protein translocase in the inner mitochondrial membrane, the OXA complex, mediates the insertion of
those inner membrane proteins that are synthesized within mitochondria by 70S ribosomes. It also helps to insert
some imported inner membrane proteins that are initially transported into the matrix space by the other complexes.

Cytosolic Presequence
Hsc70
C N

Cytosol
b-barrel
20 70
TOM40 protein
complex
Outer SAM
membrane Tom40

Small TIM
Intermembrane
50
space 21 TIM23
complex
Tim23 +++++
Inner
TIM22 Dy
membrane PAM –––––

MPP
Matrix
Matrix
protein

Figure 3.51 Two main protein import pathways in mitochondria. N-terminal cleavable presequence direct proteins
through the TOM complex, and TIM23 complex to the matrix. Precursor proteins with internal targeting signals are
recognized by the receptor Tom70 and a different inner membrane translocation complex (TIM22) is used.

The multipass inner-membrane proteins contain internal targeting sequences that are recognized by the Tom70
import receptor. A TIM22 complex is used in this pathway. Components of the TIM22 translocation system also
include the small Tim proteins (Tim8, Tim9, Tim10 and Tim13). The small Tim proteins assemble in hexameric
complexes (referred to as the small Tim complexes) in the intermembrane space in which three Tim9 polypeptides
partner with three Tim10 and three Tim8 polypeptides partner with three Tim13. Tim8–Tim13 or Tim9–Tim10 guide
the precursor through the intermembrane space to the TIM22 complex.
Targeting to outer-membrane: The proteins of outer mitochondrial membrane (such as β-barrel proteins) are
initially imported through the TOM complex, interact with small Tim complexes (Tim9–Tim10 complex or Tim8–
Tim13 complex) in the intermembrane space and are inserted into the outer membrane by the SAM (Sorting and
Assembly Machinery) complex present on outer membrane.

3.12 Plastids
Plastids are double membrane bound semi-autonomous organelles present in all living plant cells and photosynthetic
protists. All plastids contain multiple copies of the dsDNA as genetic materials and 70S ribosomes for proteins
synthesis. Plastids differentiate from proplastids. Proplastids are inherited with cytoplasm of plant egg cells. As
294 Cell Structure and Functions
immature plant cells differentiate, the proplastids develop according to the needs of the specialized cell: they
can become chloroplast, leucoplasts or chromoplasts. A collective term used for different kinds of organelles, all
derived from proplastids, is plastid. Chloroplast is the most important member of plastid family. It occurs in all
photosynthetic eukaryotes and acts as site of photosynthesis. It has a double membrane which encloses a fluid-
filled region called the stroma. Embedded in the stroma is a complex network of stacked sacs. Each stack is called a
granum and each of the flattened sacs which makes up the granum is called a thylakoid. The thylakoid membrane,
that encloses a fluid-filled thylakoid interior space, contains photosynthetic pigments. There are many grana in
each chloroplast (usually 10 to 100 grana) which are interconnected by unstacked stromal lamellae. The lipids of
the thylakoid membrane have a distinctive composition. About 80% lipids are uncharged mono- and digalactosyl
diacylglycerol and only about 10% are phospholipids.

Granum Thylakoid membrane


Granum

}
}
Stroma

Thylakoid lumen

Stroma
Outer Inner
lamella
membrane membrane

Figure 3.52 The two envelope membranes enclose the stroma. The stacks of the thylakoid termed grana are con-
nected by tubes, forming a continuous thylakoid lumen.

In the dark grown plants, proplastids develop into etioplasts, which have a yellow chlorophyll precursor pigment
protochlorophyll instead of chlorophyll. When exposed to light, the etioplasts rapidly change into chloroplasts by
converting this precursor to chlorophyll.
Chromoplasts are plastids responsible for pigment synthesis and storage. They are rich in carotenoids and mainly
responsible for the yellow, orange, or red colors of many fruits and flowers, as well as of autumn leaves. Leucoplasts
are colorless (non-pigmented) plastids and act as storage organelles. Based on the kind of substance they store,
they are further classified into amyloplasts (for starch storage), elaioplasts (for fat storage) and proteinoplasts or
aleuroplasts (for storing and modifying proteins).

3.13 Peroxisome
Peroxisome (discovered by Christian de Duve in 1965) is a single membrane bound small organelle (approximately
0.5–1 μm in diameter) present in all eukaryotes. The term, peroxisome, was proposed by de Duve because it
produced and consumed hydrogen peroxide. Peroxisomes lack DNA and ribosomes. Thus, all peroxisomal proteins
(peroxisomal matrix and membrane proteins) are encoded by nuclear genes, synthesized on ribosomes present in
the cytosol and then incorporated into pre-existing peroxisomes.
The ability of peroxisomes to divide themselves suggests that the peroxisome may have had an endosymbiotic
origin similar to mitochondria. However, the localization of peroxisomal proteins to the endoplasmic reticulum and
the similarity of some peroxisomal proteins to those localized in the ER suggest an alternative hypothesis: that the
peroxisome was developed from the ER (de novo origin). Aspects of both views may be true. Most peroxisomal
membrane proteins are made in the cytosol by membrane free ribosomes and insert into the membrane of preexisting
ones. However, few others are first synthesized by membrane bound ribosomes of ER and then integrated into the
ER membrane from where they may bud in specialized peroxisomal precursor vesicles. New peroxisome precursor
vesicles may then fuse with one another and begin importing additional peroxisomal proteins synthesized by membrane
free cytosolic ribosomes to grow into mature peroxisomes, which can enter into a cycle of growth and fission.
This page intentionally left blank.
Cell Structure and Functions 299

Telomere: Telomeres are specialized structures, which cap the ends of eukaryotic chromosomes. It consists of a
long array of short, tandemly repeated sequences. Sequencing of telomeres from different organisms has shown
that most are repetitive sequences with a high G content in the strand with its 3’ end.

Origin of replication: The origin of replication (also called the replication origin) is a particular sequence in a
chromosome at which replication is initiated. One chromosome contains multiple origin of replication.

Chromosome number
All eukaryotic cells have multiple linear chromosomes. Chromosomes number in eukaryotes are species specific. Every
species maintains a characteristic number of chromosomes. Depending on the eukaryotic organism, the number of
chromosomes varies from 2 to several hundreds. Eukaryotes can be monoploid (having one set of chromosomes),
diploid (having two sets of chromosomes) and polyploid (having more than two sets of chromosomes). The majority
of eukaryotes are diploid.

Table 3.11 Chromosome number in different eukaryotic organisms

Species Haploid number of chromosome

Saccharomyces cerevisiae (budding yeast) 16

Schizosaccharomyces pombe (fission yeast) 03

Caenorhabditis elegans 06

Arabidopsis thaliana 05

Drosophila melanogaster 04

Tetrahymena thermophila 05 (in micronucleus)

Homo sapiens (human) 23

Tetrahymena thermophila is a ciliated protozoan. It has two nuclei – somatic macronucleus (responsible for
vegetative function) and germline micronucleus (responsible for sexual reproduction). The micronuclus is diploid
and contains five pairs of chromosomes. Macronuclear chromosomes are derived from site-specific fragmentation
of the five original micronuclear chromosomes.

3.15 Cytoskeleton
Cytoskeleton is an intracellular network of protein filaments present in the cytoplasm. There are three types of
cytoskeletal fibers—microfilaments, intermediate filaments, and microtubules. All these fibers are polymers built
from small protein subunits held together by noncovalent bonds.

3.15.1 Microtubules
Microtubules are hollow, cylindrical structure, approximately 25 nm in diameter. They are present in all eukaryotic
cells and play crucial roles in the determination of cell shape and cellular motility. The basic building block of the
microtubule is αβ-tubulin heterodimer, composed of two 50 kDa subunits known as α-tubulin and β-tubulin. A
large number of additional cellular molecules, called Microtubule-Associated Proteins (MAP), bind to the surface
of the microtubule and determine and control its many functions. It is widely accepted that many of these MAPs
play crucial roles in the regulation of microtubule polymerization and dynamics, in the organization of microtubule
arrays, and in the functional interactions of microtubules with other cellular components.
300 Cell Structure and Functions
Microtubule structures
Microtubule polymers are composed of αβ-tubulin heterodimers arranged in a uniformly repeated head-to-tail
fashion. Each linear array of tubulin heterodimers is called a protofilament and in cells generally 13 protofilaments
arranged into a tube structure called microtubules. However, many examples exist in which microtubules have a
different number of protofilaments (from 11 to as many as 15). The 13 protofilaments are aligned in parallel with
the same polarity. β-tubulin subunits of all protofilaments face one end of the microtubule, and the α-subunits face
the opposite end. The ends are designated either plus or minus, based upon their different dynamic and structural
properties. β-subunits are exposed at the plus ends (fast-growing) while the α-subunits are exposed at the minus
ends (slow-growing).

Plus end Plus end


Lumen

b Polymerization 11 to 15
a
Tubulin
heterodimer

Minus end Minus end

Protofilament Microtubles

Figure 3.55 Structure of tubulin heterodimers and their organization in microtubules. The tubulin dimer is aligned
end to end into protofilaments, which pack side by side to form the wall of the microtubule. The microtubule displays
a structural polarity. The rate of polymerization and depolymerization is faster at the plus end as compared to the
minus end.

Singlet microtubule (made up of 13 protofilaments) in cytosol is very common, but many specialized microtubule
arrays also exist in which microtubules are fused along their lengths into doublet or triplet microtubule structures.
Doublet microtubules consist of 10 or 11 protofilaments of a second microtubule fused to an entire 13 protofilament
first microtubule, and triplet microtubules have an additional 10 or 11 protofilaments of a third microtubule fused
to the second microtubule. Doublet microtubules occur in the long shafts of motile cell appendages called cilia and
flagella. Triplet microtubules occur in the basal body structures that in most animal cells are situated at the base
of cilia and flagella and in centrioles, which are situated at the center of animal cell centrosomes.

10 9 10 9
8 9 8 9 9 8
10 8 10 8 10 11
7 11 7 7 7
7
6 12 6 6 6 12 6
B A C B A
5 13 5 5 5 13 5
4 1 1 4 4 1 4 1 1 4
3 2 2 3 3 2 3 2 2 3

Doublet microtubule Triplet microtubule

Figure 3.56 Each doublet or triplet contains one complete 13-protofilament microtubule (A tubule) and one or two
additional tubules (B and C) consisting of 10 protofilaments.
This page intentionally left blank.
302 Cell Structure and Functions
to one end and dissociating tubulin heterodimers from the opposite end. This special state in polymer dynamics,
known as treadmilling, keeps the polymer length unchanged.

Dynamic instability
Dynamic instability is characterized by phase transitions, or switching, between phases of relatively slow growing
and rapid shortening at the ends of individual microtubules. Dynamic instability is supposed to be a result of the
hydrolysis of GTP bound to β-tubulin shortly after assembly. The kinetics of GDP-tubulin are different from those
of GTP tubulin. GDP-bound tubulin heterodimers is prone to depolymerization. Since tubulin adds onto the end
of the microtubule only in the GTP-bound state, there is generally a cap of GTP-bound tubulin at the tip of the
microtubule, protecting it from disassembly. When hydrolysis catches up to the tip of the microtubule, it begins
a rapid depolymerization and shrinkage. This switch from growth to shrinking is called a catastrophe. Addition
of GTP-bound tubulin to the tip of the microtubule again, provides a new cap and protects the microtubule from
shrinking. This is referred to as rescue.

3.15.2 Kinesins and Dyneins


Kinesin is a motor protein that moves along microtubules. The genomes of mammals encode more than 40 kinesin
proteins, organized into at least 14 families named kinesin-1 through kinesin-14. Members of the kinesin superfamily
are characterized by a common ATP-binding domain. Since most members of the kinesin superfamily are involved
in active transport and movement, this domain is usually called motor domain. According to the position of the
motor domain, the kinesin family can be classified into N-type kinesins (with the motor domain at or near to the
N-terminus, as in Kinesin-1 to 12), M-type kinesins (with the motor domain internally located, as in kinesin-13) and
C-type kinesins (with the motor domain close to the C-terminus, as in kinesin-14). Most of kinesins are microtubule
based plus end-directed motor proteins. Kinesin-14, by contrast, are minus end directed motors.
The conventional kinesin, kinesin-1, consists of two heavy chains and two light chains. The heavy chain has three
functional domains: the motor head domain, the α-helical stalk domain and the globular tail domain. The kinesin-1
molecule comprises a pair of large globular head domains connected by an elongated coiled-coil stalk domain to a
pair of small globular tail domains, which contain the light chains. Each head has two separate binding sites: one
for the microtubule and the other for ATP. The tail domain is responsible for binding to receptors on the membrane
of cargoes. It is a plus end-directed motor protein involved in organelle transport. Although most kinesins have
two heavy chains (e.g. kinesin-1), others may have a single heavy chain or four heavy chains. The kinesin-5 family
members have four heavy chains and act as bipolar motor proteins. Kinesin-5 protein contains two motor domains
that interact with two antiparallel microtubules and move towards the plus end.

a-helical stalk domain (coiled-coil structure)

Globular tail domain


(C-terminus)
Motor head domains
ATPase domain
Microtubule binding domain

Figure 3.57 Kinesin, consists of two heavy chains and two light chains. The heavy chain has three functional do-
mains: the motor head domain, the α-helical stalk domain and the globular tail domain.

Dyneins are a family of minus-end directed microtubule based motors proteins. They are composed of two or three
heavy chains (that include the motor domain) and a large and variable number of associated light chains. Heavy
Cell Structure and Functions 303

chains are present in the form of homo- or heterodimer and heterotrimer. Dynein motors exist as either two-headed
or three-headed structures. Dyneins are the largest of the known molecular motors and they are also among the
fastest. Unlike kinesin, dynein cannot mediate cargo transport by itself. Rather, dynein-related transport requires
dynactin, a large protein complex consists of 11 subunits that links vesicles and chromosomes to the dynein light
chains. The dynein family can be classified into two major groups:
Cytoplasmic dyneins are probably found in all eukaryotic cells, and they are important for vesicle trafficking, as well as
for localization of the Golgi apparatus near the center of the cell. They mediate retrograde transport of vesicles toward
the minus end of microtubules. Cytoplasmic dynein contains approximately twelve polypeptide subunits: two identical
heavy chains (homodimer), which contain the ATPase activity and several light chains which are less understood.
Axonemal dyneins, highly specialized for the rapid and efficient sliding movements of microtubules that drive the
beating of cilia and flagella. Heavy chains in axonemal dyneins are heterodimers or heterotrimers.

3.15.3 Cilia and Flagella


Flagella is a locomotary organ present in many prokaryotes and eukaryotes. Eukaryotic flagella is different from
prokaryotic flagella. Eukaryotic flagella (singular, flagellum) have a specialized arrangement of microtubules. A
flagellum has two components–a filament and a basal body (or kinetosome). A filament possess a bundle of
microtubules, called the axoneme, in which nine outer doublet microtubules surround a central pair of singlet
microtubules. This is termed as 9 + 2 arrangement. Each doublet microtubule consists of A- and B-tubules. The
A-tubule is a complete microtubule with 13 protofilaments, while the B tubule contains 10 protofilaments. The
junction between A and B tubules of one doublet is probably strengthened by the protein tektin, a highly α-helical
protein that is similar in structure to intermediate-filament proteins. The axoneme is surrounded by the plasma
membrane. The axoneme of filament connects with the basal body. Like centrioles, basal bodies are cylindrical
structures which contain nine triplet microtubules. Each triplet contains one complete 13 protofilament microtubule,
the A tubule, fused to the incomplete B tubule, which in turn is fused to the incomplete C tubule. The A and B
tubules of basal bodies continue into the axonemal shaft, whereas the C tubule terminates within the transition
zone between the basal body and the shaft.

Nexin
Dynein arm
C1 C2

9 1

9 + 2 arrangement Plasma
Axoneme

2 membrane

8
Singlet
microtubules
C1 C2
3
Plasma
7
membrane
Radial spoke
4
Outer doublet
A B
6 microtubule
5

Basal body

Figure 3.58 Arrangement of microtubules in a flagellum. A cross section of the filament of a sperm flagellum. It has
a bundle of microtubules called the axoneme which is surrounded by the plasma membrane. In axoneme, nine outer
doublet microtubules surround a central pair of singlet microtubules. The outer doublets are joined to each other by
nexin protein and to the central pair of singlet microtubules by radial spokes.
304 Cell Structure and Functions
Within the axoneme, the two central singlet and nine outer doublet microtubules are continuous for the entire length
of the structure. Permanently attached to the A tubule of each doublet microtubule is an inner and an outer row
of dynein arms. These dyneins reach out to the B tubule of the neighboring doublet. A linker made up of protein
nexin, joins adjacent outer doublet microtubules. A second linker radial spokes, which radiate from the central
singlets to each A tubule of the outer doublets, form the third linkage system. The radial spoke and central pair
complex are involved in beat regulation.
The basis for axonemal bending is the sliding of doublet microtubules relative to one another. The doublet
microtubules are arranged with their (+) end at the outer tip of the axoneme. The dynein arms on the A-tubule of
one doublet slide along the adjacent doublet’s B-tubule toward its base, the (–) end. The force producing active
sliding requires ATP and is caused by successive formation and breakage of cross-bridges between the dynein arm
and the B-tubule. Successive binding and hydrolysis of ATP causes the dynein arms to successively release from
and attach to the adjacent doublet.
Eukaryotic flagella and cilia share a common structural organization. They differ in length, number per cell and
beating pattern. There are two types of cilia: motile cilia and non-motile cilia. Motile cilia are usually present in
large numbers per cell, for example, on the surface of epithelial cells of the oviduct and the respiratory tract or on
surface of protists such as Paramecium and Tetrahymena.

Microtubule Slide
doublet

Dynein
arms

Microtubules
doublets

Sliding causes bending

Figure 3.59 The dynein arms are attached to A-tubules, and its motor head interact with the B-tubules of adjacent
doublets. Movement of the dynein head groups in the minus end direction (toward the base of the filament) then caus-
es the A-tubule of one doublet to slide toward the base of the adjacent B-tubule. Because both microtubule doublets
are connected by nexin links, this sliding movement forces them to bend. Successive binding and hydrolysis of ATP
causes the dynein arms to successively release from and attach to the adjacent doublet.

Non-motile cilia are classified as primary or sensory cilia. Motile cilia consist of an axoneme of nine doublet
microtubules and central pair of singlet microtubules (9+2 arrangement). In contrast to those of motile 9+2 cilia,
axonemes of non-motile primary cilia lack the central pair of singlet microtubules.
Beating pattern of flagella and motile cilia differs. A flagellum has a wave-like motion that drives a cell in the same
direction as the axis of the flagellum. In contrast, motile cilia generate alternating power and recovery strokes. The
rapid power stroke moves the cell in a direction perpendicular to the axis of the cilium.
This page intentionally left blank.
306 Cell Structure and Functions
There are interesting parallels between centrosome duplication and chromosome duplication. Both use a semi-
conservative mechanism of duplication, in which the two halves separate and serve as templates for construction
of a new half. Centrosomes, like chromosomes, must replicate once and only once per cell cycle, to ensure that
the cell enters mitosis with only two copies.

Duplication Maturation and


and separation
elongation

G1 phase S phase G2 phase M phase

Figure 3.60 The centrosome cycle of an animal cell. After the pair of maternal centrioles separates slightly, a small
procentriole next to each preexisting centriole (maternal) appear which is oriented at right angle to maternal one. By
G2, growth of the daughter centrioles is complete, but the two pairs remain within a single centrosomal complex. Early
in mitosis, the centrosome splits, and each centriole pair migrates to opposite ends of the cell.

3.15.5 Actin filament


Actin filament (microfilament) is made up of G-actins. The globular (G)-actin is a 43 kDa ATPase and like tubulin,
it has a binding site for a nucleotide, but the nucleotide is ATP (or ADP) rather than GTP (or GDP). Each G-actin
contains a Mg2+ ion complexed with either ATP or ADP. G-actin is a highly conserved cytoskeletal protein that is
present at high concentrations in nearly all eucaryotic cells. It is the most abundant intracellular protein in most
eukaryotic cells. As for tubulin, the G-actin subunits assemble head-to-tail to generate filamentous (F)-actin with
a distinct structural polarity. Two parallel F-actins twist around each other in a right-handed helix to form an actin
filament. The structural polarity of actin filament is created by the regular, parallel orientation of all of their subunits.
Actin filaments are thinner and more flexible, and usually much shorter, than microtubules.

