You are on page 1of 368

ALAGAPPA UNIVERSITY

[Accredited with ‘A+’ Grade by NAAC (CGPA:3.64) in the Third Cycle


and Graded as Category–I University by MHRD-UGC]
(A State University Established by the Government of Tamil Nadu)
KARAIKUDI – 630 003

Directorate of Distance Education

M.Sc. [Home Science – Nutrition and Dietetics]


II - Semester
365 21

NUTRITIONAL BIOCHEMISTRY
Authors
Rahul Dev Ambedkar, Assistant Professor, Sri Aurobindo College, Delhi
Dr. Pradeep Kumar, Research Associate-III, Department of Pediatrics, Army Hospital Research & Referral, New Delhi
Units: (1, 2.5, 7.0-7.2, 7.4-7.8, 9.0-9.3, 9.5-9.9, 11.0-11.2)
Pratyusha Kar, M.Sc. (Biochemistry)
Units: (2.0-2.4, 2.6-2.10, 5.4, 5.6-5.12, 6.0-6.1, 6.3, 6.5-6.9, 10, 11.3-11.8 )
Varsha Srivastava, Assistant Professor, Department of Nutrition and Dietetics, Manav Rachna International Institute of
Research and Studies, Faridabad
Units: (3.0-3.2, 3.4-3.9, 5.0-5.3, 5.5)
Dr Rakhi Mittal, Associate Professor, Ginni Devi Modi Girls PG College, Modi Nagar, Ghaziabad
Units (3.3, 4.3-4.10)
Vijay Kumar Jha, Assistant Manager, Quest Diagnostics, Gurgaon
Units (4.0-4.2, 12 )
Dr Kanchan Sandhu, Assistant Professor (Home Science), Punjab Agricultural University, Ludhiana
Unit (8)
Dr Vijay Kumar Jha, Assistant Manager, Quest Diagnostics, Gurgaon
Dr Sherry Khanna, Head, Laboratory Operations, Dr. Khanna’s Pathcare Pvt. Limited, Delhi
Units (13, 14)
Vikas Publishing House,
Units: (6.2, 6.4, 9.4, 7.3)

"The copyright shall be vested with Alagappa University"

All rights reserved. No part of this publication which is material protected by this copyright notice
may be reproduced or transmitted or utilized or stored in any form or by any means now known or
hereinafter invented, electronic, digital or mechanical, including photocopying, scanning, recording
or by any information storage or retrieval system, without prior written permission from the Alagappa
University, Karaikudi, Tamil Nadu.

Information contained in this book has been published by VIKAS® Publishing House Pvt. Ltd. and has
been obtained by its Authors from sources believed to be reliable and are correct to the best of their
knowledge. However, the Alagappa University, Publisher and its Authors shall in no event be liable for
any errors, omissions or damages arising out of use of this information and specifically disclaim any
implied warranties or merchantability or fitness for any particular use.

Vikas® is the registered trademark of Vikas® Publishing House Pvt. Ltd.


VIKAS® PUBLISHING HOUSE PVT. LTD.
E-28, Sector-8, Noida - 201301 (UP)
Phone: 0120-4078900 x Fax: 0120-4078999
Regd. Office: 7361, Ravindra Mansion, Ram Nagar, New Delhi 110 055
x Website: www.vikaspublishing.com x Email: helpline@vikaspublishing.com

Work Order No. AU/DDE/DE1-291/Preparation and Printing of Course Materials/2018 Dated 19.11.2018 Copies - 500
SYLLABI-BOOK MAPPING TABLE
NUTRITIONAL BIOCHEMISTRY
Syllabi Mapping in Book

BLOCK-I: CARBOHYDRATES AND PROTEINS


Unit I Unit 1: Carbohydrates
Carbohydrates - Classification, Physical and Chemical Properties, Sources, (Pages 1-30)
Biological Role. Unit 2: Carbohydrate
Unit II Metabolism
Carbohydrate Metabolism - Glycolytic Pathway, Deficiency Diseases, Inborn (Pages 31-64)
Errors of Carbohydrate Metabolism. Nutritional Aspects of Carbohydrate. Unit 3: Proteins
Unit III (Pages 65-90)
Proteins - Classification, Physical and Chemical Properties, Sources, Biological Unit 4: Protein Metabolism
Role and Value of Protein. (Pages 91-114)
Unit IV
Protein Metabolism - Protein Synthesis, Deficiency Diseases and Inborn Errors
of Protein Metabolism.

BLOCK-II: LIPIDS, VITAMINS AND MINERALS


Unit V
Lipids - Classification, Physical and Chemical Properties, Sources, Biological Unit 5: Lipids
Role. (Pages 115-156)
Unit VI Unit 6: Lipid Metabolism
Lipid metabolism - E-Oxidation. Nutritional Aspects of Lipids, Lipid Based (Pages 157-172)
Metabolic Diseases, In-Born Errors of Lipid Metabolism. Unit 7: Vitamins
Unit VII (Pages 173-202)
Vitamins - Classification, Characteristics, Role of Vitamins in Metabolism, Unit 8: Minerals
Deficiency Diseases. (Pages 203-220)
Unit VIII
Minerals - Types, Absorption and Role of Minerals in Metabolism, Minerals
Deficiency Diseases.

BLOCK-III: NUCLEIC ACIDS AND ENZYMES


Unit IX
Nucleic Acids - DNA & RNA, Structure, Function and Metabolism, Genetic Unit 9: Nucleic Acids
Disorders. (Pages 221-258)
Unit X Unit 10: Enzymes
Enzymes - Classification, Nomenclature, Mechanism of Enzyme Action, (Pages 259-270)
Enzyme Specificity, Application of Enzymes in Clinical Diagnosis. Unit 11-Enzyme Activity
Unit XI (Pages 271-294)
Enzyme Activity - Factors Affecting Enzyme Activity, Co- Enzymes and Co-
Factors.
BLOCK-IV: HORMONES, BUFFERS AND ELECTROLYTES
Unit XII
Hormones - Role of Hormones. Interrelation Between Hormones and Nutrients. Unit 12: Hormones
Hormone Deficiency Diseases. (Pages 295-324)
Unit XIII Unit 13: Acid-Base Balance
Acid Base Balance - Normal Health, Major Sources of Acid Produced in the (Pages 325-338)
Body, Buffers, Physiological Role of Different Buffer Systems. Unit 14: Fluid and
Unit XIV Electrolyte Balance
Fluid and Electrolyte Balance - Maintenance in Normal Health. Diseases of (Pages 339-358)
Electrolytes Imbalance. Role of Nutrients in Maintenance of Fluid and
Electrolyte Balance during Disease Condition.
CONTENTS
INTRODUCTION

BLOCK I: CARBOHYDRATES AND PROTEINS


UNIT 1 CARBOHYDRATES 1-30
1.0 Introduction
1.1 Objectives
1.2 Carbohydrates: Structure and Classification
1.2.1 Monosaccharides
1.2.2 Anomeric Carbon
1.2.3 Oligosaccharides
1.2.4 Polysaccharides
1.2.5 Glycoconjugates
1.3 Chemical Properties of Carbohydrates
1.3.1 Functions of Carbohydrates
1.4 Answers to Check Your Progress Questions
1.5 Summary
1.6 Key Words
1.7 Self Assessment Questions and Exercises
1.8 Further Readings
UNIT CARBOHYDRATE METABOLISM 31-64
2.0 Introduction
2.1 Objectives
2.2 Metabolism of Carbohydrates
2.2.1 Fate of Carbohydrates
2.2.2 Fate of Pyruvate
2.2.3 Citric Acid Cycle, Krebs Cycle or Tricarboxylic Acid (TCA) Cycle
2.2.4 Gluconeogenesis
2.3 Glycogen Metabolism
2.3.1 Glycogenolysis
2.3.2 Glycogenesis
2.3.3 Pathway of HMP
2.3.4 Uronic Acid Pathway
2.3.5 Metabolism of Galactose
2.3.6 Metabolism of Fructose
2.3.7 Futile Cycle
2.4 Metabolic Defects Related to Carbohydrates
2.5 Entner Doudroff Pathway
2.6 Answers to Check Your Progress Questions
2.7 Summary
2.8 Key Words
2.9 Self Assessment Questions and Exercises
2.10 Further Readings
UNIT 3 PROTEINS 65-90
3.0 Introduction
3.1 Objectives
3.2 Protein and Its Structure
3.3 Classification of Proteins
3.4 Physical and Chemical Properties of Proteins
3.5 Answers to Check Your Progress Questions
3.5 Summary
3.7 Key Words
3.8 Self Assessment Questions and Exercises
3.9 Further Readings

UNIT 4 PROTEIN METABOLISM 91-114


4.1 Objectives
4.2 Protein Metabolism: An Introduction
4.2.1 Classification of Protein
4.2.2 Protein Synthesis
4.3 Deficiency Diseases of Proteins
4.4 Inborn Errors of Protein Metabolism
4.5 Entner Doudoroff Pathway
4.6 Answers to Check Your Progress Questions
4.7 Summary
4.8 Key Words
4.9 Self Assessment Questions and Exercises
4.10 Further Readings

BLOCK II: LIPIDS, VITAMINS AND MINERALS


UNIT 5 LIPIDS 115-156
5.0 Introduction
5.1 Objectives
5.2 Classification of Lipids
5.3 Properties of Lipids
5.4 Classfication of Fatty Acids
5.4.1 Types of Fats
5.4.2 Fatty Acyls
5.5 Properties of Fatty Acids
5.5.1 Saturated Fatty Acids
5.5.2 Unsaturated Fatty Acids
5.6 Phospholipid
5.7 Cholesterol Synthesis in E Coli
5.8 Answers to Check Your Progress Questions
5.9 Summary
5.10 Key Words
5.11 Self Assessment Questions and Exercises
5.12 Further Readings

UNIT 6 LIPID METABOLISM 157-172


6.0 Introduction
6.1 Objectives
6.2 Lipids
6.2.1 Lipid Metabolism
6.2.2 Lipid Peroxidation
6.3 Fatty Acid: Oxidation
6.4 Fatty Acid Metabolism
6.5 Answers to Check Your Progress Questions
6.6 Summary
6.7 Key Words
6.8 Self Assessment Questions and Exercises
6.9 Further Readings

UNIT 7 VITAMINS 173-202


7.0 Introduction
7.1 Objectives
7.2 Classification, Properties, and Functions of Vitamins
7.2.1 Fat-Soluble Vitamins
7.2.2 Water-Soluble Vitamins
7.3 Vitamins as Co-Factors and Co-Enzymes
7.4 Answers to Check Your Progress Questions
7.5 Summary
7.6 Key Words
7.7 Self Assessment Questions and Exercises
7.8 Further Readings

UNIT 8 MINERALS 203-220


8.0 Introduction
8.1 Objectives
8.2 Minerals in Food
8.3 Classification of Minerals
8.3.1 Major Elements
8.3.2 Trace Elements
8.4 Bioavailability and Deficiency of Minerals
8.4.1 Calcium
8.4.2 Iron
8.4.3 Iodine
8.4.4 Fluorine
8.4.5 Sodium
8.4.6 Potassium
8.5 Minerals in Food Processing
8.6 Answers to Check Your Progress Questions
8.7 Summary
8.8 Key Words
8.9 Self Assessment Questions and Exercises
8.10 Further Readings

BLOCK III: NUCLEIC ACIDS AND ENZYMES


UNIT 9 NUCLEIC ACIDS 221-258
9.0 Introduction
9.1 Objectives
9.2 Nucleic Acids: Structure
9.2.1 Nitrogenous Bases (Pyrimidine and Purine)
9.2.2 Nucleosides and Nucleotides
9.3 Ribonucleic Acid (RNA) and Deoxyribonucleic Acid (DNA)
9.3.1 Structure and Properties of DNA
9.3.2 Structure and Properties of RNA
9.3.3 DNA is Genetic Material
9.4 Synthesis and Degradation of Purines and Pyrimidines
9.4.1 Purine Nucleotide Synthesis
9.4.2 Pyrimidine Synthesis
9.4.3 Degradation of Purines and Pyrimidines
9.5 Answers to Check Your Progress Questions
9.6 Summary
9.7 Key Words
9.8 Self Assessment Questions and Exercises
9.9 Further Readings

UNIT 10 ENZYME 259-270


10.0 Introduction
10.1 Objectives
10.2 Classification, Chemical Nature and Properties of Enzymes
10.3 Factors Affecting Enzyme Activity
10.3.1 Factors Affecting the Rate of Enzyme Activity
10.4 Active Site of Enzyme
10.4.1 Structure of Active Sites
10.4.2 Models of Active Sites
10.4.3 Overview of Ligands
10.5 Answers to Check Your Progress Questions
10.6 Summary
10.7 Key Words
10.8 Self Assessment Questions and Exercises
10.9 Further Readings
UNIT 11 ENZYME ACTIVITY 271-294
11.0 Introduction
11.1 Objectives
11.2 Enzyme Activity
11.2.1 Factors Affecting Enzyme Activity
11.2.2 Michaelis-Menten Hypothesis
11.2.3 Models of Enzyme Action
11.3 Co-Enzymes and Co-Factors
11.4 Answers to Check Your Progress Questions
11.5 Summary
11.6 Key Words
11.7 Self Assessment Questions and Exercises
11.8 Further Readings

BLOCK IV: HORMONES, BUFFERS AND ELECTROLYTES


UNIT 12 HORMONES 295-324
12.0 Introduction
12.1 Unit Objectives
12.2 Introduction of Hormones
12.2.1 Structure and Function of Hormones
12.3 General Classification of Hormones
12.4 Steroid Hormone Receptors
12.5 Peptide Hormones Receptor
12.6 Hormone Receptors and Intracellular Messengers
12.7 Insulin
12.8 Role of Glucagon
12.9 Epinephrine and their Mechanism for Various Endocrine
12.10 Regulatory Systems Mediated by Cyclic AMP
12.11 Answers To Check Your Progress Questions
12.12 Summary
12.13 Key Terms
12.14 Self Assessment Questions And Exercises
12.15 Further Readings

UNIT 13 ACID-BASE BALANCE 325-338


13.0 Introduction
13.1 Objectives
13.2 Henderson–Hasselbalch Equation
13.3 Regulation of Acid–Base Balance
13.3.1 Blood Buffer System
13.3.2 Respiratory and Renal Regulation
13.4 Acid–Base Disorders
13.5 Anion Gap
13.6 Assessment of Acid–Base Homeostasis
13.7 Answers to Check Your Progress Questions
13.8 Summary
13.9 Key Words
13.10 Self Assessment Questions and Exercises
13.11 Further Readings

UNIT FLUID AND ELECTROLYTE BALANCE 339-358


14.0 Introduction
14.1 Objectives
14.2 Electrolytes
14.2.1 Sodium
14.2.2 Potassium
14.2.3 Calcium
14.2.4 Magnesium
14.2.5 Chloride
14.2.6 Bicarbonate
14.3 Electrolyte and Fluid Balance
14.4 Answers to Check Your Progress Questions
14.5 Summary
14.6 Key Words
14.7 Self Assessment Questions and Exercises
14.8 Further Reading
Introduction
INTRODUCTION

Biochemistry is a branch of science which is aimed at answering the questions,


NOTES such as ‘What is life made of?’, ‘How does it work?’ While the eye works at the
gross level of visible objects, the microscope reaches down to the cellular level,
exposing details of the various cell organelles, including nuclei and other particles
and the metabolic pathways of biomolecules in chemical language. In simple words,
‘Biochemistry’ is the study of living organisms.
Food is one of the basic needs of life and is required for growth, to maintain
good health, and recovery from illness. Food is composed of nutrients required
for our body. Upon consumption of food, organisms assimilate the nutrients present
in them and use it for growth and replacement of tissues. The process of assimilation
of nutrients is called ‘Nutrition’. In humans, nutrition specifically refers to the
consumption, absorption, utilization and excretion of essential chemical compounds
found in foods and drinks that are required by the body to produce energy as well
as to assist the body to grow and develop. There are six major classes of nutrients
which include carbohydrates, fats, proteins, vitamins, minerals and water.
Nutritional science is the study of nutrients, their function and how they are
involved in health and disease. The goal is to ensure specific nutritional guidelines
suitable for different groups of people depending on their age, sex, activity level
and special groups, such as in pregnancy or disease. It is a relatively new discipline
and began to evolve the last 100 years, even though the importance of diet to
maintain health was recognised a lot earlier. It is an applied subject that draws
information from many other biological areas particularly biochemistry, therefore
a good understanding of biochemistry is required to fully understand nutrition.
Nutritional b nic fruits and vegetables. It specifically focuses on nutrient
chemical components, and how they function metabolically, physiologically, and
biochemically. It also uses science such as physics, chemistry and biology. It is
study of nutrition as a science. Nutritional biochemical therapy refers to specific
nutrition procedures including assessment and interventions in the treatment of an
illness, injury or disease condition.
This book, Nutritional Biochemistry, is divided into four blocks that are
further divided into fourteen units which will help to understand the basics of
carbohydrates, their classification, physical and chemical properties, sources,
biological role, carbohydrate metabolism, Glycolytic pathway, deficiency diseases,
proteins, classification, physical and chemical properties, sources, biological role
and value of protein, protein metabolism, protein synthesis, deficiency diseases
and inborn errors of protein metabolism, lipids, classification, physical and chemical
properties, sources, biological role, lipid metabolism, b-oxidation, vitamins, minerals,
their types, absorption and role of minerals in metabolism, nucleic acids, DNA
and RNA, enzymes, enzyme activity, co-enzymes and co-factors, hormones, role
of hormones, interrelation between hormones and nutrients, acid base balance,
normal health, major sources of acid produced in the body, buffers, physiological
role of different buffer systems, fluid and electrolyte balance, diseases of electrolytes
imbalance, role of nutrients in maintenance of fluid and electrolyte balance during
disease conditions.
Self-Instructional
10 Material
Carbohydrates
BLOCK - I
CARBOHYDRATES AND PROTEINS

NOTES
UNIT 1 CARBOHYDRATES
Structure
1.0 Introduction
1.1 Objectives
1.2 Carbohydrates: Structure and Classification
1.2.1 Monosaccharides
1.2.2 Anomeric Carbon
1.2.3 Oligosaccharides
1.2.4 Polysaccharides
1.2.5 Glycoconjugates
1.3 Chemical Properties of Carbohydrates
1.3.1 Functions of Carbohydrates
1.4 Answers to Check Your Progress Questions
1.5 Summary
1.6 Key Words
1.7 Self Assessment Questions and Exercises
1.8 Further Readings

1.0 INTRODUCTION

Carbohydrates are polyhydroxy compounds that contain a carbonyl (C=O) group


and have the general formula (CH2O)n. It is an organic compound which consists
of the elements carbon (C), hydrogen (H) and oxygen (O) with a ratio of hydrogen
twice that of carbon and oxygen, i.e., 1:2:1. Sugars, starches, cellulose and many
other compounds are some examples of carbohydrates found in living organisms.
Carbohydrates are widely distributed in plants and animals. They are classified
on the basis of the number of monomers they contain. They are categorized mainly
into four categories; monosaccharides, oligosaccharides and glycoconjugates.
In this unit, you will study about the structure, properties, chemical reactions
and functions of these different types of carbohydrates in detail.

1.1 OBJECTIVES

After going through this unit, you will be able to:


x Understand carbohydrates
x Classify carbohydrates
x Know the reactions of carbohydrates
Self-Instructional
Material 1
Carbohydrates
1.2 CARBOHYDRATES: STRUCTURE AND
CLASSIFICATION

NOTES Carbohydrates have important structural and metabolic roles. In plants, glucose is
synthesized from carbon dioxide and water by photosynthesis and stored as starch
or used to synthesize cellulose for plant framework. Animals can also synthesize
carbohydrates from lipids and amino acids but most animal carbohydrates are
derived ultimately from plants. Glucose is the most important carbohydrate as it is
the most dietary carbohydrate absorbed in the bloodstream. The other sugars are
converted into glucose in liver. Glucose is the precursor for the synthesis of all
other carbohydrates in the body including glycogen (storage carbohydrate in animal
cells), ribose and deoxy ribose in nucleic acids and lactose in milk.
Carbohydrates are classified on the basis of the number of monomers they
contain. Their categories are as follows:
x Monosaccharides: Their formula is (CH2O)n where n = 3 – 9 and n
denotes the number of carbon atoms.
x Oligosaccharides: These are polymers from 2 – 20 molecules of
monosaccharides.
x Polysaccharides: These are polymers containing more than 20 sugar
residues.
x Glycoconjugates: These are derivatives of carbohydrates containing
proteins and lipids.
1.2.1 Monosaccharides
Monosaccharides or simple sugars are those carbohydrates that cannot be
hydrolysed into simpler carbohydrate containing a single polyhydroxy aldehyde
or ketone unit. Six-carbon sugar D-glucose (sometimes referred as Dextrose) is
the abundant monosaccharide found in nature. More than four carbons presented
in monosaccharide tend to have cyclic structures. Their general formula is Cn(H2O)n
or CnH2nOn where n is the number of carbon atoms which are from 3 to 9 and
cannot be less than 3.
These colourless, crystalline solids are freely soluble in water, but insoluble
in non-polar solvents like ether, benzene etc., Most of them have a sweet taste.
The simplest monosaccharide contains either aldehydes or ketones with two or
more hydroxyl groups. The most common six-carbon monosaccharide glucose
and fructose have five hydroxyl groups. Most of the carbon atoms presented in
monosaccharide remain attached to hydroxyl groups, called chiral centers or
chiral carbon (the carbon atom having four different functional groups) , which
give rise to many stereoisomers of sugar found in nature.
Self-Instructional
2 Material
The common monosaccharide molecules are made up of unbranched carbon Carbohydrates

chains, wherein all the carbon atoms are linked by single bonds. In the open-chain
structure of monosaccharide, one of the carbon atoms is double-bonded to an
oxygen atom to form a carbonyl group. Here, each of the other carbon atoms has
a hydroxyl group. The monosaccharide is known as an aldose sugar, if the carbonyl NOTES
group is at an end of the carbon chain, that is, in an aldehyde group, and if the
carbonyl group is at any other position, that is, in a ketone group, it is known as a
ketose sugar. The three-carbon trioses sugar comprises simplest monosaccharides;
glyceraldehyde, an aldotriose, and dihydroxyacetone, a ketotriose. This is illustrated
by Figure 1.1.

Fig. 1.1 Structure of Triose Sugar

Tetroses, pentoses, hexoses and heptoses are monosaccharides having four,


five, six, and seven carbon atoms in their framework respectively. There are aldoses
and ketoses of each of these chain lengths: aldotetroses and ketotetroses,
aldopentoses and ketopentoses, and so on as represented in Table 1.1. The two
most common monosaccharides found in nature are aldohexose D-glucose and
the ketohexose D-fructose. The aldopentoses D-ribose and ketopentose 2-deoxy-
D-ribose are major components of nucleotides and nucleic acids. Their structures
are shown in Figure 1.2

Ribose Deoxyribose

Fig. 1.2 Structure of Ribose and Deoxyribose

Sometimes, a distinction in naming between aldoses and ketoses is also


maintained. The suffix ‘oses’, is kept reserved for the aldoses and the suffix, ‘uloses’
is used for ketoses. Thus, glucose is a hexose and fructose is a hexulose.

Self-Instructional
Material 3
Carbohydrates Table 1.1 Nomenclature of Aldose and Ketose Sugar
No. of Category Formula Aldose Ketose
Carbon name
atoms
NOTES 3 Trioses C3H6O3 Glycerose Dihydroxyacetone
(Glyceraldehyde)
4 Tetroses C4H8O4 Erythrose Erythrulose

5 Pentoses C5H10O5 Ribose Ribulose

6 Hexoses C6H12O6 Glucose Fructose


7 Heptoses C7H14O7 Glucoheptose Sedoheptulose

A molecule with n chiral centers can have 2n stereoisomers commonly.


Stereoisomers are those isomeric molecules that have the same molecular formula
and have constituent sequence of bonded atoms, but differ only in the three-
dimensional orientations in space of their atoms. Structural isomers have the same
molecular formula, but they differ in the bond order between different atoms/
groups.The monosaccharide stereoisomers of each carbon–chain length can be
divided into two groups which differ in the configuration about the chiral center
most distant from the carbonyl carbon.
When the hydroxyl group, on the reference carbon is on the right in the
projection formula, the sugar is the Dextrorotatory (D) isomer and when the
hydroxyl group is on the left, it is the Levorotatory (L) isomer. The isomers in
which the configuration at this reference carbon is the same as that of D-
glyceraldehyde are called D isomers. And the isomers with the same configuration
as L-glyceraldehyde are called L isomers. Figure 1.3 shows the open chain
structure of D and L-isomer of glyceraldehydes.

D-Glyceraldehyde L-Glyceraldehyde
Fig. 1.3 Structure of D and L-isomer of Glyceraldehyde

Of the sixteen possible aldohexose–glucose configurations, eight occur in


D- isomeric form and the other eight occur in L- isomeric form. It is shown in
Figure 1.4. Most of the hexoses found in living organisms have D-isomeric form.
The numbering of carbon atoms of a sugar starts at the end of the carbon chain
nearest the carbonyl group. Stereo-isomeric form of each of the eight
D-aldohexoses differing at C-2, C-3, or C-4, are named as D-glucose, D-
Self-Instructional
4 Material
galactose, D-mannose, and so forth. The naming of four- and five-carbon ketoses Carbohydrates

are designated by inserting suffix ‘ul’ into the name of a corresponding aldose.
For example, D-ribulose is the ketopentose corresponding to the aldopentose
D-ribose.
NOTES

Fig. 1.4 Stereo-isomeric Possibilities of Glucose

In aqueous solutions, aldotetroses and all monosaccharides with framework


of carbon atoms in five or more carbon atoms occur predominantly as cyclic (ring)
structures. In these structures, the carbonyl group creates a covalent bond through
the oxygen atom of a hydroxyl group alongside the chain. These ring structures are
created as a result of a common reaction taking place between alcohols and
aldehydes or ketones to form derivative called hemiacetals or hemiketals, which
contain an extra asymmetric carbon atom and thus can exist in two stereoisomeric
forms. For example, D-glucose exists in solution as an intramolecular hemiacetal in
which the free hydroxyl group at C-5 has reacted with the aldehydic C-1, account
the latter carbon asymmetric and producing two stereoisomers, designated D and
E. This is shown in Figure 1.5.
These six-membered ring compounds are called pyranoses because they
resemble the six membered ring compound pyran. The methodical names for the
two ring forms of D-glucose are D-D-glucopyranose and E-D-glucopyranose.
Aldohexoses also exist in cyclic forms having five membered rings, which, because
they resemble the five membered ring compound furan, are known as furanoses.
However, the six-membered aldopyranose ring is much steadier than the
Self-Instructional
Material 5
Carbohydrates aldofuranose ring and predominates in aldohexose solutions. Aldoses having five
or more carbon atoms can form pyranose rings only.

NOTES

Fig. 1.5 Furan and Pyran Ring Structures of Glucose

Anomers are the monosaccharide isomeric forms that differ only in their
configuration about the hemiacetal or hemiketal carbon atom. The hemiacetal (or
carbonyl) carbon atom is called the anomeric carbon. In aqueous solution, a process
called mutarotation inter-converts D and E anomers of D-glucose. Thus, a solution
of D-D-glucose and E-D-glucose eventually form the same equilibrium mixtures
having indistinguishable optical properties. This mixture consists of about one-
third D -D-glucose, two-thirds E -D-glucose, and very small amounts of the linear
and five-membered ring (glucofuranose) forms. Ketohexoses also occur in D and
E anomeric forms. In these compounds, the hydroxyl group at C-5 (or C-6)
reacts with the keto group at C-2, forming a furanose (or pyranose) ring containing
a hemiketal linkage. D-Fructose eagerly forms the furanose ring. The more ordinary
anomer of this sugar in combined forms or in derivatives is E -D-fructofuranose.
The two sugars that vary only in the configuration around one carbon atom are
known as epimers. For example, D-glucose, and D-mannose. D-glucose and
which vary only in the stereochemistry at C-4 are D-galactose. This is shown in
Figure 1.6. Most of the sugars occur naturally in their D-isomeric form. However,
a few sugars may occur naturally in their L-isomeric form.

Self-Instructional
6 Material
Carbohydrates

NOTES

C-2 epimer C-4 epimer

Fig. 1.6 Structure of Epimeric Form of Glucose

Comparatively mild oxidizing agents such as ferric (Fe3+) or cupric (Cu2+)


ion convert carbonyl carbon present in monosaccharides to a carboxyl group by
oxidation. On the basis of oxidizing activity, monosaccharides are categorized into
two classes. Monosaccharides (such as glucose) which are capable of reducing
ferric or cupric ion are known as reducing sugars and those sugars which are
incapable of reducing ferric or cupric ion are known as non-reducing sugars. The
differences between reducing and non-reducing sugars are presented in Table
1.2.
Fehling’s reaction, a qualitative test for the presence of reducing sugar is
based on the property of reduction. Thus, it is possible to estimate the concentration
of sugar by measuring the amount of oxidizing agent reduced by a solution of sugar.
Table 1.2 Differences between Reducing and Non-Reducing Sugar

Reducing Sugar Non-Reducing Sugar


1. Carbohydrates with a free aldehyde (at C-1). 1. Aldehyde or ketone group is not free
but or a free ketone (at C-2) group.
instead utilized in bond formation.
2. They are in hemiacetal or hemiketal form. 2. They are in acetal or ketal form.
3. Do exhibit mutarotation. 3. Do not exhiblit mutarotation.
4. Do form osazones with phenyl hydrazine. 4. Do not form osazones.
5. Do form oximes with hydroxylamine. 5. Do not form oximes.
Examples — Glucose, Fructose, Examples — Sucrose, Glycogen, Inulin,
Lactose, Maltose, Cellobiose, etc. etc.
Self-Instructional
Material 7
Carbohydrates 1.2.2 Anomeric Carbon
An anomer is a special type of epimer. It is one of two stereoisomers of a cyclic
saccharine that differ in configuration at the hemiacetal or hemiketal carbon, also
NOTES known as the anomeric carbon (Refer Figure 1.7). The conversion of one anomer
to another is known as anomerization. Anomer is either of a pair of the cyclic
stereoisomer possessing different physical properties (called D or E) of a sugar or
glycoside. These differ only in configuration at the reducing carbon atom.

Fig. 1.7 Structure of Anomeric Carbon

Anomerization is a process that converts one anomer to another and takes


place in solutions for reducing sugars. It is a reversible that results in an anomeric
mixture wherein an equilibrium is eventually achieved between the two single
anomers. There is a specific ratio of the two single anomers for the concerning
sugar. If a configuration of the starting D-glucose, a solution will gradually move
towards being a mixture of approximately 64% E-D-glucopyranoside and 36%
of D-D-glucopyranose (Refer Figure 1.8). If the ratio changes, the optical rotations
will also change and this phenomenon is known as mutarotation.

Fig. 1.8 Anomeric Structure of D-Glucose

Anomerization Mechanism
The cyclic forms of sugars are generally profoundly favoured, liquid monosaccharides
(monosaccharides in aqueous solution) are always in equilibrium with their open-
chain forms. In aldohexoses, this equilibrium is seen as the hemiacetal bond between
C-1 (the only carbon bound to 2 oxygen) and C-5 is carbon cleaved (forming open
chain compound) and then reformed (forming cyclic compound).
Self-Instructional
8 Material
Once a hemiacetal bond is reformed, the OH group on C-5 may attack Carbohydrates
either of the two stereochemically distinct sides of the aldehyde group on C-1,
and depending on the side it attacks, it is decided whether D or E anomer is
formed. When the OH group is on the downside, the two stereoisomers at the
hemiacetal (anomeric) carbon are called alpha anomer and when OH group is on NOTES
the up side, it is called beta anomer.
1.2.3 Oligosaccharides
These are compound sugars that yield two to ten molecules of the identical or
different monosaccharides on hydrolysis (removal of water molecules). Accordingly,
an oligosaccharide yielding two molecules of monosaccharide on hydrolysis is
designated as a disaccharide, and the one yielding three molecules of
monosaccharide as a trisaccharide and so on. The general formula of disaccharides
is Cn(H2O) n –1 and that of trisaccharides is Cn (H2O)n – 2 and so on.
Disaccharides
The condensation of two molecules of monosaccharides via glycosidic bond
between the C-1 of one sugar and the -OH of another carbon leads to the formation
of disaccharide. They are water soluble, sweet in taste and used to carry
monosaccharides. Maltose, lactose and sucrose are few examples of disaccharide
which consist of two molecules of monosaccharide joined covalently by an O-
glycosidic bond which gets formed when a hydroxyl group of one sugar reacts
with the anomeric carbon of the other sugar. Figure 1.9 illustrates this. Glycosidic
bonds presented in disaccharides can be hydrolyzed by dilute acid to yield their
free monosaccharide components but resist cleavage by base.

Fig. 1.9 Formation of Glycosidic Bond

Some examples of disaccharides are as follows:


x Maltose: It is formed by the condensation of two molecules of D-D glucose
joined at C-1 of one residue and C-4 of second residue by glycosidic bond.
Its structure is shown in Figure 1.10.

Fig. 1.10 Structure of Maltose


Self-Instructional
Material 9
Carbohydrates x Lactose: It is formed by the condensation of one molecule of glucose and
galactose. Its structure is shown in Figure 1.11. It is the chief constituent
found in milk. Intestinal distress may be caused by a deficiency of lactase,
an intestinal enzyme needed to digest and absorb lactose in milk. Abdominal
NOTES pain, bloating, gas and diarrhoea are caused due to undigested lactose that
ferments in the colon.

Fig. 1.11 Structure of Lactose

x Sucrose: It is formed by the condensation of one molecule of glucose and


fructose. Its structure is shown in Figure 1.12. It is most commonly known
as table sugar or saccharose. Sucrose is the main ingredient found in dried
cane juice and brown sugar.

Fig. 1.12 Structure of Sucrose

Trisaccharide
Trisaccharide, such as Raffinose, is broadly found in legumes and cruciferous
vegetables including, beans, peas, cabbage, Brussels sprouts and broccoli. Raffinose
is also called melitose and it consists of galactose connected to sucrose. Its structure
is shown in Figure 1.13. Due to lack of enzyme in humans, this trisaccharide
cannot be digested. So, Raffinose is fermented in the large intestine of humans by
gas-producing bacteria.

Fig. 1.13 Structure of Raffinose


Self-Instructional
10 Material
1.2.4 Polysaccharides Carbohydrates

Polysaccharides are condensation products of more than twenty monosaccharide


units and few may have hundreds or thousands of units. They are also known as
Glycans. Most carbohydrates found in nature occur as polysaccharides, polymers NOTES
of medium to high molecular weight. Depending upon the type of monosaccharide
involved in formation, polysaccharides are categorized into two categories, which
include homopolysaccharides and heteropolysaccharides. The polysaccharides
with single type of monomer of monosaccharide are referred to as
homopolysaccharide whereas those which contain two or more different types of
monosaccharide are referred to as heteropolysaccharides. Some
homopolysaccharides are stored as fuels such as starch and glycogen which occur
intracellularly as granules or in large clusters form. Heteropolysaccharide forms a
mixture of monosaccharides on hydrolysis. They are found in many number both
in plants and as well as in animals. The strain of pneumococcus type III containing
soluble sugar is one of the simplest heteropolysaccharides which is the mixture of
repeating units of a disaccharide consisting of glucose and glucuronic acid. It yields
equimolar concentration of glucose and glucuronic acid on hydrolysis.
Starch

Starch is a homopolymer of glucose forming an D-glycosidic chain called glucosan


or glucan. It is the most abundant dietary carbohydrate in cereals, potatoes, legumes
and other vegetables. Plant cells have the capability to form starch. Starch contains
two types of glucose polymer, amylose (15–20 per cent) and amylopectin (80–85
per cent). Figure 1.12 shows the structure of these two.
Amylose consists of long, unbranched chains of D-glucose residues
connected by D-1o 4 glycosidic linkages having variable molecular weight from
a few thousand to more than a million. Amylopectin is highly branched and has a
high molecular weight up to 100 million. The glycosidic linkages joining successive
glucose residues in Amylopectin chains are D-1o 4 glycosidic bond and in the
branch points which occurs in every twenty-four to thirty residues are linked by
D-1o 6 glycosidic linkage. Dextrins are intermediates in the hydrolysis of starch.

Amylose

Self-Instructional
Material 11
Carbohydrates

NOTES

Amylopectin
Fig. 1.14 Structure of Amylase and Amylopectin

Glycogen
The main storage polysaccharide of animal cells is glycogen. Glycogen is a polymer
of subunits of glucose with D-1o 4 glycosidic linkage at the linear site and D-1o
6 glycosidic linkage at the branching site. Glycogen is more extensively branched
on an average at every eight to twelve residues and it is more compact than starch.
It is found in abundance in the liver, where it may constitute as much as 7 per cent
of the wet weight. It is found in large granules in hepatocytes. It also exists in the
skeletal muscle. Glucose units are removed one at a time from the nonreducing
ends, when glycogen is used as an energy source. The conversion of glycogen
polymer to its monosaccharides is accomplished by degradative enzymes that act
only at non-reducing ends and work simultaneously on the many branch sites.
Glycogen and starch molecules are highly hydrated as they have many
exposed hydroxyl groups available to hydrogen-bond with water. Glycogen and
starch ingested in the diet are hydrolyzed by D-amylases enzymes present in saliva
and intestinal secretions that break D-1-4 glycosidic bonds between glucose units.
Other homopolysaccharides serve as structural elements in plant cell walls (cellulose)
and animal exoskeletons (chitin).
Cellulose
Cellulose is a water-insoluble, fibrous, tough substance found mainly in the cell
walls of plants and primarily in stalks, stems, trunks and every wooded part of the
plant body. It comprises a majority of the mass of wood and cotton. The cellulose
molecule is a linear, homopolysaccharide with no branches, consisting of 10,000
to 15,000 D-glucose units which is similar to starch and glycogen. The vital
difference in cellulose, starch (amylose and Amylopectin) and glycogen is that the
glucose repeating units in cellulose have the E configuration (Refer Figure 1.15),
while in amylose, amylopectin and glycogen the glucose repeating units are in the
Self-Instructional
12 Material
E configuration. The linking of glucose residues in cellulose is done by E-1o 4 Carbohydrates

glycosidic bonds as compared to the E-1-4 glycosidic bonds of amylose in starch


and glycogen. This dissimilarity creates very different structures and physical
properties of cellulose and amylose. Due to lacking of an enzyme to hydrolyse the
E-1o 4 glycosidic linkages, most animals cannot use cellulose as a fuel source. NOTES
Termites can readily digest cellulose because their intestinal tract harbors a symbiotic
microorganism with Trichonympha that secretes cellulase enzyme which hydrolyzes
the D-1o 4 linkages. Wood-rot fungi and bacteria also produce cellulase.
Hemicelluloses
Hemicelluloses constitute xylan and other related substances. They are neither
structurally associated to cellulose nor do they contain the same building blocks.
But they are less partially soluble in water or alkali. The hemicelluloses consist
of either pentose sugars such as xylose, arabinose or hexose sugar such as
glucose, mannose, and galactose along with uronic acids. The rich sources of
hemicelluloses include unrefined cereals, some fruits and vegetables and whole
wheat. They adsorb water and are partially digestible. They mostly serve as
storage and supporting substances in plants. The term ‘hemicelluloses’ has been
discontinued in use as a large number of analogous polysaccharides were revealed
in fungi and bacteria. Xylan is a delegate of the group and is being discussed in
the subsequent section.

Fig. 1.15 Structures of Cellulose, Pectin and Hemicelluloses

Xylan
Xylan is the most abundant and extensively spread carbohydrate after cellulose.
Straw and bark consist of upto 30 per cent xylan, the residues of sugarcane also
contain about 30 per cent xylan. It also occurs in conifer wood constitute between
7 – 12 per cent and deciduous wood constitute nearby 20 – 25 per cent xylan.
Xylan is a linear homopolymer of an aldopentose called D-xylose having E-1o 4
Self-Instructional
Material 13
Carbohydrates linkage bearing side chains of 4–O–methylglucuronic acid or arabinose. Xylan
can be derived from a cellulose chain by substituting hydrogen atoms for the —
CH2OH groups, but the number of units per polymer is considerably lower and in
the range of 30 – 100 (Refer Figure 1.16).
NOTES

Fig. 1.16 Structure of Xylan

The digestion of Xylan by a large number of microorganisms is faster in


comparison to cellulose. Many cellulose-degrading organisms such as
myxococcoides, sporocytophaga, etc. also produce xylanase, an enzyme
responsible for the digestion of xylan. The ability to utilize Xylan is very frequent to
be found among fungi and is admirable substrate for the nurturing of mushrooms.
The enzyme Xylanase is formed constitutively in some bacteria e.g., Clostridia
and in others bacterial cell, it is inducible by xylan.
Chitin
Chitin is a linear homopolysaccharide comprising N-acetylglucosamine residues
in E linkage (Refer Figure 1.17). Chitin varies from cellulose in only the chemical
replacement of the hydroxyl group at C-2 with an acetylated amino group. The
formation of extended fibres by Chitin is similar to that of cellulose. However,
vertebrates cannot digest it, just as they cannot digest cellulose. Chitin is a vital
component of the rigid exoskeletons of almost a million species of arthropods
which also includes insects, lobsters and crabs. It is nearly certain tha it is the
second polysaccharide to be found most abundantly in nature, after cellulose.

Fig. 1.17 Structure of Chitin

Self-Instructional
14 Material
Peptidoglycan Carbohydrates

Peptidoglycan is the most inflexible layer of the bacterial cell envelope. It is partly
made up of a heteropolysaccharide made from two corresponding monosaccharide
units that are heteropolymer of corresponding E 1-4-linked N-AcetylGlucosamine NOTES
(NAG) and N-AcetyLmuramic acid (NAM) residues (Refer Figure 1.18). The
precise structure of peptidoglycan is actually dependent on the bacterial species
with linear polymers stretching out adjacent to each other in the cell wall, with
short peptides cross linking it. The polysaccharide chains are welded into a tough
sheath by peptide cross-links. This tough sheath engulfs the complete cell, thereby
preventing cellular swelling and lysis because of the osmotic entrance of water.
The enzyme lysozyme destroys bacteria by carrying out a hydrolysis of the E 1-4
glycosidic bond between N-AcetylGlucosamine (NAG) and N-AcetylMuramic
acid (NAM). Lysozyme is present to a great extent in tears, mainly as a protection
for bacterial infections of the eye. It is even made by some bacterial viruses for
ensuring their liberation from the host bacterial cell, a necessary step of the viral
infection cycle. Penicillin and associated antibiotics destroy bacteria by stopping
the synthesis of the cross-links, that leaves the cell wall so weak that they cannot
withstand osmotic lysis.

Fig. 1.18 Structure of Peptidoglycans

Different kinds of heteropolysaccharide occupy the extracellular space in


animal tissues, resulting in the formation of a matrix. This is known to hold the
different cells as one, providing protection, shape and support to cells, tissues and
organs.
Self-Instructional
Material 15
Carbohydrates Mucopolysaccharides
Polysaccharides which are comprised not only of a mixture of simple sugars but
also of derivatives of sugars such as amino sugars and uronic sugars are termed as
NOTES mucopolysaccharides. They are gelatinous in nature and have high molecular weights
in the range of up to 5 × 106. They mostly act as structural support material for
connective tissue or mucous substances of the body. They serve both as a lubricant
and a cementing substance. They consist mainly of disaccharide units in which a
uronic acid is bound by a glycosidic bond to the C-3 of an acetylated amino acid
in 1o 3 linkage. These disaccharide residues are polymerized by 1o 4 linkages
to give a linear macromolecule. The uronic and sulphuric acid residues impart a
strong acidic character to these substances. The lists the structural features of
some common mucopolysaccharides are illustrated in Table 1.3.
Table 1.3 Common Mucopolysaccharides
Mucopolysaccharides Two Components of the Disaccharide Units

Hyaluronic Acid. D-Glucuronic Acid +N-Acetyl-D-


Glucosamine.
Chondroitin Sulphate A. D-Glucuronic Acid + N-Acetyl-D-
Galactosamine-4-Sulphate.
Chondroitin Sulphate C. D-Glucuronic Acid + N-Acetyl-D-
Galactosamine-6-Sulphate.
Dermatan Sulphate L-iduronic Acid + N-Acetyl-D-Galactosamine-
(Chondroitin Sulphate B). 4-Sulphate.
Keratosulphate. D-Galactose + N-Acetyl-D-Glucosamine-6-
Sulphate.

Hyaluronic Acid
Hyaluronic acid is the most abundant member of mucopolysaccharides and is
found mainly in higher animals as a component of various tissues such as the vitreous
body of the eye, the umbilical cord and the synovial fluid of joints. The high viscosity
of the synovial fluid and its role as biological lubricant is largely due to the presence
of its hyaluronic acid content of about 0.03 per cent). Frequently, it is prepared
from umbilical cord.
Hyaluronic acid (Refer Figure 1.19) has a slightly complex structure as
compared to the remaining mucopolysaccharides. It is a straight-chain polymer of
D-glucuronic acid and N-Acetyl- D-Glucosamine (NAG) alternating in the chain.
Its molecular weight is approximately 5,000,000. The two glycosidic linkages
which are invloved in the formation of these mucopolysaccharides include E-1o
3 and E-1o 4 linkage. Hyaluronic acid is an acidic substance at cellular pH
because the carboxyl groups are largely ionized.

Self-Instructional
16 Material
Carbohydrates

NOTES

Fig. 1.19 Structure of Hyaluronic Acid

Upon hydrolysis, hyaluronic acid produces an equimolar amount of mixture


of D-glucuronic acid, D-glucosamine and acetic acid. Hyaluronic acid is broken
down or digested rapidly by the enzyme. The enzyme hyaluronidase brings about
the depolymerization of hyaluronic acid and binds it to smaller fragments. This
enzyme is a ‘spreading source’ of skin and connective tissue and also has a
physiologic role in fertilization. The sperm is rich in hyaluronidase and hence they
can move forward better in the cervical canal and finally can fertilize the ovum.
Chondroitin
Chondroitin is of limited distribution as it is mainly found in cartilage. It is also a
component of cell coats. It is a parent substance for two more widely distributed
mucopolysaccharides, chondroitin sulphate A and chondroitin sulphate B. The
structure of chondroitin is similar to hyaluronic acid except that it contains
galactosamine in spite of glucosamine. It is a polymer of E-D-glucuronido- 1, 3-
N-Acetyl D-Galactosamine joined by E- 1 o 4 glycosidic linkages. Chondroitin
produces equimolar mixture of D-glucuronic acid, D-galactosamine and acetic
acid on hydrolysis (Refer Figure 1.20).

Fig. 1.20 Structure of Chondroitin


Self-Instructional
Material 17
Carbohydrates Chondroitin Sulphates
The two chondroitin sulphates A and C are widely distributed in the body and
form foremost structural components of cartilage, tendons and bones. They are
NOTES more frequently associated with collagen and probably with other proteins too.
Chondroitin sulphates may be regarded as derivatives of chondroitin where, in the
galactosamine moiety, a sulphate group is esterified either at carbon 4 as in
chondroitin sulphate A or at carbon 6 as in chondroitin sulphate C (Refer
Figure 1.21). The two linkages includes E-1o 3 and E-1o 4 which are involved
in both types of chondroitin sulphates A and C. Chondroitin sulphates A and C
both yield roughly equivalent amounts of D-glucuronic acid, D-galactosamine,
acetic acid and sulphuric acid on hydrolysis.

Fig. 1.21 Structure Comparison between Different Mucopolysaccharide

Dermatan Sulphate
Dermatan sulphate is mucopolysaccharides structurally similar to chondroitin
sulphate A excluding that the D-glucuronic acid is replaced by L-iduronic acid at
C-5. The bonding involved in Dermatan sulphate structure formation includes D-
1o 3 and E-1o 4 glycosidic linkage. Dermatan sulphate is even recognized by
its common name, chondroitin sulphate B. Dermatan sulphate varies from both
chondroitin sulphate A and C in the constitution of their repeating disaccharide unit
(Refer Figure 1.22).

Fig. 1.22 Structure of Dermatan Sulphate

Self-Instructional
18 Material
Keratosulphate Carbohydrates

Keratosulphate varies from other mucopolysaccharides in that the D-galactose


replaces the uronic acid component. In this, the second acetylated amino sugar
constituent which is N-Acetyl-D-Glucosamine undergoes esterification by a sulphate NOTES
group at carbon number 6. In spite of the two corresponding linkages participating
are E-1 o 4 and E-1 o 3, here the linkage between the repeating disaccharide
units is E-1o 3 rather than E-1o4 (Refer Figure 1.23).

Fig. 1.23 Structure of Keratosulphate

Heparin
Heparin is the sulphated mucopolysaccharides which is mainly found in liver, lung,
arterial walls and, certainly wherever mast cell are found, possibly for the purpose
of neutralizing biogenic amines, for example, histamine. Heparin (Refer Figure
1.24) is a heteropolysaccharide composed of D-glucuronic acid units, most of the
seven out of every eight undergo esterification at C2 and D-glucosamine-N-
sulphate units with an additional O-sulphate group at C6. Both the bondings of the
polymer are corresponding D-1o 4 glycosidic linkage. Thus, the content of
sulphate is extremely great corresponding to approximately five to six molecules
per tetrasaccharide repeating unit. The relative positions of the sulphate residues
may also vary. Its molecular weight ranges between 17,000 and 20,000.