ADP

7 nm

– end + end
Actin filament

Figure 3.61 Actin filament made up of two strings of monomeric G actin subunits coiled about each other in a
right-handed helix.

Like a microtubule, an actin filament is a polar structure, with two structurally different ends - a slow-growing
minus end and a faster-growing plus end. The minus end of an actin filament is also referred to as the ‘pointed
end’ and the plus end as the ‘barbed end.’ The plus end polymerizes at up to 10 times the rate of the minus end.
The role of ATP hydrolysis in actin polymerization is similar to the role of GTP hydrolysis in tubulin polymerization.
In neither case is hydrolysis required to form the filament; instead, it serves to weaken the bonds in the polymer
and thereby promotes depolymerization.
The in vitro polymerization of G-actin occurs in three sequential phases. The first phase, termed nucleation phase,
is a lag phase in which G-actin aggregates into short, oligomers. In the second phase called elongation phase,
these nuclei are rapidly elongated by the addition of G-actins to both ends of the oligomers. As F-actin filaments
grow, the concentration of soluble G-actin monomers decreases. In third phase, steady-state phase, equilibrium
Cell Structure and Functions 307

is reached between filaments and monomers. G-actin monomers exchange with subunits at the filament ends, but
there is no net change in the total length of filaments.
The spontaneous initiation of actin-filament assembly requires the formation of a trimeric nucleus in a process that
is called nucleation. Spontaneous nucleation is kinetically unfavourable. Two classes of protein have been identified
that bypass the need for spontaneous nucleation and nucleate actin assembly. These proteins, commonly referred
to as actin nucleating proteins, are the actin-related protein-2/3 (Arp2/3) and formins. The Arp2 and Arp3 and five
other proteins (p40, p35, p19, p18 and p14) form a complex of ~220 kDa called Arp2/3 complex. This complex
has been found in all eukaryotes and is highly conserved across species. The complex binds to the sides of existing
filaments and initiates polymerization of a new filament at a distinctive 70° angle from the existing filaments.
Formins are multi-domain proteins. They are characterised by the presence of FH domains (FH1 and FH2). The
FH1 domain interacts with the G-actin binding protein profilin, providing G-actin for filament assembly, while the
adjacent FH2 domain nucleates F-actin. FH2 domain remains persistently associated with the fast growing barbed
end. Formin function is regulated by binding with Rho GTP to its Rho binding domain.

Table 3.12 Properties of microtubules and microfilaments

Microfilaments Microtubules

Structure Two intertwined chains of F-actin Hollow, cylindrical, consists of 13 protofilaments

Diameter 7 nm Outer : 25 nm, Inner : 15 nm

Monomers G-actin α-tubulin, β-tubulin

Polarity Plus and minus ends Plus and minus ends

Nucleotide bound ATP GTP

Drugs that affect microtubules and actin filament


Microtubule and actin filaments are the targets of a variety of drugs that act by interfering with the polymerization
and depolymerization of subunits. Example of some common drugs that affect microtubules and actin filament:

Actin filament
Cytochalasin D : Binds plus ends of actin filaments and prevents elongation.
Latrunculin : Binds G actin monomers and prevents them from polymerizing into filaments.
Phalloidin : Binds tightly all along the sides of actin filaments and stabilize them against depolymerization.
Jasplakinolide : Induces actin polymerization by stimulating actin filament nucleation; stabilizes F-actin.
Swinholide : Severs filaments.

Microtubules
Nocodazole : Causes microtubules to depolymerize to tubulin subunits.
Colchicine : Binds tubulin subunits and prevents their polymerization.
Colcemid : Binds tubulin subunits and prevents their polymerization.
Taxol : Binds to and stabilizes microtubules, preventing their depolymerization.
Vinblastine : Binds β-tubulin to prevent polymerization.
Vincristine : Binds free tubulin to prevent polymerization.

3.15.6 Myosin
Myosin is a actin filament based motor protein. The form of myosin found in muscle and first identified was myosin-II.
Each myosin-II molecule is an oligomer composed of one pair of identical heavy chains and two pairs of non-identical
light chains. Each of the heavy chains has a globular head domain at its N-terminus that contains an actin-binding
This page intentionally left blank.
312 Cell Structure and Functions
— + Actin filament

Myosin thick filament

ATP

— +

ATP

Hydrolysis of ATP

— +
ADP
Pi

Pi

— +
ADP Figure 3.65
Coupling between ATP hydrolysis and
conformational changes for myosin.
ATP binding with myosin releases the
head from the filament. ATP hydrolysis
Power stroke occurs while the myosin head is detached
ADP
from the filament, causing the head to
assume different conformation. When the
— +
head rebinds to filament, the release of
phosphate, followed by the release of ADP,
triggers the power stroke that moves the
filament relative to the motor protein.

Smooth muscle contraction


The skeletal muscle contraction is regulated by the tropomyosin-troponin complex bound to the thin actin filament
switching between the contraction-inducing state in the presence of calcium and the relaxed state in its absence.
In contrast, contraction of vertebrate smooth muscle is regulated primarily by a pathway in which the myosin
regulatory light chain associated with the myosin II undergoes phosphorylation and dephosphorylation. The smooth
muscle contracts when the regulatory light chain is phosphorylated by the enzyme myosin light chain kinase.
Because this enzyme is activated by calcium, the cytosolic calcium level indirectly regulates the extent of light chain
phosphorylation and hence contraction. The calcium dependent regulation of myosin light chain kinase activity
is mediated through the calcium binding protein calmodulin. Calcium first binds to calmodulin, and the calcium
calmodulin complex then binds to myosin light chain kinase and activates it. Because this regulation involves myosin,
it is known as thick filament regulation. Smooth muscle contraction also occurs in a calcium independent manner.
In this situation, Rho kinase activates the regulatory light chain by phosphorylation.

3.15.8 Intermediate filaments


Intermediate filaments are ropelike cytoplasmic filaments of about 10 nm diameter. These filaments are found
in many metazoans, including vertebrates, nematodes and molluscs but not in plants and fungi. Unlike the actin
and tubulin proteins, the intermediate filament proteins are chemically heterogenous and show species-specific
variations in molecular weight. More than 50 different intermediate filament proteins have been identified and
classified into six groups (mentioned below).
Cell Structure and Functions 313

The principal functions of intermediate filaments are structural to reinforce cells and to organize cells into tissues.
Unlike microfilaments and microtubules, intermediate filaments do not participate in cell motility. All intermediate
filaments share a common structural organization. The individual polypeptide of intermediate filament is an elongated
molecule consisting of a non-α-helical N-terminal head domain, a central α-helical rod domain and a non-α-helical
C-terminal tail domain. The central rod domain consists of long tandem repeats of a distinctive seven amino acid
sequence called the heptad repeat. Polypeptide chain forms a parallel coiled coil dimeric structure with another. Two
dimers then line up side by side to form an antiparallel tetramer of four polypeptide chains. Tetramer, the soluble
subunit of intermediate filament, further organizes to form higher level organization. Tetramer is analogous to
the αβ-tubulin heterodimer or G-actin. Unlike the actin or tubulin subunits, the intermediate filament subunits do
not contain a binding site for a nucleoside triphosphate. The antiparallel arrangement of dimers implies that the
tetramer, and hence the intermediate filament that it forms, is a non-polarized structure.

N C
Monomer
(Single intermediate filament protein)
a-helical rod domain

N C
Dimer
(Hetero or homodimer,
Parallel coiled coil structure) N Coiled-coil dimer C

N C C
N
Tetramer
(Staggered arrangement of dimer,
Lacks structural polarity) N C C
N
Staggered tetramer of two coiled-coil dimers

Protofilament
(Two tetramers packed together)

Two tetramers packed together

Intermediate filament
(about 10 nm diameter)

Figure 3.66 A model of intermediate filament construction.

Intermediate filament proteins are classified into six major types based on their sequences and tissue distribution:
Type Protein Site of expression
I Acidic keratins Epithelial cells
II Neutral or basic keratins Epithelial cells
III Vimentin Most widely distributed of all intermediate filament proteins is vimentin, which is
typically expressed in leukocytes, blood vessel endothelial cells, some epithelial
cells, and mesenchymal cells such as fibroblasts.
Desmin Muscle cells
Glial fibrillary acidic protein Glial cells
IV Neurofilament proteins Neurons
In mammals, three different neurofilament proteins have been recognized:
NF-L, NF-M and NF-H, for low, middle and high molecular weight, respectively.
V Nuclear lamins Most ubiquitous group of intermediate filaments are found exclusively in the nu-
cleus. Lamins form a network structure that lines the inside surface of the inner
nuclear membrane termed nuclear lamina.
VI Nestin Stem cells of central nervous system.
314 Cell Structure and Functions

3.16 Cell junctions


Many cells in tissues are linked to one another and to the extracellular matrix at specialized contact sites called cell
junctions. The cell junctions are critical to the development and functions of multicellular organisms. Cell junctions
can be classified into three functional groups: occluding junctions, anchoring junctions and communicating junctions.

Occluding junctions
Occluding junctions seal cells together in an epithelium in a way that prevents even small molecules from leaking
from one side of the sheet to the other (i.e. forms permeability barrier across epithelial cell sheets). These junctions
are of two types– tight junction and septate junction.
Tight junctions (or zonula occludens) are cell-cell occluding junctions mediated by two major transmembrane
proteins-claudins and occludin. Claudins and occludins associate with intracellular peripheral membrane proteins
called ZO proteins. Tight junctions make the closest contact between adjacent cells and prevent the free passage
of molecules (including ions) across an epithelial sheet in the spaces between cells. They also maintain the polarity
of epithelial cells by preventing the diffusion of molecules between the apical and the basolateral regions of the
plasma membrane. Septate junctions are the main occluding junctions in invertebrates.

Lumen

Tight
junction

Cell 1 Cell 2 Cell 3 Cell 4

Figure 3.67 Tight junctions allow cell sheets to serve as barriers to solute diffusion. Schematic drawing showing
how a small extracellular molecule present on one side of an epithelial cell sheet cannot traverse the tight junctions
that seal adjacent cells together.

Anchoring junctions
Anchoring junctions mechanically attach cells (and their cytoskeletons) to their neighbours or to the extracellular
matrix and perform the key task of holding cells together into tissues. It includes two main types of junctions -
adherens junction and desmosome.

Adherens junctions
Adherens junctions connect bundles of actin filaments from cell to cell or from cell to the extracellular matrix.
Adhesion belt (or zonula adherens): It is a cell to cell junction, mediated by actin filaments and proteins belonging
to the cadherin family. Adhesion belts are usually located near the apical surface, just below the tight junctions.
Focal contact (or adhesion plaque): It is a cell-matrix junction which is mediated by transmembrane adhesion
proteins of the integrin family and by actin filament.

Desmosomes
Desmosomes are buttonlike points of intercellular contacts which bond neighbouring cells together. It has a dense
cytoplasmic plaque which is composed of a mixture of intracellular attachment proteins, including plakoglobin and
Cell Structure and Functions 315

desmoplakins. The cytoplasmic plaque is responsible for connecting the cytoskeleton to the transmembrane linker
proteins of the cadherin family of cell-cell adhesion molecules. Desmosomes contain two specialized cadherin
proteins, desmoglein and desmocollin. Through extracellular domains, cadherins are responsible for holding the
adjacent membranes together. Each plaque is associated with a thick network of keratin intermediate filaments (in
most epithelial cells) and desmin intermediate filaments (in heart muscle cells), which are attached to the surface
of the plaque.
Hemidesmosomes, or half-desmosomes, resemble desmosomes, but instead of joining adjacent epithelial cell
membranes, they connect the basal surface of epithelial cells to the underlying basal lamina- a specialized mat
of extracellular matrix at the interface between the epithelium and connective tissue. The transmembrane linker
proteins in hemidesmosomes belong to the integrin family of extracellular matrix receptors, rather than to the
cadherin family of cell-cell adhesion proteins used in desmosomes.

Cell Keratin Cell


intermediate filaments Intermediate filaments

Plasma Plasma
membrane membrane
Cytoplasmic plaque Cytoplasmic plaque

Intercellular Cadherin Integrin


space

Cytoplasmic plaque

Plasma
membrane

Cell Extracellular matrix

A. Desmosomes B. Hemidesmosomes

Figure 3.68 A. Schematic drawing of a desmosome. On the cytoplasmic surface of each interacting plasma mem-
brane is a cytoplasmic plaque. Each plaque is associated with a network of keratin intermediate filaments. Transmem-
brane adhesion proteins, which belong to the cadherin family of cell-cell adhesion molecules, bind to the plaques and
interact through their extracellular domains to hold the adjacent membranes together. B. Schematic drawing of a
hemidesmosome, joining adjacent epithelial cell membranes to the underlying basal lamina.

Table 3.15 Characteristics of anchoring junctions

Junction type Transmembrane protein Intracellular linkage

Zonula adherens Cadherin Actin filaments

Focal contact Integrin Actin filaments

Desmosome Cadherin Intermediate filaments

Hemi-desmosome Integrin Intermediate filaments

Communicating junctions
Communicating junctions mediate the passage of chemical or electrical signals from one interacting cell to its partner.

Gap junction
Gap junctions serve as direct connections between the cytoplasms of adjacent cells. They act as channels, which
allow inorganic ions and other small water-soluble molecules up to 1000 Da to pass directly from the cytoplasm of
one cell to the cytoplasm of the other, thereby coupling the cells both electrically and metabolically. In electrically
This page intentionally left blank.
Cell Structure and Functions 317

3.17 Cell adhesion molecules


Cells adhere to each other and to the extracellular matrix through cell surface proteins called cell adhesion molecules
(CAMs). CAMs include transmembrane proteins that fall into two categories of cell-cell adhesion molecules or cell-
matrix adhesion molecules.
Cadherins (Calcium adhering) are cell-cell adhesion molecules. Most cadherins are single-pass transmembrane
glycoprotein composed of about 700–750 amino acid residues. They are present in the plasma membrane in
the form of dimers or oligomers. The large extracellular part of the polypeptide chain is usually folded into five
extracellular cadherin (EC) domains, each containing about 100 amino acid residues. The cadherins are calcium-
binding proteins which hold cells together by homophilic interactions (molecules on one cell bind to other molecules
of the same kind on adjacent cell). The extracellular domains of cadherins mediate homophilic cell-cell adhesion
and the cytoplasmic tails of typical cadherins, including all classical and some nonclassical ones interact with the
cytoskeleton (actin or intermediate filaments) via a number of intracellular anchor or adaptor proteins (such as
β-catenin, α-catenin, vinculin and plakoglobin).

N N

2+
Ca

Extracellular

Plasma
membrane {
Cytosol
C C
Cadherin dimer

Figure 3.71 A typical classical cadherin molecule. The principle feature common to all cadherins is the presence of
extracellular cadherin repeats (EC domains) in the extracellular region of a membrane protein. There are Ca2+-binding
sites between each pair of repeats.

Members of cadherin superfamily

Classical cadherins
Type I cadherins: E-cadherin, N-cadherin, R-cadherin
Type II cadherins: VE-cadherin

Non-classical cadherins
Desmosomal cadherins: Desmocollin and Desmoglein
Atypical cadherins: T-cadherin, LI-cadherin
Proto-cadherins: Pcdhα, Pcdhβ, and Pcdhγ

The type I and type II classical cadherins were originally named on the basis of the tissues within which they were
first identified e.g. epithelial (E)-cadherin, neural (N)-cadherin and vascular endothelial (VE)-cadherin. Desmosomal
cadherins such as desmocollins and desmogleins are components of desmosomes, whose extracellular cadherin
repeats are responsible for adhesion and intracellular regions interact with intermediate filaments via desmosomal
This page intentionally left blank.
Cell Structure and Functions 319

• Structural proteins such as collagens and elastins.


The major structural protein of the extracellular matrix is collagen, which is the single most abundant protein
in animal tissues. Connective tissues also contain elastic fibers, composed principally of a protein called elastin
(collagens and elastins are discussed in chapter 1 - Biomolecules and Catalysis).

• Proteoglycan such as heparan sulfate, chondroitin sulfate.


Polysaccharide glycosaminoglycans, which are usually found covalently linked to protein in the form of
proteoglycans. Proteoglycans differ from glycoproteins with respect to the nature, quantity, and arrangement
of their sugar side chains. Whereas glycoproteins contain 1-60% carbohydrate by weight, proteoglycans can
contain as much as 95% carbohydrate by weight, mostly in the form of long, unbranched GAG chains.
The fibrous structural proteins of the extracellular matrix are embedded in gels formed from polysaccharides
called glycosaminoglycans, or GAGs, which consist of repeating units of disaccharides. One sugar of the
disaccharide is either N-acetylglucosamine or N-acetylgalactosamine, which in most cases is sulfated and the
second is usually acidic (either glucuronic acid or iduronic acid) sugar. GAGs are highly negatively charged.
There are four main groups of GAGs : hyaluronan, chondroitin sulfate and dermatan sulfate, heparan sulfate
and heparin, and keratan sulfate. Hyaluronan is the simplest of the GAGs. Except hyaluronan, all of the other
GAGs are linked to proteins in the form of proteoglycans. The polysaccharide chains are mainly assembled on
the core protein in the Golgi apparatus before delivery to the exterior of the cell by exocytosis.

• Adhesion proteins such as laminins and fibronectins.


Adhesion proteins, the third class of extracellular matrix constituents, are responsible for linking the components
of the matrix both to one another and to the surface of cells.

Laminins are extracellular heterotrimeric matrix glycoproteins composed of three polypeptide chains (α, β
and γ) that are disulfide-bonded. Each of the polypeptide chains is more than 1500 amino acid residues long.
The α-chain of laminin contains globular LG domain which mediates calcium dependent binding to specific
carbohydrates. Laminins play the central role in organizing and establishing the basal lamina (formerly called
basement membrane). Their primary role is in cell-matrix attachment, but many additional biological activities,
including promoting cell growth and migration, tumour growth, nerve regeneration and wound repair have
been demonstrated.

Fibronectin is an extracellular matrix glycoprotein, present in all vertebrates. It exists as a dimer, consisting
of two nearly identical monomers linked by a pair of disulfide bonds. Both are encoded by a single gene but
differences appear due to alternative splicing. Each chain is about 2500 amino acids long and is folded into
five or six domains. Each domain is specialized for binding to a particular molecule or to a cell. Each chain
consists of three types of repeating units (termed FN repeats): FN I (12 copies), FN II (2 copies) and FN III
(15 copies). FN I repeating unit is about 40 amino-acid residues in length and contain two disulfide bonds;
FN II repeating unit is approximately 60 amino acid residues long and with two intrachain disulfide bonds; and
FN III repeating unit is about 90 amino acid residues long and without any disulfide bonds.
Fibronectins help attach cells to the extracellular matrix by binding to ECM components (particularly collagens),
proteoglycans and to cell surface adhesion molecules such as integrins. A specific Arg-Gly-Asp tripeptide sequence
(termed as RGD sequence) present on type III fibronectin repeat is required for integrin binding and mediates
cell adhesion. Fibronectin plays a major role in cell adhesion, growth, migration and differentiation. Fibronectins
are of two types– soluble plasma fibronectin and insoluble cellular fibronectin. Soluble plasma fibronectin is
a major protein component of blood plasma and is produced in the liver by hepatocytes. Insoluble cellular
fibronectin is secreted by various cells, particularly fibroblast, as soluble protein dimer and then assembled
into an insoluble matrix.
This page intentionally left blank.
322 Cell Structure and Functions
Middle lamella
A thin layer of material, the middle lamella, is present at the junction, where the walls of neighboring cells come
into contact. It acts as cementing material. The composition of the middle lamella differs from the rest of the wall.
It is high in pectin (as calcium pectate) and may be complexed with hydroxyproline-rich glycoproteins.

3.20 Cell signaling


All cells receive and respond to signals from their surroundings. This is accomplished by a variety of signal
molecules that are secreted or expressed on the surface of one cell and bind to receptors expressed by other cells,
thereby integrating and coordinating the functions of the many individual cells that make up organisms. Each cell
is programmed to respond to specific extracellular signal molecules. Extracellular signaling usually involves the
following steps:
1. Synthesis and release of the signaling molecule by the signaling cell;
2. Transport of the signal to the target cell;
3. Binding of the signal by a specific receptor leading to its activation;
4. Initiation of signal-transduction pathways.