Fig. 1.24 Structure of Heparin

Heparin acts as an anticoagulant. It prevents coagulation of blood by inhibiting


the prothrombinthrombin conversion. This eliminates the effect of thrombin on
fibrinogen.
Self-Instructional
Material 19
Carbohydrates The heteropolysaccharides described above contain two different sugars in
their repeating units or monomers. However, there are many heteropolysaccharides
which contain more than two carbohydrates in their repeating units. Vegetable
‘gums’ and agar – agar are two prominent examples which are explained as follows:
NOTES
Vegetable ‘Gums’
Vegetable ‘gums’ contain as many as four different monosaccharide units. Most
common monosaccharide units include D-glucuronic acid, D-mannose, D-xylose
(the second most abundant sugar in the biosphere) and L-rhamnose.
Agar – Agar
Agar is a gelatinous polysaccharide produced by certain marine red algae of
rhodophycean members such as species of Gelidium, Gracilaria, Hypnea, etc.
The chief producer country for the production of agar – agar is Japan. It consists
of D- galactose and L-galactose, predominantly with 1o 3 glycosidic bonds and
always contains some amount of sulphuric acid. It yields D- and L-galactose in a
ratio of 9: 1 on hydrolysis. It forms highly viscous gels and has melting point between
90 and 100°F. At lower temperature, it solidifies. It is soluble in hot water but is
insoluble in cold water (Refer Figure 1.25).

Fig. 1.25 Structure of Agar

Agar – agar is of immense value in the preparation of foodstuffs, but


principally it is used extensively in biological laboratories as the base material for
the preparation of culture media, especially for bacteria and fungi. It is largely
used as a solidifying agent in desserts, as a laxative and as a sizing material for
textiles. It is also used in the preparation of some medicines, and in cosmetics and
leather industry. It also finds application as an emulsifier in dairy products. Agar –
agar is not utilized by man and hence adds to the bulk of the faeces and helps in its
propulsion.
1.2.5 Glycoconjugates
Glycoconjugates is the biologically active molecule which constitutes the
informational carbohydrate covalently linked to a protein or a lipid. They play the
role of being target labels for few proteins and also as intermediaries of specific
Self-Instructional
20 Material
cell – cell interactions and also interactions between cells and the extracellular Carbohydrates

matrix, cell – cell identification and adhesion, cell migration at the time of
development, blood clotting, the immune response and wound healing.
Glycoconjugates include proteoglycans, glycoprotein and glycolipids. Types of
glycoconjugates are as follows: NOTES

Proteoglycans
The macromolecules of the cell surface or extracellular matrix in which one or
more glycosaminoglycans chains are joined covalently to a membrane protein or a
secreted protein are referred as proteoglycans. The fundamental proteoglycan
unit consists of a ‘core protein’ with covalently attached glycosaminoglycans. The
glycosaminoglycans moiety commonly forms the greater portion of the
proteoglycans molecule by mass. Glycosaminoglycans, governs the structure, and
is often the primary site of biological functions. In several instances, the biological
function is the condition of several binding sites, with abundant prospects for
hydrogen bonding and electrostatic interactions with different proteins of the cell
surface or the extracellular matrix. Proteoglycans are the main constituents of
connective tissue like cartilage, which involves several non-covalent interactions
with other proteoglycans, proteins and glycosaminoglycans providing power and
making them flexible. Mammalian cells can produce at least thirty types of molecules
that are members of the proteoglycan super family. These molecules may act as
tissue organizers, manipulate the development of specialized tissues, arbitrate the
activities of various growth factors, and control the extracellular assemblage of
collagen fibrils.
Glycoproteins
The carbohydrate – protein conjugates wherein the carbohydrate moieties are
much small with more structural difference as compared to the glycosaminoglycans
of proteoglycans are known as glycoproteins or mucoproteins (Refer Figure 1.26).
The attachment of a carbohydrate at its anomeric carbon is with the help of a
glycosidic linkage to the –hydroxyl (OH) group of a Serine residue (Ser) or
Threonine (Thr) residue by an O-link or through an N- link glycosyl linkage to the
amide nitrogen of an Asn residue (N-linked). Only a small number of glycoproteins
possess a single oligosaccharide chain. However, several contain a number of
oligosaccharide chains that can comprise from 1 per cent to 70 per cent or more
of the glycoprotein by mass. The structures of several O- and N-linked
oligosaccharides from a number of glycoproteins are recognized. The outer surface
of the plasma membrane contains several membrane glycoproteins having a range
of oligosaccharides of differing complexity with covalent attachments. Glycoproteins
are rich in information, and form extremely particular sites to be recognized with
high-affinity linking by other proteins.

Self-Instructional
Material 21
Carbohydrates

NOTES

Fig. 1.26 Structure of Glycoprotein

Glycolipids
The membrane lipids wherein the hydrophilic head groups are oligosaccharides
are called glycolipids (Refer Figure 1.27). The dominating surface feature of the
external membrane of gram-negative bacteria like Escherichia coli (E.coli) and
Salmonella typhimurium contain glycoprotein. These molecules are the primarily
targeted by the antibodies which the vertebrate immune system produces
responding to bacterial infection. They are thus vital factors that determine the
serotype of bacterial strains (serotypes are strains that are differentiated, based on
the antigenic properties). The lipopolysaccharides of S. typhimurium involve six
fatty acids bound to two glucosamine residues, out of which one is the point of
bonding for a complicated oligosaccharide. E. coli contains similar but different
lipopolysaccharides. The lipopolysaccharides of some bacteria are dangerous for
humans and other animals. For instance, they are responsible for the dangerously
reduced blood pressure which takes place in toxic shock syndrome which results
from gram negative bacterial infections.

Fig. 1.27 Structure of Glycolipid


Self-Instructional
22 Material
Carbohydrates

Check Your Progress


1. Name the process by which plants synthesize glucose.
2. What form do most of the hexoses found in living organisms have? NOTES

3. What property is the Fehling’s reaction, a qualititative test based on?


4. Name the two categories of polysaccharides.
5. Which is the main storage polysaccharide of animal cells.
6. Which is the most inflexible layer of the bacterial cell envelope?
7. Why is chondroitin of limited distribution?
8. What are glycoconjugates?
9. What does the fundamental proteoglycan unit consist of?
10. Define glycolipids.

1.3 CHEMICAL PROPERTIES OF


CARBOHYDRATES

Carbohydrates possess active groups which are responsible for their chemical
behaviour. These groups are glycosidic (OH), alcoholic (OH) and aldehyde (CHO)
or ketonic (CO). Most of the reactions of mutarotating sugars take place in aqueous
solution so that all the three forms, D-form, aldehyde/keto form and E-form become
available for the reaction. Besides, all these three forms are interconvertible.
1. Reaction of Glycosidic (OH)- Group
(i) Reaction with Alcohol
The glycosidic (OH) group of mutarotating sugars reacts with alcohols to form D-
and E- glycosides or acetals. Thus, glucose forms glucosides and fructose forms
fructosides. The glucosides or glycosides in general, do not exhibit mutarotation
as the aldehyde group in them is converted to the acetal group. A number of
glycosides occur in nature, for example, phlorhizin (glucose + phloretin) in rose
bark. They are useful as medicaments.
α-D Glucose + Methyl Alcohol o α-Methyl D-Glucoside
(ii) Reaction with Acetic Anhydride (Esterification)
The glycosidic and alcoholic OH groups of monosaccharides and disaccharides
react with acetylating agents (like acetic anhydride in pyridine) to form acetate
derivatives called esters.
Self-Instructional
Material 23
Carbohydrates The ability of sugars to form esters indicates the presence of alcohol groups
in their molecule. As glucose upon acetylation yields a penta-acetate derivative
which obviously contains 5 OH groups.
α-D Glucose + Acetic Anhydride o α-D Glucose Penta-Acetate + Water
NOTES
(iii) Reaction with Methyl Iodide (Etherification)
The alcoholic OH groups of monosaccharides and disaccharides are converted
to ether groups upon treatment with methylating agents. The glycosidic OH group
of the sugars is, however, first protected with an R group by converting these
sugars to glycosides which are, then, allowed to react with the methylating agent.
α-Methyl Glucose + Methyl Iodide + Silver Oxide o Tetramethyl Ether of α-
Methyl Glucoside+ + Silver Iodide +Water
2. Oxidation with Acids
The alcoholic OH group and CHO group (or CO group) are oxidized to carboxyl
groups by certain oxidizing agents. The oxidation may be brought about under
mild or vigorous oxidizing conditions.
(i) With Mild Oxidants (like HOBr)—Only the aldehyde group of
monosaccharide or disaccharide is oxidized to produce monocarboxylic acids.
Ketoses, however, do not respond to this reaction. Hence, this reaction is used to
distinguish aldoses from ketoses.
Glucose + HOBr o Gluconic Acid + HBr
(ii) With Strong Oxidants (like conc. HNO3)—Both the aldehyde group or
ketone group and the primary alcohol group are oxidized to yield dicarboxylic
acids. With aldoses, acids with same number of carbon atoms are obtained whereas
ketoses react to produce acids with fewer number of carbon atoms.
Glucose + HNO3 o Glucaric Acid + Water
Fructose + HNO3 o Trihydoxyglucaric Acid + Tartaric Acid +
Glycolic Acid
3. Oxidation with Metal Hydroxides
Metal hydroxides like Cu (OH)2, AgOH and Bi(OH)3 oxidize the free aldehyde
(or ketone) group of mutarotating mono- and di-saccharides and are, at the same
time, themselves reduced to the lower oxides or to the free metals. Thus,
Cu (OH)2 is reduced to CuO and AgOH and Bi(OH)3 are reduced to the free
metal, Ag and Bi. The reaction with cupric hydroxide is as follows:
Reducing sugar + 2 Cu++o Oxidized Sugar + 2 Cu++
Cupric ion (Cu++) is, in fact, the most common oxidizing agent of these. It is the
active ingredient in Fehling’s, Benedict’s and Barfoed’s reagents.
Fehling’s reagent consists of two solutions. Solution A contains 7 per cent Cu.SO4
and solution B contains 25 per cent KOH and 35 per cent sodium potassium
tartarate. When the two solutions are mixed in equal amounts, a clear blue solution
Self-Instructional
24 Material
results for the tartarate forms a soluble complex with the copper hydroxide Carbohydrates
produced. This solution is widely used as an oxidizing agent preferably in quantitative
sugar determinations.
With Fehling’s reagent, the overall reaction occurs as follows:
NOTES
Glucose + Fehling Solution o Gluconic Acid + Cuprous Oxide + Water
In clinical laboratories, however, a modification of Fehling’s reagent is used for the
qualitative determination of glucose in urine. This is known as Benedict’s reagent
and consists of a single solution containing CuSO4, Na2.CO3 (in place of KOH)
and sodium citrate (in place of sodium potassium tartarate). Sodium carbonate
reduces alkalinity to such an extent that, unlike the Fehling’s reagent, the two
solutions (Cu.SO4 and sodium citrate) here are kept mixed indefinitely as one
solution.
Barfoed’s reagent utilizes a weakly acid solution for oxidation. It contains 7 per
cent cupric acetate and 1 per cent acetic acid. This reagent can be used to
distinguish monosaccharides from disaccharides as the former give a cuprous oxide
precipitate in dilute acid more quickly than the latter. Monosaccharides are, thus,
more active reducing agents than the disaccharides.
The Molisch test employs sulphuric acid for dehydration and D-naphthol
as the phenol. The acid also hydrolyses acetal groups present and thus makes this
test probably the most general of all the tests for carbohydrates.
4. Reaction with Hydrogen Cyanide (Kiliani Synthesis)
The addition of HCN to mutarotating sugars creates a new asymmetric carbon
atom, producing two new compounds namely, cyanohydrin A and cyanohydrin B.
These are then hydrolysed with dilute mineral acids to form hydroxy acids which
are later converted to D-lactones and finally reduced to two new sugars containing
one more carbon atom than the parent sugar.
5. Reaction with Alanine
The aldehyde group of carbohydrates undergoes condensation with the amino
group of alanine for forming Schiff’s base. The browning reaction that occurs
when bread is being baked and rest of the mixtures of carbohydrates and proteins
is thought to be because of the formation of Schiff’s base between the amino
groups of proteins and the aldehyde groups of carbohydrates.
Glucose + Alanine o Schiff base + Water
6. Reaction with Phenyl Hydrazine
Reaction with phenyl hydrazine involves only two carbon atoms viz., the carbonyl
(i.e., the aldehyde or ketone group) and the adjacent one. One mole of phenyl
hydrazine reacts with one mole of aldose or ketose to form a mole of hydrazone.
With a second mole of phenyl hydrazine, the hydrazone is oxidized to aldohydrazone
Self-Instructional
Material 25
Carbohydrates and the phenyl hydrazine itself is reduced to aniline and ammonia. Finally, a third
mole of phenyl hydrazine reacts with the aldohydrazone to produce osazone.
(i) Glucose + Phenyl Hydrazine o Glucose Hydrazone + Water
NOTES (ii) Glucose Hydrazone + Phenyl hydrazine o Aldohydrazone + Aniline +
Water
(iii) Aldohydrazone+ + Phenyl hydrazine o Osazone +Water
7. Reaction with Hydroxylamine
Simple sugars react with hydroxylamine to yield oximes.
Glucose + Hydroxylamine o Aldooxime + Water
8. Fermentation
Monosaccharides such as glucose, fructose and mannose are readily fermented
by yeast. The process of yeast fermentation is very complex. During this process,
sugar phosphate and sugar acid phosphate are formed. Ordinarily, this process
results in the formation of alcohol and carbon dioxide.
1.3.1 Functions of Carbohydrates
The functions of carbohydrates are given as follows:
x Carbohydrates give the primary energy molecules to the body and leave
protein to be used up as a molecule for energy production, instead of this
protein concentrates in the body and repairing and maintain the body.
x They are the only energy source of energy for the brain. It is necessary for
the nervous tissue regulation.
x They are necessary for the complete metabolization of fats.
x Some carbohydrates encourage the growth of healthy bacteria in the
intestine.
x They are sometimes used to add flavors and as sweeteners.
x They prevent ketosis that is caused due to high metabolism of fat.
x They regulate the sugar in the blood.
x They help in the fertilization, growth and development of cells.
x Complex carbohydrates are a good source of fiber.
x In excess amount, carbohydrates are stored in the body as fats. Hence, it is
recommended to take a minimum daily amount of carbohydrates in the
body so that they are not converted to fats and lead to weight gain.
x Cellulose and other carbohydrates form the cell wall of plant cells along
with hemicelluloses and pectin.
x Chitin and other carbohydrates form the cell wall of fungal cells and the
exoskeleton of arthropods.
Self-Instructional
26 Material
x Peptidoglycans and other carbohydrates form the cell wall of bacteria and Carbohydrates

cyanobacteria.
x Heparin is a form of a carbohydrate that prevents intravascular clotting of
blood.
NOTES
x They function as hormones, such as Follicular Stimulating Hormone (FSH)
in ovulation in females) and Leutinizing Hormone (LH).
x Carbohydrates are also used in industries like paper, textile and breweries.
x Hyaluronic acid is a glycoprotein, which acts as a lubricant between joints
to provide frictionless movement.
x Many antigens are glycoproteins that give immunological properties to blood.
x Agar is a polysaccharide which is used in the culture media.
x Different forms of carbohydrates are stored in living organisms as food,
such as polysaccharides, starch in plants and glycogen in liver and muscles
in animals.

Check Your Progress


11. Which groups in carbohydrates are responsible for their chemical
behaviour?
12. What do the aldehyde group and the primary alcohol group yield when
oxidized?
13. What do simple sugars yield when they are made to react with
hydroxylamine?

1.4 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. In plants, glucose is synthesized from carbon dioxide and water by


photosynthesis.
2. Most of the hexoses found in living organisms have the D-isomeric form.
3. Fehling’s reaction, a qualitative test for the presence of reducing sugar is
based on the property of reduction.
4. The two categories of polysaccharides are homopolysaccharides and
heteropolysaccharides.
5. The main storage polysaccharide of animal cells is glycogen.
6. Peptidoglycan is the most inflexible layer of the bacterial cell envelope.
7. Chondroitin is of limited distribution as it is mainly found in cartilage.
Self-Instructional
Material 27
Carbohydrates 8. Glycoconjugates is the biologically active molecule which constitutes the
informational carbohydrate covalently linked to a protein or a lipid.
9. The fundamental proteoglycan unit consists of a ‘core protein’ with covalently
attached glycosaminoglycans.
NOTES
10. The membrane lipids wherein the hydrophilic head groups are
oligosaccharides are called glycolipids.
11. Carbohydrates possess active groups which are responsible for their
chemical behaviour.
12. Both the aldehyde group or ketone group and the primary alcohol group
are oxidized to yield dicarboxylic acids.
13. Simple sugars react with hydroxylamine to yield oximes.

1.5 SUMMARY

x Carbohydrates have important structural and metabolic roles. In plants,


glucose is synthesized from carbon dioxide and water by
photosynthesis and stored as starch or used to synthesize cellulose
for plant framework.
x The different types of carbohydrates are monosaccharides, oligosaccharides,
polysaccharides and glycoconjugates.
x Monosaccharides or simple sugars are those carbohydrates that cannot be
hydrolysed into simpler carbohydrates containing a single polyhydroxy
aldehyde or ketone unit.
x Oligosaccharides are compound sugars that yield two to ten molecules of
the identical or different monosaccharides on hydrolysis (removal of water
molecules).
x Polysaccharides are condensation yield of more than twenty monosaccharide
units and few may have hundreds or thousands of units.
x The main storage polysaccharide of animal cells is glycogen. Glycogen is a
polymer of subunits of glucose with D-1o 4 glycosidic linkage at the linear
site and D-1o 6 glycosidic linkage at the branching site.
x Peptidoglycan is the most inflexible layer of the bacterial cell envelope. It
is partly made up of a heteropolysaccharide made from two corresponding
monosaccharide units that are heteropolymer of corresponding E 1-4-
linked N-Acetyl-Glucosamine (NAG) and N-AcetylMuramic acid (NAM)
residues.
x Glycoconjugate is the biologically active molecule which constitutes the
informational carbohydrate covalently linked to a protein or a lipid.
Glycoconjugates include proteoglycans, glycoprotein, and glycolipids.
Self-Instructional
28 Material
x Carbohydrates possess active groups which are responsible for their Carbohydrates

chemical behaviour. These groups are glycosidic (OH), alcoholic (OH) and
aldehyde (CHO) or ketonic (CO).

NOTES
1.6 KEY WORDS

x Carbohydrates: These are polyhydroxy compounds that contain a


carbonyl (C=O) group of the general formula, (CH2O)n.
x Monosaccharide: Also known as simple sugars, these are those
carbohydrates that cannot be hydrolysed into simpler carbohydrates
containing a single polyhydroxy aldehyde or ketone unit.
x Starch: It is a homopolymer of glucose forming an D-glycosidic chain called
glucosan or glucan.
x Cellulose: It is a water-insoluble, fibrous, tough substance found mainly in
the cell walls of plants and primarily in stalks, stems, trunks and every wooded
part of the plant body.
x Chitin: It is a linear homopolysaccharide comprising N-acetylglucosamine
residues in E linkage.
x Agar: It is a gelatinous polysaccharide produced by certain marine red
algae of rhodophycean members such as species of Gelidium, Gracilaria,
Hypnea, etc.

1.7 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. Classify carbohydrates on the basis of the monomers they contain.
2. State some examples of disaccharides.
3. What is hyaluronic acid?
4. State the reaction of the glycosidic group with alcohol.
5. What is Fehling’s reagent? Write the overall reaction that occurs with
Fehling’s reagent.
Long Answer Questions
1. Describe monosaccharides.
2. Write short notes on
x Glycogen
x Peptidoglycan
Self-Instructional
Material 29
Carbohydrates 3. What are glycoconjugates? Discuss.
4. Explain the reaction of glycosidic (OH)- group when oxidized with metal
hydroxides.
NOTES
1.8 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
30 Material
Carbohydrate

UNIT 2 CARBOHYDRATE Metabolism

METABOLISM
NOTES
Structure
2.0 Introduction
2.1 Objectives
2.2 Metabolism of Carbohydrates
2.2.1 Fate of Carbohydrates
2.2.2 Fate of Pyruvate
2.2.3 Citric Acid Cycle, Krebs Cycle or Tricarboxylic Acid (TCA) Cycle
2.2.4 Gluconeogenesis
2.3 Glycogen Metabolism
2.3.1 Glycogenolysis
2.3.2 Glycogenesis
2.3.3 Pathway of HMP
2.3.4 Uronic Acid Pathway
2.3.5 Metabolism of Galactose
2.3.6 Metabolism of Fructose
2.3.7 Futile Cycle
2.4 Metabolic Defects Related to Carbohydrates
2.5 Entner Doudroff Pathway
2.6 Answers to Check Your Progress Questions
2.7 Summary
2.8 Key Words
2.9 Self Assessment Questions and Exercises
2.10 Further Readings

2.0 INTRODUCTION

Carbohydrate metabolism refers to various biochemical processes responsible for


the metabolic formation, breakdown, and inter-conversion of carbohydrates
in living organisms. Carbohydrate metabolism begins with digestion in the small
intestine where monosaccharides are absorbed into the blood stream. Blood sugar
concentrations are controlled by three hormones: insulin, glucagon, and epinephrine.
There are top four stages in reregulation of carbohydrate metabolism. First stage
is regulation of Carbohydrate Metabolism at the Cellular and Enzymatic Level.
Second stage is regulation of Glycolysis, Gluconeo-Genesis and Hexose
Monophosphate Shunt. Third stage is regulation of Glycogen Metabolism. Fourth
stage is regulation of the Citric Acid Cycle.
Carbohydrates are central to many essential metabolic pathways. Plants
synthesize carbohydrates from carbon dioxide and water through photosynthesis,
allowing them to store energy absorbed from sunlight internally.
When animals and fungi consume plants, they use cellular respiration to break down
these stored carbohydrates to make energy available to cells. Both animals and
Self-Instructional
Material 31
Carbohydrate plants temporarily store the released energy in the form of high energy molecules,
Metabolism
such as ATP, for use in various cellular processes.
Although humans consume a variety of carbohydrates, digestion breaks
down complex carbohydrates into a few simple monomers (monosaccharides)
NOTES
for metabolism: glucose, fructose, and galactose. Glucose constitutes about 80%
of the products, and is the primary structure that is distributed to cells in the tissues,
where it is broken down or stored as glycogen. In aerobic respiration, the main
form of cellular respiration used by humans, glucose and oxygen are metabolized
to release energy, with carbon dioxide and water as byproducts. Most of the
fructose and galactose travel to the liver, where they can be converted to glucose.
Some simple carbohydrates have their own enzymatic oxidation pathways, as do
only a few of the more complex carbohydrates. The disaccharide lactose, for
instance, requires the enzyme lactase to be broken into its monosaccharide
components, glucose and galactose.
In this unit, you will study about carbohydrate metabolism - Glycolytic
pathway, deficiency diseases, inborn errors of carbohydrate metabolism and
nutritional aspects of carbohydrate.

2.1 OBJECTIVES

After going through this unit, you will be able to:


x Discuss metabolism of carbohydrates
x Discuss regulation of carbohydrates
x Describe glycogen metabolism
x Understand metabolic defects related to carbohydrates
x Explain Entner-Doudoroff pathway

2.2 METABOLISM OF CARBOHYDRATES

Following are the various important pathways of carbohydrate metabolism:


x Glcolysis (Embden – Meyerhof Pathway): Pyruvate and lactate are
formed by oxidation of glucose.
x Citric Acid Cycle [Krebs Cycle or Tricarboxylic Acid (TCA) Cycle]—
Acetyl CoA undergoes oxidation to form CO2. TCA cycle is the ultimate
common oxidative pathway for carbohydrates, fats or amino acids, by acetyl
CoA.
x Gluconeogenesis: It is the process of formation of glucose from non-
carbohydrate precursors (for example, amino acids, glycerol, etc.)
x Glycogenesis: It is the process of glycogen synthesis from glucose.
x Glycogenolysis: It is the process of breaking down of glycogen to
Self-Instructional
glucose.
32 Material
x Hexose Monophosphate Shunt( Pentose Phosphate Pathway or Carbohydrate
Metabolism
Direct Oxidative Pathway): It is an alternative pathway to glycolysis and
TCA cycle for oxidation of glucose to carbon dioxide and water.
x Uronic Acid Pathway (an Alternative to Oxidative Pathway of
NOTES
Glucose): In this pathway, glucuronic acid, peptose and sometimes ascorbic
acid are formed from glucose.
x Galactose Metabolism: This pathway involves formation of glucose from
galactose and finally, formation of lactose.
x Fructose Metabolism: This pathway involves pyruvate formation by
oxidation of fructose and the connection between glucose and fructose
metabolism.
x Amino Sugar and Mucopolysaccharide Metabolism: This pathway
involves the formation of amino sugar and other sugars for the formation of
mucopolysaccharides and glycoproteins.
2.2.1 Fate of Carbohydrates

Intake of Carbohydrates
Our regular diet contains carbohydrates, proteins, lipids, vitamins and minerals.
Most of these macromolecules are in complex forms which need to be converted
into simpler form before they get absorbed in our digestive tract. The dietary food
contains polysaccharides (such as starch, glycogen), disaccharides (such as lactose,
sucrose) and in very less amount monosaccharides (such as glucose, fructose).
These carbohydrates undergo digestion in our digestive system, i.e., gastro-intestinal
tract. The glycoside bonds are hydrolyzed by a glucose entry into the cell which
can be of two types. These types are as follows:
x Insulin-independent transport system of glucose occurring in hepatocytes,
erythrocytes and brain.
x Insulin-dependent transport system occurring in muscle and adipose tissue.
Group of enzymes known as glycosidases present are salivary a-amylase,
pancreatic D- amylase, maltase, sucrose and lactase.
Table 2.1 Process of Carbohydrate Digestion

Site/ Location Enzymes Substrate Product


Mouth Salivary Amylase Starch Dectrins Isomaltase
and Maltase

Stomach Enzyme Initiated by – –


Acidic pH
Small Intestine Pancreatic D-amylase, Starch Glucose
D-glucoamylase,
Isomaltase, Maltase
Lactase Lactose Glucose+ Galactose
Sucrase Sucrose Glucose + Fructose

Self-Instructional
Material 33
Carbohydrate After digestion, the sugar absorption occurs in the duodenum and upper
Metabolism
jejunum of the small intestine in the blood capillaries. From the capillaries, it flows
through blood vessels and is supplied to the cells. It is in the cells that the process
of glucose metabolism starts.
NOTES
Glycolysis (Greek meaning: glycose for sweet or sugar, lysis for dissolution)
was discovered in 1940 and was known as Embden Meyerhof pathway (E.M.
pathway) after the name of the scientist who discovered it. Glycolysis is defined as
the sequence of reaction converting glucose (or glycogen) to pyruvate or lactate
with the production of ATP (Refer Figure 2.1).

Fig. 2.1 Glycolysis

Pathway of Glycolysis
The pathway of glycolysis contains three steps which are as follows:
1. Preparatory Stage: This stage prepares the molecule of glucose by adding
phosphate group from ATP molecule for breaking it into two equal parts.
Self-Instructional
34 Material
Stage 1: Formation of Glucose–6–Phosphate: Glucose is phosphorylated Carbohydrate
Metabolism
to glucose – 6 – phosphate in the presence of the enzyme Hexokinase. This
stage requires ATP B and Mg2+.
Stage 2: Formation of Fructose–6–Phosphate: On isomerization by NOTES
phosphofructoisomerase, Glucose-6- phosphate is converted to Fructose
– 6 – phosphate. Mg2+ is also required.
Stage 3: Formation of Fructose–1, 6–Phosphate: Fructose–6–phosphate
undergoes phosphorylation to form fructose – 1,6 – Phosphate by enzyme
Phosphofructokinase. This utilizes one ATP molecule and requires Mg2+ also.
2. Splitting Stage: In this stage the energized intermediate molecule having
six carbon atoms will be split into two equal parts of compound containing
three carbon atoms.
Stage 4: Formation of Glyceraldehyde–3–Phosphate and
Dihydroxyacetone Phosphate: The six carbon fructose–1, 6–phosphate
is split to form two three carbon compounds. The two three carbon
compounds are glyceraldehyde phosphate and dihydroxyacetone phosphate.
The reaction is catalysed by the aldolase or fructose –1, 6–phosphate
aldolase.
Stage 5: Formation of Two Molecule Glyceraldehyde–3–Phosphate:
The dihydroxyacetone phosphate is then converted to its isomer
glyceraldehyde–3–phosphate by the enzyme phosphotriose isomerase.
Hence, two molecules of gylceraldehyde–3–phosphate are acquired by
splitting of one glucose molecule. Hereafter every intermediate will be double
in number.
3. Pay-Off Stage: This stage gives one ATP.
Stage 6: Formation of 1, 3–Bisphosphoglycerate: Glyceraldehyde–3–
phosphate is converted to 1, 3–bisphosphoglycerate in the presence of the
enzyme glyceraldehyde–3–phosphate dehyrogenase with the formation of
NADH + H+. This step utilizes one inorganic phosphate to form 1,3–
bisphosphoglycerate, a high energy compound. The NADH formed enter
into the Electron Transport Chain (ETC) and synthesize 6 ATP (3 ATP per
molecule of NADH formed from 2 glyceraldehyde–3–Phosphate to two
1.3–biphosphoglycerate) by oxidative phosphorylation.
Stage 7: Fomation of 3–Phosphoglycerate: 1.3–biphospoglycerate is
converted to 3–phosphoglycerate with the formation of ATP. The reaction
occurs in the presence of the enzyme phosphoglycerate kinase. When ATP
is generated from the substrate without the help of ETC, it is known as a
substrate level phosphorylation. Thus, this reaction is an example of substrate
level phosphorylation.
Self-Instructional
Material 35
Carbohydrate Stage 8: Formation of 2–Phosphoglycerate: 2–phosphoglycerate is
Metabolism
formed from 3–phosphoglycerate with the action of the enzyme
phosphoglycerate mutase. This is a type of isomerization reaction.
Stage 9: Formation of Phosphoenol pyruvate: The enzyme Enolase
NOTES
acts on 2– phosphoglycerate and converts it to phosphoenol pyruvate. This
step requires Mg2+ or Mn2+. This is the step which is inhibited by the fluoride
and this principle is used to stop glycolysis to estimate blood glucose level.
Stage 10: Formation of Pyruvate: Pyruvate is synthesized from
phosphoenolpyruvate by the action of pyruvate kinase with generation of
ATP from ADP. K+ and Mg2+ or Mn2+ are required by the enzyme pyruvate
kinase. This step is also a good example of substrate level phosphorylation.
2.2.2 Fate of Pyruvate

Intake of Pyruvate
The fate of pyruvate will be decided on the basis of presence or absence of oxygen.
In anaerobic condition or less oxygen tension, pyruvate will form lactate.
Otherwise in aerobic condition or normal oxygen condition, pyruvate will form
Acetyl CoA and will enter into the Citric acid cycle (Refer Figure 2.2).

Fig. 2.2 Fate of Pyruvate

Anaerobic Condition
In the absence of oxygen or in anaerobic condition, pyruvate is reduced to
lactate by the activity of the enzyme lactate dehydrogenase. This step utilizes
NADH from the reaction of Stage 6 (in which glyceraldehydes–3–
phosphate dehydrogenase convert gylceraldehyde –3–phosphate to 1, 3–
bisphosphoglycerate. The formation of NAD+ from NADH, is used to continue
the gylcolysis by supplying NAD+ to stage 6 continuously. There is a loss of ATP
in this step because of utilization of NADH. Thus, 6 less number of ATP molecules
are formed in lactate formation. Lactate formation occurs in skeletal muscles during
heavy exercise due to lack of oxygen supply. This step also occurs in RBCs which
lack mitochondria to continue the glycolysis.
Self-Instructional
36 Material
Energetics of Glycolysis Carbohydrate
Metabolism
The energetics of glycolysis takes places in two conditions, namely, aerobic condition
and anaerobic condition. These are as follows:
In aerobic conditions: The energetics of glycolysis in aerobic conditions is shown NOTES
in Table 2.2.
Table 2.2 Energetics of Glycolysis in Aerobic Conditions

Enzyme No. of ATPs Synthesized


Glyceraldehyde–3–phosphate 6
dehydrogenase (2 NADH, etc.
oxidative phosphorylation).
Phosphoglycerate kinase 2
(substrate level phosphorylation).
Pyruvate kinase (substrate level 2
kinase).
Two ATPs are consumed in the -2
reaction catalysed by hexokinase
and phophofructokinase.
Net ATP synthesized in glycolysis. 8

In anaerobic condition
6ATP consumed by lactate dehydrogenase. -6
Net ATP (synthesized). 2

Conversion of Pyruvate to Acetyl CoA


Acetyl CoA can arise from many sources. But Acetyl CoA is formed readily from
pyruvate of glcolytic pathway. The multienzyme pyruvate dehydrogenase complex
converts pyruvate to Acetyl CoA by oxidative decarboxylation reaction.
The pyruvate dehydrogenase enzyme has the following three catalytic sites:
x Pyruvate Dehyrogenase (E1)
x Dihydrolipoyl Transacetylase (E2)
x Dihydrolipoyl Dehyrogenase (E3)
The enzyme pyruvate dehydrogenase requires five cofactors (coenzymes),
which are as follows;
x TPP (Thiamine Pyrophosphate)
x Lipoamide
x FAD (Flavine Adenine Dinucleotide)
x Coenzyme A
x NAD (Nicotinamide Adenine Dinucleotide)
The coenzymes and prosthetic groups of pyruvate dehydrogenase are shown in
Table 2.3.
Self-Instructional
Material 37
Carbohydrate Table 2.3 Coenzymes and Prosthetic Groups of Pyruvate Dehydrogenase
Metabolism
Cofactors Location Function
Thiamine Pyrophosphate Bound to E1 Decarboxylate pyruvate yielding
a hydroxyethyl TPP carbanion
NOTES
Lipoic Acid Covalently linked to a Accepts the hydroxyethyl
Lys on E2 (lipoamide) carbanion from TPP as an acetyl
group
Coenzyme A (CoA) Substrate for E2 Accepts the acetyl group from
lipoamide
Flavin Adenine Bound to E3 Reduced by lipoamide
Dinucleotide (FAD)
Nicotinamide Adeninbe Substrate for E3 Reduced by FADH2
Dinucleotide (NAD+)

In this reaction, there is a formation of 2 NADH per glucose molecule.


Thus, 6 ATP (2 × 3 ATP) molecules are formed in this step.
2.2.3 Citric Acid Cycle, Krebs Cycle or Tricarboxylic Acid (TCA) Cycle
This is the major pathway for energy supply to the body. This cycle involves
oxidation of acetyl CoA to CO2 and H2O (Refer Figure 2.3).
The cycle was discovered by Hans Adolf Krebs in 1937 for which he was
awarded the Noble prize for Physiology and Medicine in 1953. Hence, the cycle
was named after him.
Location of TCA cycle
Enzymes are found in mitochondrial matrix just near to ETC, so that ATP can be
synthesized readily without any delay.

Fig 2.3 TCA Cycle


Self-Instructional
38 Material
Step 1: Formation of Citrate: The condensation reaction between Carbohydrate
Metabolism
acetylCoA and oxalacetate occurs in the first step. This reaction is catalysed by
the enzyme citrate synthase. Since, citrate is the first compound formed in the
cycle, the cycle is known as citric acid cycle. The citrate contains three carboxylate
NOTES
groups. Therefore, it is known as TCA cycle.
Steps 2 and 3: Formation of Isocitrate: This step consists of two stages.
First, dehydration occurs and after that rehydration occurs. Citrate is first converted
to cis-aconitate by dehydration reaction and then forms isocitrate by rehydration
reaction. The enzyme involved in both the reactions is Aconitase.
Steps 4 and 5: Formation of D – Ketoglutarate: Isocitrate is converted
to a-ketoglutarate via formation of oxalosuccinate. This reaction is carried out
by isocitrate dehydrogenase. NADH and CO2 are released at this stage. The
NADH formed enters into ETC and produces 3 ATP by oxidative
phosphorylation.
Step 6: Formation of Succinyl CoA: D-Ketoglutaryl dehydrogenase
oxidative decarboxylate D – ketoglutarate to succinyl CoA. This enzyme requires
five cofactors—TPP, Lipoamide, NAD+, FAD and CoA. NADH and CO2 are
formed for the second time in this cycle. Therefore, again 3 ATP molecules are
formed in this reaction.
Step 7: Formation of Succinate: Succinyl CoA synthatase converts succinyl
CoA to succinate with simultaneous formation of GTP from GDP and Pi. This
GTP ultimately results in the formation ofATP by the enzyme nucleoside diphosphate
kinase:
GTP + ADP o ATP + GDP
Step 8: Formation of Fumarate: Succinate dehydrogenase oxidizes
succimate to fumarate. This results in the formation of FADH2 instead of NADH.
The FADH2 enters ETC and releases a molecule of ATP through oxidative
phosphorylation.
Step 9: Formation of Malate: Fumarate is hydrated to form malate in the
reaction catalysed by fumarase enzyme .
Step 10: Regeneration of Oxaloacetate: Malate undergoes oxidation
reaction with the formation of oxaloacetate. This reaction occurs in the presence
of the enzyme malate dehydrogenase and the third and last NADH forms. Thus,
the oxaloacetate formed is ready to condense with another Acetyl CoA.
The final reaction of TCA cycle is as follows:
Acetyl CoA + 3NAD+ + FAD + GDP + Pi + 2H2O o 2 CO2 + 3 NADH
+ 3H+ + FADH + GTP + CoA
This cycle repeats itself twice per molecule of glucose because two molecules
of Acetyl CoA are formed from one glucose molecule.
Self-Instructional
Material 39
Carbohydrate Energetics of TCA Cycle
Metabolism
The energetics of TCA cycle is shown in Table 2.4.
Table 2.4 Energetics of TCA Cycle
NOTES
Enzyme No. of ATPs Synthesized
Isocitrate Dehydrogenase (2 NADH, ETC, 6
Oxidative Phosphorylation)
α–ketoglutarate Dehydrogenase (2 NADH, 6
ETC, Oxidative Phosphorylation)
Succinate Thiokinase (Substrate Level 2
Phosphorylation)
Succinate Dehydrogenase (2 FADH2, ETC, 4
Oxidative Phosphorylation)
Malate Dehydrogenase (2 NADH, ETC, 6
Oxidative Phosphorylation)
Total ATP Formed 24

Table 2.5 shows the energetics of glucose metabolism in aerobic conditions.


Table 2.5 Energetics of Glucose Metabolism

Source No. of ATPs Formed


From Glycolysis 8
From Pyruvate Dehydrogenase 6
(2 NADH, ETC, Oxidative Phosphorylation)
From TCA Cycle 24
Total ATP Per Molecule of Glucose Under 38
Aerobic Condition
Total ATP Per Molecule of Glucose Under 2
Anaerobic Condition

Energetics of Glucose Oxidation


The reaction of glucose oxidation is as follows:
C6H12O6 + 6 O2 + 38 ADP + 38 Pi o 6CO2 + 6H2O + 38 ATP
You know that one ATP has a high energy bond of 30.5 KJ. Therefore, 38
ATP store energy of = 30 × 30.5 = 1159 KJ. You also know that if a glucose
molecule is burnt in a calorimeter, approximately 2780 KJ of heat is released.
Therefore, it could be concluded that 48 per cent of the energy in glucose
burning is used to synthesize 38 ATP molecules.
2.2.4 Gluconeogenesis
When glucose is synthesized from non-carbohydrate compound, it is known as
gluconeogenesis. The main precursors are as follows:
x Lactate
x Pyruvate
Self-Instructional
40 Material
x Glucogenic Amino Acid Carbohydrate
Metabolism
x Propionate
x Glycerol
Site of Gluconeogenesis NOTES
Gluconeogenesis occurs in the cytosol of cell. It mostly occurs in liver and sometimes
in the kidney matrix.
Reaction of gluconeogenesis
The reaction of gluconeogenesis is shown in Figure 2.4.

Fig. 2.4 Gluconeogenesis

The pathway for gluconeogenesis is reverse of glycolysis except three steps which
are irreversible. The steps which are catalysed by the following enzymes are
irreversible:
x Hexokinase
x Phosphofructokinase
x Pyruvate Kinase
Thus, these three steps are bypassed by other enzymes leading to glucose synthesis,
i.e., gluconeogenesis.
x Formation of Phosphoenolpyruvate from Pyruvate: Since, pyruvate is
present in mitochondria and the phosphoenolpyruvate forms in cytosol, this
step is divided into following three stages:
o Formation of Oxaloacetate: In mitochondria, pyruvate carboxylase
converts pyruvate to oxaloacetate by utilization of one ATP and one
CO2.
Self-Instructional
Material 41
Carbohydrate o Transportation of Oxaloacetate from Mitochondria to Cytosol
Metabolism
via the Formation of Malate: Oxaloacetate formed in the
mitochondria is required to be transported across the cytosol. But the
mitochondria membrane is impermeable to oxaloacetate . So,
NOTES oxaloacetate is first converted to malate in the presence of an enzyme
named malate dehydrogenase. The malate dehydrogenase is such an
enzyme which exists on the inner side of mitochondria as well as
cytosol. Malate is transported outside the cytosol by malate transporter.
Then, oxaloacetate is regenerated from malate by the same enzyme
malate dehydrogenase.
o Formation of Phosphoenolpyruvate: In the cytosol, oxaloacetate
is converted to phosphoenolpyruvate. This reaction occurs in the
presence of phosphoenolpyruvate carboxykinase with the utilization
of GTP or ITP and not ATP. The CO2 fixed in reaction catalyzed by
pyruvat carboxylase is removed by the phosphoenolpyruvate
carboxykinase.
x Formation of Fructose-6-Phosphate: The next irreversible step of
glycolysis is converting fructose-6-phosphate to fructose-1,6-bisphosphate.
For the formation of fructose-1,6- bisphosphate from phosphoenolpyruvate,
it is reversed. The enzyme fructose-1,6-bisphosphatase reverses this reaction
and fructose-6-phosphate is formed from fructose-1,6-bisphosphate. This
is not present in smooth muscle and heart muscle. This enzyme require
Mg2+ for its functioning.
x Formation of Glucose– 6-Phosphate: This step of conversion of glucose-
6-phosphate to glucose is catalyzed by glucose-6-phosphatase. Enzymes
for this conversion are present in liver and kidney but they are absent in
muscles, brain and adipose tissue.
Overall Reaction of Gluconeogenesis:
2 pyruvate + 4 ATP + 2 GTP + 2NADH + 2H+ + 6 H2O o Glucose + 2 NAD+
+ 4 ADP + 2 GDP+ 6Pi + 6 H+
Substrates for Gluconeogenesis
1. Glucose from Lactate/Cori Cycle
Lactate is one of the most important sources of carbon atoms for glucose synthesis
by gluconeogenesis. In the anaerobic glycolysis of skeletal muscle, pyruvate is
reduced to lactate by Lactate Dehydrogenase (LDH) enzyme. This reaction serves
two important functions during anaerobic glycolysis. These are as follows:
x During lactate formation, the LDH reaction requires NADH and yields
NAD + which is then used by the glyceraldehyde-3-phosphate
dehydrogenase reaction of glycolysis. Thus, these two reaction are, intimately
coupled during anaerobic glycolysis.
Self-Instructional
42 Material
x The lactate produced by the LDH reaction is released to the blood stream Carbohydrate
Metabolism
and transported to the liver where it is converted to glucose. Thus, the
glucose is then returned to the blood for use by muscle as an energy source
and to replenish glycogen stores. This cycle is termed as the cori cycle.
NOTES
The Cori cycle consumes energy during glyoclysis by costing four molecule of ATP
(Refer Figure 2.5).

Fig. 2.5 The Cori Cycle

2. Pyruvate
Pyruvate generated in the muscle and other peripheral tissues can be transaminated
to alanine. This reaction happens when an enzyme named Alanine Transaminase
(ALT) is present. This enzyme is also known as Serum Glutamate–Pyruvate
Transaminase (SGPT). After the reaction occurs, the alanine gets returned to the
liver for gluconeogenesis. The transamination reaction needs an D-amino acid
(glutamate) as a donor for the amino group, leading to the formation of an D-keto
acid (pyruvate) in the process. This pathway is known as the glucose–alanine
cycle. Though most of the amino acids get degraded in liver some even get de-
aminated. Therefore, the glucose–alanine cycle is, an indirect mechanism for the
muscle to eliminate nitrogen while replenishing its energy supply. This generally
occurs during body fasting condition. But, the glucose–alanine cycle’s major function
is allowing the non-hepatic tissues to deliver the amino portion of the catabolized
amino acids to the liver for the process of excretion in the form of urea. In the liver
the alanine gets converted back to pyruvate and then it is used as a gluconeogenic
substrate (if that is the hepatic requirement) or it is oxidized in the TCA cycle
(Refer Figure 2.6).

Fig. 2.6 Glucose–Alanine Cycle


Self-Instructional
Material 43
Carbohydrate 3. Amino Acids
Metabolism
All the amino acids (except leucine and lysine) can be degraded to TCA cycle
intermediates. This cycle helps in the formation of oxaloacetate and after that into
NOTES pyruvate from the carbon skeleton of the amino acid. Therefore, the pyruvate can
be used by the gluconeogenic pathway. When the glycogen that is stored decreases
in muscle due to excessive exertion and liver being in fasting condition, catabolism
of muscle proteins to amino acids acts as the major source of carbon for the
maintenance of blood glucose levels.
4. Glycerol
Oxidation of fatty acids lead to the formation of glycerol. It yields huge amounts of
energy on a molar basis. But the fatty acid carbons cannot be used for the final
synthesis of glucose. The two carbon units of acetyl-CoA are obtained from b-
oxidation of fatty acids. This oxidation is incorporated in TCA cycle. Thus, the
acetyl CoA is lost in the TCA cycle of two carbons as CO2. Therefore, it can be
concluded that fatty acids do not undergo net conversion for carbohydrate.
The glycerol backbone of lipids can be used for gluconeogenesis. The steps
involved in this process need phosphorylation of glycerol-3-phosphate by glycerol
kinase and dehydrogenation to DiHydroxyAcetone Phosphate (DHAP) by
Glyceraldehyde-3-Phosphate DeHydrogenase (G3PDH). The glyceraldehyde-
3-phosphate dehydrogenase reaction is identical to the one used in the transport
of cytosolic reducing equivalents into the mitochondrion for use in oxidative
phosphorylation. This transport pathway is known as glycerol phosphate shuttle
(Refer Figure 2.7).

Fig. 2.7 Glycerol Phosphate Shuttle

The glycerol phosphate shuttle is a secondary mechanism for the transport


of electrons from cytosolic NADH to mitochondrial carriers of the oxidative
phosphorylation pathway. The primary cytoplasmic NADH electron shuttle is the
malate-aspartate shuttle. This shuttle involves two enzymes. One shuttle is the
Self-Instructional
44 Material
cytosolic version of the enzyme glycerol-3-phosphate dehydrogenase (glycerol- Carbohydrate
Metabolism
3-PDH) which has a substrate, named, NADH whereas the other shuttle is the
mitochondrial form of the enzyme which has FAD+ as one of its substrates. The
final result is that there is a regular conversion of the glycolytic intermediate, DHAP
NOTES
and glycerol-3-phosphate with the concomitant transfer of the electrons from the
reduced cytosolic NADH to mitochondrial oxidized FAD+. As, the electrons from
mitochondrial FADH2 feed into the oxidative phosphorylation pathway at co-enzyme
Q (as opposed to NADH-ubiquinone oxidoreductase [complex I]) only two moles
of ATP will be generated from glycolysis.
The glycerol backbone of adipose tissue stored triacylgycerols is ensured
of being used as a gluconeogenic substrate since adipose cells lack glycerol kinase.
In fact, adipocytes require a basal level of glycolysis in order to provide them with
DHAP as an intermediate in the synthesis of triacyglycerols.
5. Propionate
Oxidation of fatty acids with an odd number of carbon atoms and amino acids
generates the terminal of oxidation product, namely, propionyl-CoA. Propionyl-
CoA is converted in TCA intermediate, succinyl-CoA. This conversion gets carried
out by the ATP-requiring enzyme, propionyl-CoA carboxylase then methylmalonyl-
CoA epimerase and finally the vitamin B12 requiring enzyme, methylmalonyl-CoA
mutase. The utilization of propionate in gluconeogenesis only has quantitative
significance in ruminants. Figure 2.8 shows the conversion of propionyl-CoA to
succinyl-CoA.

Fig. 2.8 Conversion of Propionyl-CoA to Succinyl-CoA

6. Glucose from Fat


Glucose and fat cannot be generated in animals because they lack glyoxylate cycle.
This cycle is present in plants and microorganisms so they can utilize fat to form
glucose.
In the absence of available carbohydrates in these organisms (for example,
in certain microbial environments or during seed germination in plants), the glyoxylate
cycle allows the synthesis of glucose from lipids through acetate which is generated
in fatty acid â-oxidation.
The glyoxylate cycle bypasses the steps in the citric acid cycle where carbon
gets lost in the form of CO2. The initial two steps of the glyoxylate cycle are
Self-Instructional
Material 45
Carbohydrate identical to those in the citric acid cycle: acetateocitrateoisocitrate. In the first
Metabolism
step, catalyzed by the first glyoxylate cycle enzyme, isocitrate lyase, isocitrate
undergoes cleavage into succinate and glyuoxylate (the latter gives the cycle its
name). Glyoxylate condenses with acetyl-CoA. It is a step catalyzed by malate
NOTES
synthase, yielding malate. Both the malate and oxaloactate can be converted into
phosphoenolpyruvate, which is a substrate of phosphoenolpyruvate
carboxikinase—the first enzyme in gluconeogenesis. Therefore, the final outcome
of the glyoxylate cycle is the production of glucose from fatty acids. Succinate
generated in the first step can enter into the citric acid cycle to eventually form
oxaloacetate.
In plants, the glyoxylate cycle occurs in special organelle peroxisomes known
as glyoxysomes. It was once thought that vertebrates cannot perform this cycle as
there was no evidence of the existence of its two key enzymes, namely, isocitrate
lyase and malate synthase (Refer Figure 2.9).