In animals, extracellular signaling by signal molecules can be classified into four categories—endocrine, paracrine,
autocrine and juxtacrine signaling.
In endocrine signaling, the signaling molecules act on target cells distantly located from their site of synthesis. It
is a long-range signaling in which signal molecule is transported by the blood stream.
In paracrine signaling, the signaling molecules released by a cell affect target cells only in close proximity. An
example of this is the action of neurotransmitters in carrying signals between nerve cells at a synapse.

Endocrine signaling

Bloodstream

Signal
molecule Target cell
Paracrine signaling

Target cell

Autocrine signaling

Figure 3.74 Long-range signaling between cells is called endocrine when the signal molecule is transported by the
bloodstream (typical for hormones), paracrine when the signal diffuses between neighboring cells across the extra-
cellular matrix (typical for neurotransmitters and many so-called tissue hormones or local mediators), and autocrine
when the signal re-acts on the transmitter cell.
Cell Structure and Functions 323

In autocrine signaling, the signaling molecules produce an effect on same cell that produces it. One important
example of such is the response of cells of the vertebrate immune system to foreign antigens. Certain types of
T-lymphocytes respond to antigenic stimulation by synthesizing a growth factor that drives their own proliferation,
thereby increasing the number of responsive T-lymphocytes and amplifying the immune response.
In juxtacrine signaling, signal molecules do not diffuse from the cell producing it and cell bearing signal molecules
interact with receptor proteins of adjacent responding cells. Unlike other modes of cell signaling, juxtacrine signaling
requires physical contact between the cells involved. Notch signalling and classical cadherin signalling are examples
of juxtacrine signaling.

3.20.1 Signal molecules


Signal molecules are chemically heterogenous compounds. These molecules are divided into two categories –
membrane bound and secretory signal molecules. Membrane bound signal molecules remain bound to the surface
of the cells and mediate contact dependent signaling. In most cases, signal molecules are secreted by signaling
cells. Secreted extracellular signal molecules are further divided into three general categories based on the distance
over which signals are transmitted: endocrine, paracrine and autocrine signal molecules.
Extracellular signal molecules are synthesized and released by signaling cells and produce a specific response
only in target cells that have either cell surface receptors or intracellular receptors for the signaling molecules.
Extracellular signal molecules fall into two broad categories - small lipophilic molecules that diffuse across the
plasma membrane and interact with intracellular receptors; and hydrophilic molecules that bind to cell-surface
receptors. Few lipophilic signal molecules bind to cell-surface receptors also. Most of these molecules are members
of eicosanoids, which include prostaglandins, prostacyclin, thromboxanes and leukotrienes. All eicosanoids are
synthesized from arachidonic acid, which is formed from phospholipids. However, most of the extracellular signal
molecules are hydrophilic and bind to the cell surface receptors of the target cell.

Examples of signal molecules that interact with cell surface receptor


Epinephrine, Non-epinephrine, Glucagons, Insulin, Gastrin, Secretin, Cholecystokinin and ACTH

Examples of signal molecules (hormones) that interact with cytosolic or nuclear receptor
Steroid hormones (Progesterone, Estradiol, Testosterone, Cortisol, Corticosterone, Aldosterone),
Steroid like hormone (α-ecdysone) and
Non-steroid hormones (Thyroid hormone and Retinoic acid).

Binding of extracellular signaling molecules to the cell surface receptor leads to increase (or decrease) in concentration
of low molecular weight intracellular signaling molecules termed secondary messengers. These low-molecular-weight
signaling molecules include cAMP, cGMP, diacylglycerol (DAG); inositol 1,4,5-trisphosphate (IP3), phosphoinositides
and calcium.

3.20.2 Receptors
The cellular response to a particular extracellular signal molecule depends on its binding to a specific receptor located
on the surface of a target cell or in its nucleus or cytosol. Receptors are chemically protein or glycoprotein molecules
which bind to signaling molecules (termed ligand). Binding of a ligand to its receptor causes a conformational change
in the receptor that initiates a sequence of reactions leading to a specific cellular response. Based on location,
receptors are classified into two broad categories - intracellular receptors and cell-surface receptors.

Intracellular receptors
Intracellular receptor proteins are located in the cytosol or the nucleus. These include receptors for steroid hormones,
thyroid hormones, retinoids and vitamin D as well as different “orphan” receptors. The intracellular receptors are all
structurally related and belong to the nuclear receptor superfamily. Within the cell, intracellular receptor – ligand
This page intentionally left blank.
Cell Structure and Functions 331

Exterior

PLC-b PIP2 DAG DAG


a b g +
Receptor protein GTP PKC
IP3
(GPCR) Gq-protein
(activated)
Binds
Open IP3-gated
2+
Ca -release channel
ER 2+
Ca

Figure 3.81 The activated receptor binds to a specific trimeric G protein (Gq), causing the α-subunit to dissociate
and activate phospholipase C-β. Two intracellular mediators are produced when PIP2 is hydrolyzed by phospholipase
C-β: IP3, which diffuses through the cytosol and releases Ca2+ from the ER, and DAG, which remains in the membrane
and helps activate the enzyme protein kinase C.

Calcium-Calmodulin complex
Many of the effects of calcium ion are mediated by the protein calmodulin, which is activated by calcium ion binding
when the concentration of cytosolic calcium ion increases. Calmodulin (17 KDa protein) is a small, highly conserved
calcium binding cytosolic acidic protein found in all eukaryotic cells. It binds up to four calcium ions and acts as
an important intracellular receptor for regulatory calcium signals. Calcium ion binds to calmodulin in a cooperative
fashion. Thus a small change in the level of cytosolic calcium ion leads to a large change in the level of active
calmodulin. As it binds calcium, calmodulin undergoes conformational changes which can increase its affinity for
target proteins. It acts both directly, through interaction with key target enzymes, and indirectly, via specific kinases.
Many effects of calcium, however, are more indirect and are mediated by protein phosphorylations catalyzed by
a family of serine-threonine protein kinases called calcium-calmodulin-dependent kinases (CaM-kinases). Some
CaM-kinases phosphorylate gene regulatory proteins, such as the CREB protein and in this way activate or inhibit
the transcription of specific genes. One of the best-studied CaM-kinases is CaM-kinase II, which is found in most
animal cells but is especially enriched in the nervous system. Calmodulin participates in the regulation of several
biological processes including energy and biosynthetic metabolism, cell motility, exocytosis, cytoskeletal assembly
and intracellular modulation of both cAMP and calcium concentrations.

Mode of action of cholera and pertussis toxins


Cholera toxin is an enterotoxin secreted by the bacterium Vibrio cholerae, which is unusual in having two circular
chromosomes rather than one. Genes for cholera toxin are present on the integrated phage genome, CTXφ. The
cholera toxin is an oligomeric complex made up of six protein subunits: a single copy of the A-subunit and five copies
of the B-subunit. The A-subunit has catalytic property that ADP-ribosylates G-proteins. The pentameric protein binds
to the ganglioside GM1 present on the surface of the intestinal epithelium. The A-subunit ribosylates the Arg residue
of the α-subunit of Gs protein. ADP ribose is provided by the intracellular NAD+. This ADP ribosylation alters the
A-subunit so that it can no longer hydrolyze its bound GTP, causing it to remain in an active state that stimulates
adenylyl cyclase indefinitely. The resulting prolonged elevation in cyclic AMP levels within intestinal epithelial cells
causes activation of PKA. PKA phosphorylates the CFTR and Na+-H+ exchanger present in the intestinal epithelial
cells. This causes a large efflux of chloride and sodium ion and water into the gut, thereby causing the severe
diarrhea that characterizes cholera.
Pertussis toxin, an exotoxin that causes pertussis (whooping cough), is secreted by Bordetella pertussis.
B. pertussis colonizes the respiratory tract, where it destroys the ciliated epithelial cells that normally sweep away
mucus. Without this ciliary action, vigorous coughing is needed to clear the tract. Pertussis toxin catalyzes the ADP
This page intentionally left blank.
Cell Structure and Functions 343

H D

Sensor histidine kinase Response regulator

Figure 3.91 Two component system.

3.20.8 Chemotaxis in bacteria


Movement of organisms in specific directions in response to chemical stimulus is called chemotaxis. Chemotaxis in
bacteria depends on a signaling pathway that terminates at the flagellar motor.
Bacterial flagellar motion is rotatory in nature. Membrane proteins MotA and MotB along with FliG create a proton
channel that drives the rotation of the flagellum. The rotation of flagella can be clockwise or counterclockwise.
When the flagella rotate clockwise, forward motion ceases and the cells tumble. But counterclockwise rotations of
flagella impart forward motion (run).
In the absence of a chemical gradient, bacteria such as E. coli move in a random fashion that includes runs, where
the cell is swimming forward and tumbles when the cell stops. Following a tumble, the direction of the next run
is random. However, if a gradient of a chemical attractant is present, these random movements become biased.
As the bacterium senses that it is moving toward higher concentrations of the attractant, runs become longer and
tumbles less frequent. If the bacterium is sensing a repellent, the same general mechanism applies, although in
this case it is the decrease in concentration of the repellent that promotes runs.

Flagella
(clockwise rotation)

Figure 3.92 The two-component signaling pathway that enables chemotaxis receptors to control the flagellar motor
during bacterial chemotaxis.

Bacterial chemotaxis is mediated by a transmembrane receptors and a phosphorylation relay system. It depends on
a two component signaling pathway activated by histidine kinase associated receptors. The chemotaxis receptors
are methylated during adaptation and so are also called Methyl-accepting Chemotaxis Proteins (MCPs). The
phosphorylation relay system enables the chemotaxis receptors to control the flagellar motor. In E. coli, binding of a
repellent increases the activity of the receptor, which binds CheW (adaptor protein) and CheA (sensor protein acts as
histidine kinase), thereby stimulating CheA to phosphorylate itself on histidine. CheA quickly transfers its covalently
bound, high-energy phosphate directly to aspartate of CheY (response regulator) to generate CheY-phosphate. The
phosphorylated CheY dissociates from the receptor, diffuses through the cytosol and binds to the flagellar motor and
344 Cell Structure and Functions
causes it to rotate clockwise, resulting in tumbling. Phosphorylated state of CheY remains only for a few seconds.
Protein CheZ accelerates the dephosphorylation of CheY-phosphate, thereby inactivating it. The binding of an
attractant has the opposite effect. It inactivates the receptor and, therefore, decreases the phosphorylation of CheA
and CheY, which results in counterclockwise flagellar rotation and run. Each of the phosphorylated intermediates
decays in about 10 seconds, enabling the bacterium to respond very quickly to changes in its environment. The
response to an increase in the concentration of an attractant or repellent is only transient, even if the higher level
of ligand is maintained, as the bacteria desensitize, or adapt, to the increased stimulus. The adaptation is mediated
by the covalent methylation. A methyltransferase, CheR, catalyzes methylation of the MCP. [In other species of
bacteria such as B. subtilis, attractants may stimulate and repellents inhibit CheA activity].

Repellent CheA Attractant

CheY CheYP

CheZ

Flagellum Counterclockwise rotation Clockwise rotation


Sensor Degree of methylation increases Degree of methylation decreases
Behavior Run Tumbling

Chemotaxis proteins
1. CheA, a cytoplasmic sensor kinase.
2. CheW, an adaptor protein linking the sensor protein with CheA.
3. CheY, the response regulator controlling the flageller motor.
4. CheZ, an Asp–specific protein phosphatase for signal termination.
5. CheR, a methyltransferase catalyzing methylation of the MCP.

3.20.9 Quorum sensing


The term quorum sensing describes a bacterial communication phenomenon that allows bacteria to communicate
using secreted signal molecules to assess their population density. This process enables a population of bacteria to
collectively regulate gene expression and, therefore, behaviour. In quorum sensing, bacteria assess their population
density by detecting the concentration of a particular signal molecule termed autoinducer, which is correlated with
cell density.
Quorum sensing is the regulation of gene expression in response to fluctuations in cell-population density. Bacteria
that use quorum sensing constantly produce and secrete certain signaling molecules (called autoinducers). These
bacteria also have a receptor for the autoinducer. When the inducer binds to the receptor, it activates the transcription
of a set of genes, including those responsible for the synthesis of the autoinducer itself. The concentration of the
autoinducer in the surrounding medium depends on cell-population density. As the bacterial population grows, the
concentration of the autoinducer in the surroundings increases, causing more autoinducer molecules to be synthesized.
The detection of a minimal threshold stimulatory concentration of an autoinducer leads to an alteration in gene
expression. Both gram-positive and gram-negative bacteria use quorum sensing communication circuits to regulate
a diverse array of physiological activities. These processes include symbiosis, virulence, competence, conjugation,
antibiotic production, motility and sporulation. In general, Gram-negative bacteria use N-Acyl-L-Homoserine Lactones
(AHLs) as autoinducers, and Gram-positive bacteria use processed oligo-peptides to communicate.
Cell Structure and Functions 345

Sensor
LuxR LuxR

RR Response regulator
Ligand binding
Phosphorylation

P P
LuxR LuxR RR RR

DNA DNA

Gene expression Gene expression

Gram-negative bacteria Gram-positive bacteria

Figure 3.93 Mechanisms of action of auto-inducers: two major pathways along which extracellular signals control
gene transcription. Depending on the chemical structure, auto-inducers (black dots) activate transcription factors
either by direct interaction or via a receptor-mediated cascade. In contrast to AHL as auto-inducer in gram negative
bacteria, the auto-inducers of gram positive species are unable to penetrate cellular membranes. Instead their effects
are mediated by two component system controlling the transcription of corresponding genes.

Examples
The first incidence of this biological phenomenon came to light with the discovery of luminescence produced
by marine bacteria Vibrio fischeri. These bacteria, when free-living in sea water (i.e. at a low cell density) are
non-luminescent. However, when grown to high cell densities, they bioluminesces with a blue-green light. This
bacterium commonly forms symbiotic relationships with some fishes and squid species. These marine organisms
carry a specialized organ called the light organ, in which bacteria luminescent appearance in dark environments
is due to the maintenance of a high-density V. fischeri population in the light organ. In the marine environment,
the bacteria only luminesce when colonising the light organs and do not emit light when in the free-living state.
Research on how V. fischeri regulates bioluminescence led to the discovery of bacterial quorum sensing via N-Acyl-
L-Homoserine Lactones (AHLs).
Synthesis of lipophilic AHL is catalyzed by an enzyme called ALH synthase, the product of the luxI gene. The luxI
gene is subject to positive autoregulation i.e. transcription of luxI increases as AHL accumulates in the cell. This
is accomplished through a transcriptional activator, LuxR, which is active only when it binds AHL. Thus, without
AHL-activated LuxR, the luxI gene will be transcribed only at basal level. AHL freely diffuses out of the cell and
accumulates in the environment. When cell density increases, the concentration of AHL also increases. Greater
concentration of AHL flows back into the cell and activates high level transcription of luxI and other genes whose
products are needed for bioluminescence.

3.20.10 Scatchard plot


The Scatchard plot is a graphical method of analyzing equilibrium ligand-binding data. It is used to determine the
number of ligand-binding sites on a receptor, whether these sites show cooperative interactions, whether more
than one class of site exists, and the respective affinities of each site.
Ligand binds specifically and non-covalently with the receptor. A receptor may possess one binding site per molecule
or more than one independent binding sites. Receptor-ligand binding is described by the equation;

ˆˆˆˆ
†
R (Receptor) + L (Ligand) ‡ˆˆˆ
ˆ R - L (Receptor - Ligand complex)
This page intentionally left blank.
Cell Structure and Functions 367

Problem

An animal cell enters in the process of cell division. Name the drugs affecting the following steps.
a. a drug that inhibits microtubule formation.
b. a drug that allows microtubules to form but prevents them from shortening.
c. a drug that inhibits cytokinesis.

Solution

a. Colchicine; b. Taxol; c. Cytochalasin

Syncytium and coenocyte


In some organisms, karyokinesis is not followed by cytokinesis as a result of which multinucleate condition arises
leading to the formation of a coenocyte. In contrast to a coenocyte, a syncytium is a multinucleate cell which forms
as a result of fusions of multiple uninuclear cells (i.e. cells with a single nucleus). A classic example of a syncytium
is the formation of skeletal muscle. In animals, well-studied examples of the coenocytic state are found in the
blastoderm of Drosophila. However, the word syncytium is used incorrectly to describe it.

3.22.2 Meiosis
Meiosis (term coined by Farmer and Moore) is a specialized form of cell division in which number of chromosomes is
reduced to half. The reduction in chromosome number is achieved by one round of DNA replication being followed
by two rounds of chromosome segregation with no intervening round of DNA replication.

n and C
n and 2C

G0 S

Meiosis I
G1 G2 Meiosis II
2n and 2C 2n and 4C

Diploid cell 2n and 4C


2n and 2C
Interphase n and 2C

Figure 3.117 Change in chromosome number and amount of DNA during meiosis I and meiosis II.

Meiosis is divided into two parts– meiosis I and meiosis II. At the end of the meiotic process, there are four daughter
cells rather than the two produced at the end of the mitotic process. Each of the resulting daughter cells has one
half of the number of chromosomes as the parent cell.

Meiosis I
Prophase I of meiosis I is divided into five sub-stages: Leptotene, Zygotene, Pachytene, Diplotene and Diakinesis.
Leptotene: Prophase I begins at the leptotene stage. Although each chromosome has replicated and consists of
two sister chromatids, these chromatids are unusually closely apposed, and each chromosome therefore appears
to be single (separate chromatids will not become visible until late in prophase, at either the diplotene stage or
in diakinesis). At this stage, chromosome ends are attached to the inner nuclear envelope. In the leptotene to
zygotene transition, the tips of the chromosomes move until most end up in a limited region near each other. This
forms of arrangement is called a bouquet stage. Bouquet formation directly facilitates homologous chromosome
pairing and synapsis.
This page intentionally left blank.
Cell Structure and Functions 371

3.22.3 Nondisjunction and aneuploidy


During mitosis, each chromosome replicates and the two sister chromatids separate, with one going to each
daughter cell. Similarly, during meiosis, the two homologous chromosomes separate during the first division, and
then the two sister chromatids move to separate cells during the second division. The result of a normal mitosis
or meiosis is that all the progeny cells receive complete sets of chromosomes. Occasionally, however, homologous
chromosomes and sister chromatids fail to separate or disjoin properly during mitosis or meiosis. The failure of
chromosomes to separate properly during mitosis or meiosis is called nondisjunction. Nondisjunction produces cells
or gametes with extra or missing chromosomes; it is the primary cause of the aneuploidy.
During meiosis, non-disjunction can occur during meiosis I or meiosis II. In meiosis I, when non-disjunction of
homologous chromosome occurs, both homologs move towards the same pole. In meiosis II, non-disjunction of
sister chromatids occurs with both chromatids going to the same pole. Depending on where in meiosis this non-
disjunction occurs, some gametes will end up with one or more extra chromosomes and the other will end up with
fewer chromosomes than a complete set.

A. Non-disjunction of homologous chromosomes in meiosis I

Normal
n–1
meiosis II

G0 Non-disjunction
in meiosis I n–1

Normal
meiosis II n+1
Cell
2n and 2C

n+1

B. Non-disjunction of sister chromatids in meiosis II

Non-disjunction
in meiosis II n+1

G0
Normal n–1
meiosis I

Normal
meiosis II n
Cell
2n and 2C

Figure 3.121 Mechanisms of formation of aneuploid gametes during meiosis. A. Non-disjunction of homologous
chromosomes in meiosis I and B. Non-disjunction of sister chromatids in meiosis II.
This page intentionally left blank.
Cell Structure and Functions 373

Stem cells
Stem cells are unspecialized (undifferentiated) cells that have the ability to differentiate into other cells and self-
regenerate. These cells divide to produce one daughter cell that remains a stem cell and one that divides and
differentiates. Because the division of stem cells produces new stem cells as well as differentiated daughter cells, stem
cells are self renewing populations of cells that can serve as a source for the production of differentiated cells throughout
life. Typically, stem cells generate an intermediate cell type or types before they achieve their fully differentiated state.
The intermediate cell is called a precursor or progenitor cell. The ability to differentiate is the potential to develop into
other cell types. Depending on the ability to differentiate into other cell types, stem cells can be classified as totipotent,
pluripotent and multipotent stem cells. Totipotent stem cells are cells that can give rise to a fully functional organism
as well as to every cell type of the body. Pluripotent stem cells can differentiate into nearly all cell types. Multipotent
stem cells can differentiate into a limited number of closely related families of cells.