Fig. 2.9 Glyoxylate Cycle

Check Your Progress


1. What is gluconeogenesis?
2. What is glycogenolysis?
3. Where is the enzyme salivary amylase found?
4. In what condition, pyruvate forms lactate?
Self-Instructional
46 Material
Carbohydrate
2.3 GLYCOGEN METABOLISM Metabolism

This has two parts. One is the catabolic part ,i.e., glycogenolysis and the other is
the anabolic part, i.e., glycogenesis. NOTES
2.3.1 Glycogenolysis
Glycogenolysis refers to excess of glucose stored in the form of glycogen in muscle
and liver. The glycogen present in muscle is a ready source of energy both for
aerobic as well as anaerobic condition. After heavy activities, the glycogen in muscle
is lost. But the glycogen in the liver serves as a reservoir of glucose for other
tissues during fasting condition, for example, nerve cell of brain which depends
only on glucose as fuel.
The catabolic pathway for glycogen to form glucose-6-phosphate is known as
glycogenolysis.
The following three enzymes are important for conversion of glycogen to glucose:
1. Glycogen Phosphorylase
2. Glycogen Debranching Enzyme
3. Phosphoglucomutase
The following are the functions of the above-mentioned three enzymes:
x Action of Glycogen Phosphoryladse: Glycogen phosphorylase along with
Inorganic Phosphate (Pi) attacks on the D-14-glycosidic linkage between
two glucose residues at a non-reducing end of glycogen. As a result, the
terminal glucose residue is removed in the form of D–D–glucose-1-
phosphate. This is a form of phosphorolysis reaction which conserves the
glycosidic bond energy in the form of phosphate bond. This reaction requires
pyridoxal phosphate in the form of cofactor. The non-reducing end of
glycogen is continuously acted upon by glycogen phosphorylase till it reaches
a D-16-branching.
x Action of Debranching Enzyme: The debranching enzyme shows its action
on the D-16-branching. The debranching enzyme has following types of
activity.
o Transferase Activity
o D-1,6-Glucosidase Activity
After debranching action, a glucose molecule is released. The remaining
section contains unbranched D-1,4 polymer.
x Action of Phosphoglucomutase: Phosphoglucomutase isomerizes the
glucose-1-phosphate to glucose -6-phosphate. This glucose-6-phosphate
enters into glycolysis pathway.

Self-Instructional
Material 47
Carbohydrate 2.3.2 Glycogenesis
Metabolism
The anabolic pathway for the formation of glycogen from glucose is known as
glycogenesis. Glycogen formation requires a lot of energy.
NOTES It was found out that glucose-1-phosphate combined with Uridine
Triphosphate (UTP) to form UDP-glucose. This was discovered by Luis Leloir in
1957.
Pathway of Glycogenesis: Glycogenesis requires three enzymes to
synthesize glycogen:
x UDP-Glucose Pyrophosphorylase
x Glycogen Synthase
x Glycogen Branching Enzyme
Following are the reactions of glycogenesis:
x UDP-Glucose Pyrophosphorylase: The enzyme UDP-glucose
pyrophosphorylase helps in attacking of phosphoryloxygen of glucose-1-
phosphate on the D-phosphorus atom of UTP to form UDP glucose with
the release of inorganic pyrophosphate (PPi). The PPi is then hydrolyzed
by enzyme Inorganic PyroPhosphatase to inform Inorganic Phosphate (Pi).
Glucose-1-phosphate+UTPoUDP-glucose+PPi+H2O + PPi +2 Pi
Glucose-1-phosphate +UTPoUDP-glucose + 2Pi
x Action of Glycogen Synthase: The glucosyl unit of UDP glucose is
transferred to the (C4)–OH group one of glycogen’s non-reducing and to
form an D-1,4 glycosidic bond. This reaction is catalyzed by enzyme glycogen
synthesis. The UDP which is released, is converted to UTP in the presence
of enzyme nucleoside diphosphate kinase and simultaneously, a molecule
of ATP is utilized.
UDP + ATPoUTP + ADP
Glycogen synthesis is initiated by the attachment of a glucose residue to a
specific Tyr–OH group of protein named glycogenin. So, for glycogen
synthesis to start, a glycogenin protein as a primer is required.
x Action of Glycogen Branching Enzyme: The name of glycogen branching
enzyme is amylo-1,4 1,6-transglycosylase. This branching first breaks the
D-1,4 glycosidic linkage and then modifies it to D-1,6 glycosidic bond.
The glucosyl residue is attached to the (C6)–OH group of glucose residues
on the same or another glycogen chain.
The pentose phosphate pathway (also known as the phosphogluconate
pathway and the hexose monophosphate shunt) is a method that generates
NADPH and pentoses (5-carbon sugar). This pathway has two distinct
phases. The first phase is known as the oxidative phase, in which NADPH
is generated, and the second phase is termed as non-oxidative synthesis of
5-carbon sugars. This pathway is another option for glycolysis and although
Self-Instructional
48 Material
it does involve oxidation of glucose, its major role is anabolic rather than Carbohydrate
Metabolism
catabolic. For most of the organisms, it takes place in the cytosol.
2.3.3 Pathway of HMP
The pathway of HMP shunt has been divided into two phases: NOTES

1. Oxidative Phase: Glucose-6-phosphate is the starting molecule in oxidative


phase. 6-phosphogluconate is synthesized from glucose-6-phosphate in
the presence of enzyme, glucose-6-phosphate dehydrogenase with the
utilization of NADP. 6-phosphogluconate is then converted to 6-
phosphogluconate in the presence of enzyme gluconolactoe hydrolase which
shows the hydrolysis reaction. In the next step, phosphogluconate
dehydrogenase acts on 6-phosphogluconate and CO2 is released. As a
result, ribulose-5-phosphate is formed with coconmitant formation of
NADPH (Refer Figure 2.8).
2. Non-Oxidative Phase: This pathway starts with the ribulose-5-phosphate
as shown in Figure 2.9.This substrate is acted upon by two enzymes. They
are as follows:
x Ribulose 5-phosphate epimerase alters the configuration with the
formation of ketopentose, xylulose 5-phosphate.
x Ribose 5-phosphate ketoisomerase acts on ribulose 5-phosphate to
form corresponding aldopentose, ribose 5-phosphate.This is the
precursor of the ribose required for nucleotide and nucleic acid
synthesis.
Transketolase transfers the two-carbon unit comprising of carbons 1 and 2 of
a ketose onto the aldehyde carbon of an aldose sugar. It therefore affects the
conversion of a ketose sugar into an aldose with two carbons less and
simultaneously converts an aldose sugar into a ketose with two carbons
more. The reaction requires Mg2+ and thiamin diphosphate (vitamin B1) as
coenzyme. Thus, transketolase catalyzes the transfer of the two-carbon unit
from xylulose 5-phosphate to ribose 5-phosphate, producing the seven-
carbon ketose sedoheptulose-7-phosphate and the aldose glyceraldehydes
3-phosphate. Transaldolase allows the transfer of a three-carbon
dihydroxyacetone moiety (carbons 1–3) from the ketose sedoheptulose
7-phosphate onto the aldose glyceraldehyde 3-phosphate to form the ketose
fructose 6-phosphate and the four-carbon aldose erythrose-4-phosphate.
In a further reaction catalyzed by transketolase, xylulose 5-phosphate donates
a two-carbon unit to erythrose 4-phosphate to form fructose 6-phosphate
and glyceraldehyde 3-phosphate. In order to oxidize glucose completely to
CO2 via the pentose phosphate pathway, there must be enzymes present in
the tissue to convert glyceraldehyde 3-phosphate to glucose 6-phosphate.
This involves the reversal of glycolysis and the gluconeogenic enzyme fructose
Self-Instructional
Material 49
Carbohydrate 1,6-bisphosphatase. In tissues that lack this enzyme, glyceraldehydes 3-
Metabolism
phosphate follows the normal pathway of glycolysis.
Figure 2.10 shows the oxidative stage of Pentose Phosphate pathway.
NOTES

Fig. 2.10 Oxidative Phase of Pentose Phosphate Pathway

Figure 2.11 shows the non-oxidative stage of Pentose Phosphate Pathway.

Fig. 2.11 Non-oxidative Phase of Pentose Phosphate Pathway


Self-Instructional
50 Material
2.3.4 Uronic Acid Pathway Carbohydrate
Metabolism
This is also known as glucoronic acid pathway. It is another alternate pathway for
the oxidation of glucose. This is used to synthesize the following:
x Glucuronic Acid NOTES
x Pentoses
x Ascorbic Acid
The reaction in uronic acid is as follows:
1. Formation of UDP (Glucuronate): The reaction starts with glucose
phosphate. The enzyme phosphoglucomutase isomerizes glucose-6-
phosphate to glucose-1-phosphate. This glucose-1-phosphate is then
converted to UDP glucose in the presence of UDP glucose
pyrophosphorylase with the utilization of UTP. After that, the UDP—glucose
is oxidized to UDP—glucuronates with the help of enzyme UDP
dehydrogenises (Refer Figure 2.12).
The UDP-glucuronate has many different fates, such as the following:
(i) UDP glucuronate helps in converting insoluble compound to soluble
compound and then detoxify it.
x It is also used for synthesis of heteropolysaccharide (such as
gylcosaminoglcans).
x It can conjugate with other substances such as bilirubin, steroid
hormones and certain drugs.
(ii) Formation of L–Gluconate: UDP-glucuronate is acted upon by
hydrolytic enzymes, glucuronidase and forms D–glucuronate with
concomitant release of UDP. The D-glucuronate is then converted L-
gulonate by an NADPH-dependent reaction.
(iii) Fate of L-Gluconate: It can enter into any of the following two
pathways:
x Formation of Ascorbic Acid: Enzyme L–gulonolactone is
absent in humans. 2-keto-L- gulonolactone further forms L-
ascorbic acid.
x Formation of D-Xylulose: L-gulonate undergoes oxidation
reaction to produce 3- ketogulonate with the formation of
NADH. L-xylulose is synthesized from 3- ketogulonate by
decarboxylation reaction. L-xylulose further forms D-xylulose
in a NADPH-dependent reaction. These D-xylulose then enter
HMP shunt by forming xylulose-5-phosphate.

Self-Instructional
Material 51
Carbohydrate
Metabolism

NOTES

Fig. 2.12 Glucuronic Acid Synthesis

2.3.5 Metabolism of Galactose


Milk and milk products contain disaccharide lactose. This lactose is acted upon
by enzyme lactase in intestine and is hydrolyzed into glucose and galactose.
Galactose is then absorbed in intestinal wall and this flows into cell via blood.
The pathway for metabolism of galactose consists of the following:
1. Formation of Galactose-1-Phosphate: Galactose undergoes
phosphorylation reaction to form galactose-1-phosphate with the action of
enzyme galactokinase.
2. Formation of UDP-Galactose: Galactose is converted to UDP-galactose
in presence of enzyme galctose-1-phosphate uridyltransferase and the UDP
is obtained from UDP-glucose. The UDP-galactose formed has two
following fates:
x It can be utilized in the formation of compound like lactose, glycolipids,
glycoproteins, glycosaminoglycans and cerebrosides.
x It can be converted to UDP glucose with the help of enzyme UDP-
hexose-4-epimerase and ether into glucose metabolism. That’s why,
galactose is not an essential dietary requirement because it can be
converted to glucose in body.
2.3.6 Metabolism of Fructose
The fructose is obtained from sucrose by the activity of enzyme sucrase in the
intestinal wall. It is transported to cell via blood (Refer Figure 2.13).
Following are the steps for the metabolism of fructose:
x Fructose is converted to fructose-1-phosphate in the presence of enzyme
fructokinase. Fructose-1-phosphate is then split into gylceraldehyde and
dihydoxyacetone phosphate by enzyme Aldolase B. Thus, the
glyceraldehyde formed is acted upon by the enzyme triokinase and as result
glycraldehyde-3-phosphate is formed. The glyceraldehyde-3-phosphate
either enters glycolysis or gluconeogenesis.
Self-Instructional
52 Material
x Fructose can also be converted to fructose-6-phosphate by the enzyme Carbohydrate
Metabolism
hexikinase and then it can enter glycolysis and gluconeogenesis.

NOTES

Fig. 2.13 Fructose

2.3.7 Futile Cycle


A futile cycle (substrate cycle) is observed when two metabolic pathways run
simultaneously in the opposite directions. This does not have any overall effect
and loss of energy in the form of heat. This is seen in case of glycolysis and
gluconeogenesis run at the same time. This results in the formation of pyruvate
from glucose by glycolsis. Thus, this pyruvate form can be converted back to
glucose in gluconeogenesis. This occurs with an overall consumption of ATP. Futile
cycles are important in the regulation of metabolic reaction by regulating cycle to
oscillate between two states. This gives a very high sensitivity to small changes in
the activity of any of the enzymes present in the metabolic pathway. The futile
cycle can only generate heat from the energy released which helps in maintaining
thermal homeostasis, for example in the brown adipose tissue of young mammals.
Giving an overall reaction of:
ATP + H2O o ADP + Pi + Heat
In the afore-mentioned equation, one ATP molecule is being hydrolyzed
without any useful metabolic work being done. Therefore, it can be concluded
that if these two reactions were allowed to proceed simultaneously at a high rate in
the same cell, a large amount of chemical energy would have been dissipated as
heat.
This process is not an economical one. Therefore, it has been called a futile
cycle.

Check Your Progress


5. What are the two part of glycogen metabolism?
6. Give three enzymes which are important for conversion of glycogen to
glucose.
7. What is futile cycle?
Self-Instructional
Material 53
Carbohydrate
Metabolism 2.4 METABOLIC DEFECTS RELATED TO
CARBOHYDRATES

NOTES Metabolic defects related to carbohydrates are as follows:


1. Diabetes Mellitus: This is a type of metabolic disorder in which the blood
glucose level increases, i.e., hyperglycemia (hyper - increases, glyce -
glucose, mia - related to blood). Blood glucose level is maintained by several
hormones. Among this all, insulin is the most important hormone. Insulin is
the hormone which is secreted by the b-cells of the islets of Langerhans of
pancreas. This is the most important hormone needed to regulate blood
glucose level. It is released in response to hyperglycemic condition. Insulin
is the only hormone which decreases blood glucose level. Insulin is the
hormone which binds to the cell surface receptor and sends information to
the cell to uptake the glucose from the blood. For blood to be in
hyperglycemic state, either insulin is not produced or if produced, is not
recognized by the cell surface receptor due to decrease in the level of
receptor.
There are two types of diabetes mellitus which are as follows:
x Insulin-Dependent Diabetes Mellitus (IDDM): This type of
diabetes mellitus is also known as type 1 diabetes. It can also be
called as juvenile onset diabetes because it is generally seen in children
of age 12–15 years. 10–20 per cent of diabetes cases are IDDM.
This diabetes is mainly caused due to absence of insulin production as
a result of destruction of E-cells of pancreas. The destruction of E-
cells may have been caused in response to certain drugs, virus and
autoimmunity. Autoimmunity may have been caused due to some
genetic changes of the cells that the E-cells are not recognized by the
immune system to get rid of the non-self recognized E-cells. Such
children lack total insulin production and as a result blood glucose
level increases after food intake. The patient is required to be given
insulin therapy. This is the reason that such diabetic patients are called
Insulin-Dependent Diabetes Mellitus (IDDM) as they depend on
binsulin externally.
x Non-Insulin Dependent Diabetes Mellitus (NIDDM): This type
of diabetes mellitus is also known as type 2 diabetes or Adult onset
diabetes. This type of diabetes is very common and is found in 80–
90 per cent of the diabetic population. It is generally found in adults
of above 35 years of age and is less severe than IDDM. The reason
behind NIDDM may be genetic and environmental. Obese people
due to overeating and sedentary lifestyle, are the most common cases
of NIDDM. One of the most important diabetogenic factors is obesity
Self-Instructional
54 Material
in individuals with a family history of diabetes. Obese people have Carbohydrate
Metabolism
increased resistance to the action of insulin because of decrease in
insulin receptors on the insulin responsive to target cells. NIDDM
people have normal or even increased serum insulin level. This is
because overeating increases the insulin production and reduces NOTES
insulin receptor synthesis. Therefore, NIDDM can be treated only
by losing weight. Table 2.6 shows the details of the two types of
diabetes.
Table 2.6 Type 1 and Type 2 Diabetes

Type 1 Type 2
Age at onset Usually under 30 Usually over 40
Symptom onset Abrupt Gradual
Body weight Normal Obese – 80%
HLA association Positive Negative
Family history Common Nearly universal
Insulin in blood Little to none Some usually present
Islet cell antibodies Present at onset Absent
Prevalence 0.2% – 0.3% 6%
Symptoms Polyuria, polydipsia, Polyuria, polydipsia,
polyphgia, weightloss, peripheral neuropathy
ketoacidosis
control Insulin, diet and Diet, exercise and often oral
exercise hypoglycemic drugs or insulin
Vascular and neural changes Eventually develop Will usually develop
Stability of condition Fluctuates, may be May be difficult to control in
difficult to control poorly motivated patients

2. Lactic Acidosis: It is a physiological condition characterized by low pH in


body tissues and blood (acidosis) accompanied by the buildup of lactate,
and is considered a distinct form of metabolic acidosis. Lactic acid has
three carbons out of which one is carboxylic acid. Normal lactose level in
plasma is 4 – 15 mg /dl. The lactic acid in plasma increases in two conditions:
x Increased Production
x Decreased Utilization
Reduction in pH due to increased lactic acid in plasma is called lactic
acidosis. This can be of two types:
(i) Mild Lactic Acidosis: This is not life threatening. This occurs due to
strenuous exercise, shock, respiratory diseases, cancers, low pyruvate
dehydrogenase activity, Von Gierke’s disease, etc.
Self-Instructional
Material 55
Carbohydrate (ii) Severe Lactic Acidosis: This is life threatening. This condition is
Metabolism
created because of the impairment, collapse or damage to
circulatory system which is seen in myocardial infarction, pulmonary
embolism, uncontrolled haemorrhage and severe shock. The main
NOTES
reason behind this condition is depletion in O2 supply to the tissues
with sudden reduction in ATP synthesis because lactic acid
production leads to the generation of only 2 ATP. This causes cell
death or necrosis.
3. Glycogen Storage Disease (GSD): This disease is also known as
glycogenosis and dextrinosis. It occurs due to defects in the processing
of glycogen synthesis or breakdown within muscles, liver and other cell
types. GSD has two types of causes: genetic and acquired. Genetic GSD
is caused by any inborn error of metabolism (genetically defective enzymes)
involved in these processes. Acquired GSD is caused by intoxication with
the alkaloid castanospermine. GSD is the type of heritable diseases which
is characterized by an abnormal storage and accumulation of glycogen in
the tissue, generally in the liver (Refer Table 2.7). This includes the following
types:
x Phosphorylase E kinase deficiency: This is a form of glycogen
storage disease caused by an x-linked deficiency of the kinase which
activates phosphorylase activity.
x Type Ia: This type of inherited disorder is seen in the first year of the
life of a child. A child suffering from this disorder will have a defective
gene which results in the deficiency of enzyme glucose 6-phosphatase.
x Type Ib: This is similar to Ia but is less dangerous. The defect is in the
enzyme glucose-6-phosphatase microsomal translocase.
x Type II: This glycogen storage disorder occurs due to deficiency in
the enzyme lysosomal a-glucosidase.
x Type III: This diseased condition of glycogen storage occurs due to
defects in debranching enzymes in the liver and muscle tissues.
x Type IV: This type of glycogen storage disease is related to congenital
defect in branching enzyme in Glycogenesis.
x Type V: This disease occurs as a result of muscle phosphorylase
deficiency.
x Type VI: This disease occurs because of genetically defective liver
phosphorylase.
x Type VII: This is a type of glycogen storage disease in which the
defect lies in the muscle phosphofructokinase enzyme.

Self-Instructional
56 Material
Table 2.7 Types of Glycogen Storage Disease Carbohydrate
Metabolism
Name Enzyme Deficiency Eponym Symptoms
GSD type I Glucose-6-phosphatase Von Gierke's disease Growth failure
GSD type II Acid maltase Pompe's disease Death by age ~2 years
(infantile variant) NOTES
GSD type III Glycogen Debrancher Cori's disease or Forbes'
disease
GSD type IV Glycogen branching enzyme Andersen disease Failure to thrive, death at
age 5 years
GSD type V Muscle glycogen McArdle disease
phosphorylase
GSD type VI Liver glycogen phosphorylase Hers' disease
GSD type VII Muscle Phosphofructokinase Tarui's disease Growth retardation
GSD type IX Phosphorylase kinase, PHKA2 - Delayed motor
development,
Growth retardation
GSD type XI Glucose transporter, GLUT2 Fanconi–Bickel syndrome

4. Glucose-6-Phosphate Deficiency: This is an inherited sex-linked disorder.


The deficiency of Glucose-6-Phosphate Dehydrogenase (G6PD) occurs in
all the cells but its effect is more in RBC. The NADPH availability in RBC
is provided by the Hexose MonoPhosphate (HMP) shunt. If the G6PD is
defective, the NADPH synthesis does not occur in the RBC. This results in
the accumulation of methemoglobin and peroxides in erythrocytes. This
ultimately leads to haemolysis.
5. Wernicke–Kosakoff Syndrome: Wernicke–Korsakoff syndrome is
associated with a genetic defect in the HMP shunt. Defect in transketolase
activity decreases the affinity with thiamine pyrophosphate. The same
condition is also seen in alcoholics whose diets are vitamin deficient.
6. Essential Pentosuria: This disease is seen in case of deficiency of an
NADP-dependent enzyme Xylitol dehydrogenase. The deficiency of this
enzyme causes blockage in the formation of Xylitol from L- Xylulose. Such
persons excrete excess amount of L- Xylulose in urine. The disease is
generally asymptomatic.
7. Classical Galactosemia: This is a rare congenital disease involving
deficiency of the enzyme galactose-1-phosphate uridyl transferase. Following
are the manifestations of this disorder:
x The galactose level increases in blood, i.e., galactosemia, and urine,
i.e., Galactosuria.
x The galactose which gets accumulated will be diverted towards the
production of galactitol (dulcitol) in the presence of the enzyme aldose
reductase. Accumulation of Galctotol in lens leads to the development
of cataract.
Self-Instructional
Material 57
Carbohydrate x The accumulation of galactose-1-phosphate and galctotol in different
Metabolism
tissues, such as, liver, nervous tissue, lens and kidney disturb their
functions.
x The clinical symptoms observed in case of galactosemia are as follows:
NOTES
R Weight Loss
R Hepatosplenomegaly
R Jaundice
R Mental Retardation
R Cataract
R Aminoaciduria
R Albuminuria
The patient should avoid Galactose and Lactose containing food.
Galactosemia can also be due to the enzyme galactokinase. This would
also lead to galactosemia, galactosuria, formation and accumulation of
galactitol. They develop all other complications, such as, deficiency of
galactose-1-phosphate uridyltransferase except hepatic and renal
complications.
8. Essential Fructosuria: Defect in the gene for enzyme hepatic fructokinase
stops the conversion of fructose-1-phosphate from fructose. As a result,
fructose accumulates in tissues and is excreted through the urine. Such patients
are suggested to consume a fructose free diet.
9. Hereditary Fructose Intolerance: This condition is created because of
the lack of enzyme aldolase B. This results in the accumulation of fructose-
1-phosphate, severe hypoglycemia, vomitting, hepatic failure and jaundice.
Hypoglycemia occurs because gylcogenolysis is inhibited due to allosteric
inhibition of liver phosphorylase by fructose-1-phosphate. This can be
avoided by taking fructose and sucrose free diet.
10. Consumption of High Fructose: Fructose is rapidly converted to fructose-
1- phosphate by the enzyme fructokinase. As a result of this, aldolase B is
less active and this results in the accumulation of fructose-1- phosphate.
The Pi remains trapped in the organic molecule and cannot be used in other
necessary metabolic functions. This is known as sequestering phosphate. It
has been observed that high consumption of fructose leads to increased
uric acid in the blood. This occurs because of excess accumulation of ADP
and AMP which gets degraded to uric acid.
11. Mucopolysaccharidoses/ Lysosomal Storage Disease: This disease
is caused because of defects in the GlycosAminoGlycans (GAG). Previously,
GAGs was known as Mucopolysaccharides. Therefore, lysosomal storage
Self-Instructional
58 Material
disease is also known as Mucopolysaccharidoses. These are caused due Carbohydrate
Metabolism
to lack of enzymes required for degradation of GAGs that results in the
accumulation of GAGs in different tissues. Accumulation of GAG s leads to
skeletal deformities and mental retardation. Different types of
NOTES
Mucopolysaccharidoses are as follows:
x Hurler’s Syndrome (MPS – I): L-Iduronidase enzyme is defective.
x Hunter’s Syndrome (MPS – II): This disease shows defective
Iduronate sulphatase enzyme.
x Sanfilippo syndrome (MPS – III): In this case, four different types
of enzymes are defective, such as, heparin sulphamidase.
x Sly syndrome (MPS – VII): Such patients lack the enzyme
E-Glucuronidase.
12. Abnormalities of carbohydrate digestion: Any defect in disaccharides
such as lactase, sucrose, can cause the movement of undigested
disaccharides to the large intestine where they will lead to withdrawal of
water from intestinal mucosa by osmosis. This results in diarrhoea. Later,
these undigested carbohydrates will be degraded by bacterial activity which
will lead to flatulence. Flatulence is a type of condition where intestinal motility,
cramps and irritations occur. These occur because of the activity of intestinal
bacteria on carbohydrates that produces hydrogen, methane and carbon
dioxide. Sometimes, lactate and short chain fatty acids are also released.
Following are the disorders related to carbohydrate digestion:
Lactose Intolerance: This is caused due to defect in the enzyme lactase.
This causes lactose to flow into the large intestine which will lead to diarrhoea
and gas formation due to microbial degradation (flatulence).
Sucrase Deficiency: This disorder occurs because of deficiency in enzyme
sucrose. It also shows the same symptoms as that of lactose intolerance. All
kinds of intolerance can be avoided simply by taking a diet that is free of
these disaccharides.