Totipotent stem cell


These cells have unlimited capability, and
have the ability to form extraembryonic
membranes and tissues, the embryo itself,
and all postembryonic tissues and organs.

Pluripotent stem cell


These cells are capable of giving rise to
most, but not all, tissues of an organism.
An example is inner mass cells.

Multipotent stem cell


These cells are committed to give rise to
cells that have a specific function.
An example is blood stem cell.
Blood stem Other committed
cells stem cells

RBCs
WBCs
Platelets

There are two broad types of stem cells: embryonic stem cells, which are isolated from the inner cell mass of blastocysts,
and adult stem cells, which are found in various tissues. Embryonic stem cells can become all cell types of the body
because they are pluripotent. An adult stem cell (also termed as somatic stem cell) is an undifferentiated cell found
among differentiated cells in a tissue or organ, can renew itself and differentiate to yield the major specialized cell
types of the tissue or organ. The primary roles of adult stem cells in a living organism are to maintain and repair the
tissue in which they are found. Unlike embryonic stem cells, which are defined by their origin (the inner cell mass of
the blastocyst), the origin of adult stem cells in mature tissues is unknown. Most adult stem cells are multipotent. The
bone marrow contains two kinds of stem cells. One population, called hematopoietic stem cells, forms all the types
of blood cells in the body. A second population called bone marrow stromal cells generates bone, cartilage, fat and
fibrous connective tissue. The adult brain also contains stem cells that are able to generate the brain’s three major
cell types—astrocytes and oligodendrocytes, which are non-neuronal cells and neurons or nerve cells.
374 Cell Structure and Functions

3.23 Apoptosis
Apoptosis (from the Greek words apo = from and ptosis = falling,) is an energy dependent biochemical mechanism of
programmed cell death. It is a genetically programmed process occurs normally during embryogenesis, metamorphosis
and aging. For example, the differentiation of human fingers in a developing embryo requires the cells between the
fingers to initiate apoptosis so that the fingers can separate. Apoptosis also occurs as a defense mechanism such
as in immune reactions or when cells are damaged by disease or noxious agents. Although apoptosis is the most
common form of programmed cell death (PCD), there are several non-apoptotic programmed cell death such as
autophagy and necroptosis have also been reported.
The demise of cells by apoptosis is marked by a well-defined sequence of morphological changes. Apoptotic cells
become more compact, blebbing occur at the membranes, chromatin becomes condensed and DNA is fragmented.
During the early stage of apoptosis, cell shrinkage and pyknosis (i.e. chromatin condensation) occur. With cell
shrinkage, the cells becomes smaller in size with dense cytoplasm. Pyknosis is the most characteristic feature of
apoptosis. Later, extensive plasma membrane blebbing occurs and separation of cell fragments occurs in the form
of small membrane-bound apoptotic bodies by a process called budding. Apoptotic bodies consist of cytoplasm
with tightly packed organelles with or without a nuclear fragment. These bodies are subsequently phagocytosed by
macrophages or surrounding cells. Chemical changes in the surface of apoptotic cells or bodies allow the surrounding
cells or macrophages to recognize and engulf them. An especially important change occurs in the plasma membrane
of apoptotic cells. The negatively charged phospholipid phosphatidylserine is normally exclusively located in the
inner leaflet of the lipid bilayer of the plasma membrane, but it flips to the outer leaflet in apoptotic cells, where it
can serve as a marker of these cells. There is essentially no inflammatory reaction associated with the process of
apoptosis nor with the removal of apoptotic cells because:
1. apoptotic cells do not release their cellular constituents into the surrounding interstitial tissue;
2. they are quickly phagocytosed by surrounding cells and,
3. the engulfing cells do not produce anti-inflammatory cytokines.

Apoptosis versus necrosis

Necrosis refers to the degradative processes that occur after cell death. It is not a mechanism of cell death. The
process that leads to necrosis is called oncosis. In contrast to necrosis, which is a form of an energy independent
cell death that results from acute tissue injury, apoptosis is carried out in an ordered process that generally confers
advantages during an organism’s life cycle.

Apoptosis Necrosis

Cell shrinkage and convolution Cell swelling

Pyknosis and karyorrhexis Karyolysis, pyknosis and karyorrhexis

Intact cell membrane Disrupted cell membrane

Cytoplasm retained in apoptotic bodies Cytoplasm released

No inflammation Inflammation usually present

Pyknosis (or karyopyknosis) is the irreversible condensation of chromatin in the nucleus of a cell undergoing necrosis
or apoptosis. Karyorrhexis is the nuclear fragmentation and karyolysis is the complete dissolution of the chromatins.

Mechanisms of apoptosis

The mechanisms of apoptosis are highly complex and regulated, involving an energy-dependent cascade of molecular
events. There are multiple apoptotic pathways. These pathways are both caspase-dependent as well as caspase-
This page intentionally left blank.
Chapter 04

Prokaryotes and Viruses

4.1 General features of Prokaryotes


Prokaryotes (pro means before and karyon means kernel or nucleus) consist of eubacteria and the archaea (also
termed as archaebacteria or archaeobacteria). The term eubacteria refer specifically to bacteria. The informal
name bacteria are occasionally used loosely in the literature to refer to all the prokaryotes, and care should be
taken to interpret its meaning in any particular context. Prokaryotes can be distinguished from eukaryotes in terms
of their cell structure and molecular make-up. Prokaryotic cells have a simpler internal structure than eukaryotic
cells. Although many structures are common to both cell types, some are unique to prokaryotes. Most prokaryotes
lack extensive, complex, internal membrane systems. The major distinguishing characteristics of prokaryotes and
eukaryotes are as follows:

Features of prokaryotic organisms


True membrane bound nucleus – Absent
DNA complexed with histone – Absent
Number of chromosomes – One (mostly)
Mitosis and meiosis – Absent
Genetic recombination – Partial (unidirectional transfer of DNA)
Sterol in plasma membrane – Absent (Except Mycoplasma)
Ribosome – 70S
Unit membrane bound organelles – Absent
Cell wall – Present in most of prokaryotic cells. In eubacteria, it is made up of
peptidoglycan.

Features of eukaryotic organisms


True membrane bound nucleus – Present
DNA complexed with histone – Present
Number of chromosomes – More than one
Mitosis and meiosis – Present
Genetic recombination – By crossing over during meiosis
Sterol in the plasma membrane – Present
Ribosome – 80S (in cytosol) and 70S (in organelles)
Unit membrane bound organelles – Present
Cell wall – Made up of cellulose in plant and chitin in fungi. Absent in animal cells.
392 Prokaryotes and Viruses
Prokaryotic cells show similarities with eukaryotic organelles like mitochondria and chloroplast. The endosymbiotic
theory (Margulis, 1993) proposes that the mitochondria and chloroplasts of eukaryotic cells originated as symbiotic
prokaryotic cells. The presence of circular, covalently closed DNA and 70S ribosomes in mitochondria and chloroplast
support this theory.

Table 4.1 Similarities between prokaryotic cells and eukaryotic organelles

Prokaryotic cells Eukaryotic organelles

Nature of DNA ds circular ds circular

Histone protein Absent Absent

Ribosome type 70S 70S

Growth Binary fission Binary fission

4.2 Phylogenetic overview


Historically, prokaryotes were classified on the basis of their phenotypic characteristics. Prokaryotic taxonomy therefore
involved measuring a large number of characteristics, including morphology and biochemical characteristics (e.g.
ability to grow on different substrates, cell wall structure, antibiotic sensitivities, and many others). This contrasts
with the classification of eukaryotic organisms, for which phylogenetic (evolution-based) classification was possible
through the availability of fossil evidence.
A major revolution occurred with the realization that evolutionary relationships could be deduced on the basis of
differences in gene sequence. The most important gene for prokaryote phylogeny is the 16S ribosomal RNA (rRNA)
gene, which is present in all cells. The gene is approximately 1500 bp in length and possesses signature sequences.
These sequences are conserved and found in the organisms of one taxonomic group but not in other groups.

Bacteria Archaea Eukaryotes

Green
filamentous Entamoebae Slime Animals
Spirochetes bacteria molds
Gram Methanosarcina Fungi
positive
Methanobacterium Halophiles
Proteobacteria Plants
Methanococcus
Cyanobacteria Ciliates
T. Celer
Planctomyces Thermoproteus Flagellates
Pyrodicticum
Cytophaga Trichomonads
Microsporidia
Thermotoga
Diplomonads
Aquifex

Phylogenetic tree of life

Figure 4.1 A phylogenetic tree of living things, based on RNA data (proposed by Carl Woese), showing the separa-
tion of bacteria, archaea, and eukaryotes from a common ancestor.

Based on ribosomal RNA signature sequences, Carl Woese proposed a radical reorganization of the five kingdoms
into three domains. In his classification system, Woese placed all four eukaryotic kingdoms (protista, fungi, plantae,
animalia) into a single domain called Eukarya, also known as the eukaryotes. He then split the former kingdom
of Monera into the Eubacteria and the Archaea domains. Unlike Whittaker’s five kingdom system, Woese’s three
domain system organizes biodiversity by evolutionary relationships.
This page intentionally left blank.
394 Prokaryotes and Viruses
negatively or positively charged at neutral pH. More accurately, dyes can be referred to as anionic or cationic. Cationic
or basic dyes (such as crystal violet, methylene blue, safranin, malachite green) will react with groups that have a
negative charge. Anionic or acidic dyes (such as eosin, acid fuchsin) will react with groups that have a positive charge.
Bacterial staining protocols can be divided into three basic types – simple, differential, and specialized. Simple
stains react uniformly with all cell types and only distinguish the organisms from their surroundings. Differential
stains do not stain all types of cells with the same color. It discriminates different cell types depending upon the
chemical or physical composition of the cells. The differential stains most frequently used for bacteria are the Gram
stain and the acid-fast stain. Specialized stains detect specific structures of cells such as flagella and endospores.
These stains are used to color and isolate specific parts of organisms.

Gram staining

Gram staining (or Gram’s method) is a differential staining method for differentiating bacterial species into two
groups based on the physical properties of their cell walls. The method is named after the inventor, the Danish
scientist Hans Christian Gram, who developed the technique in 1884.
The Gram staining procedure involves four basic steps:
1. The bacteria are first stained with the basic dye crystal violet. Crystal violet (it is referred to as a primary stain)
imparts purple color to all cells.
2. The bacteria are then treated with Gram’s iodine solution. This allows the stain to be retained better by forming
an insoluble crystal violet-iodine complex. Iodine is used as a mordant. A mordant is used to increase the
affinity of a stain for a biological specimen.
3. Gram’s decolorizer, a mixture of ethyl alcohol and acetone, is then added. A decolorizer or decoloring agent
removes the stain from the specimen. This is the differential step. After this step some bacteria retain the
purple color while some other loose purple color. Bacteria that retain color are classified as Gram-positive and
bacteria that lose the color after decolorization are classified as Gram negative.
4. Because Gram-negative bacteria are colorless after the treatment with decolorizer, they are no longer visible.
Thus, the counterstain safranin (also a basic dye) is applied. Since the Gram-positive bacteria are already stained
purple, they are not affected by the counterstain. Gram-negative bacteria, that are now colorless, become
directly stained by the safranin. Thus, Gram-positive appear purple, while Gram-negative appear red or pink.

Gram positive
Gram negative

Application of Application of Alcohol wash Application of


crystal violet iodine (mordant) (decolorization) safranin
(counterstain)

Figure 4.2 The Gram-staining procedure. In the first step of the Gram-staining procedure, the smear is stained
with the basic dye crystal violet, the primary stain. It is followed by treatment with an iodine solution functioning as
a mordant. The decolorization with ethanol or acetone removes crystal violet from Gram-negative cells but not from
Gram-positive cells. The Gram-negative cells then turn pink to red when counterstained with safranin.

Acid-fast staining

The acid-fast stain is a differential stain used to identify acid-fast organisms such as members of the genus
Mycobacterium. The acid-fast staining procedure involves heating of bacteria with a mixture of basic fuchsin and
phenol (also known as Ziehl-Neelsen stain). The presence of phenol and heat treatment helps the stain penetrate
the cell wall. Once basic fuchsin has penetrated cell wall, acid-fast cells are not easily decolorized by an acid-alcohol
treatment and hence remain red. It occurs due to the presence of large amounts of mycolic acid, a branched
chain hydroxy fatty acid. Non-acid-fast bacteria are decolorized by acid-alcohol. Because non-acid-fast bacteria
are colorless after the treatment with decolorizer, they are no longer visible. Finally, the counterstain methylene
blue is applied. Methylene blue colors non-acid-fast bacteria blue.
Prokaryotes and Viruses 395

Capsule Cell wall Ribosome

Chromosome Flagellum

Plasma Mesosome Inclusion


membrane body

Figure 4.3 Structure of a bacterium. Bacterial cells almost always are bounded by a chemically complex cell wall.
Inside this wall, and separated from it by a periplasmic space, lies the plasma membrane. Bacterial cell does not
contain internal membrane-bound organelles, hence, its interior appears morphologically simple. The genetic material
is localized in a discrete region, the nucleoid, and is not separated from the surrounding cytoplasm by nuclear mem-
branes. Ribosomes and inclusion bodies are scattered in the cytoplasmic matrix. Both Gram-positive and Gram-neg-
ative cells can use flagella for locomotion. In addition, many cells are surrounded by a capsule or slime layer external
to the cell wall.

Cell Wall
Bacterial cells almost always are bounded by a chemically complex cell wall. The cell wall protects bacteria against
osmotic lysis. The cell wall is chemically composed of peptidoglycans (also termed as murein). Peptidoglycans are
unique to bacterial cells. In Gram-positive bacteria, peptidoglycans consist of a single 20 to 80 nm thick homogeneous
layer lying outside the plasma membrane. In contrast, the Gram-negative bacterial cell wall consists of 2 to 7 nm
thick peptidoglycan layer covered by a 7 to 8 nm thick outer membrane.
Peptidoglycan is a polymer containing two sugar derivatives N-acetylglucosamine (NAG) and N-acetylmuramic
acid (NAM) joined through β-1,4 glycosidic bond. NAM is NAG with lactic acid attached by ether linkage. A peptide
chain of four alternating D- and L- amino acids called tetrapeptide is connected to the carboxyl group of the NAM.

NAM NAG

CH2OH CH2OH
H O H O
O H O H O
H OH H
H H

O NH — C — CH3 H NH — C — CH3
n

H3C—CH—C O O O

L-Ala

D-Glu

L-Lys or Meso-diaminopimelic acid

D-Ala

Figure 4.4 Structure of peptidoglycan. Heteropolymer chains of alternating N-acetylglucosamine (NAG) and
N-acetylmuramic acid (NAM) are connected by glycosidic bond. The tetrapeptide side chain is composed of alternating
D- and L-amino acids.
This page intentionally left blank.
410 Prokaryotes and Viruses
cells so that, when they multiply, each will form an individually distinct colony, which may then be used to
establish new cultures with the assurance that only one type of organism will be present. Pure cultures may be
more easily isolated if the growth medium of the original mixed culture favours the growth of one organism to
the exclusion of others.

4.5.2 Bacterial growth


Bacterial growth is an orderly increase in the quantity of cellular constituents (i.e. cell mass) and number. It depends
upon the ability of the cell to form new protoplasm from nutrients available in the environment. The increase in
bacterial cell number occurs by cell division which is known as binary fission. Binary fission begins when the DNA of
the cell is replicated. Each circular strand of DNA then attaches to the plasma membrane. The cell elongates, causing
the two replicated DNA to separate. The plasma membrane then invaginates (grows inward) and splits the cell into
two daughter cells through a process called cytokinesis. The principles that underlie chromosome segregation in
bacteria and eukaryotes share similarities, although bacteria segregate DNA as it replicates and lack a eukaryote-
like mitotic apparatus for segregating chromosomes. A few bacterial species reproduce by budding; they form a
small bud that enlarges until its size approaches that of the parent cell, and then it separates.

Bacterial
chromosome

DNA replication and


separation of two DNA copies

Cross-wall formation and


cell separation

Figure 4.15 Binary fission. A method of asexual reproduction in which the cell divides into two nearly equal sized
daughter cells. The genetic material is also equally partitioned, therefore, the daughter cells are genetically identical.

FtsZ protein plays an important role in cell division. FtsZ is homologous to tubulin, the building block of the microtubule
in eukaryotes. The cellular concentration of FtsZ regulates the frequency of division. At the time of onset of septal
invagination, the FtsZ protein is recruited from the cytoplasm to the division site, where it assembles into a ring
that remains associated with the leading edge of the invaginating septum until the separation is completed.
When a bacterial cell is inoculated into a flask containing fresh culture medium and incubated, it enters into a
rapid growth phase during which the bacterial cell divides and increases its population in the flask medium. Since
the bacteria are not transferred to a new medium or no fresh nutrients are added to the medium, the increasing
population of bacterial cells, after sometime, enters into a stationary-phase with the exhaustion of the required
nutrients and the accumulation of inhibitory end products in the medium. Eventually, the stationary phase of
bacterial population culminates into death-phase when the viable bacterial cells begin to die. A batch culture can
be considered to be a closed system. Growth in batch culture can be divided into four distinct phases – these are
lag phase, log phase, stationary phase and death phase.
Prokaryotes and Viruses 411

Lag phase

During the lag phase, there is no increase in cell number. It is a period of adaptation of cells to a new environment.
There is no change in number, but an increase in mass. Thus in this phase, the bacterial cells are not dormant.
The length of the lag phase is determined in part by the characteristics of the bacterial species and in part by
conditions in the media.

Log or growth phase

In this phase, cell number increase exponentially with time. The log phase is also known as the exponential phase.
During exponential growth, the rate of increase of cells in the culture is proportional to the number of cells present
at any particular time. During exponential growth, the number of cells increases in the geometric progression
20, 21, 22, 23 until, after n divisions, the number of cells is 2n. In reality, exponential growth is only part of the
bacterial life cycle and not representative of the normal pattern of growth of bacteria in nature. The cells divide
at a constant rate depending upon the composition of the growth medium and the conditions of incubation. The
rate of exponential growth of a bacterial culture is expressed as generation time, also the doubling time of the
bacterial population. The time interval required for the cells (or population) to divide is called the generation time
or the doubling time.

Stationary phase

Exponential growth cannot be continued forever in a batch culture (e.g. a closed system). In this phase, the cell
growth rate has leveled off and become constant. The number of cells multiplying equals the number of cells dying.
Population growth is limited by one of three factors:
• Exhaustion of available nutrients;
• Accumulation of inhibitory metabolites or end products;
• Exhaustion of space, in this case called a lack of biological space.

Death phase

If incubation continues after the population reaches stationary phase, a death phase follows, in which the viable cell
population declines. During the death phase, the number of viable cells decreases geometrically (exponentially),
which essentially is reverse of the growth during the log phase.

Lag Log Stationary Death

9.0
Log10 viable cells/ml

8.0

7.0

6.0

5.0

Time

Figure 4.16 Bacterial growth curve in a closed system. When bacteria are introduced into a fresh culture medium,
usually no immediate increase in cell number occurs, and therefore this period is called the lag phase. This phase var-
ies considerably in length with the condition of the bacteria and the nature of the medium. During log phase, bacteria
divide exponentially. In a closed system, population growth remains exponential for only a few generations and then
enters a stationary phase due to factors such as nutrient limitation and waste accumulation. The final phase of the
growth curve is the death phase, which is characterized by a net loss of cells.
This page intentionally left blank.
420 Prokaryotes and Viruses
The likelihood that two genes will be co-transduced depends on how close they lie. If two genes are far apart
along the bacterial chromosome, they will never be co-transduced, because the bacteriophage cannot physically
package a DNA fragment that is too large. The co-transduction can also be used to map genes present in the
bacterial chromosome. Mapping of genes by the process of co-transduction is based on the fact that the closer the
genes to each other, greater the probability of their co-transduction. That is, the frequency of co-transduction is
inversely proportional to the distance between two genes. For example, consider three genes a, b and c for which
the frequencies of co-transduction determined by two factor crosses are: a-b, 90%; a-c, 33%; and b-c, 42%. The
values indicate that the b is closer to a than to c. A mathematical formula (proposed by Wu) is used to relate co-
transduction frequencies to map distance:

C = (1–d/L)3

Where, C is the co-transduction frequency, d is the distance in minutes between two genes and L is the size of the
transducing fragment in minutes.