2.5 ENTNER DOUDROFF PATHWAY

Entner-Doudoroff (ED) Pathway


x This pathway occurs in both aerobic and anaerobic condition.
x Occur in prokaryotes only.
x It occurs in cytoplasm.
x Pyruvate and glyceraldehyde-3-phosphate produced from glucose by ED.
Self-Instructional
Material 59
Carbohydrate
Metabolism

NOTES

Fig. 2.15 ED Pathway

x At first glucose is phosphorylated to glucose -6-phosphate by the enzyme


hexokinase.
x Glucose-6-phosphate is then oxidized to 6-phosphogluconolactone releasing
a molecule of NADPH. This reaction is catalyzed by the enzyme glucose-
6-phosphate dehydrogenase.
x Hydrolase enzyme converts 6-phopshogluconolactone to 6-
phosphogluconate.
x 6-phosphogluconate undergoes dehydration reaction catalyzed by 6-
phosphogluconate dehydratase to form 2-Keto 3-Deoxy 6-
PhosphoGluconate (KDPG).
x KDPG splits to form pyruvate and glceraldehyde-3-phosphate. It is
catalyzed by KDPG aldolase enzyme
Self-Instructional
60 Material
Carbohydrate
x Glyceraldehyde-3-phosphate is then metabolized by glycolysis to form Metabolism
pyruvate.
Significance of ED Pathway
NOTES
x This pathway used two specific enzymes, ie., 6-phosphogluconate
dehydratase and KDPG aldolase.
x This pathway generates 1 ATP, 1 NADH and 1 NADPH from one glucose
molecule.

Check Your Progress


8. What is diabetes mellitus?
9. What is lactose intolerance?
10. What is the significance of ED pathway?

2.6 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. It is the process of formation of glucose from non- carbohydrate precursors


(for example, amino acids, glycerol, etc.)
2. Glycogenolysis is the process of breaking down of glycogen to glucose.
3. Enzyme salivary amylase found in Mouth.
4. In anaerobic condition or less oxygen tension, pyruvate will form lactate.
5. Glycogen metabolism has two parts. One is the catabolic part, i.e.,
glycogenolysis and the other is the anabolic part, i.e., glycogenesis.
6. The three enzymes that are important for convension of glycogen to glucose
are: 1. Glycogen phosphorylase 2. Glycogen debranching enzyme 3.
Phosphoglucomutase
7. A futile cycle (substrate cycle) is observed when two metabolic pathways
run simultaneously in the opposite directions. This does not have any overall
effect and loss of energy in the form of heat. This is seen in case of glycolysis
and gluconeogenesis run at the same time.
8. Diabetes mellitus is a type of metabolic disorder in which the blood glucose
level increases, i.e., hyperglycemia (hyper - increases, glyce - glucose, mia
- related to blood). Blood glucose level is maintained by several hormones.
Among this all, insulin is the most important hormone.
9. Lactose intolerance is caused due to defect in the enzyme lactase. This
causes lactose to flow into the large intestine which will lead to diarrhoea
and gas formation due to microbial degradation (flatulence).
Self-Instructional
Material 61
Carbohydrate 10. ED pathway used two specific enzymes, i.e., 6-phosphogluconate
Metabolism
dehydratase and KDPG aldolase. This pathway generates 1 ATP, 1 NADH
and 1 NADPH from one glucose molecule.

NOTES
2.7 SUMMARY

x Gluconeogenesis is the process of formation of glucose from non-


carbohydrate precursors (for example, amino acids, glycerol, etc.)
x Glycogenesis is the process of glycogen synthesis from glucose.
x Glycogenolysis is the process of breaking down of glycogen to glucose.
x Hexose monophosphate shunt (Pentose phosphate pathway or direct
oxidative pathway) is an alternative pathway to glycolysis and TCA cycle
for oxidation of glucose to carbon dioxide and water.
x The dietary food contains polysaccharides (such as starch, glycogen),
disaccharides (such as lactose, sucrose) and in very less amount
monosaccharides (such as glucose, fructose).
x The fate of pyruvate will be decided on the basis of presence or absence of
oxygen. In anaerobic condition or less oxygen tension, pyruvate will form
lactate. Otherwise in aerobic condition or normal oxygen condition, pyruvate
will form Acetyl CoA and will enter into the Citric acid cycle.
x Lactate is one of the most important sources of carbon atoms for glucose
synthesis by gluconeogenesis. In the anaerobic glycolysis of skeletal muscle,
pyruvate is reduced to lactate by Lactate DeHydrogenase (LDH) enzyme.
x Pyruvate generated in the muscle and other peripheral tissues can be
transaminated to alanine. This reaction happens when an enzyme named
Alanine Transaminase (ALT) is present. This enzyme is also known as Serum
Glutamate–Pyruvate Transaminase (SGPT).
x All the amino acids (except leucine and lysine) can be degraded to TCA
cycle intermediates. This cycle helps in the formation of oxaloacetate and
after that into pyruvate from the carbon skeleton of the amino acid. Therefore,
the pyruvate can be used by the gluconeogenic pathway.
x Glucose and fat cannot be generated in animals because they lack glyoxylate
cycle. This cycle is present in plants and microorganisms so they can utilize
fat to form glucose.
x Glycogen metabolism has two parts. One is the catabolic part, i.e.,
glycogenolysis and the other is the anabolic part, i.e., glycogenesis.
Self-Instructional
62 Material
x Entner-Doudoroff (ED) pathway occurs in both aerobic and anaerobic Carbohydrate
Metabolism
condition.

2.8 KEY WORDS NOTES

x Carbohydrates: Any of a large group of organic compounds occurring in


foods and living tissues and including sugars, starch, and cellulose. They
contain hydrogen and oxygen in the same ratio as water (2:1) and typically
can be broken down to release energy in the animal body.
x Metabolism: The chemical processes that occur within a living organism
in order to maintain life.
x Anaerobic: Relating to or requiring an absence of free oxygen.
x Glucose: A simple sugar which is an important energy source in living
organisms and is a component of many carbohydrates.
x Pyruvate: A salt or ester of pyruvic acid called as pyruvate.

2.9 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions

1. How is pyruvate converted to acetyl CoA?


2. Write a short note on gluconeogenesis.
3. Write a short note on glycerol.
4. Draw a diagram to show glyoxylate cycle.
5. Discuss in brief the pathway of HMP.
6. What is glycogen storage disease?
Long Answer Questions

1. Discuss important pathways of carbohydrates metabolism.


2. Describe pathway of glycolysis.
3. Explain fate of pyruvate.
4. Describe tricarboxylic acid cycle.
5. What are metabolic defects related to carbohydrates?
6. Explain Entner-Doudroff pathway.
Self-Instructional
Material 63
Carbohydrate
Metabolism 2.10 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
NOTES S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
64 Material
Proteins

UNIT 3 PROTEINS
Structure NOTES
3.0 Introduction
3.1 Objectives
3.2 Protein and Its Structure
3.3 Classification of Proteins
3.4 Physical and Chemical Properties of Proteins
3.5 Answers to Check Your Progress Questions
3.5 Summary
3.7 Key Words
3.8 Self Assessment Questions and Exercises
3.9 Further Readings

3.0 INTRODUCTION

Protein, highly complex substance that is present in all living organisms. Proteins
are of great nutritional value and are directly involved in the chemical processes
essential for life. The importance of proteins was recognized by chemists in the
early 19th century, including Swedish chemist Jöns Jacob Berzelius, who in 1838
coined the term protein, a word derived from the Greek proteios, meaning ‘holding
first place’. Proteins are species-specific; that is, the proteins of one species differ
from those of another species. They are also organ-specific; for instance, within a
single organism, muscle proteins differ from those of the brain and liver.
A protein molecule is very large compared with molecules of sugar or salt
and consists of many amino acids joined together to form long chains, much as
beads are arranged on a string. There are about 20 different amino acids that
occur naturally in proteins. Proteins of similar function have similar amino acid
composition and sequence. Although it is not yet possible to explain all of the
functions of a protein from its amino acid sequence, established correlations
between structure and function can be attributed to the properties of the amino
acids that compose proteins.
Most proteins consist of linear polymers built from series of up to 20
different L-D- amino acids. All proteinogenic amino acids possess common
structural features, including an D-carbon to which an amino group,
a carboxyl group, and a variable side chain are bonded. Only proline differs from
this basic structure as it contains an unusual ring to the N-end amine group, which
forces the CO–NH amide moiety into a fixed conformation. The side chains of the
standard amino acids, detailed in the list of standard amino acids, have a great
variety of chemical structures and properties; it is the combined effect of all of the
amino acid side chains in a protein that ultimately determines its three-dimensional
structure and its chemical reactivity. The amino acids in a polypeptide chain are
Self-Instructional
Material 65
Proteins linked by peptide bonds. Once linked in the protein chain, an individual amino
acid is called a residue, and the linked series of carbon, nitrogen, and oxygen
atoms are known as the main chain or protein backbone.
In this unit, you will study about proteins, their classification, physical and
NOTES
chemical properties, sources, biological role and value of protein in detail.

3.1 OBJECTIVES

After going through this unit, you will be able to:


x Understand what proteins are
x Classify proteins
x Analyse the physical and chemical properties of proteins
x Discuss the sources, biological role and value of protein

3.2 PROTEIN AND ITS STRUCTURE

Proteins are large biomolecules, or macromolecules, consisting of one or more


long chains of amino acid residues. Proteins perform a vast array of functions
within organisms, including catalysing metabolic reactions, DNA replication,
responding to stimuli, providing structure to cells and organisms, and transporting
molecules from one location to another. Proteins differ from one another primarily
in their sequence of amino acids, which is dictated by the nucleotide sequence of
their genes, and which usually results in protein folding into a specific three-
dimensional structure that determines its activity.
A linear chain of amino acid residues is called a polypeptide. A protein
contains at least one long polypeptide. Short polypeptides, containing less than
20–30 residues, are rarely considered to be proteins and are commonly called
peptides, or sometimes oligopeptides. The individual amino acid residues are
bonded together by peptide bonds and adjacent amino acid residues.
The sequence of amino acid residues in a protein is defined by the sequence of
a gene, which is encoded in the genetic code. In general, the genetic code specifies
20 standard amino acids; however, in certain organisms the genetic code can
include selenocysteine and—in certain archaea—pyrrolysine. Shortly after or even
during synthesis, the residues in a protein are often chemically modified by post-
translational modification, which alters the physical and chemical properties, folding,
stability, activity, and ultimately, the function of the proteins. Sometimes proteins
have non-peptide groups attached, which can be called prosthetic
groups or cofactors. Proteins can also work together to achieve a particular function,
and they often associate to form stable protein complexes.
Once formed, proteins only exist for a certain period and are
then degraded and recycled by the cell’s machinery through the process of protein
Self-Instructional
66 Material
turnover. A protein’s lifespan is measured in terms of its half-life and covers a wide Proteins

range. They can exist for minutes or years with an average lifespan of 1–2 days in
mammalian cells. Abnormal or misfolded proteins are degraded more rapidly either
due to being targeted for destruction or due to being unstable.
NOTES
Like other biological macromolecules such as polysaccharides and nucleic
acids, proteins are essential parts of organisms and participate in virtually every
process within cells. Many proteins are enzymes that catalyse biochemical reactions
and are vital to metabolism. Proteins also have structural or mechanical functions,
such as actin and myosin in muscle and the proteins in the cytoskeleton, which
form a system of scaffolding that maintains cell shape. Other proteins are important
in cell signaling, immune responses, cell adhesion, and the cell cycle. In animals,
proteins are needed in the diet to provide the essential amino acids that cannot
be synthesized. Digestion breaks the proteins down for use in the metabolism.
Proteins may be purified from other cellular components using a variety of
techniques such as ultracentrifugation, precipitation, electrophoresis,
and chromatography; the advent of genetic engineering has made possible a number
of methods to facilitate purification. Methods commonly used to study protein
structure and function include immunohistochemistry, site-directed mutagenesis,
X-Ray crystallography, nuclear magnetic resonance and mass spectrometry.
The structure of proteins is studied under following :
x Primary Structure
x Secondary Structure
x Tertiary Structure
x Quaternary Structure
Primary Structure
The linear sequence of amino acids in the polypeptide chain is known as the primary
structure. The determination of the protein structure helps in the analysis of amino
acids. The properties of proteins are dependent on the sequence of the amino-
acids in the polypeptide chain. Figure 3.1 displays the primary structure of a protein.

Fig 3.1 Primary Structure of a Protein


Self-Instructional
Material 67
Proteins Secondary Structure
When the peptide chain folds itself in a three-dimensional structure, it is known as
Secondary Structure. The secondary structure is dependent on the structural
NOTES characteristics of the peptide bond. Apart form the peptide bonding, hydrogen
bonding that occurs between the nitrogen of one peptide bond and oxygen of
another peptide bond, also enhances the stability of the secondary structure of
proteins. Proteins fold themselves in two broad classes of structure:
x Globular Proteins
x Fibrous Proteins
The globular proteins are compactly folded and coiled whereas the fibrous proteins
are filamentous and elongated. Figure 3.2 displays the secondary structure of a
protein

Fig. 3.2 Secondary Structure of a Protein


Tertiary Structure
The tertiary structure of protein shows a specific three-dimensional structure. The
folding regular units of the secondary structure as well as of areas of the peptide
chain devoid of secondary structure result in the formation of the tertiary structure.
The folded portions are held together by the hydrogen bond formed between the
R-group of the oppositely charged ends. The bonds that stabilize the tertiary
structure of proteins are:
x Van der Waal bonds (if side groups are non-polar).
x Hydrogen bonds (if side groups are amino or hydroxyl groups).
x Ionic bonds (if side groups are acids or bases).
x Covalent bonds (if side groups are cysteine residues).
Figure 3.3 displays the tertiary structure of a protein.

Self-Instructional
68 Material
Proteins

NOTES

Fig. 3.3 Tertiary Structure of a Protein


Quaternary Structure
When the proteins contain two or more polypeptide chains, the structure that is
formed on the native combination of these polypeptide chains is known as quaternary
structure. The primary, secondary and tertiary structures are found in proteins that
contain only single polypeptide chain. Figure 3.4 displays the quaternary structure
of a protein.

Fig. 3.4 Quarternary Structure of a Protein


Studies of the conformation, function, and evolution of proteins have also revealed
the central importance of a unit of organization distinct from the four just described.
This is the protein domain, a substructure produced by any part of a
polypeptide chain that can fold independently into a compact, stable structure. A
domain usually contains between 40 and 350 amino acids, and it is the modular
unit from which many larger proteins are constructed.
The smallest protein molecules contain only a single domain, whereas larger
proteins can contain as many as several dozen domains, usually connected to each
other by short, relatively unstructured lengths of polypeptide chain.
The central core of a domain can be constructed from a helices, from b
sheets, or from various combinations of these two fundamental folding elements.
Each different combination is known as a protein fold. So far, about 1000 different
protein folds have been identified among the ten thousand proteins whose detailed
Self-Instructional
Material 69
Proteins conformations are known.Many protein molecules are either attached to the outside
of a cell’s plasma membrane or secreted as part of the extracellular matrix. All
such proteins are directly exposed to extracellular conditions. To help maintain
their structures, the polypeptide chains in such proteins are often stabilized by
NOTES covalent cross-linkages. These linkages can either tie two amino acids in the same
protein together, or connect different polypeptide chains in a multi-subunit protein.
The most common cross-linkages in proteins are covalent sulfur–sulfur bonds.
These disulfide bonds (also called S–S bonds) form as proteins are being prepared
for export from cells. Their formation is catalyzed in the endoplasmic reticulum by
an enzyme that links together two pairs of –SH groups of cysteine side chains that
are adjacent in the folded protein. Disulfide bonds do not change the conformation
of a protein but instead act as atomic staples to reinforce its most favored
conformation. For example, lysozyme—an enzyme in tears that dissolves bacterial
cell walls—retains its antibacterial activity for a long time because it is stabilized
by such cross-linkages.
Disulfide bonds generally fail to form in the cell cytosol, where a high
concentration of reducing agents converts S–S bonds back to cysteine –SH groups.
Apparently, proteins do not require this type of reinforcement in the relatively mild
environment inside the cell.
Amino Acid Sequence and Protein Folding
There are an enormous number of ways to make proteins with the same three-
dimensional structure. Random mutations can cause amino acid sequences to change
without a major change in the conformation of a protein. Due to this reason, a
current goal of structural biologists is to determine all the different protein folds
that proteins have in nature, and to devise computer-based methods to test the
amino acid sequence of a domain to identify which one of these previously
determined conformations the domain is likely to adopt.
A computational technique called threading can be used to fit an amino
acid sequence to a particular protein fold. For each possible fold known, the
computer searches for the best fit of the particular amino acid sequence to that
structure. In many cases, one particular three-dimensional structure will stand out
as a good fit for the amino acid sequence, suggesting an approximate conformation
for the protein domain. In other cases, none of the known folds will seem possible.
By applying x-ray and NMR studies to the latter class of proteins, structural
biologists hope to able to expand the number of known folds rapidly, aiming for a
database that contains the complete library of protein folds that exist in nature.
With such a library, plus expected improvements in the computational methods
used for threading, it may eventually become possible to obtain an approximate
three-dimensional structure for a protein as soon as its amino acid sequence is
known.
Self-Instructional
70 Material
Proteins
3.3 CLASSIFICATION OF PROTEINS

Proteins are the macromolecules responsible for the biological processes in the
cell. They consist at their most basic level of a chain of amino acids, determined by NOTES
the sequence of nucleotides in a gene. Depending on the amino acid sequence
(different amino acids have different biochemical properties) and interactions with
their environment, proteins fold into a three-dimensional structure, which allows
them to interact with other proteins and molecules and perform their function (Refer
Figure 3.5). Proteins consist of one or more polypeptides. A polypeptide is a
chain of amino acids. The polypeptide chains fold into their final three-dimensional
structure to constitute a functional protein. The amino acid sequence and structure
in this example correspond to ribosomal protein L2.

Fig. 3.5 Proteins: Three-Dimensional Structure

Proteins that have diverged from a common ancestral gene are known
as homologous. Proteins with similar sequences are assumed to be homologous
and usually (within certain limits) have similar structures and functions.
Different methods of protein classification have been proposed, but currently
none of them is universally valid. Below, some examples based on chemical
composition, structure, functions, and solubility in different solvents.
Protein Classification Based on Chemical Composition
On the basis of their chemical composition, proteins may be divided into two
classes: simple and complex.
Simple Proteins: Also known as homoproteins, they are made up of only amino
acids. Examples are plasma albumin, collagen, and keratin.
Conjugated Proteins: Sometimes also called heteroproteins, they contain in their
structure a non-protein portion. Three examples are glycoproteins, chromoproteins,
and phosphoproteins.
x Glycoproteins: They are proteins that covalently bind one or more
carbohydrate units to the polypeptide backbone. Typically, the branches
consist of not more than 15-20 carbohydrate units, where you can
Self-Instructional
Material 71
Proteins find arabinose, fucose (6-deoxygalactose), galactose, glucose,
mannose, N-acetylglucosamine (GlcNAc, or NAG), and N-
acetylneuraminic acid (Neu5Ac or NANA).
Examples of glycoproteins are: glycophorin, the best known among
NOTES
erythrocyte membrane glycoproteins; fibronectin (Refer Figure 3.6), that
anchors cells to the extracellular matrix through interactions on one side
with collagen or other fibrous proteins, while on the other side with cell
membranes; all blood plasma proteins, except albumin; immunoglobulins
or antibodies.

Fig. 3.6 Human Fibronectin

x Chromoproteins: They are proteins that contain colored prosthetic groups.


Typical examples are: hemoglobin and myoglobin, which bind, respectively,
one and four heme groups; chlorophylls, which bind a porphyrin ring with a
magnesium atom at its centre; rhodopsins, which bind retinal.
x Phosphoproteins: They are proteins that bind phosphoric acid to serine
and threonine residues. Generally, they have a structural function, such as
tooth dentin, or reserve function, such as milk caseins (alpha, beta, gamma
and delta), and egg yolk phosvitin.
Protein Classification Based on Shape
On the basis of their shape, proteins may be divided into two classes: fibrous and
globular.
Fibrous Proteins
They have primarily mechanical and structural functions, providing support to
the cells as well as the whole organism. These proteins are insoluble in water as
they contain, both internally and on their surface, many hydrophobic amino acids.
The presence on their surface of hydrophobic amino acids facilitates their packaging
Self-Instructional
72 Material
into very complex supramolecular structures. Their polypeptide chains form long Proteins

filaments or sheets, where in most cases only one type of secondary structure, that
repeats itself, is found.
In vertebrates, these proteins provide external protection, support and shape;
NOTES
in fact, thanks to their structural properties, they ensure flexibility and/or strength.
Some fibrous proteins, such as D-keratins, are only partially hydrolyzed in
the intestine. Following are some examples.
x Fibroin: It is produced by spiders and insects. An example is that produced
by the silkworm, Bombyx mori.
x Collagen: The term ‘collagen’ indicates not a single protein but a family of
structurally related proteins (at least 29 different types), which constitute
the main protein component of connective tissue, and more generally, the
extracellular scaffolding of multicellular organisms (Refer Figure 3.7). In
vertebrates, they represent about 25-30% of all proteins. They are found in
different tissues and organs, such as tendons and the organic matrix of bone,
where they are present in very high percentages, but also in cartilage and in
the cornea of the eye.
x In the different tissues, they form different structures, each capable of
satisfying a particular need. For example, in the cornea, the molecules are
arranged in an almost crystalline array, so that they are virtually transparent,
while in the skin they form fibers not very intertwined and directed in all
directions, which ensure the tensile strength of the skin itself.
x The different types of collagen have low nutritional value as deficient in
several amino acids (in fact, they contain no tryptophan and low amount of
the other essential amino acids). The gelatin used in food preparation is a
derivative of collagen.

Fig. 3.7 Collagen

x D-Keratins: They constitute almost the entire dry weight of nails, claws,
beak, hooves, horns, hair, wool, and a large part of the outer layer of the
skin.
Self-Instructional
Material 73
Proteins The different stiffness and flexibility of these structures is a consequence
of the number of disulfide bonds that contribute, together with other binding
forces, to stabilize the protein structure. And this is the reason why wool
keratins, which have a low number of disulfide bonds, are flexible, soft
NOTES and extensible, unlike claw and beak keratins that are rich in disulfide
bonds.
x Elastin: This protein provides elasticity to the skin and blood vessels, a
consequence of its random coiled structure that differs from the structures
of the ±-keratins and collagens.
Globular Proteins
Most of the proteins belong to this class. They have a compact and more or less
spherical structure, more complex than fibrous proteins. In this regard, motifs,
domains, tertiary and quaternary structures are found, in addition to the secondary
structures. They are generally soluble in water but can also be found inserted into
biological membranes (transmembrane proteins), thus in a hydrophobic
environment. Unlike fibrous proteins, that have structural and mechanical functions,
they act as:
x Enzymes
x Hormones
x Membrane Transporters and Receptors
x Transporters of Triglycerides, Fatty Acids and Oxygen in the Blood
x Immunoglobulins or Antibodies
x Grain and Legume Storage Proteins
Examples of globular proteins are myoglobin, hemoglobin, and
cytochrome c.
At the intestinal level, most of the globular proteins of animal origin are
hydrolyzed almost entirely to amino acids.
Protein Classification Based on Biological Functions
The multitude of functions that proteins perform is the consequence of both
the folding of the polypeptide chain, therefore of their three-dimensional
structure, and the presence of many different functional groups in the amino acid
side chains, such as thiols, alcohols, thioethers, carboxamides, carboxylic acids
and different basic groups.
From the functional point of view, they may be divided into several groups.
x Enzymes (Biochemical Catalysts): In living organisms, almost all reactions
are catalyzed by specific proteins called enzymes. They have a high catalytic
power, increasing the rate of the reaction in which they are involved at least
Self-Instructional
74 Material
by factor 106. Therefore, life as we know could not exist without their Proteins

‘facilitating action’. Almost all known enzymes, and in the human body they
are thousand, are proteins (except some catalytic RNA molecules called
ribozymes, that is, ribonucleic acid enzymes).
NOTES
x Transport Proteins: Many small molecules, organic and inorganic, are
transported in the bloodstream and extracellular fluids, across the cell
membranes, and inside the cells from one compartment to another, by
specific proteins. Examples include haemoglobin that carries oxygen from
the alveolar blood vessels to tissue capillaries; transferrin, which carries
iron in the blood; membrane carriers; Fatty Acid Binding Proteins (FABP),
that is, the proteins involved in the intracellular transport of fatty acids;
proteins of plasma lipoproteins, macromolecular complexes of proteins and
lipids responsible for the transport of triglycerides, which are otherwise
insoluble in water; albumin, that carries free fatty acids, bilirubin, thyroid
hormones, and certain medications, such as aspirin and penicillin, in the
blood.
Many of these proteins also play a protective role, since the bound molecules,
such as fatty acids, may be harmful for the organism when present in free form.
x Storage Proteins: These proteins store the given different types. Examples
include ferritin that stores iron intracellularly in a non-toxic form; milk caseins
that act as a reserve of amino acids for the milk; egg yolk phosvitin that
contains high amounts of phosphorus; prolamins and glutelins, the
storage proteins of cereals.
x Mechanical Support: Proteins have a pivotal role in the stabilization of
many structures. Examples are D-keratins, collagen and elastin. The same
cytoskeletal system, the scaffold of the cell, is made of proteins.
o They generate movement. They are responsible, among others, for the
contraction of the muscle fibers (of which myosin is the main component);
the propulsion of spermatozoa and microorganisms with flagella; the
separation of chromosomes during mitosis.
o They are involved in nerve transmission. An example is the receptor for
acetylcholine at synapses.
o They control development and differentiation. Some proteins are involved
in the regulation of gene expression. An example is the Nerve Growth
Factor (NGF), discovered by Rita Levi-Montalcini that plays a leading
role in the formation of neural networks.
x Hormones: Many hormones are proteins. They are regulatory molecules
involved in the control of many cellular functions, from metabolism to
reproduction. Examples are insulin, glucagon, and Thyroid-Stimulating
Hormone (TSH).
Self-Instructional
Material 75
Proteins x Protection Against Harmful Agents: The antibodies or immunoglobulins
are glycoproteins that recognize antigens expressed on the surface of viruses,
bacteria and other infectious agents. Interferon, fibrinogen, and factors of
blood coagulation are other members of this group.
NOTES
x Storage of Energy: Proteins, and in particular the amino acids that constitute
them, act as energy storage, second in size only to the adipose tissue, that in
particular conditions, such as prolonged fasting, may become essential for
survival. However, their reduction of more than 30% leads to a decrease of
the contraction capacity of respiratory muscle, immune function, and organ
function that are not compatible with life. Therefore, proteins are an
extremely valuable fuel.
Protein Classification Based on Polarity
Proteins are the macromolecules responsible for the biological processes in the
cell. They consist at their most basic level of a chain of amino acids, determined by
the sequence of nucleotides in a gene. In a watery environment, proteins fold so
that nonpolar side chains are mostly in the interior of the protein, and hydrophilic
side chains are on the outside. Inside the nonpolar core of a membrane, a
polypeptide must also maintain a hydrophobic core.
Since proteins have nonpolar side chains their reaction in a watery
environment is similar to that of oil in water. The nonpolar side chains are pushed
to the interior of the protein allowing them to avoid water molecule and giving the
protein a globular shape.
Classification Based on Solubility of Proteins and their Physical Properties
Proteins are classified based on their solubility and nutritional properties.
The American Physiological Survey and American Society of Biological chemists
have classified proteins as follows:
x Simple Proteins
x Conjugated Proteins
x Derived Proteins

Self-Instructional
76 Material
Table 3.1 Classification of Proteins Based on their Solubility Proteins

S. Types Properties Examples


no.

1) SIMPLE PROTEINS or Yield only amino acids or NOTES


HOLOPROTEINS their derivatives on
hydrolysis

A Albumins 1. Soluble in pure water, Ovalbumin (egg white),


dilute solutions of serum albumin (blood
acids, bases and salts plasma), lactalbumin
2. Precipitated by (milk whey), leucosine
ammonium sulphate (cereals)

3. Coagulated by heat
4. Widely distributed in
nature

B Globulins 1. Insoluble in pure water Euglobulins: Serum


(euglobulins) globulin (blood), tuberin
2. Soluble in water (potato), ovovglobulin
(pseudoglobulin) (blood plasma), pomeline
(oranges)
Pseudoglobulins:
pseudoglobulin (milk
whey)

C Glutelins 1. In soluble in all neutral Glutenin (wheat), glutelin


solvents (corn), oryzenin (rice)
2. Soluble in dilute acids
and alkalis

D Prolamines 1. Soluble in alcohol Glaidin (wheat), zein


(maize), hordein (oat)

E Fibrous Proteins 1. They are characteristic of Collagen (muscles),


skeletal muscles and other keratin (skin)
protective tissues.

F Histones 1. Soluble in water Proteins of thymus and


haemoglobin

G Protamine 1. Soluble in water Proteins found in sperm


2. Not coagulated by heat of salmon (salmine)

Self-Instructional
Material 77
Proteins
2. CONJUGATED PROTEINS Proteins conjugated with
or COMPLEX PROTEINS some other molecules
OR HETEROPROTEINS

A Nucleoproteins Associated with nucleic acids Found in genetic


NOTES materials

B Glycoproteins and Associated with Glycoproteins: egg


Mucoproteins carbohydrates albumin, serum
globulins and serum
albumins
Mucoproteins:
ovomucoid (egg white),
osseomucoid (bone),
tendomucoid (tendon)

C Phosphoproteins Associated with phosphorus Caseinogen (milk);


ovovitellin (egg yolk)

D Haemoglobins Associated with haem Haemoglobin

E Lecithoproteins Associated with lecithin Eggs

F Metalloproteins Associated with various Collagen, casein,


metals albumin (bound to Hg,
Ag, Cu, Zn);
Siderophilin or
transferrin (bound to
iron); Ceruloplasmin
(bound to copper)

3 DERIVED PROTEINS Derivatives of protein


molecules

A Primary Protein Derivatives Size of protein molecule is


not altered

1 Proteans 1. Insoluble in water Edestan (edestin);


2. Produced by action myosin (myosin)
of enzymes, acids or
water on proteins

2 Metaproteins 1. Result from further Acid and alkali in


action of acids or metaproteins
alkalis
2. Insoluble in water
3. Soluble in dilute
acids or alkalis

Self-Instructional
78 Material
Proteins
3 Conjugated proteins 1. Result from action of heat or Egg albumin (cooked)
alcohol

B Secondary Derived 1. Size smaller than


Proteins original protein
molecules NOTES
2. Derivatives of proteins
in which hydrolysis has
occured

1 Proteoses 1) Soluble in water Albumose (albumin);


2) Coagulated by heat globulose (globulin)

2 Peptones 1) Soluble in water Products obtained on


2) Does not coagulate by heat digestion of proteins

3) Produced by action of acids


or enzymes

Nutritional Classification of Proteins


Based on their nutritional properties proteins are classified as complete proteins,
partially complete proteins and incomplete proteins.
Table 3.2 Classification of Proteins Based on their Nutritional Properties

S.No. Type Function Limiting amino Example


acids*
1 Complete Proteins Promote Nil Egg
Growth Proteins
2 Partially Complete Promote Partially lacking in Wheat
Proteins Moderate one or more essential
Growth amino acids
3 Incomplete Proteins Do not Completely lacking in Gelatin or
Promote one or more essential Zein
Growth amino acids

Protein Source for Limiting Amino Acids: The essential amino acids found in
the smallest quantity in the food stuff are known as limiting amino acids. Table 3.3
lists the limiting amino acids and their protein source.
Table 3.3 Limiting Amino Acids and their Protein Source

Protein Source Limiting Amino Acids

Wheat Lysine
Rice Lysine
Legumes Tryptophan or Methionine (or Cysteine)
Maize Lysine and Tryptophan
Egg Nil
Chicken Nil
Self-Instructional
Material 79
Proteins
3.4 PHYSICAL AND CHEMICAL PROPERTIES OF
PROTEINS

NOTES Physical Properties of Proteins


Colour and Taste
 Colourless
 Tasteless
 Homogenous
 Crystalline
Shape and Size
 The shape varies from a simple crystalloid spherical structure to long fibrillar
structures.
 Spherical shaped proteins (Globular proteins) are formed by folding and
crumpling of protein chains found in plants (cells of leaves and seeds).
 Common examples of globular proteins are pepsin, inulin, ribonuclease etc.
 Thread-like and ellipsoidal shaped proteins (fibrillar proteins) are found in
animal tissues. Common examples of fibrillar proteins are fibrinogen, myosin,
etc.
Table 3.4 Size of Proteins

S. No. Example of Protein Size


1 Haemoglobin 55 Å
2 Catalase 80*64*54 Å
3 Collagen 3000 Å

 Some protein molecules can assemble to form filaments that may span the
entire length of a cell.
 A long chain of identical protein molecules can be constructed if each
molecule has a binding site complementary to another region of the surface
of the same molecule. An actin filament, for example, is a long helical structure
produced from many molecules of the protein actin. Actin is very abundant
in eucaryotic cells, where it constitutes one of the major filament systems of
the cytoskeleton.
 The biological structures are often formed by linking subunits that are very
similar to each other—such as amino acids or protein molecules—into long,
repetitive chains.
 If all the subunits are identical, the neighbouring subunits in the chain can
often fit together in only one way, adjusting their relative positions to minimize
the free energy of the contact between them. As a result, each subunit is
Self-Instructional
80 Material
positioned in exactly the same way in relation to the next, so that subunit Proteins

three fits onto subunit two in the same way that subunit two fits into subunit
one, and so on.
 As it is very rare for subunits to join in a straight line, this arrangement
NOTES
generally results in a helix—a regular structure that resembles a spiral
staircase.
 Helices occur commonly in biological structures, whether the subunits are
small molecules linked together by covalent bonds (for example, the amino
acids in a helix) or large protein molecules that are linked by noncovalent
forces (for example, the actin molecules in actin filaments). A helix is an
unexceptional structure, and is generated simply by placing many similar
subunits next to each other, each in the same strictly repeated relationship
to the one before.
 A protein molecule can have an elongated and fibrous shape.
 Most of the proteins we have discussed so far are globular proteins, in
which the polypeptide chain folds into a compact shape like a ball with an
irregular surface.
 Enzymes tend to be globular proteins, even though most of them are large
and complicated, with multiple subunits, they have an overall rounded shape.
In contrast, other proteins have essential functions in the cell requiring each
individual protein molecule to span a large distance. These proteins generally
have a relatively simple, elongated three-dimensional structure and are
commonly referred to as fibrous proteins.
 A large family of intracellular fibrous proteins consists of -keratin, and its
relatives. Keratin filaments are extremely stable and are the main component
in long-lived structures such as hair, horn, and nails.
 An -keratin molecule is a dimer of two identical subunits, with the long a
helices of each subunit forming a coiled-coil. The coiled-coil regions are
capped at each end by globular domains containing binding sites. This enables
this class of protein to assemble into ropelike intermediate filaments—an
important component of the cytoskeleton that creates the cell’s internal
structural scaffold.
 Fibrous proteins are especially abundant outside the cell, where they are a
main component of the gel-like extracellular matrix that helps to bind
collections of cells together to form tissues.
 Extracellular matrix proteins are secreted by the cells into their surroundings,
where they often assemble into sheets or long fibrils. Collagen is the most
abundant of these proteins in animal tissues.
 A collagen molecule consists of three long polypeptide chains, each
containing the nonpolar amino acid glycine at every third position.

Self-Instructional
Material 81
Proteins x This regular structure allows the chains to wind around one another to
generate a long regular triple helix. Many collagen molecules then bind to
one another side-by-side and end-to-end to create long overlapping arrays—
thereby generating the extremely tough collagen fibrils that give connective
NOTES tissues their tensile strength.
x In contrast to collagen is another protein in the extracellular matrix, i.e.,
elastin. Elastin molecules are formed from relatively loose and unstructured
polypeptide chains that are covalently cross-linked into a rubber-like elastic
meshwork. Unlike most proteins, they do not have a uniquely defined stable
structure, but can be reversibly pulled from one conformation to another.The
resulting elastic fibers enable skin and other tissues, such as arteries and
lungs, to stretch and recoil without tearing.
Molecular Weight
The molecular weight of protein ranges between 5 x 103 and 1 x 106
Molecular weights of proteins lies close to or multiples of 35,000 and 70,000.
Colloidal Nature
Proteins show many colloidal properties because of their large shape and size.
They have extremely slow diffusion rates and produce scattering of light in solutions.
Denaturation
Proteins have a three dimensional structure. This structure though very complicated,
is delicate. That is, it undergoes alternations even if subjected to some physical or
chemical agents. It is important to note that the alterations in the protein structure
do not break the peptide bonds. The loss of native conformation of proteins that
subsequently alters its characterizing properties is known as ‘denaturation’ (Refer
Figure 3.8).

Fig. 3.8 Denaturation of Proteins


Changes brought About by Denaturation of Proteins
Following are the changes brought about by denaturation of proteins:
x Hydrolysis of the peptide bonds becomes easier.
x Solubility of the proteins decreases.
x Proteins lose their catalytic properties.
Self-Instructional
82 Material
x Proteins lose their hormonal properties. Proteins

x Viscosity of the proteins increase.


Causes of Denaturation
Following are the causes of denaturation: NOTES
x Modification in the structures of the proteins.
x Breaking of hydrogen bonds.
x Breaking of hydrophobic bonds.
x Breaking of salt linkages.
It is important to note that the covalent bonds and the peptide bonds do not
break. The proteins unfold and assume a random coil structure. This is followed
by the coagulation of the proteins.
Physical and Chemical Agents that Cause Denaturation of Proteins
Following are the physical and chemical agents that cause denaturation of proteins:
x Heat (physical agent) is one of the most important causative factors for
denaturation. The rate of denaturation increases with an increase in the
temperature of the medium.
x Denaturation of proteins due to heat can be observed in stirring, shaking,
high pressure treatment and subjecting the proteins to ultra-violet radiations.
x The pH of the medium acts as a chemical agent in the denaturation of proteins.
The rate of denaturation increases with increase in the pH of the medium.
Other chemical agents include urea, guanine hydrochloride, Sodium Dodecyl
Sulphate (SDS).
During processing of food, most of the proteins undergo coagulation or
denaturation. However, this property of proteins is of great concern during the
processing of milk.
Renaturation
Proteins of high molecular weight can resume their structure again if they are
subjected to mild treatments. This process is known as reversible denaturation or
renaturation.
Amphoteric Nature of Proteins
Two-Dimensional gel Electrophoresis (2DE) separates proteins by molecular charge
and molecular size. Proteins are first solubilized in a denaturing buffer containing a
neutral chaotrope, a zwitterionic or neutral detergent, and a reducing agent. First-
dimension isoelectric keywords, focusing, then subjects proteins to a high voltage
within a pH gradient. The amphoteric nature of proteins means that each migrates
to the pH where the net molecular charge is zero. After equilibration, to ensure
Self-Instructional
Material 83
Proteins complete protein unfolding, the second dimension separates by molecular size.
Each protein is therefore resolved at a unique isoelectric point/molecular size
coordinate. After visualization by staining, proteome changes are revealed by gel
image analysis and protein spots of interest excised and identified by mass
NOTES spectrometry sequence analysis combined with a database comparison. Variations
to this procedure include staining or radio-labelling prior to electrophoresis.
Although 2DE does have limitations, the most significant being the resolution of
membrane and/or hydrophobic proteins, the potential solutions offered by pre-
fractionation or adjustments to the electrophoresis regimen imply that this technique
is likely to remain central to proteomic research.
Ion Binding Capacity
A research was carried out by the United States department of agriculture to
study the ion binding capacity of proteins. This preliminary investigation tests
the premise that biologically relevant (1) peptide-metal ion interactions, and (2)
metal ion-dependent macromolecular recognition events (for example, peptide-
peptide interactions) may be modeled by biomimetic affinity chromatography.
Divinylsulfone-activated agarose (6 per cent) was used to immobilize three
different synthetic peptides representing metal-binding protein surface domains
from the human plasma metal transport protein Histidine-Rich Glycoprotein
(HRG). The synthetic peptides represented 1-3 multiple repeat units of the 5-
residue sequence (Gly-His-His-Pro-His) found in the C-terminal of HRG. By
frontal analyses, immobilized HRG peptides of the type (GHHPH)nG, where n
= 1-3, were each found to have a similar binding capacity for both Cu(II) ions
and Zn(II) ions (31-38 mumol/ml gel). The metal ion-dependent interaction of a
variety of model peptides with each of the immobilized HRG peptide affinity
columns demonstrated differences in selectivity despite the similar internal
sequence homology and metal ion binding capacity. The immobilized 11-residue
HRG peptide was loaded with Cu(II) ions and used to demonstrate the selective
adsorption and isolation of proteins from human plasma. These results suggest
that immobilized metal-binding peptides selected from known solvent-exposed
protein surface metal-binding domains may be useful model systems to evaluate
the specificity of biologically relevant metal ion-dependent interaction and transfer
events in vitro.
Solubility of Proteins
The surface of a protein has a net charge that depends on the number and identities
of the charged amino acids, and pH. At a specific pH the positive and negative
charges will balance and the net charge will be zero. This pH is called the isoelectric
point, and for most proteins it occurs in the pH range of 5.5 to 8. A protein has its
lowest solubility at its isoelectric point. If there is a charge at the protein surface,
the protein prefers to interact with water, rather than with other protein molecules.
This charge makes it more soluble. Without a net charge, protein-protein
interactions and precipitation are more likely.
Self-Instructional
84 Material
Optical Activity Proteins

Polarized light is rotated to the left by all protein solutions. This concludes that they
are levorotatory.
Chemical Properties NOTES
The chemical properties of proteins are described as follows:
Hydrolysis
Protein hydrolysis is the process by which peptide bonds are broken under acidic
conditions at high temperatures, generating free amino acid residues. The resulting
hydrolysates can then be analysed by amino acid analysis to determine the amino
acid composition and concentration of proteins. The hydrolysis procedure is the
rate-limiting step for amino acid analysis. Microwave-assisted protein hydrolysis
offers the potential to reduce the hydrolysis time significantly.
Reaction of Proteins by Acidic Agents
Proteins have regions which resist hydrolysis with mineral acid. The presence of a
strong organic acid was found to be efficient for hydrolysis of a hydrophobic
peptide bond. The proposed condition, a 2:1 (by vol.) mixture of concentrated
hydrochloric acid and trifluoroacetic acid at 166° C for 25 minutes was observed
to be equivalent to the conventional conditions (6 M HCl at 110 ° C for more than
24 hours) without any significant decomposition of amino acids. The method was
shown to be superior to conventional conditions, especially for hydrophobic proteins.
The present method also destroys tryptophan like the conventional acid hydrolysis.
Reaction of Proteins by Alkaline Agents
It has been observed that proteins are hydrolysed by 2N NaOH (alkaline
hydrolysis). Alkaline hydrolysis of proteins leads to destruction of amino acids like
arginine, cysteine, cystine, serine, threonine, etc. Racemization of the amino acids
is also observed during alkaline hydrolysis.
Reaction of Proteins by Proteolytic Enzymes
Enzymes like pepsin and trypsin hydrolyse proteins under suitable and pH. This
type of hydrolysis is useful in isolation of proteins. Despite of this advantage, enzyme
hydrolysis of proteins has various drawbacks. This process requires a long
incubation period. Moreover, the hydrolysis is incomplete.

Check Your Progress


1. Define the term protein.
2. How many types of structure does protein have?
3. What is protein fold?
4. Write in short about storage proteins.
5. What is the shape and size of protein?
Self-Instructional
Material 85
Proteins
3.5 ANSWERS TO CHECK YOUR PROGRESS
QUESTIONS

NOTES 1. Proteins are large biomolecules, or macromolecules, consisting of one or


more long chains of amino acid residues. Proteins perform a vast array of
functions within organisms, including catalysing metabolic reactions, DNA
replication, responding to stimuli, providing structure to cells and organisms,
and transporting molecules from one location to another.
2. The structure of proteins is studied under following:
x Primary structure
x Secondary structure
x Tertiary structure
x Quaternary structure
3. The central core of a domain can be constructed from a helices, from b
sheets, or from various combinations of these two fundamental folding
elements. Each different combination is known as a protein fold.
4. Storage Proteins: These proteins store the given different types. Examples
include ferritin that stores iron intracellularly in a non-toxic form; milk caseins
that act as a reserve of amino acids for the milk; egg yolk phosvitin that
contains high amounts of phosphorus; prolamins and glutelins, the
storage proteins of cereals.
5. Shape and size of proteins are as follows:
x The shape varies from a simple crystalloid spherical structure to long
fibrillar structures.
x Spherical shaped proteins (Globular proteins) are formed by folding
and crumpling of protein chains found in plants (cells of leaves and
seeds).
x Common examples of globular proteins are pepsin, inulin, ribonuclease
etc.
x Thread-like and ellipsoidal shaped proteins (fibrillar proteins) are found
in animal tissues. Common examples of fibrillar proteins are fibrinogen,
myosin, etc.
x Some protein molecules can assemble to form filaments that may span
the entire length of a cell.
x A long chain of identical protein molecules can be constructed if each
molecule has a binding site complementary to another region of the
surface of the same molecule. An actin filament, for example, is a long
helical structure produced from many molecules of the protein actin.
Actin is very abundant in eucaryotic cells, where it constitutes one of
Self-Instructional
the major filament systems of the cytoskeleton.
86 Material
x The biological structures are often formed by linking subunits that are Proteins

very similar to each other—such as amino acids or protein molecules—


into long, repetitive chains.

NOTES
3.5 SUMMARY

x Proteins are large biomolecules, or macromolecules, consisting of one or


more long chains of amino acid residues.
x Proteins perform a vast array of functions within organisms,
including catalysing metabolic reactions, DNA replication, responding to
stimuli, providing structure to cells and organisms, and transporting
molecules from one location to another.
x Proteins differ from one another primarily in their sequence of amino acids,
which is dictated by the nucleotide sequence of their genes, and which usually
results in protein folding into a specific three-dimensional structure that
determines its activity.
x A linear chain of amino acid residues is called a polypeptide. A protein
contains at least one long polypeptide.
x Short polypeptides, containing less than 20–30 residues, are rarely
considered to be proteins and are commonly called peptides, or
sometimes oligopeptides.
x The individual amino acid residues are bonded together by peptide bonds and
adjacent amino acid residues.
x The sequence of amino acid residues in a protein is defined by the sequence
of a gene, which is encoded in the genetic code.
x In general, the genetic code specifies 20 standard amino acids; however, in
certain organisms the genetic code can include selenocysteine and—in
certain archaea—pyrrolysine.
x Shortly after or even during synthesis, the residues in a protein are often
chemically modified by post-translational modification, which alters the
physical and chemical properties, folding, stability, activity, and ultimately,
the function of the proteins.
x Sometimes proteins have non-peptide groups attached, which can be
called prosthetic groups or cofactors.
x Proteins can also work together to achieve a particular function, and they
often associate to form stable protein complexes.
x Once formed, proteins only exist for a certain period and are
then degraded and recycled by the cell’s machinery through the process
of protein turnover.
Self-Instructional
Material 87
Proteins x A protein’s lifespan is measured in terms of its half-life and covers a wide
range. They can exist for minutes or years with an average lifespan of 1–2
days in mammalian cells.
x Abnormal or misfolded proteins are degraded more rapidly either due to
NOTES
being targeted for destruction or due to being unstable.
x Like other biological macromolecules such as polysaccharides and nucleic
acids, proteins are essential parts of organisms and participate in virtually
every process within cells.
x Many proteins are enzymes that catalyse biochemical reactions and are vital
to metabolism. Proteins also have structural or mechanical functions, such
as actin and myosin in muscle and the proteins in the cytoskeleton, which
form a system of scaffolding that maintains cell shape.
x Other proteins are important in cell signaling, immune responses, cell
adhesion, and the cell cycle. In animals, proteins are needed in the diet to
provide the essential amino acids that cannot be synthesized.
x Digestion breaks the proteins down for use in the metabolism.
x Proteins may be purified from other cellular components using a variety of
techniques such as ultracentrifugation, precipitation, electrophoresis,
and chromatography; the advent of genetic engineering has made possible
a number of methods to facilitate purification.
x Methods commonly used to study protein structure and function
include immunohistochemistry, site-directed mutagenesis, X-ray
crystallography, nuclear magnetic resonance and mass spectrometry.
x The linear sequence of amino acids in the polypeptide chain is known as the
primary structure.
x The determination of the protein structure helps in the analysis of amino
acids.
x The properties of proteins are dependent on the sequence of the amino-
acids in the polypeptide chain.
x When the peptide chain folds itself in a three-dimensional structure, it is
known as Secondary Structure.
x The secondary structure is dependent on the structural characteristics of
the peptide bond.
x Apart form the peptide bonding, hydrogen bonding that occurs between
the nitrogen of one peptide bond and oxygen of another peptide bond, also
enhances the stability of the secondary structure of proteins.
x The tertiary structure of protein shows a specific three-dimensional structure.
x The folding regular units of the secondary structure as well as of areas of
the peptide chain devoid of secondary structure result in the formation of
the tertiary structure.
Self-Instructional
88 Material
x The folded portions are held together by the hydrogen bond formed between Proteins

the R-group of the oppositely charged ends.


x The central core of a domain can be constructed from a helices, from b
sheets, or from various combinations of these two fundamental folding NOTES
elements. Each different combination is known as a protein fold.
x The most common cross-linkages in proteins are covalent sulfur–sulfur bonds.
These disulfide bonds (also called S–S bonds) form as proteins are being
prepared for export from cells.
x The term ‘collagen’ indicates not a single protein but a family of structurally
related proteins (at least 29 different types), which constitute the main protein
component of connective tissue, and more generally, the extracellular
scaffolding of multicellular organisms.
x Helices occur commonly in biological structures, whether the subunits are
small molecules linked together by covalent bonds or large protein molecules
that are linked by noncovalent forces.
x Enzymes tend to be globular proteins, even though most of them are large
and complicated, with multiple subunits, they have an overall rounded shape.