4.6.3 Conjugation
In 1946, Joshua Lederberg and Edward Tatum showed that bacteria undergo conjugation, a process in which the
genetic information from one bacteria is transferred to, and recombined with, that of other bacteria. In conjugation,
direct contact between the donor and recipient bacteria leads to the establishment of a cytoplasmic bridge between
them and transfer of a part or all of the donor genome to the recipient takes place. Donor ability is determined by
the presence of self transmissible /conjugative plasmids called fertility plasmids or sex plasmids. Self transmissible
plasmids exist in both Gram positive and Gram negative bacteria. A recipient cell that has received DNA as a
result of conjugation is called transconjugant. Transfer of plasmids is generally intraspecific; however, many
plasmids have transfer system that enable them to transfer DNA between unrelated species. These are known as
promiscuous plasmids.

F+ – F— conjugation
The F-plasmid (also called F-factor) of E. coli is the prototype for fertility plasmids in Gram-negative bacteria.
Strains of E. coli with an extrachromosomal F plasmid are called F+ and function as donors, whereas strains that
lack the F-plasmid are F– and behave as recipients. The conjugative functions of the F-plasmid are specified by a
cluster of at least 25 transfer (tra) genes which determine the expression of sex pili, synthesis and transfer of DNA
during mating, interference with the ability of F+ bacteria to serve as recipients, and other functions. The tra genes
can be divided into two groups: those whose products are involved in the mating pair formation (Mpf) and those
whose products are involved in processing the plasmid DNA for transfer (Dtr). The Mpf component includes a sex
pilus that extrudes from the cell and holds mating cells together. The Mpf system also includes a type IV secretion
system (T4SS) which act as a channel through which DNA and protein pass.
Each F+ bacterium has 1 to 3 sex pili that bind to a specific outer membrane protein on recipient bacteria to initiate
mating. An intercellular cytoplasmic bridge is formed, and one strand of the F-plasmid DNA is transferred from
donor to recipient, beginning at a unique origin and progressing in the 5’ to 3’ direction. The site on the plasmid
DNA at which transfer initiates is called the origin of transfer (oriT). Relaxase protein makes a single strand cut at
the ori T site in the plasmid. The transferred strand is converted to circular double-stranded F-plasmid DNA in the
recipient bacterium. Both of the exconjugant bacteria are F+, and the F-plasmid can, therefore, spread by infection
among genetically compatible populations of bacteria.
This page intentionally left blank.
436 Prokaryotes and Viruses
Tetracyclines
The tetracyclines are a family of antibiotics with a common four-ring structure to which a variety of side chains are
attached. Tetracyclines are broad-spectrum antibiotics effective against Gram-negative bacteria, Gram-positive
bacteria, rickettsias, chlamydiae, and mycoplasmas.

Aminoglycoside antibiotics
The aminoglycoside antibiotics are inhibitors of protein synthesis in bacteria. All members of this family of antibiotics
have amino groups and carbohydrate molecules associated with the main molecules. The members include gentamicin,
kanamycin, tobramycin, and streptomycin. The aminoglycosides are most active against Gram-negative pathogens.

Erythromycin
Erythromycin, a macrolide antibiotic, is synthesized by Streptomyces erythraeus. The macrolides contain a 12- to
22-carbon lactone ring linked to one or more sugars. Erythromycin is useful against Gram-positive bacteria and
acts as an inhibitor of protein synthesis.

Chloramphenicol
Chloramphenicol was first produced from cultures of Streptomyces venezuelae, it is now made through chemical
synthesis. Antibiotic has a very broad spectrum of activity.

4.12 Virus
Viruses are simple, noncellular entities consisting of one or more molecules of either DNA or RNA enclosed in a coat
of protein. They can reproduce only within living cells and are obligate intracellular parasites. Viruses are smaller
than prokaryotic cells ranging in size from 0.02 to 0.3 μm (smallpox virus is largest virus about 200 nm in diameter
and polio virus is the smallest virus about 28 nm in diameter). A fully assembled infectious virus is called a virion.
The main function of the virion is to deliver its DNA or RNA genome into the host cell so that the genome can be
expressed (transcribed and translated) by the host cell. Each viral species has a very limited host range; i.e. it can
reproduce in only a small group of closely related species.

Viral structure
The structure of virions are very diverse, varying widely in size, shape and chemical composition. All viruses have
a nucleocapsid composed of nucleic acid surrounded by a protein capsid.
A protein coat, the capsid, which functions as a shell to protect the viral genome from nucleases and which during
infection attaches the virion to specific receptors exposed on the prospective host cell. Capsids are formed as single
or double protein shells and consist of only one or a few structural protein species. The proteins used to build the
capsid are called capsomeres. The nucleic acid together with the capsid forms the nucleocapsid. Some viruses have
a membranous envelope that lies outside the nucleocapsid. Those virions having an envelope are called enveloped
viruses; whereas those lacking an envelope are called naked viruses. In enveloped viruses, the nucleocapsid is
surrounded by a lipid bilayer and glycoprotein derived from the modified host cell membrane. Enveloped viruses
often exhibit a fringe of glycoprotein spikes, also called peplomers. In viruses that acquire their envelope by budding
through the plasma membrane or another intracellular cell membrane, the lipid composition of the viral envelope
closely reflects that of the particular host membrane.
Viral genomes are smaller in size. The largest known viral genome, that of bacteriophage G, is 670 kbs. The
genome of a virus may consist of DNA or RNA, which may be single stranded (ss) or double stranded (ds), linear
or circular. The genomic RNA strand of single-stranded RNA viruses is called sense (positive sense, plus sense)
in orientation if it can serve as mRNA, and antisense (negative sense, minus sense) if a complementary strand
synthesized by a viral RNA transcriptase serves as mRNA.
RNA genomes of certain viruses may be segmented in nature. The segmented genomes are those which are divided
into two or more physically separate molecules of nucleic acid, all of which are then packaged into a single viral particle.
Prokaryotes and Viruses 437

The segmented genome is different from multipartite genome. Multipartite genomes are also segmented, but each
genome segment is packaged into a separate virus particle. These discrete particles are structurally similar and
may contain the same component proteins, but often differ in size depending on the length of the genome segment
packaged. Multipartite viruses are only found in plants.

Table 4.18 Types of viral nucleic acids

Nucleic acid type Nucleic acid structure


DNA

Single stranded Linear, single-stranded DNA


Circular, single-stranded DNA
Double stranded Linear, double-stranded DNA
Linear double-strand DNA with single chain breaks
Circular, double-strand DNA
RNA

Single stranded Linear, single-stranded, positive-strand RNA


Linear, single-stranded, negative-strand RNA
Linear, single-stranded, segmented RNA
Double stranded Linear, double-stranded, segmented RNA

Shape/symmetry
All viruses have a nucleocapsid (nucleic acid and protein) structure. The symmetry (refers to the way in which the
capsomeres are arranged in the virus capsid) may be icosahedral (spherical shape) or helical (rod shape).
Helical symmetry is seen in nucleocapsids of many filamentous and pleomorphic viruses. Helical nucleocapsids
consist of a helical array of capsomeres wrapped around a helical filament of nucleic acid. A typical virus with
helical symmetry is TMV.
Icosahedral morphology is characteristic of the nucleocapsids of many spherical viruses. An icosahedron is a regular
polyhedron with 20 equilateral triangular faces and 12 vertices. Complex structures have capsid symmetry that is
neither purely icosahedral nor helical. As for example T4 virus of E. coli.

Helical symmetry Icosahedral symmetry

Figure 4.32 Diagrammatic representation of helical and icosahedral symmetry.

Classification of viruses

Viruses are classified into different taxonomic groups based on their host, virion structure and composition, mode of
reproduction, and the nature of any diseases caused. Currently viruses are classified with a taxonomic system placing
primary emphasis on the host, type and strandedness of viral nucleic acids, and on the presence or absence of an envelope.
Based on the nature of the host, viruses are classified into animal virus, plant virus, bacterial virus, insect virus and others.
This page intentionally left blank.
458 Prokaryotes and Viruses
Hepatitis D virus, classified as a hepatitis delta virus, is considered to be a subviral satellite because it can propa-
gate only in the presence of the hepatitis B virus. Transmission of hepatitis D virus can occur either via simultane-
ous infection with hepatitis B virus (coinfection) or via infection of an individual previously infected with hepatitis
B virus (superinfection). The hepatitis D virus genome consists of a single stranded, negative sense, circular RNA.

4.12.6 Plant viruses


Plant viruses exist in rod and polyhedral shape. Most plant viruses have genomes consisting of a single RNA strand
of plus (+) sense type. The best-known plant virus is the rod-shaped tobacco mosaic virus (TMV). Relatively few
plant viruses have DNA genomes. There are only two classes of DNA containing plant viruses. The cauliflower mosaic
virus belongs to the first class, which contains a double-stranded DNA genome in a polyhedral capsule. The second
class of DNA containing plant viruses are the geminiviruses (gemini = twins), characterized by a connected pair of
capsids, each containing a circular, single-stranded DNA molecule of about 2500 nucleotides.
Tobacco Mosaic Virus (TMV) causes leaf mottling and discoloration in tobacco and many other plants. It was the
first virus to be discovered (by Dmitri Iwanowasky) and first virus to be crystallized (by W. Stanley). TMV is a rod
shaped virus with ~2130 capsomeres arranged in a hollow right handed helix. It contains a single genetic RNA (ss,
plus sense) of ~6400 nucleotides.

RNA

Capsid

Figure 4.51 Tobacco mosaic virus has a rod-like appearance. Its capsid is made up of ~2130 capsomeres. One mol-
ecule of genomic ssRNA, 6400 nucleotides long, present in the centre of the capsid. The capsomere self-assembles
into the rod like helical structure (16.3 capsomeres per helical turn) around the RNA.

4.13 Prions and Viroid


Prions are proteins. Prion proteins are designated as PrP. The word prion, coined in 1982 by Stanley B. Prusiner, is
derived from the words protein and infection. The endogenous, normal cellular form is denoted PrPC (for Cellular)
while the disease-causing, infectious and misfolded form is denoted PrPSc (for Scrapie, after one of the diseases
first linked to prions). Prions are glycosylated proteins and linked to the membrane by a GPI-linkage. The infectious
form, PrPSc, is responsible for neurodegenerative diseases in animals including human. The normal cellular PrPC
form is converted into PrPSc through a process whereby a portion of its α-helical and coil structure is refolded into
a β-sheet. This structural transition is accompanied by profound changes in the physicochemical properties of the
PrP. PrPC is sensitive to proteases whereas PrPSc is protease resistant. High content of β-sheet in PrPSc results in
the formation of amyloid fibrillous structure that is absent from the PrPC form. The PrPSc form can perpetuate itself
by causing the newly synthesized PrP protein to take up the PrPSc form instead of the PrPC form.
Prions are novel transmissible agents causing a group of neuro-degenerative diseases that can be perpetuated by
inoculating animal with tissue extracts from infected one. Collectively, prion diseases are described as spongiform
encephalopathies. No prion diseases of plants are known.
This page intentionally left blank.
460 Prokaryotes and Viruses
encapsidated by the protein coat of these helper viruses. The nature of nucleic acid in virusoid is circular RNA
which possesses ribozyme domain. Virusoids replicate via rolling circle mechanism. The strand of RNA that is
packaged into the helper virus is called the plus strand. Similar to viroid, it forms a hammerhead structure that
has a self-cleaving activity.

4.13.1 Bacterial and viral disease


Any organism or agent that causes a disease is a pathogen. Its ability to cause disease is called pathogenicity. The
term disease broadly refers to any condition that impairs normal function of an organism. Infectious diseases, (also
known as transmissible diseases or communicable diseases) comprise illness resulting from the infection, presence
and growth of pathogens in a host organism.

Table 4.25 Some important human disease and their causative agent

Bacterial disease

Disease Causative agent

Anthrax Bacillus anthracis

Botulism Clostridium botulinum

Cholera Vibrio cholerae

Legionnaire’s disease Legionella pneumophila

Typhoid fever Salmonella typhi

Diphtheria Corynebacterium diphtheriae

Meningococcal meningitis Neisseria meningitidis

Pneumococcal pneumonia Streptococcus pneumoniae

Tuberculosis Mycobacterium tuberculosis

Whooping cough Bordetella pertussis

Gonorrhea Neisseria gonorrhoeae

Syphilis Treponema pallidum

Tetanus Clostridium tetani

Viral disease

Disease Causative agent

Hepatitis Hepatitis A, B, C, D, E

Influenza Influenza virus

Measles Measles virus

AIDS Human immunodeficiency (HIV) virus

Rabies Rabies virus

Severe Acute Respiratory Syndrome (SARS) is a respiratory disease in humans which is caused by the SARS coro-
navirus that was first identified in 2003. Avian influenza (known informally as avian flu or bird flu) is flu infection
in birds. All known viruses that cause influenza in birds belong to the species influenza A virus. There are many
subtypes of avian influenza A viruses such as H5N1, H7N3, H7N7 and H9N2. Swine influenza (also called pig influ-
enza, swine flu, hog flu and pig flu) is an infection caused by swine influenza viruses. Swine influenza virus (SIV)
is any strain of the influenza family of viruses that is endemic in pigs. The known SIV strains include influenza C
and the subtypes of influenza A known as H1N1, H1N2, H2N1 and others.
This page intentionally left blank.
Chapter 05

Immunology

Immunology is the science that is concerned with immune response to foreign challenges. Immunity (derived from
Latin term immunis, meaning exempt), is the ability of an organism to resist infections by pathogens or state of
protection against foreign organisms or substances. The array of cells, tissues and organs which carry out this activity
constitute the immune system. Immunity is typically divided into two categories—innate and adaptive immunity.

5.1 Innate immunity


Innate (native/natural) immunity is present since birth and consists of many factors that are relatively nonspecific—
that is, it operates against almost any foreign molecules and pathogens. It provides the first line of defense against
pathogens. It is not specific to any one pathogen but rather acts against all foreign molecules and pathogens. It
also does not rely on previous exposure to a pathogen and response is functional since birth and has no memory.

Elements of innate immunity


Physical barriers
Physical barriers are the first line of defense against microorganisms. It includes skin and mucous membrane.
Most organisms and foreign substances cannot penetrate intact skin but can enter the body if the skin is damaged.
Secondly, the acidic pH of sweat and sebaceous secretions and the presence of various fatty acids and hydrolytic
enzymes like lysozyme inhibit the growth of most microorganisms. Similarly, respiratory and gastrointestinal tracts
are lined by mucous membranes. Mucus membranes entrap foreign microorganisms. The respiratory tract is also
covered by cilia, which are hair like projections of the epithelial-cell membranes. The synchronous movement of
the cilia propels mucus-entrapped microorganisms out of these tracts. Similarly, the conjunctiva is a specialized,
mucus-secreting epithelial membrane that lines the interior surface of each eyelid. It is kept moist by the continuous
flushing action of tears (lacrimal fluid) from the lacrimal glands. Tears contain lysozyme, lactoferrin, IgA and thus
provide chemical as well as physical protection.
Microorganisms do occasionally breach the epithelial barricades. It is then up to the innate and adaptive immune
systems to recognize and destroy them, without harming the host. In case of innate immune response several
antimicrobial chemicals and phagocytic cells provide protection against pathogens.

Chemical mediator
A variety of chemicals mediate protection against microbes during the period before adaptive immunity develops.
The molecules of the innate immune system include complement proteins, cytokines, pattern recognition molecules,
acute-phase proteins, cationic peptides, enzyme like lysozyme and many others.

Complement proteins
The complement proteins are soluble proteins/glycoproteins that are mainly synthesized by liver and circulate in the
blood and extracellular fluid. They were originally identified by their ability to amplify and complement the action
464 Immunology
of antibodies; hence, the name complement. It also bridges innate and adaptive immunity and removes immune
complexes. The complement system is composed of over 30 serum proteins. Activation of complement proteins
in response to certain microorganisms results in a controlled enzymatic cascade, which targets the membrane of
pathogenic organisms and leads to their destruction.

Cytokines
The term cytokine is a generic term for any low molecular weight soluble protein or glycoprotein released by one
cell population which acts as an intercellular mediator. It includes monokines, lymphokines, interleukins, interferons
and others. Cytokines are required for immunoregulation of both innate as well as adaptive immune responses
Interferons are cytokines made by cells in response to virus infection, which essentially induce a generalized
antiviral state in surrounding cells.
Chemokines are small, positively charged secreted proteins that have a central role in guiding the migrations of various
types of white blood cells. They bind to the surface of endothelial cells, and to negatively charged proteoglycans of
the extracellular matrix in organs. By binding to G-protein-linked receptors on the surface of specific blood cells,
chemokines attract these cells from the bloodstream into an organ, guide them to specific locations within the
organ, and then help stop migration.

Pattern recognition molecule


Many molecules involved in innate immunity have the ability to recognize Pathogen-Associated Molecular Pattern
(PAMP) for the initial detection of microbes. PAMPs are microbe-specific molecular signatures. PAMPs are recognized
by pattern-recognition receptors (PRRs). Mammals have several distinct classes of pattern recognition receptors
(PRRs) including Toll-like receptors (TLRs), RIG-I-like receptors (RLRs), Nod-like receptors (NLRs), AIM2-like
receptors (ALRs) and C-type lectin receptors (CLRs). Among these, TLRs were the first to be identified, and are
the best characterized.
Toll-Like Receptors (TLRs) are a class of pattern recognition receptors that function exclusively as signaling
receptors. It was originally identified as a protein involved in the establishment of dorso-ventral polarity in developing
fly embryos. It is also involved, however, in the adult fly’s resistance to fungal infections. TLRs are type I integral
trans-membrane glycoproteins. Each TLR is composed of an extracellular N-terminal domain and an intracellular
C-terminal region (human TLRs are 700–1100 amino acids long). The N-terminal domain is constituted by about
16–28 leucine rich repeats (LRRs) and has the function to recognize PAMPs, whereas the second, also called Toll/
IL-1 receptor (TIR) domain that initiates downstream signaling. All TLRs are synthesized in the ER, traffic to the
Golgi, and are recruited to the cell surface or to intracellular compartments such as endosomes.
The TLR family comprises 10 members (TLR1–TLR10) in human and 12 (TLR1–TLR9 and TLR11-TLR13) in mouse. TLRs
localize to the cell surface or to intracellular compartments such as the ER, endosome, lysosome or endolysosome
and they recognize distinct or overlapping PAMPs such as lipid, lipoprotein, protein and nucleic acid. TLRs are
expressed in innate immune cells such as dendritic cells and macrophages as well as non-immune cells such as
fibroblast cells and epithelial cells. TLRs are largely classified into two subfamilies based on their localization, cell
surface TLRs and intracellular TLRs. In human, cell surface TLRs include TLR1, TLR2, TLR4, TLR5, TLR6 and TLR10,
whereas intracellular TLRs are localized in the intracellular compartments such as endosome include TLR3, TLR7,
TLR8 and TLR9. Different TLRs recognize different ligands. Cell surface TLRs mainly recognize microbial membrane
components such as lipids, lipoproteins and proteins.

TLRs and their ligands


Bacterial lipopolysaccharide TLR4
Bacterial diacyl lipopeptides TLR2/TLR6
Bacterial triacyl lipopeptides TLR2/TLR1
Bacterial peptidoglycans TLR2(?)
Bacterial lipoteichoic acid TLR2/TLR6
Bacterial flagellin TLR5
This page intentionally left blank.
472 Immunology
Bone marrow

In most mammals including humans and mice, bone marrow is the site of B-cell synthesis and maturation. Before
birth, the yolk sac, fetal liver, and fetal bone marrow are the major sites of B-cell maturation; after birth, generation
of mature B-cells occurs in the bone marrow. After maturation, the mature B-cells are transported by the circulating
blood to the secondary lymphoid organs, where they encounter and respond to foreign antigens. Bone marrow is
not the site of B-cell maturation in all species. In birds, B-cells undergo maturation in the bursa of fabricius. This
organ, situated near the cloaca, consists of lymphoid centers that contain epithelial cells and lymphocytes. These
lymphocytes consist solely of antibody-producing B-cells. Mammals do not have a bursa of fabricius.

Hematopoietic tissue Thymus Lymphoid organ

T-cell
precursor
T-cell

Cell mediated
immune response
Hematopoietic Thymus
stem cells lymphocyte Antigen

Antibody
response
Bone marrow B cell
lymphocyte

Figure 5.3 The development and activation.

Thymus

Thymus is the site where T-cells mature. Progenitor cells from the bone marrow migrate into the thymus gland, where
they differentiate into T-cells. It is a flat, bilobed organ situated above the heart. Each lobe is surrounded by a capsule
and is divided into lobules, which are separated from each other by strands of connective tissue called trabeculae.
Each lobule is organized into two compartments: the outer compartment, or cortex, and the inner compartment, or
medulla. T-lymphocytes mature in the cortex and migrate to the medulla, where they encounter macrophages and
dendritic cells. Here, they undergo thymic selection, which results in the development of mature, functional T-cells,
which then leave to enter the peripheral blood circulation, through which they are transported to the secondary
lymphoid organs. It is in these secondary lymphoid organs where the T-cells encounter and respond to foreign antigens.

5.4.2 Secondary lymphoid organs/tissues


Mature B and T-lymphocytes migrate from bone marrow and thymus, respectively, through the bloodstream to the
secondary (peripheral) lymphoid organs. These secondary (peripheral) lymphoid organs are those organs in which
antigen-driven proliferation and differentiation take place.
The major secondary lymphoid organs are the spleen, the lymph nodes and mucosa associated lymphoid tissue
(MALT). Spleen and lymph nodes are the highly organized secondary lymphoid organs. The secondary lymphoid
organs have two major functions: They are highly efficient in trapping and concentrating foreign substances, and
they are the main sites of production of antibodies and the induction of antigen-specific T-lymphocytes.

Spleen

The spleen is the largest of the secondary lymphoid organs. It is highly efficient in trapping and concentrating
foreign substances carried in the blood. It is the major organ in the body in which antibodies are synthesized and
from which they are released into the circulation.
Immunology 473

The interior of the spleen is a compartmentalized structure. The compartments are of two types– Red pulp and white
pulp. Red pulp is the site where old and defective RBCs are destroyed and removed, whereas white pulp forms PALS
(Periarteriolar lymphoid sheath) which are rich in T-cells. The marginal zone, located peripheral to the PALS, is rich
in lymphocyte and macrophage. Approximately 50% of spleen cells are B-lymphocytes; 30-40% are T-lymphocytes.

Lymph nodes

Lymph nodes are small encapsulated bean shaped structures (normally <1cm in diameter) found in various regions
throughout the body. The lymph nodes are composed of a medulla and a cortex, which is surrounded by a capsule
of connective tissue. They are packed with lymphocytes, macrophages, and dendritic cells. The cortical region
contains primary lymphoid follicles. After antigenic stimulation, these structures enlarge to form secondary lymphoid
follicles with germinal centers containing dense populations of lymphocytes (mostly B-cells). The deep cortical area
or paracortical region contains T-cells and dendritic cells. Antigens are brought into these areas by dendritic cells,
which present antigen fragments to T-cells. The medullary area of the lymph node contains antibody-secreting
plasma cells that have traveled from the cortex to the medulla via lymphatic vessels.
Lymph nodes are highly efficient in trapping antigen that enters through the afferent lymphatic vessels. In the
node, the antigen interacts with macrophages, T-cells, and B-cells, and that interaction brings about in immune
response, manifested by the generation of antibodies and antigen-specific T-cells.

Mucosa associated lymphoid tissue

The majority of secondary lymphoid tissue in the human body is located within the lining of respiratory, digestive and
genitourinary tracts. These are collectively called as Mucosa Associated Lymphoid Tissue (MALT). There are several
types of MALT. Two major MALT includes Bronchial Associated Lymphoid Tissue (BALT) and Gut-Associated Lymphoid
Tissue (GALT). GALT includes the tonsils, adenoids, and specialized regions in the small intestine called Peyer’s patches.

5.5 Antigens
Adaptive immune responses arise as a result of exposure to foreign compounds. The compound that evokes the
response is referred to as antigen, a term initially coined due to the ability of these compounds to cause antibody
responses to be generated. An antigen is any agent capable of binding specifically to T-cell receptor (TCR) or an
antibody molecule (membrane bound or soluble). The ability of a compound to bind with an antibody or a TCR is
referred to as antigenicity. There is a functional distinction between the term antigen and immunogen. An immunogen
is any agent capable of inducing an immune response and is therefore immunogenic. The distinction between the
terms is necessary because there are many compounds that are incapable of inducing an immune response, yet
they are capable of binding with components of the immune system that have been induced specifically against
them. Thus all immunogens are antigens, but not all antigens are immunogens.

Requirements for immunogenicity


A substance must possess the following characteristics to be immunogenic:

Foreignness
The most important feature of an immunogen is that an effective immunogen must be foreign with respect to the
host. The adaptive immune system recognizes and eliminates only foreign (nonself) antigens. Self antigens are
not recognized and thus individuals are tolerant to their own self molecules, even though these same molecules
have the capacity to act as immunogens in other individuals of the same species.

Size
The second requirement for being immunogenic is that the compound must have a certain minimal molecular weight.
There is a relationship between the size of immunogen and its immunogenicity. In general, small compounds with a
molecular weight <1000 Da (e.g. penicillin, aspirin) are not immunogenic; those of molecular weight between 1000
This page intentionally left blank.
482 Immunology
A newly synthesized class II MHC protein must avoid clogging its binding groove prematurely in the ER lumen with
peptide derived from endogenously synthesized proteins. A special trimeric protein, called the invariant chain,
ensures this by associating with newly synthesized class II MHC in the ER. Part of its polypeptide chain lies within
the peptide-binding groove of the MHC protein, thereby blocking the groove from binding other peptides in the
lumen of the ER. The invariant chain is cleaved by proteases in the late endosome. However, a short fragment of
the invariant chain termed CLIP (for class II-associated invariant chain peptide) remain bound to the class II MHC,
preventing any premature binding of antigenic peptide. This fragment is then released (mediated by a non-classical
class II-MHC like protein called HLA-DM), freeing the MHC protein to bind peptides derived from endocytosed
proteins. Another non-classical class II MHC HLA-DO may act as a negative regulator of class II antigen processing
by binding to HLA–DM and inhibiting its role in the dissociation of CLIP from MHC.

Exogenous Endocytic Peptides Peptide-class II


antigens Endocytosis compartments MHC complex
or
phagocytosis

Exogenous antigen

4 Plasma membrane
CLIP

3 5
CLIP

1. Class II MHC binds invariant chain blocking


Golgi apparatus
2
binding of endogenous antigen
2. MHC complex is routed through Golgi to endocytic
Invariant chain pathway compartments
3. Invariant chain is degraded, leaving CLIP fragment
ER
4. Exogenous antigen is taken up, degraded
1 Class II MHC fragments routed to endocytic pathway
compartments
5. Class II-MHC peptide is transported to PM

Figure 5.11 The processing of an exogenous protein antigen for presentation to a helper T-cell.

5.6.3 Laboratory mice


Mice are the most commonly used mammalian research model. They are common experimental animals in biology,
primarily because they are mammals, are relatively easy to maintain and handle, reproduce quickly, and share a high
degree of homology with humans. Laboratory mice include several inbred, outbred, knockout and transgenic mice
strains. Many laboratory strains are inbred. An inbred strain is one that is produced using at least 20 consecutive
generations of sister and brother or parent and offspring matings. The mating of two genetically related parents
is called inbreeding. Inbreeding results in increased homozygosity. In contrast to inbred mice, outbred mice are
usually heterozygous at many loci.
If mice are inbred (that is, have identical alleles at all loci), each H-2 locus will be homozygous because the maternal
and paternal haplotypes are identical, and all offspring therefore express identical haplotypes. Inbred mouse strains
are syngeneic or identical at all genetic loci. Two strains are considered congenic if they are genetically identical
except at a single genetic locus.
Immunology 483

Some inbred mouse strains have been designated as prototype strains and the MHC haplotype expressed by these
strains is designated by an arbitrary italic superscript (e.g. H-2a, H-2b). If another inbred strain has the same set
of alleles as the prototype strain, its MHC haplotype is the same as the prototype strain.

Table 5.5 H-2 haplotypes of some mouse strains

H-2 alleles
Prototype strain Other strains with the same haplotype Haplotype K IA IE S D
CBA AKR, C3H, C57BR k k k k k k
DBA/2 BALB/c, SEA, YBR d d d d d d
C57BL/10 (B10) C57BL/6, C57L b b b b b b
A A/He, A/Sn a k k k d d

5.7 Immunoglobulins: Structure and function


Antibodies, the antigen-binding glycoproteins are synthesized exclusively by B-cells and in billions of forms, each
with a different amino acid sequence and a different antigen binding site. Collectively called immunoglobulins (Ig),
they are among the most abundant protein components in the blood, constituting about 20% of the total protein
components in the blood plasma. Antibodies (Ab) are present on the B-cell membrane and also are secreted by
plasma cells.

5.7.1 Basic structure of antibody molecule


The simplest antibodies are Y-shaped molecules with two identical antigen-binding sites, one at the tip of each arm
of the Y. Because of their two antigen-binding sites, they are described as bivalent.
Antibody has a common structure of 4 polypeptide chains. It is a heterodimer and consists of two identical light
(L) chains (each containing about 220 amino acids residues, about 25000 MW) and two identical heavy (H) chains
(each usually containing about 440 amino acids residues, about 50,000 MW). Each light chain is bound to heavy
chain by disulfide bridges and other non-covalent linkages. Thus, antibody is a dimer of H—L chain.
All species studied have the two major classes of light chains: κ and λ. Any one individual of a species produces both
types of light chain. However, in any one immunoglobulin molecule, the light chains are always either both κ or both
λ, never one of each. While there are two types of light chains, the immunoglobulins of virtually all species have been
shown to consist of five different types of heavy chains- α, γ, δ, ε and μ. These five different types of heavy chains
are called isotypes. The heavy-chains of a given antibody molecule determine the class of that antibody: IgM (μ),
IgG (γ), IgA (α), IgD (δ) or IgE (ε). Each class can have either κ or λ light chains. Any individual of a species makes
all heavy chains, but in any one antibody molecule, both heavy chains are identical. Thus an antibody molecule
of the IgG class could have the structure κ2γ2 with two identical κ light chains and two identical γ heavy chains.
Alternatively, it could have the structure λ2γ2 with two identical λ light chains and two identical γ heavy chains.

Immunoglobulin heavy chain isotypes


Isotype Heavy chain
IgM μ
IgD δ
IgG γ
IgA α
IgE ε

Minor differences in the amino-acid sequences of the α and the γ heavy chains led to further classification of the
heavy chains into subclasses. In humans, there are two subclasses of α heavy chains (α1 and α2) and four subclasses
of γ heavy chains (γ1, γ2, γ3 and γ4).
This page intentionally left blank.
Immunology 495

According to this theory, an animal first randomly generates a vast diversity of lymphocytes, and then those
lymphocytes that can react against the foreign antigens that the animal actually encounters are specifically selected
for proliferation. As each lymphocyte develops in a central lymphoid organ, it becomes committed to react with
a particular antigen before ever being exposed to the antigen. It expresses this commitment in the form of cell-
surface receptor proteins that specifically fit the antigen.

Precursor cell

B1 B2 B3 Different
non-activated B-cells

Antigen binding to
Ag
specific B-cell

B2

Proliferation and
differentiation

B2 B2 B2 B2

Figure 5.23 The clonal selection theory of B-cells leading to antibody production. Adapted from Molecular Biology
of the cell, Albert et al., Garland Science.

When a lymphocyte encounters its antigen in a peripheral lymphoid organ, the binding of the antigen to the
receptors activates the lymphocyte, causing it both to proliferate and to differentiate into an effector cell. An antigen
therefore selectively stimulates those cells that express complementary antigen-specific receptors and are thus
already committed to respond to it. This arrangement is what makes adaptive immune responses antigen-specific.
According to the clonal selection theory, then, the immune system functions on the ready-made principle rather
than the made-to-order one.
The term clonal in clonal selection theory derives from the postulate that the adaptive immune system is composed
of millions of different families, or clones, of lymphocytes, each consisting of T or B-cells descended from a common
ancestor. Each ancestral cell was already committed to make one particular antigen-specific receptor protein, and
so all cells in a clone have the same antigen specificity.

5.9 Kinetics of the antibody response


Humoral immunity is mediated by serum antibodies which are the proteins secreted by the B-cells. B-cells are
initially activated to secrete antibodies after the binding of antigens to specific membrane immunoglobulin molecules
(B-cells receptors), which are expressed by these cells. Once bound, the B-cell receives signals to begin making
the secreted form of this immunoglobulin, a process that initiates the full-blown antibody response whose purpose
is to eliminate the antigen from the host. Antibodies are a heterogeneous mixture of serum globulins, all of which
share the ability to bind individually to specific antigens.
496 Immunology
Primary and secondary responses

The first exposure of an individual to an immunogen is referred to as the primary immunization, which generates
a primary response. The primary antibody response may be divided into several phases, as follows:

1. Lag or latent phase: It is the immediate stage following antigenic stimulus during which no antibody is detectable
in circulation. The length of this period is generally one to two weeks.
2. Log or exponential phase: In this phase there is a steady rise in the titer of antibody and the concentration of
antibody in the serum increases exponentially.
3. Plateau or steady state: During this phase there is an equilibrium between antibody synthesis and degradation.
4. Declining phase: The concentration of antibody in serum declines rapidly.

A second exposure to the same immunogen results in a secondary response. This second exposure may occur
after the response to the first immune event has leveled off or has totally subsided. The secondary response is also
called the memory or anamnestic response and the B-and T-lymphocytes that participate in the memory response
are termed memory cells.
The primary response is slow and short lived with a long lag phase and low titer of antibodies that do not persist
for long. However the secondary response is prompt, powerful and prolonged, with a short or negligible lag phase
and a much higher level of Ab that lasts for long periods.
Antibody concentration in serum

Primary response Secondary response

IgG
De

IgG
cli
ne

IgM
ph

IgM
as
e

Latent period

10 days 15 days 5 days 10 days

First exposure to antigen Second exposure to antigen

Figure 5.24 Antibody production and kinetics.

In the primary response, the first class of antibody detected is generally IgM, then IgG, or another antibody class.
There is a marked change in the type and quality of antibody produced in the secondary response. There is a shift in
class response, known as class switching, with IgG antibodies appearing at higher concentrations and with greater
persistence than IgM, which may be greatly reduced or disappear altogether. This may be also accompanied by
the appearance of IgA and IgE. The IgG, IgE, and IgA molecules are collectively referred to as secondary classes
of antibodies because they are thought to be produced only after antigen stimulation and because they dominate
secondary antibody responses.
With the passage of time after immunization, there is usually a progressive increase in the affinity of the antibodies
produced against the immunizing antigen. This phenomenon, known as affinity maturation, is due to the accumulation
of point mutations specifically in both heavy-chain and light-chain V-region coding sequences.
Immunology 497

Table 5.9 Features of primary and secondary antibody responses

Feature Primary response Secondary response

Time lag after immunization Usually 5-10 days Usually 1-3 days

Peak response Smaller Larger

Antibody isotype Usually lgM > lgG Relative increase in lgG

Antibody affinity Lower Higher

Induced by All immunogens Only protein antigens

Required immunization Relatively high doses of antigens, optimally Low doses of antigens; adjuvants may
with adjuvants not be necessary

Responding B-cell Naive B-cell Memory B-cell

Antigens Thymus-dependent and thymus-independent Thymus-dependent

5.10 Monoclonal antibodies and Hybridoma technology


Antibodies produced ordinarily by infection or immunization are polyclonal because natural antigens have multiple
epitopes, each of which generates clones of lymphocytes. This results in antisera containing antibodies from different
clones of lymphocytes with specificities against different epitopes of the antigens. On the other hand, monoclonal
antibodies (mAb) are monospecific antibodies. These antibodies are produced from clone of single lymphocyte
directed against a single antigenic determinant or epitope. Such antibodies produced by a single clone and directed
against a single epitope.

Hybridoma technology is a method of forming hybrid cell lines (called hybridomas) by fusing a specific antibody-
producing B-cell with a myeloma cell (cancerous B-cell). The antibodies produced by the hybridoma are all of a
single specificity and are therefore monoclonal antibodies. The production of monoclonal antibodies was invented by
Cesar Milstein and Georges J. F. Köhler in 1975. They shared the Nobel Prize in 1984 for Medicine and Physiology.
Hybridomas are somatic cell hybrids produced by fusing antibodies forming spleen cells with myeloma cells.
Antibody-producing B-cells normally die after several weeks in cell culture in vitro. Therefore, antibody-producing
B-cells are fused with B-cell tumors called myelomas. These myelomas are capable of dividing indefinitely and
are, therefore, often called immortal cell lines. The immortal cell lines that result from the B cell-myeloma fusion
are hybrid cell lines called hybridomas. The hybridoma cell lines share the properties of both fusion partners. They
grow indefinitely in vitro and produce antibodies. To produce a monoclonal antibody, a mouse is immunized with
the antigen of interest. During the next several weeks, antigen-specific B-cells proliferate and begin producing
antibodies in the mouse. Spleen tissue, rich in B-cells, is then removed from the mouse, and the B-cells are fused
with myeloma cells.
Hybridomas are selected by the use of a selective medium in which the myeloma cells die, but hybridomas survive.
The most widely used selective systems involve the inclusion of the antibiotic aminopterin in the growth medium.
Aminopterin is a synthetic derivative of pterin. This folate analog acts as a competitive inhibitor for enzyme
dihydrofolate reductase which catalyzes the reduction of dihydrofolate into tetrahydrofolate. Normal animal cells
synthesize purine nucleotides and thymidylate for DNA synthesis by a de novo pathway requiring tetrahydrofolate.
Thus, the addition of aminopterin inhibits the de novo nucleotide synthesis pathway. However, normal cells survive
in this medium as they are able to use the salvage pathway for nucleic acid synthesis. But if the cells are unable to
produce the enzyme Hypoxanthine-Guanine Phospho Ribosyl Transferase (HGPRT), they are unable to utilize the
salvage pathway and, therefore, die in the aminopterin-containing medium.
This page intentionally left blank.
Immunology 517

Class I Inhibitory
MHC receptor


No killing
+

Ligand
Activating
Normal cell receptor NK cell

Killing
+

Ligand
Activating
Altered self cell receptor NK cell

Figure 5.46 An activating receptor on NK cells interacts with its ligand on normal and altered self cells, inducing an
activation signal that results in killing. However, interaction of inhibitory NK-cell receptors with class I MHC molecules
delivers an inhibition signal that counteracts the activation signal. Expression of class I molecules on normal cells
thus prevents their destruction by NK cells. Because class I expression is often decreased on altered self cells (virus
infected cells and tumor cells), the killing signal predominates, leading to their destruction.

5.13.1 Superantigens
Superantigens are viral or bacterial proteins that bind simultaneously to the variable domain of β of a T-cell receptor
(TCR) and to the α-chain of a class II MHC molecule (i.e. outside the peptide-binding groove). Because of their
unique binding ability, superantigens can activate large numbers of T-cells irrespective of their antigenic specificity.
Superantigens can be exogenous and endogenous. Exogenous superantigens are soluble proteins secreted by
bacteria whereas endogenous superantigens are cell-membrane proteins encoded by certain viruses that infect
mammalian cells.

b Ag
a
MHC TCR
a b

Superantigen

APC TH cell

Figure 5.47 Superantigen-mediated cross-linkage of T-cell receptor (TCR) and class II MHC molecules. Superanti-
gen binds to class II MHC molecule and a part of the Vβ chain of the T-cell receptor that is outside the normal anti-
gen-binding site and this binding is sufficient to trigger T-cell activation. A superantigen binds to all TCRs bearing a
particular V sequence regardless of their antigen specificity.
518 Immunology

5.14 Cytokines
Cytokines are low-molecular-mass (generally less than 30 kDa) soluble proteins/glycoproteins, non-immunoglobulin
in nature, secreted by a variety of cell types and act nonenzymatically through specific receptors to regulate host
cell function. They do not include the peptide and steroid hormones of the endocrine system. Cytokines play major
roles in the development of cellular and humoral immune responses, induction of the inflammatory response,
regulation of hematopoiesis, control of cellular proliferation and differentiation.
Cytokines can affect the same cell responsible for their production (an autocrine function) or nearby cells (a paracrine
function), or they can be distributed by the circulatory system to distant target cells (an endocrine function). They
are highly potent hormone-like substances, active even at femto molar concentration. However, they differ from
endocrine hormones as being not produced by glands but by widely distributed cells. Cytokines produce biological
actions only when they bind to specific, high-affinity receptors on the surface of target cells. The biological activities
of cytokines exhibit pleiotropy (a given cytokines that has different biological effect on different target cells),
redundancy (two or more cytokines that mediates similar functions), synergy (combined effect of two cytokines
on cellular activity is greater than the additive effect of the individual cytokines) and antagonism (effect of one
cytokines inhibit the effect of another cytokines).

Target cell Effect

Pleiotropy
B-cell Activation, proliferation, differentiation
Activated TH cells IL-4 Thymocyte Proliferation
Mast cell Proliferation

Redundancy
IL-2
Activated TH cells IL-4 B-cell Proliferation
IL-5

Synergy
IL-4
Activated TH cells + B-cell Induces class switch to IgE
IL-5
Antagonism

Activated TH cells IL-4 B-cell Blocks class switch of IgE induced by IL-4

IFN-g

Figure 5.48 Cytokine attributes of pleiotropy, redundancy, synergy (synergism), antagonism.

Cytokines differ from hormones and growth factors. All three are secretory proteins that elicit their biological effects
at very low concentrations by binding to receptors on target cells. Growth factors tend to be produced constitutively,
whereas cytokines and hormones are secreted in response to discrete stimuli. Unlike hormones, which generally act
long range in an endocrine fashion, most cytokines act over a short distance in an autocrine or paracrine fashion.
In addition, most hormones are produced by specialized glands and tend to have a unique action on one or a few
types of target cell. In contrast, cytokines are often produced by, and bind to, a variety of cells.
There are over 100 different cytokines. The generic name of cytokines includes all proteins with a small molecular
weight, released by cells of the immune system, especially by monocytes and T-lymphocytes. But they are also
secreted by many cells in addition to those of the immune system, such as endothelial cells and fibroblasts. They
used to have different names depending either on their origin, such as lymphokines (produced by lymphocytes),
monokines (substances produced by monocytes or macrophages) or on their activity: chemokines, interleukins,
interferons. Cytokines may be grouped into following categories: hematopoietins, interleukins, interferons, chemokines
and members of the tumor necrosis factor (TNF) family.
This page intentionally left blank.
Immunology 525

Biologically active functions Complement component


Cell lysis C5b–9 (membrane-attack complex)
Inflammatory response C3a, C4a, and C5a (anaphylatoxins)
Chemotaxis of leukocytes C3a, C5a
Opsonization of particulate antigens C3b, C4b
Viral neutralization C3b, C5b–9 (membrane-attack complex)
Solubilization and immune clearance C3b

5.16 Hypersensitivity
Hypersensitivity is an exaggerated immune response that results in tissue damage and is manifested in the individual
on a second or subsequent contact with an antigen.
Hypersensitivity has been traditionally classified into immediate and delayed types based on the time required for
a sensitized host to develop clinical reactions on re-exposure to the antigen. Later, Gell and Coombs proposed a
classification scheme which defined four types of hypersensitivity reactions.

Type I Hypersensitivity
Type I hypersensitivity (also known as allergic reaction) is induced by antigens referred to as allergens. The term
allergen refers specifically to nonparasitic antigens capable of stimulating type I hypersensitive responses. Type I
hypersensitive reactions are IgE-mediated humoral antibody responses. These IgE-mediated reactions are stimulated
by the binding of IgE (via its Fc region) to high-affinity IgE-specific FC receptors expressed on mast cells and
basophils. When cross linked by antigens, the IgE antibodies trigger the mast cells and basophils to release primary
mediators, vasoactive amines, stored in the granules (degranulation). The most significant primary mediators are
histamine, proteases, eosinophil chemotactic factor, neutrophil chemotactic factor, and heparin.
These mediators cause all the normal consequences of an acute inflammatory reaction – increased vascular
permeability, smooth muscle contraction, granulocyte chemotaxis and extravasation etc. Mast cell activation via FC
also leads to the production of two other types of mediators. These secondary mediators, unlike the stored granule
contents, must be synthesized de novo and comprise arachidonic acid derivatives (prostaglandins and leukotrienes),
platelet-activating factor, bradykinins, and various cytokines.

Allergen Allergen

FC receptor
IgE for IgE

Subsequent Degranulation
exposures
to allergen
Mast cell
binding
fragment
Sensitized Release of allergic
Mast cell mediators

Figure 5.53 Ag induces crosslinking of IgE bound to mast cells and basophils with release of vasoactive mediators.

Type I hypersensitivity can be anaphylaxis or atopy. Anaphylaxis is a very rapid, life-threatening, severe whole
body allergic reaction. It is caused by re-exposure to a previously encountered antigen. Atopy (atopic allergy) is a
hereditary tendency to develop allergic reaction to substances such as pollen, food, insect venom etc.

Type II Hypersensitivity
Type II hypersensitivity is generally called a cytolytic or cytotoxic reaction because it results in the destruction of
host cells, either by lysis or toxic mediators. Type II Hypersensitivity is caused by antibodies binding to cells or
526 Immunology
tissue antigens. The antibodies are of the IgM or IgG classes and cause cell destruction by FC dependent mechanisms
either directly or by recruiting complement via the classical pathway. Classical examples of type II hypersensitivity
reactions are the response exhibited by a person who receives a transfusion with blood from a donor with a different
blood group and erythroblastosis fetalis.
Two different antibody-mediated mechanisms are involved in these cytotoxic reactions. In complement-mediated
hypersensitivity reactions, the antibodies react with a cell membrane component, leading to complement fixation.
This activates the complement cascade and leads either to lysis of the cell or opsonization. Blood cells are most
commonly affected by this mechanism.

+ Complement
activation

Target cell

Figure 5.54 Antibody subclasses activate the complement system, creating pores in the membrane of a foreign cell.

Antibody-dependent cell mediated cytotoxicity (ADCC) used FC receptors expressed on many cell types (e.g. natural
killer cells, macrophages, neutrophils, eosinophils) as a means of bringing these cells into contact with antibody-
coated target cells. Lysis of these target cells requires contact but does not involve phagocytosis or complement
fixation. Instead, ADCC lysis of target cells is analogous to that of cytotoxic T cells and involves the release of
cytoplasmic granules containing perforin and granzymes that activate events leading to apoptosis. ADCC reactions
involve IgG and IgG Fc receptors.

FC receptor

+ Apoptosis of
target cell

Target cell NK cells

Figure 5.55 Antibodies mediate cell destruction by antibody dependent cell-mediated cytotoxicity (ADCC), in which
cytotoxic cells bearing FC receptors bind to the FC region of antibodies on target cells and promote killing of the cells.

Type III Hypersensitivity


Type III hypersensitivity is mediated by immune complexes essentially of IgG antibodies with soluble antigens.
Circulating immune complexes may lodge at various tissue sites where they activate complement and subsequently
cause tissue cell lysis. Normally these complexes are phagocytosed effectively by the fixed monocytes and
macrophages of the monocyte-macrophage system. In the presence of excess amounts of some soluble antigens,
the antigen-antibody complexes may not be efficiently removed. Their accumulation can lead to a hypersensitivity
reaction from complement that triggers a variety of inflammatory process. This form of hypersensitivity has a lot
in common with type I except that the antibody involved is IgG and therefore not prebound to mast cells.

Type IV Hypersensitivity
Type IV hypersensitivity, commonly called delayed type hypersensitivity, is the only class of hypersensitive
reactions which are triggered by antigen-specific T-cells (cell-mediated immunity reactions). This is mediated by
T-cell dependent effector mechanisms involving TH cells, primarily of the TH1 subtype, but in a few cases TC-cells.
Immunology 527

Antibodies do not play a role in type IV hypersensitivity reactions. On activation, the TH1 cells release cytokines
that cause accumulation and activation of macrophages, which, in turn, cause local damage. The tuberculin skin
test is an example of a type IV hypersensitivity. This test is done by putting a small amount of tuberculin purified
protein derivative (PPD) under the top layer of skin. If any person has ever been exposed to the Mycobacterium
tuberculosis, skin will react to the antigens by developing a firm red bump at the site within 2 days. It is a standard
method of determining whether a person is infected with Mycobacterium tuberculosis.

Overview of Hypersensitivity
IgE-mediated

Type I hypersensitivity
Ab-mediated

IgG/IgM-mediated
Immediate
Type II hypersensitivity
Symptoms are manifested
within minutes or hours Ag-Ab mediated IgG-mediated
after exposure
Type III hypersensitivity
Hypersensitivity

Delayed T-cell mediated


Symptoms are manifested Type IV hypersensitivity
within days after exposure

5.17 Autoimmunity
The body is normally able to distinguish its own self-antigens from foreign nonself antigens and does not mount an
immunologic attack against itself. This phenomenon is called immune tolerance. Autoimmunity is a condition in
which structural or functional damage is produced by the action of immunologically competent cells or Ab against
self antigen. Autoimmunity literally means protection against self, but actually it implies injury to self, and therefore
sometimes the term is also under criticism.
Autoimmune disease results from the activation of self-reactive T and B-cells that, following stimulation by genetic
or environmental triggers, cause actual tissue damage. Four factors influence the development of autoimmune
disease. These factors are genetic, viral, hormonal and psycho-neuro-immunological (the influence of stress and
neurochemicals). All four of these factors can affect gene expression, which directly or indirectly interferes with
important immunoregulatory actions. Based on the site of involvement and nature of lesions autoimmune diseases
may be classified as hemocytolytic, localized (or organ specific), systemic (or non-specific) and transitory diseases.
Important examples of autoimmune diseases in human and their respective autoantigen are given below in the table.

Table 5.14 Some autoimmune diseases in humans

Disease Autoantigen

Autoimmune hemolytic anemia Rh blood group

Graves disease Thyroid-stimulating hormone receptor

Multiple sclerosis Myelin basic protein

Myasthenia gravis Acetylcholine receptor

Rheumatoid arthritis Unknown synovial joint antigen

Systemic lupus erythematosus DNA, histones, snRNP

Type 1 diabetes mellitus Pancreatic beta cell antigen


528 Immunology

5.18 Transplantation
The immune system has evolved as a way of discriminating between self and non-self. This discriminating power
of the immune system between self and non-self is undesirable in the case of tissue transplant from one individual
to another for therapeutic purposes. Indeed, result of transplants culminates in the phenomenon of graft rejection.
Before the discussion about the immunological mechanisms associated with graft rejection, it is important to
understand the various gradations in relationship from donor to recipient.
Isograft : Graft between genetically identical individuals (syngeneic). In humans, an isograft (or syngraft) can
be performed between monozygotic twins.
Allograft : Transplants between genetically different individuals within a species.
Xenograft : A graft between individuals from different species.
Autograft : A graft or transplant from one body part to another on the same individual.

Transplanting tissue that is not immunologically privileged generates the possibility that the recipient’s cells will
recognize the donor’s tissue as foreign. This triggers the recipient’s immune mechanisms, which may destroy the
donor tissue. Such a response is called a graft rejection reaction. Some transplanted tissues do not stimulate an
immune response. For example, a transplanted cornea is rarely rejected because lymphocytes do not circulate into
the anterior chamber of the eye. This site is considered an immunologically privileged site. Another example of a
privileged tissue is the heart valve.
A tissue rejection reaction can occur by two different mechanisms. First, foreign class II MHC molecules on
transplanted tissue, or the graft is recognized by host T-helper cells, which aid cytotoxic T-cells in graft destruction.
Cytotoxic T-cells then recognize the graft through the foreign class I MHC molecules. This response is much like the
activation of CTLs by virally infected host cells. A second mechanism involves the T-helper cells reacting to the graft
and releasing cytokines. The cytokines stimulate macrophages to enter, accumulate within the graft, and destroy
it. The MHC molecules play a dominant role in the tissue rejection reaction because of their unique association
with the recognition system of T-cells.

5.19 Immunodeficiency diseases


Immunodeficiencies occur when one or more components of the immune system is defective. Immunodeficiencies
may be primary and secondary.

A deficiency caused by a defect in one or more genes involved in the development or function of the immune system
is called primary immunodeficiency.

A deficiency in the immune system that is acquired after birth, usually because of infection and that is not related
to a genetic defect is called secondary or acquired immunodeficiency.

Example of primary immunodeficiency diseases:

Severe combined immunodeficiency (SCID)


SCID is a genetic disorder which is characterized by a very low number of circulating lymphocytes. Both arms
(B-cells and T-cells) of the adaptive immune system become non-functional. As such patients make neither specific
T-cell dependent antibody responses nor cell-mediated immune responses, and thus cannot develop immunological
memory. Several different defects can lead to the SCID phenotype. In X-linked SCID, which is the commonest
form of SCID, T-cells fail to develop because of defect in the genes code for several cytokine receptors, including
those for the interleukins IL-2, IL-4, IL-7, IL-9 and IL-15. The autosomally inherited SCID occurs due to adenosine
deaminase deficiency. Adenosine deaminase catalyzes conversion of adenosine to inosine, and its deficiency results
in accumulation of adenosine, which interferes with purine metabolism which result in an accumulation of nucleotide
metabolites that are particularly toxic to developing T-cells.
This page intentionally left blank.
Chapter 06

Diversity of Life

6.1 Taxonomy
Taxonomy (arrangement by the rules) is the branch of biology that deals with identification (placement of a new
organism into a previously described group), nomenclature (the naming of organisms) and classification (ordering
of organisms into groups- can be phenetic or phylogenetic) of organisms. Systematics is the process of organizing
taxonomic information about organisms into a logical classification that provides the framework for all comparative
studies. It is the scientific study of biological diversity and its evolutionary history. Systematics and taxonomy are
collectively referred to as the systematic biology.

Levels of taxonomy
There are three levels of taxonomy:
Alpha taxonomy : It is concerned with finding, describing and naming of organisms. This is the first and most
basic step in taxonomy.
Beta taxonomy : It includes identification of natural groups and biological classes.
Gamma taxonomy : It includes study of evolutionary processes and patterns.
Organisms were first classified more than 2,000 years ago by Greek philosopher Aristotle. He classified organisms
as either plant or animal. Modern biological classification began with the eighteenth century Swedish naturalist
C. Linnaeus. He established a simple system for classifying and naming organisms. He developed a hierarchy
(a ranking system) for classifying organisms that is the basis for modern taxonomy.

6.1.1 Nomenclature
Nomenclature is the formal naming of a particular organism according to some standardized system. The fundamental
principle of nomenclature is that each organism must have only one scientific name. In contrast to scientific names,
many organisms also bear common names (also called vernacular names), which are generally used by people
within a limited geographic region. Presently, the criteria for scientific naming of plants, algae and fungi are based
on the rules and recommendations of the International Code of Nomenclature or ICN. It was formerly called the
International Code of Botanical Nomenclature (ICBN); the name was changed at the International Botanical Congress
in July 2011. Similarly, International Code of Zoological Nomenclature (ICZN) provides rules for naming animals. The
scientific names of species are binomials (literally meaning two names). Swedish botanist Linnaeus (referred to as
the father of taxonomy) first proposed the system of binomial nomenclature (or binary nomenclature). According
to binomial nomenclature, the first name of the binomial is the genus name (generic name). The second name of
the binomial is the specific epithet or species epithet (specific name - a term used only in zoology). The first word
denoting the genus starts with a capital letter while the specific epithet starts with a small letter. Binomial names
are always either italicized or underlined to indicate their Latin origin. For example, Mangifera indica is the scientific
name of mango. In this name Mangifera represents the genus while indica, is a specific epithet. A binomial name
in which the name of the genus and that of the species are identical is called tautonym.
538 Diversity of Life
Nomenclature types

The scientific names must be associated with some physical entity, known as a nomenclatural type or simply type.
A nomenclatural type is one particular specimen of an organism to which the scientific name of that organism is
formally attached. Types are usually physical specimens that are kept in a museum or herbarium research collection,
but it may also be an illustration. The type serves the purpose of acting as a reference for the name, upon which
the name is based. There are different types of nomenclatural type:
Holotype : A holotype is the one specimen or illustration upon which a name is based.
Isotype : An isotype is a duplicate specimen of the holotype, collected at the same time by the same person
from the same population.
Lectotype : A lectotype is a specimen that is selected from the original material to serve as the type when no
holotype was designated at the time of publication, if the holotype is missing, or if the original
type consisted of more than one specimen or taxon.
Neotype : A specimen later selected to serve as the single type specimen when an original holotype has been
lost or destroyed, or where the original author never cited a specimen.

6.1.2 Classification
Classification is the arrangement of taxa into some type of order. The purpose of classification is to provide a system
for cataloguing and expressing relationships between taxa. Classification is not a single step process but involves
hierarchy of steps in which each step represents a category termed rank. Rarely, the term taxonomic category is
used instead of rank. Every organism can be classified at seven taxonomic ranks- kingdom, phylum, class, order,
family, genus and species. Each rank contains organisms with similar characteristics. The kingdom is the largest
unit of classification. It splits into smaller units called phyla (singular, phylum). Phyla splits into classes, classes
into orders, orders into families, families into genera (singular, genus) and genera into species.

Example:
Seven taxonomic ranks Taxa
Kingdom Plantae
Phylum (Division) Magnoliophyta
Class Liliopsida
Order Arecales
Family Arecaceae
Genus Cocos
Species Cocos nucifera

The various units of classification – kingdom, phylum, class and so on — are called taxonomic categories and together
they make up the taxonomic hierarchy. A taxon is a defined group of organisms typically treated at a given rank.
In the above example, Magnoliophyta is a taxon placed at the rank of phylum; Liliopsida is a taxon placed at the
rank of class. As one progresses down the hierarchy, the number of organisms in each taxon decreases, and the
similarities between them increases.

6.1.3 Biological species concept


Biologists almost universally agree that the species is a fundamental natural unit. However, biologists have not
been able to agree on exactly how species should be defined. Taxonomists practically define species by means of
morphological or phenetic characters. If one group of organisms consistently differs from other organisms, it will
be defined as a separate species.
However, the most widely accepted species concept is the biological species concept. It defines species in terms of
interbreeding. According to Ernst Mayr, species are groups of interbreeding natural populations that are reproductively
This page intentionally left blank.
Diversity of Life 541

0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1.0

Coefficient of association

Figure 6.1 Phenogram. The higher the coefficient of association, the more closely related are the OTUs.

Measurement of similarity for binary characters (also called two-state characters)

The simplest and most common type of character used by taxonomist is one that can occur as one of only two
states and designated as 1 and 0. ‘1’ might indicate the presence of a feature, and ‘0’ its absence or ‘1’ and ‘0’
may represent two alternative forms of a character, such as ‘red’ and ‘white.’

0 1

0 M00 M10
B
1 M01 M11

Given below are two OTUs A and B, each with n binary characters. The character states are compared from
one OTU to the next.
M11 represents the total number of characters positive for both A and B.
M01 represents the total number of characters that are negative for A and positive for B
M10 represents the total number of characters that are positive for A and negative for B.
M00 represents the total number of characters negative for both A and B.
Many similarity coefficients have been proposed for binary data of this kind but two have been commonly used
in taxonomy – simple matching coefficient and Jaccard’s coefficients.

Simple matching coefficient

It is the ratio of the total number of character states for each character which are common between two OTUs
to the total number of characters.
M00 + M11
Simple matching coefficient (SMC) =
M00 + M01 + M10 + M11

Jaccard’s coefficient (or Jaccard similarity coefficient or Jaccard index)

It is the ratio of the number of positive matches to the total number of characters minus the number of negative
matches.
M11
Jaccard' s coefficient =
M01 + M10 + M11

When used for binary characters, the Jaccard coefficient is very similar to the simple matching coefficient. The
main difference is that the simple matching coefficient has the term M00 in its numerator and denominator,
whereas the Jaccard coefficient does not. Thus, the simple matching coefficient compares the number of matches
with the entire set of the possible attributes, whereas the Jaccard coefficient only compares it to the attributes
that have been chosen by at least A or B.
542 Diversity of Life

Cladistics
The evolutionary relationship of organisms is referred to as phylogeny. Some biologists feel that classification
should be based on phylogeny, and in particular on the points at which different groups have diverged from each
other. The phylogenetic principle classifies species according to how recently they share a common ancestor. Two
species that share a more recent common ancestor will be put in a group at a lower taxonomic rank than two
species sharing a more distant common ancestor.
The branch of systematics concerned with inferring phylogeny is termed as cladistics. It can be defined as the
concepts and methods for the determination of branching patterns of evolution. Cladistic relationship refers to the
paths of the ancestral lineages and therefore describes the sequence of branching of the ancestral lines. Some
workers, however, have preferred the term phylogenetic systematic or phylogenetic taxonomy to emphasize the
reliance on phylogeny for classification. This is the most widely used method of classifying organisms today. The
approach focuses on the branching of one lineage from another in the course of evolution.

Synapomorphies are the basis for cladistics


‘Character’ refers to any heritable trait possessed by an organism. Biologists distinguish two different types of
characters: homoplasies and homologies. A homoplasy (or a homoplasious character) is a character that species
share through convergent evolution. It is a similarity in appearance but not in origin. The wings of birds and bats
are an example of a homoplasy. The homoplasious characters shared between species that were not present in
the common ancestor. These characters are not an indicator of phylogenetic relations. Homoplasy also refers to
analogous structures which is similarity in function but not in origin.
A homology (or a homologous character) is a character shared between species that was also present in their
common ancestor (i.e. similarity in origin). Homologous characters in turn are divided into shared derived (advance)
characters and shared ancestral (primitive) characters. A shared ancestral character (or symplesiomorphic character)
is shared with the species ancestral to more than one group. A shared derived character (or synapomorphic
character) is a trait that is shared by two or more taxa and their most recent common ancestor, whose ancestor
in turn does not possess the trait. Characters that are unchanged from those of the ancestors are called ancestral
or plesiomorphic whereas characters that are changed in the descendants are called derived or apomorphic.

Homoplasious characters
All characters
that are similar Shared ancestral
in different species or primitive character

Homologous characters

Shared derived
or advance character

The phylogenetic relation comes from shared derived homologies. For example, all mammals have backbones, but
the presence of a backbone does not distinguish mammals from other vertebrates because all vertebrates have
backbones. Thus, we say that for mammals, the backbone is a shared ancestral character. In contrast, hair is a
character shared by all mammals but not found in their ancestors. Thus, in mammals, hair is considered a shared
derived character, unique to a particular clade. Note that a backbone can also qualify as a shared derived character,
but only at a node that distinguishes all vertebrates from other animals.
The basic idea behind cladistics is that members of a group share a common evolutionary history, and are closely
related, more so to members of the same group than to other organisms. These groups are recognized by sharing
unique features which were not present in distant ancestors (shared derived characters). Hence the first step in
basic cladistic analysis is to determine which character states are primitive or ancestral and which are derived.
The outgroup comparison method is generally used to determine this. Outgroup comparison is based on the
assumption that homologies present in both the outgroup and ingroup must be primitive characters that predate
the divergence of both groups from a common ancestor.
This page intentionally left blank.
550 Diversity of Life
The cellular slime molds exist in two forms. When nutrients are abundant, these slime molds exist as solitary
free-living amoeba-like cells. Each amoeboid cell has a haploid nucleus and divides by mitosis. When nutrients
become inadequate, many of these single-celled uninucleated, haploid organisms aggregate and move as a single
body. The aggregated form is called a pseudoplasmodium, or slug. Eventually, slug forms fruiting body containing
spores. After being released, each spore germinates and a single haploid amoeboid cell emerges. Cellular slime
molds differ from plasmodial slime molds in being haploid organisms (only the zygote is diploid) and in having
fruiting bodies that function in asexual rather than sexual reproduction. Dictyostelium discoideum, a cellular slime
mold, is a model organism for studying the evolution of multicellularity.

6.3.4 Oomycetes
Oomycetes or oomycota (oomycete means egg fungus) include the water molds, the white rusts and the downy
mildews. Although oomycetes have traditionally been placed in kingdom fungi and are still studied by mycologists,
they are not true fungi. Unlike true fungi, the oomycetes do not have cell walls made of chitin. In the five-kingdom
system, they are currently placed in the kingdom protista. Although oomycetes descended from plastid-bearing
ancestors (closely related to brown algae), they no longer have plastids and do not perform photosynthesis. Instead,
they typically acquire nutrients as decomposers or parasites. The most notable and best-studied oomycete species
is Phytophthora infestans, causing late blight of potato.

6.4 Fungi
Fungi are heterotrophic, spore bearing eukaryotes. Fungi may be single-celled or multicellular. Multicellular fungi
are composed of networks of long filamentous, branched structure called hyphae. The hyphae often aggregate
in a dense network known as a mycelium. The hyphae may be without cross walls (as in the case of lower fungi)
or divided into compartments by formation of septa (in the higher fungi). However, even in the septate hyphae,
the cytoplasm of the cells is continuous via a central pore in the septum. In certain stages, often during transition
to the sexual or asexual reproduction phase, the mycelium forms tissue like aggregates, called plectenchyma. A
typical plectenchyma is the flesh of the mushroom. Although fungi lack true organs, the mycelia of ascomycetes and
basidiomycetes may become organized into more complex reproductive structures called fruiting bodies, or sporocarps.

Table 6.2 Some differentiating characteristics of plants, animals and fungi

Character Animals Plants Fungi

Mode of nutrition Heterotrophy Autotrophy Heterotrophy

Chloroplasts No Yes No

Cell wall (constituent) Cell wall absent Cellulose and lignin Mainly chitin and glucans

Sterols Cholesterol Sitosterol/stigmasterol Ergosterol

Food storage Glycogen Starch Glycogen

Alternation of generations No Yes Yes

The major divisions of fungi are mainly classified based on their sexual spores and sexual reproductive structures.
Currently, four major divisions are recognized:
• The chytridiomycota are commonly known as chytrids. These fungi produce zoospores that are capable of
moving on their own through a liquid medium by simple flagella.
• The zygomycota are known as zygomycetes and reproduce sexually with meiospores called zygospores and asex-
ually with sporangiospores. Black bread mold (Rhizopus stolonifer) is a common species that belongs to this group.
• The ascomycota, commonly known as sac fungi or ascomycetes, form meiotic spores called ascospores,
within a special sac-like structure called an ascus. Unlike zygomycetes, most ascomycetes bear their sexual
This page intentionally left blank.
552 Diversity of Life
Sexual reproduction in basidiomycetes is similar to that of ascomycetes. Sexually compatible haploid hyphae
fuse to produce a dikaryotic mycelium. This leads to the production of a basidiocarp. The most commonly known
basidiocarps are mushrooms, but they may also take many other forms. Club-like structures known as basidia
generate haploid basidiospores following karyogamy and meiosis. These basidiospores then germinate to produce
new haploid mycelia.

Table 6.3 Characteristics of the different phyla of fungi

Character Chytridiomycota Zygomycota Ascomycota Basidiomycota

Habitat Aquatic/terrestrial Terrestrial Terrestrial Terrestrial

Ploidy Diploid Monoploid Monoploid Dikaryotic, Monoploid

Motile stage Present Absent Absent Absent

Sexual spore Not confirmed Zygospore Ascospores Basidiospores

Asexual spore Holocarpic Sporangiospores/ Conidia Arthrospores, oidia,


Chlamydospores conidia

Pathogenic Obligate parasites Facultative parasites Facultative or obligate Facultative or obligate


relationship parasites parasites

6.4.1 Mycorrhiza
Mutualistic association between fungi and plant roots are called mycorrhizae (mycorrhizae means fungus roots).
Mycorrhizal fungi (fungi that form mycorrhizae) can deliver inorganic nutrients such as calcium and phosphate. In
exchange, the plants supply the fungi with organic nutrients.
There are several different types of mycorrhizal fungi. Ectomycorrhizal fungi form sheaths of hyphae over the
surface of a root and also grow into the extracellular spaces of the root cortex. Endomycorrhizal fungi extend their
hyphae into the root cells. When the fungal hyphae penetrate the cortical cells of the plant root, they form highly
branched structures called arbuscules, hence these mycorrhizas commonly are called arbuscular mycorrhizas
(AM). In many arbuscular mycorrhizae, roots harbouring arbuscules may also contain terminal balloon-like swelling
called vesicles. Such mycorrhizas are called vesicular-arbuscular mycorrhizas (VAM). In ectomycorrhizas, the fungus
surrounds but does not penetrate living cells in the roots. In the conifers, the hyphae grow between the cells of the
root epidermis and cortex, forming a characteristic highly branched network, called the Hartig net.

6.4.2 Lichens
Lichens are a mutualistic association of photosynthetic algae and fungi. The photosynthetic partners are typically
unicellular or multicellular green algae or cyanobacteria. The fungal component may be a member of ascomycetes
(mostly) or member of basidiomycetes (in a few cases). The fungus usually gives a lichen its overall shape and
structure, and tissues formed by hyphae account for most of the lichen’s mass. The algal partners generally occupy
an inner layer below the lichen surface.
The algae provide carbon compounds; the cyanobacteria also fix nitrogen and provide organic nitrogen. The fungi
provide their photosynthetic partners with a suitable environment for growth. The physical arrangement of hyphae
allows gas exchange, protects the photosynthetic partner, and retains water and minerals. The fungi of many
lichens reproduce sexually by forming ascocarps or basidiocarps whereas algal partner reproduce independently
of the fungus by asexual cell division.
Diversity of Life 553

6.5 Plantae
The kingdom plantae are multicellular eukaryotes with photosynthetic nutrition. Cells typically have cellulose wall,
vacuole, plastids and several photosynthetic pigments which always include chlorophyll a. It can be broadly divided into:

Cryptogamae (plant without seeds): Algae (Thallophyta), Bryophytes and Pteridophytes.


Phanerogamae (plant with seeds): Gymnosperm and Angiosperm.

Bryophyta, pteridophyta, gymnosperm and angiosperm are termed as the land plants or embryophytes. One major
innovation of land plants was the evolution of the embryo. The embryo is defined as an immature sporophyte. Land
plants can be vascular (or tracheophyta) and non-vascular. During the early evolution of land plants, nonvascular
land plants diverged before the vascular plants. Non-vascular plants are often informally called bryophytes (from
the Greek bryon, moss, and phyton, plant). Bryophytes include the liverworts, hornworts and mosses. The terms
bryophyta and bryophyte are not synonymous. Bryophyta is the formal taxonomic name for the phylum that
consists solely of mosses.

6.5.1 Plant life cycle


The sexual life cycle in plants alternate between diploid (sporophyte) and haploid (gametophyte) phases. Sporophyte
literally means spore-plant, and gametophyte means gamete-plant. The haploid gametophytic body produces
gametes by mitosis whereas the diploid sporophytic phase produces meiospores (sexual spores) by the process of
meiosis. The diploid phase produces the haploid phase by meiosis; the haploid gametes then fuse to make a zygote
that starts another diploid phase. Three distinct versions of a generalized sexual life cycle occur among plants:

Haplontic life cycle


In haplontic life cycle, diploid sporophytic phase is represented only by the one-celled zygote. There are no free-
living sporophytes. The dominant, photosynthetic phase is the free-living gametophyte. The haploid gametophyte
produce gametes by mitosis. Fusion of gametes (termed fertilization or syngamy) forms diploid zygote. Meiosis
occurs in the zygote (zygotic meiosis) which results in the formation of haploid meiospores. These spores in turn,
germinate and divide by mitosis to form a haploid gametophyte body once again. Most green algae such as Volvox,
Spirogyra and some species of Chlamydomonas represent this pattern of life cycle.

Haploid
(n)

Zoospores (n) Gametes (n)


(Male and Female)

Zygotic Fertilization
meiosis

(2n)
Zygote

Figure 6.6 In haplontic life cycle, the zygote divides by meiosis (zygotic meiosis) to form haploid cells. Each of these
cells divides by mitosis to produce a multicellular haploid individual that eventually gives rise to gametes by mitosis.
This page intentionally left blank.
Diversity of Life 561

Spores

Gametophyte
(Prothallus)
Antheridium

Archegonium

Sperms

Mature
sporangium

Embryo

Sorus
(Cluster of sporangia)

Young
sporophyte

Gametophyte

Rhizome
Adult sporophyte

Figure 6.11 Fern life cycle.

True roots arise from these rhizomes and absorb water and nutrients from the soil in which the fern is growing. The
leaves, which are called fronds, are borne on the rhizomes. The immature leaves are coiled and known as croziers.
When leaves in early stage are coiled, this type of development is called circinate vernation. The leaves bear
sporangia which differentiate on the lower surface of leaves, in clusters called sori. Meiosis takes place within these
sporangia, followed by differentiation of the haploid spores, which ultimately are released from the sporangia. Most
ferns are homosporous, producing single types of spore. Only a few genera of ferns are heterosporous producing
two different types of spores – micro and megaspores. The fern life cycle differs from that of a moss primarily in
the much greater development, independence, and dominance of the fern’s sporophyte.

6.5.6 Gymnosperm
The gymnosperms are a group of vascular seed plants. In these plants, the ovules are not enclosed by ovary wall
and remain exposed, both before and after fertilization. The term gymnosperm derives from the Greek word gymnos
meaning ‘naked’ and sperma, meaning ‘seed’. The seeds that develop after fertilization from ovule, are not covered,
i.e. are naked. Seeds are exposed on modified leaves (sporophylls) that usually form cones or strobili. In contrast,
angiosperm seeds are enclosed in fruits, which are mature ovaries. Thus gymnosperms are all fruitless plants.
562 Diversity of Life
General features
1. Terrestrial adaptation and dominant sporophytic phase.
2. Mostly woody plants.
3. Most gymnosperms lack vessels in their xylem (with the exception of the gnetophytes) and companion cells
in the phloem.
4. An ovule is produced, but this is not enclosed within ovary wall.
5. All are heterosporous (producing mega- and microspores). Microspores termed pollen grains make direct
contact with the ovule. Microgametophyte develops within the pollen wall (i.e. endosporic) and without male
sex organ, antheridium. Megaspore bears megagametophyte which contains multicellular female sex organ
(archegonium).
6. Gymnosperms differ from the seedless plants in not requiring water for sperm to swim in to reach the egg.
Thus, except cycads and the maidenhair tree, all gymnosperms have non-flagellated sperms.
7. No double fertilization to produce endosperm (although double fertilization has been documented in Ephedra
and Gnetum). The endosperm, nutritive tissue for the embryo, is haploid.

6.5.7 Angiosperms
Angiosperms (also called flowering plants) are a major group of land plants. The angiosperms, being seed plants,
exhibit a life cycle that is in many respects similar to that found in gymnosperms.

General features
1. Dominant sporophytic phase.
2. Vessels present in their xylem and companion cells in the phloem.
3. The ovule develops within ovary of a carpel and enclosed by ovary wall.
4. All are heterosporous (producing mega- and microspores). Micro or male gametophyte develops within
microspore (pollen grain) and without antheridium. Megaspores bears mega or female gametophyte and
without archegonium. The gametophytes of angiosperms are much reduced in size and cell number. The mature
microgametophyte within the pollen grain consists of only three cells, and the mature megagametophyte within
the ovule, in most angiosperms, consists of only seven cells and eight nuclei.
5. Pollination in angiosperms is indirect in which pollen is deposited on the stigma of the carpel rather than directly
on the ovule as is the situation in gymnosperms.
6. Double fertilization characteristically occurs in angiosperms to produce embryo as well as endosperm. The
endosperm, nutritive tissue for the future embryo, is not haploid (n) but triploid (3n) and forms after fertilization.

Table 6.4 Comparative features of different land plants

Bryophyta Pteridophyta Gymnosperm Angiosperm

Dominant phase Gametophyte Sporophyte Sporophyte Sporophyte

Vascular system Absent Present Present Present

Embryo Present Present Present Present

Sex organ Multicellular Multicellular Multicellular Absent

Male sex organ Present Present Absent Absent

Female sex organ Present Present Present Absent

Gamete (male) Motile Motile Non-motile Non-motile


Diversity of Life 563

Flower
Flower is a modified shoot of limited growth. Flower may be pedicilate (with stalk which is called pedicel) or sessile
(without stalk). The tip of the pedicel swells up to form a cone like structure, the thalamus or receptacle or torus. The
thalamus consists of nodes and highly reduced or indistinct internodes. In a typical flower, four types of structures
called floral leaves are arranged in four whorls. These four whorls of floral leaves are divided into two groups:
Non-essential or accessory whorls (calyx and corolla) and Essential whorls (androecium and gynoecium or pistil)
Calyx is the outermost whorl. It is a collection of sepals. It is generally green and leaf like and protects the flower
bud. Corolla is a collection of petals. The petals are generally large, brightly colored and may have fragrance and
help in pollination by attracting insects. The number of sepals and petals is usually constant for a species. Flowers of
monocotyledons usually have three sepals and three petals; those of dicotyledons usually have four or five of each.
Androecium is a collection of stamens (microsporophyll). A stamen has two parts, the filament and the anther. A
typical anther is a bilobed structure present at the tip of the filament. It bears four pollen sacs which are filled with
tiny grains called pollen or pollen grains. On the basis of the length of the stamens, there may be two different
conditions – didynamous (in this case out of four stamens two are long and two short, example, Ocimum) and
tetradynamous (in this case out of six, four are longer forming an inner whorl while two are short forming an
outer whorl, example, mustard family).
Gynoecium (or pistil) is a collection of carpels. A sterile gynoecium or pistil is known as pistillode. Each carpel has
three parts:
Ovary : It is the swollen basal part of the carpel that contains one or more ovules which later become seeds.
Each ovule encloses an embryo sac.
Stigma : It is attached to the style and receives the pollen grain.
Style : It forms the neck of the carpel to which stigma is attached. Styles may be described as terminal style
(if a style is lying in the same straight line with the ovary; e.g. China rose), lateral style (if a style
arises from the side of the ovary; e.g. Strawberry) and gynobasic style (if a style arising from the
depression in the centre of the ovary or directly from thalamus; e.g. Ocimum). When the base of the
style is swollen to form a pad-like structure it is termed as stylopodium (e.g. Coriandrum).

On the basis of a number of carpels, gynoecium may be simple (or monocarpellary) and compound. When the
gynoecium is made up of only one carpel (e.g. Pea), it is known as simple gynoecium. When the gynoecium is
made up of two or more carpels then it is known as compound gynoecium. Compound gynoecium may be bi-, tri-,
tetra- and pentacarpellary. The multicarpellary condition may be syncarpous (a pistil of two or more carpels; which
are fused, e.g. Melia) or apocarpous (a pistil of two or more carpels; which are free; e.g. Clematis) in nature.

Union of flower parts

In some flowers, all the parts are attached directly to the receptacle and are entirely free from each other. Thus
there may be separate sepals (polysepaly), separate petals (polypetaly), separate stamens and separate pistils. In
other, the members of a whorl may be more or less united with each other, a condition referred to as a coalescence
of parts. Coalescence may involve sepals (synsepaly or gamosepaly), petals (synpetaly or gamopetaly), stamens
or carpels. Stamens are named differently depending upon the parts of stamens fused. It may be adelphous (if
the filaments of different stamens are united and anthers remain free), syngenesious (if the anthers of different
stamens are united, but the filaments are free e.g. Helianthus spp.) and synandrous (if anthers as well as filaments
of the stamens are united throughout their whole length e.g. Cucurbita). In adelphous condition, the stamens
may be united by their filaments into one (monadelphous), two (diadelphous) or several (polyadelphous). The
attachment of stamens with the sepals, petals or gynoecium is called adhesion. It may be epiphyllous (stamens are
attached to perianth, example, Liliaceae), epipetalous (the stamens are attached to petals, example, sunflower)
and gynandrous (stamens remain attached to the carpels, example, Calotropis).
This page intentionally left blank.

S-ar putea să vă placă și