3.7 KEY WORDS

x Proteins: Proteins are large biomolecules, or macromolecules, consisting


of one or more long chains of amino acid residues.
x Primary structure: The linear sequence of amino acids in the polypeptide
chain is known as the primary structure.
x Secondary structure: When the peptide chain folds itself in a three-
dimensional structure, it is known as secondary Structure.
x Protein fold: The central core of a domain can be constructed from a
helices, from b sheets, or from various combinations of these two fundamental
folding elements. Each different combination is known as a protein fold.

3.8 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. What are proteins?
2. What is the tertiary structure of protein?
3. Draw a well-labelled diagram of secondary structure of protein.
4. What is protein domain?
Self-Instructional
Material 89
Proteins 5. Define protein fold.
6. Write a short note on fibrous protein.
Long Answer Questions
NOTES
1. Discuss in detail about proteins.
2. Distinguish between the types of proteins structure with the help of diagram.
3. Explain the amino acid sequence and protein folding.
4. Discuss in detail about classification of proteins.
5. Write about physical and chemical properties of proteins.

3.9 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
90 Material
Protein Metabolism

UNIT 4 PROTEIN METABOLISM


Structure NOTES
4.1 Objectives
4.2 Protein Metabolism: An Introduction
4.2.1 Classification of Protein
4.2.2 Protein Synthesis
4.3 Deficiency Diseases of Proteins
4.4 Inborn Errors of Protein Metabolism
4.5 Entner Doudoroff Pathway
4.6 Answers to Check Your Progress Questions
4.7 Summary
4.8 Key Words
4.9 Self Assessment Questions and Exercises
4.10 Further Readings

4.0 INTRODUCTION

Protein metabolism denotes the various biochemical processes responsible for the
synthesis of proteins and amino acids (anabolism), and the breakdown of proteins
by catabolism. The steps of protein synthesis include transcription, translation,
and post translational modifications. During transcription, RNA polymerase
transcribes a coding region of the DNA in a cell producing a sequence of RNA,
specifically messenger RNA (mRNA). This mRNA sequence contains codons: 3
nucleotide long segments that code for a specific amino acid. Ribosomes translate
the codons to their respective amino acids. In humans, non-essential amino acids are
synthesized from intermediates in major metabolic pathways such as the Citric
Acid Cycle. Essential amino acids must be consumed and are made in other
organisms. The amino acids are joined by peptide bonds making a polypeptide
chain. This polypeptide chain then goes through post translational modifications
and is sometimes joined with other polypeptide chains to form a fully functional
protein.
Dietary proteins are first broken down to individual amino acids by various
enzymes and hydrochloric acid present in the gastrointestinal tract. These amino
acids are further broken down to á-keto acids which can be recycled in the body
for generation of energy, and production of glucose or fat or other amino acids.
Proteins can be broken down by enzymes known as peptidases or can break
down as a result of denaturation. Proteins can denature in environmental conditions
the protein is not made for.
Protein synthesis is the process whereby biological cells generate
new proteins; it is balanced by the loss of cellular proteins via degradation or export.
Translation, the assembly of amino acids by ribosomes, is an essential part of the
Self-Instructional
Material 91
Protein Metabolism biosynthetic pathway, along with generation of messenger RNA (mRNA),
aminoacylation of transfer RNA (tRNA), co-translational transport, and post-
translational modification. Protein biosynthesis is strictly regulated at multiple steps.
They are principally during transcription (phenomena of RNA synthesis from DNA
NOTES template) and translation (phenomena of amino acid assembly from RNA).
The cistron DNA is transcribed into the first of a series of RNA intermediates.
The last version is used as a template in synthesis of a polypeptide chain. Protein
will often be synthesized directly from genes by translating mRNA. However, when
a protein must be available on short notice or in large quantities, a protein
precursor is produced. A proprotein is an inactive protein containing one or
more inhibitory peptides that can be activated when the inhibitory sequence is
removed by proteolysis during posttranslational modification. A preprotein is a
form that contains a signal that specifies its insertion into or through membranes,
i.e., targets them for secretion. The signal peptide is cleaved off in the endoplasmic
reticulum. Preproproteins have both sequences (inhibitory and signal) still present.
In protein synthesis, a succession of tRNA molecules charged with
appropriate amino acids are brought together with an mRNA molecule and
matched up by base-pairing through the anti-codons of the tRNA with successive
codons of the mRNA. The amino acids are then linked together to extend the
growing protein chain, and the tRNAs, no longer carrying amino acids, are released.
This whole complex of processes is carried out by the ribosome, formed of two
main chains of RNA, called ribosomal RNA (rRNA), and more than 50 different
proteins. The ribosome latches onto the end of an mRNA molecule and moves
along it, capturing loaded tRNA molecules and joining together their amino acids
to form a new protein chain.
In this unit, you will study about protein metabolism, protein synthesis,
deficiency diseases and inborn errors of protein metabolism in detail.

4.1 OBJECTIVES

After going through this unit, you will be able to:


x Discuss protein metabolism
x Explain protein synthesis
x Analyse the deficiency diseases of proteins
x Discuss inborn errors of protein metabolism

4.2 PROTEIN METABOLISM: AN INTRODUCTION

In 1939, G. J. Muller investigated the substances found in milk and egg. J. J.


Berzelius suggested that these substances should be called proteins. Now, these
are regarded as fundamental structural components of the body. They are the
Self-Instructional
92 Material
nitrogenous macromolecules composed of many amino acids joined by peptide Protein Metabolism

linkages. Proteins are defined as the high molecular weight mixed polymers of
amino acids joined by peptide linkages. Proteins are the essence of life processes.
They are the fundamental constituents of all protoplasm and are involved in the
structural integrity and functions of living cells. They are at the centre of the action NOTES
in the biological processes. They function as enzymes, which catalyse the complex
set of chemical reactions that are collectively referred to as life. Proteins serve as
regulators of these reactions both directly as components of enzymes and indirectly
in the form of chemical messengers known as hormones as well as the receptors
for these hormones. They act to transport and store biologically important substances
such as metal ions, O2, glucose, lipids and many other molecules. In the form of
muscle fiber and other contractile assemblies, proteins generate the coordinated
mechanical motion of numerous biological processes, including the separation of
chromosomes during cell division and the movement of eyes. Proteins, such as
rhodopsin in the retina of eye, acquire sensory information that is processed through
the action of nerve cell proteins. The proteins of immune system such as
immunoglobulin form an essential biological defense system in higher animals. Clearly
there is considerable validity to the old clinch that proteins are the ‘building blocks’
of life.
Biological Significance of Proteins
Several types of proteins have been characterized and isolated from the cellular
components of living organisms. Depending upon their chemical and physical
structure and location inside the cell, various proteins perform various functions.
Structural proteins are generally inert to biochemical reactions. They maintain
the natural form and position of the organs, the cell wall and primary fibrous
constituent of the cell’ have structural proteins.
Collagen is the most abundant known protein in animals, forming a major
part of the skin, cartilage, ligament, tendon and bone. The scales of fish and reptiles
and hairs, feathers, horns hoofs and claws are made up of the protein keratin.
Capacity of motion and flexibility m the organisms is the organisms are the virtue
of certain proteins with great tensile strength. There are contractile proteins. Catalytic
proteins are the most active proteins of an organism.
x The properties of living cells depend upon the biological processes, which
are regulated by enzymes. The major portion of an enzyme is made up of
proteins. These proteins are mostly spherical in shape and are termed globular
proteins. Some enzymes are simple proteins containing only ammo acids,
others are complex proteins.
x Enzymes catalyse a variety of reactions in the living cells. Certain proteins,
especially in the animals are involved in the transport of many essential
biological factors to different parts of the organisms.
Self-Instructional
Material 93
Protein Metabolism x Haemoglobin transporting O2 from one part of the body to the other is an
example of carrier protein.
Structural Organization of Proteins
NOTES The proteins have the following four structural forms:
x Primary Structure
x Secondary Structure
x Tertiary Structure
x Quaternary Structure
Primary Structure: This structural level is in the linear sequence in which the
amino acids are held together by peptide bonds in the peptide chain. The peptide
bonds form the backbone and side chains of amino acid residues project outside
the backbone chain.
Secondary Structure: In the secondary structure the peptide chain assumes a
three-dimensional structure by folding or coiling, zigzag linear or mixed forms. The
linkages or bonds involved in the secondary structure formation are as follows:
x Hydrogen Bonds: These are weak, low energy, non covalent bonds sharing
single hydrogen by two electronegative atoms, i.e., O and N. The H bonding
in secondary structure occurs regularly. This regularity allows the protein to
assume helical configuration or sheeted structure.
x Disulphide Bonds: These are formed between two cysteine residues. They
are strong, high energy, covalent bonds.
The secondary structures are of the following two types:
x D-Helix
x E-Pleated Sheet Structure
D-Helix: A peptide chain when forms regular helical coils is called a helix. These
coils are stabilized by hydrogen bonds. The helixes can be either right handed or
left handed. But only right handed a helix has been found in protein structure.
Each amino acid residue advances by 0.15 nm along the helix. 3.6 amino
acid residues are present in a complete turn. The distance between two equivalent
points on turn is 0.54 nm and is called as pitch. Proline is never found in a helix.
The proteins of hair, nail, skin contains a group of proteins called keratins, which
is rich in u helical structure.
E-Pleated Sheet Structure: This structure is formed owing to hydrogen bonds
between carbonyl oxygen and amide hydrogens of two or more adjacent
extended polypeptide chains (Refer Figure 4.1). Thus, there is inter-chain hydrogen
bonding in E-pleated structure. The structure is not absolutely planar but is slightly
pleated owing to the angles of bonds. The adjacent chains in E-pleated sheet
structure are either parallel or anti-parallel depending on whether the amino to
Self-Instructional
94 Material
carbonyl peptide linkage of the chains runs in the same or opposite direction. In Protein Metabolism

both parallel and anti-parallel E-pleated sheet structures, the side chains are on
opposite sides of the sheet. Generally, glycine, serine and alanine are most common
to form E-pleated sheet. Proline occurs in E-pleated sheet although it tends to
disrupt the sheets by producing rings. NOTES

Fig. 4.1 E-Pleated Sheet of Amino Acid

Tertiary Structure: Tertiary structure of a protein is its three-dimensional folding


of its arrangement i.e. polypeptide chain with the secondary structure which may
be further folded forming many sizes. Such structural conformation is called as
tertiary structure. It is only such conformations, which are biologically active and
protein in this state is called as native protein. The tertiary structure is constituted
by steric relationship between the amino acids located far apart but brought closer
by folding. The bonds responsible for folding are as follows:
x Hydrogen Bonds: There are weak bonds formed by the polar side chains
of ammo acids. The internal hydrogen bonding groups of a protein are
arranged such that nearly all-possible hydrogen bonds are formed. Clearly,
H bonding has a major influence on structures of proteins.
x Hydrophobic inTeractions: These interactions occur between non-polar
side chains of ammo acids as leucine, alanine and isoleucine. They constitute
the major stabilizing forces for tertiary structure forming a compact three-
dimensional structure.
x Ionic or Electrostatic Interactions: These are formed between oppositely
charged polar side chains of amino acids such as acidic and basic amino
acids.
x Disulphide Bonds: These bonds are formed between two cysteine amino
acids. Van der wall forces- These occur between non-polar side chains.
Quaternary Structure: Proteins, because of their multiple polar and non-polar
groups, stick to almost anything, anything that is but other proteins. The Svedberg
Self-Instructional
Material 95
Protein Metabolism discovered that some proteins are composed of more than one polypeptide chain.
Furthermore, these polypeptide subunits associate in a geometrically specific
manner. The special arrangement of these subunits is known as quaternary
structure.
NOTES
A protein with two or more identical subunits is called oligomer. A protomer
is the structural unit of an oligomeric protein. The proteins with identical subunits
are referred to as oligomers and these identical subunits as protomers. A protomer
may therefore consist of one polypeptide chain or several unlike polypeptide chains.
In this sense, haemoglobin is a dimer (oligomer of two protomers).
4.2.1 Classification of Protein
Proteins are classified on the following basis:
x Shape and Size
x Functional Properties
x Solubility and Physical Properties
Classification on the Basis of Shape and Size
Proteins are differentiated on the basis of there different shapes and sizes.
x Fibrous Proteins: This is simple protein. When the axial ratio of length,
width of a protein molecule is more than 10, it is called as fibrous protein
(for example, collagen, scleroprotein).
x Globular Proteins: When the axial ratio of length, width of a protein
molecule is less than 10, it is called as globular protein (for example,
myoglobin, haemoglobin, ribonuclease, etc).
Classification on the Basis of Functional Properties
The proteins are differentiated with regard to the functions they perform as follows:
x Defense Proteins: Involved in defense mechanisms, for example,
immunoglobulins.
x Respiratory Proteins: Involved in the function of respiratory, for example,
haemoglobin, myoglobulin, cytochromes.
x Contractile Proteins: Involved in muscle contractions and relaxation, for
example, protein of skeletal muscles.
x Hormones: A protein released by a cell or a gland in one part of the body
to sends out messages that affect cells in other parts of the organism.
x Enzymes: Proteins that catalyse the cellular reactions.
x Structural proteins: Involved in structural integrity of cells, for example,
proteins of skin, cartilage, nail.

Self-Instructional
96 Material
Classification on the Basis of Solubility and Physical Properties Protein Metabolism

This is the most acceptable scheme of classification of proteins. According to this


scheme, proteins are classified on the basis of their solubility and physical properties
and are divided into the following three classes: NOTES
x Simple Proteins
x Conjugated Proteins
x Derived Proteins Acids
Simple Proteins

These are the proteins, which on complete hydrolysis yield only amino acids. These
are further subclassified on the basis of the solubility and heat coagulability. Their
properties depend on the shape and size of the molecule. Major subclasses are as
follows:
x Albumen: It is found in egg, blood and milk. Its main properties are as
follows:
R It is heat coagulable.
R It is water soluble.
R It is soluble in weak salt solution.
R It is insoluble in strong salt solution.
x Globulin: It is found in egg, muscle, blood and milk. Its main properties are
as follows:
R It coagulates on heating.
R It is insoluble in water.
R It is soluble in weak salt solution.
R It is insoluble in strong salt solution.
x Glutelin: It is found in wheat and maize. Its main properties are as follows:
R It is soluble in dilute acids and alkalies.
R It is not coagulated by heat.
x Prolamine: It is found in wheat, barley and corn. It is soluble in 70-90%
alcohol.
x Histone: It is rich in basic amino-acids, soluble in water and associated
with nucleic acids.
x Protamines: It is rich in basic amino acids and soluble in water found in
nucleic acids.
Self-Instructional
Material 97
Protein Metabolism Conjugated Proteins
Conjugated proteins are simple proteins combined with a non-protein group called
prosthetic group. Protein part is called apoprotein, and the entire molecule is called
NOTES holoprotein.
x Nucleoproteins: The nucleoproteins are compounds made up of simple
basic proteins such as protamine or histone with nucleic acid as prosthetic
group. They are proteins of cell nuclei and chief constituents of chromatin.
Deoxyribonucleo proteins contain DNA as prosthetic group and are found
in nuclei, mitochondria and chloroplasts. Ribonucleoproteins occur in nuclei
and ribosome granules. They have RNA as prosthetic group. The examples
are nucleohistone and nucleo prolamine.
x Glycoproteins: The glycoproteins have the carbohydrate moiety as
prosthetic group. The polypeptide chain is attached to one or more
heterosaccharide units by covalent bonds. About 600-700 units are attached
to the peptide chain, one per 6.4 amino acid residue. The glycoproteins are
secreted by the submaxillary glands of various animals.
Derived Proteins
These are the proteins formed from native protein by the action of heat, physical
forces or chemical factors. This class of proteins includes those products formed
from the simple and conjugated proteins. It is not a well-defined class of proteins.
These are produced by various physical and chemical factors and derived into
two major groups.
x Primary Derived Proteins: These are the denatured or coagulated
proteins. Their molecular weight is same as that of native proteins, but they
differ in solubility, precipitation and crystallization. The heat, X-ray vigorous
shaking, acid, alkali cause denaturation of proteins and they form primary
derived proteins. These are synonymous with denatured proteins.
x Secondary Derived Proteins: These are the proteins, which are formed
from the hydrolysis of proteins. They are called proteoses, peptones and
peptides. They differ in their molecular sizes.
x Peptides: These are composed of only a small number of amino acids
joined by peptide bonds. These are water-soluble and are not coagulated
by heat and are not salted out of solution, but they can be precipitated by
phosphotungstic acid. These are named according to the number of amino
acids present in them. Dipeptides are made up of two amino acids, tripeptides
are made of three ammo acids and polypeptides are made up of more than
three amino acids. The number of amino acids depends on the
4.2.2 Protein Synthesis
Since proteolytic enzymes can degrade proteins to polypeptides, it seemed possible
that these might be the substrates for protein synthesis. Indeed, in view of the
Self-Instructional
98 Material
similarity of the domains within the different proteins, even those possessing different Protein Metabolism

functions, the possibility was considered that the primary structures of proteins
might be modified by the exchange of amino acids within existing polypeptide
chains. It is now apparent that the polypeptide backbone of all proteins is
synthesized from free amino acids. NOTES

Mechanism of Protein Synthesis


Peptides are artificially synthesized for the following purposes:
x To check whether the sequence analysis is correct or not.
x To alter the primary structure of a peptide for one or two amino acids in
order to determine the biologically important area or the active centre.
x To get pure preparations for medical or diagnostic purpose. For example,
HIV antibody in the blood of AIDS patients is detected by ELISA method.
For this, pure antigen from HIV is to be coated in the test tubes. Preparation
of enough quantity of antigen from the virus is tedious and hazardous.
Commercially, it is cheaper to synthesize smaller peptides than isolating
them from biological sources.
Emil Fischer in 1890 developed the basic mechanism to protect or activate
reactive groups of amino acids. Bruce Merrifield in 1961 introduced solid phase
peptide synthesis He was awarded the Nobel prize in 1984. He simplified the
process by adding; the carboxy terminal end amino acid to insoluble polystyrene
beads, so that washing and purification processes become rapid. Insulin was the
first major protein chemically synthesized. In principle, carboxyl group of the last
amino acid is fixed on the resin, and other amino acids are added sequentially.
Synthesis of Multichain Proteins
As you have seen, some proteins comprise several different polypeptide chains.
The association of such chains may be by means of weak non-covalent bonds, as
with haemoglobin, in which case the chains associate spontaneously so no novel
mechanism is required. In other cases, association may be promoted by the
presence of another protein, known as a molecular chaperone. An example is that
of the immunoglobulins where immunoglobulin-binding protein, present within the
rough endoplasmic reticulum of the plasma cells, assists in combining heavy and
light chains. Proteins that are destined to be exported from a cell are usually
synthesized in a precursor form with an additional peptide at the N-terminus. Such
proteins (for example, serum albumin) are known as proproteins and the peptide
as a propeptide. The propeptide is removed by an enzyme called furin—a member
of a superfamily of PC proteases, during the passage of the protein through the
Golgi apparatus to give rise to the mature protein. Many propeptides, including
those in the blood coagulation cascade and numerous viral proteins, are linked to
the mature protein through two adjacent basic amino acids (Refer Figure 4.2).

Self-Instructional
Material 99
Protein Metabolism

NOTES

Figure 4.2 Biosynthesis of Multichain Proteins

Synthesis of Polyproteins
In the case of some peptide hormones, the active peptide is split from a larger
protein. When the precursor protein gives rise to more than one mature protein it
is known as a polyprotein. The cleavage pattern of a particular polyprotein may
vary among different tissues; thus the same gene product can yield different sets of
polypeptide hormones. An example of this is proopiomelanocortin, which is the
precursor of AdrenoCorTicotropic Hormone (ACTH) and other hormones. The
initial translation product in the pituitary contains a signal peptide at the N-terminus,
which is removed to give proopiomelanocortin with an MT of 31 000. The dark
vertical bars represent proteolytic cleavage sites for specific enzymes, which
comprise a. family of proteases named Kex2, PC2 and PC3.The cleavage sites
are arg-lys, lys-arg or lys-lys. In the corticotropic cell of the anterior pituitary,
enzymes are present that cleave at sites 3 and 5, releasing the major products
ACTH and (3-lipotropin into the general circulation. In the pars intermedia,
especially in vertebrates below humans, these products are further cleaved at
major sites 4, 6 and 7 to release a-MSH (Melanocyte-Stimulating Hormone),
CLIP (Corticotropin-Like Intermediate Lobe Peptide), y-lipotropin and P-
endorphin into the general circulation. Some (S-lipotropin may be further degraded
to form P-endorphin. This contains a pentapeptide, enkephalin. which may be
released by hydrolysis at 8. P-Endorphin has morphine-like properties (endogenous
morphine) in that it and enkephalin bind to opiate receptors in nervous tissue. p-
Lipotropin was once thought, erroneously, to mobilize tatty acids from adipose
tissue.
Although, as indicated. P-endorphin can give rise to met-enkephalin; it was
subsequently shown that the enkephalins in the adrenals and brain arise from
proenkephalins, which contain multiple copies (often six) of the enkephalins
sandwiched between pairs of basic amino acid residues.
Self-Instructional
100 Material
All tissues are able to synthesize the non-essential amino acid, remodel Protein Metabolism

amino acids and convert non-amino acid carbon skeletons into amino acids and
other derivatives that contain nitrogen. However, the liver is the principal site of
nitrogen metabolism in the body. When nitrogen is in excess in diet, the potentially
toxic nitrogen of amino acids is excreted via the following processes: NOTES
x Transaminations
x Deamination
x Urea Formation
The carbon skeletons of proteins are normally conserved via the following processes:
x As fatty acid by fatty acid synthesis pathway.
x As carbohydrate by gluconeogenesis.

4.3 DEFICIENCY DISEASES OF PROTEINS

Protein is the building block of the muscles, skin, enzymes and hormones of our
body. It plays an essential role in the functioning of all body tissues and organs.
Most foods contain some form of protein. Deficiency of protein leads to various
health problems, specifically the low protein intake may cause changes in body
composition that develop over a long period of time, such as muscle wasting.
The most severe form of protein deficiency is known as kwashiorkor. It
most often occurs in children in developing countries where famine and imbalanced
diets are common.
Protein not only helps maintain muscle and bone mass, but it’s also essential
for body growth.
How Much Protein Do You Need?
Not everyone has the same protein requirement. It depends on many factors,
including body weight, muscle mass, physical activity and age.
The Recommended Daily Allowance (RDA) is 0.4 grams of protein for
each pound of body weight (0.8 grams per kg). This translates to 66 grams of
protein per day for an adult weighing 165 pounds (75 kg).
The richest sources of protein include fish, meat, eggs, dairy products and
legumes.
Worldwide, however, a lack of protein in the diet is a matter of concern,
especially when it affects children. It can lead to problems of malnutrition, such as
kwashiorkor and marasmus. These can be life-threatening.
The protein deficiency can also arise if a person has a health condition, such as:
x An eating disorder, for example, anorexia nervosa.
x Certain genetic conditions.
Self-Instructional
Material 101
Protein Metabolism x Later stages of cancer.
x Difficulty absorbing nutrients, for example, due to Irritable Bowel Syndrome
(IBS) or Gastric Bypass Surgery.
NOTES Very low protein intake can lead to:
x Weak Muscle Tone.
x Oedema, which is swelling due to fluid retention.
x Thin and brittle hair.
x Skin lesions.
x In adults, loss of muscle mass.
x In children, stunted growth.
Following are the protein deficiency diseases:
Kwashiorkor
Kwashiorkor is a disease caused by a severe deficiency of protein in diets that
contain calories mostly from carbohydrates, such as yams, rice and bananas. It
usually affects older children. People with kwashiorkor appear puffy in the abdomen
area from retention of fluid (Researched by University of Maryland Medical Center).
Signs and Symptoms: The defining sign of kwashiorkor in a malnourished child
is pitting oedema (swelling of the ankles and feet). Other signs include a distended
abdomen, an enlarged liver with fatty infiltrates, thinning of hair, loss of teeth, skin
depigmentation and dermatitis. Children with kwashiorkor often develop irritability
and anorexia. Generally, the disease can be treated by adding protein to the diet;
however, it can have a long-term impact on a child’s physical and mental
development, and in severe cases may lead to death.
Causes: Kwashiorkor is a severe form of malnutrition, caused by a deficiency in
dietary protein. The extreme lack of protein causes an osmotic imbalance in the
gastro-intestinal system causing swelling of the gut diagnosed as an oedema or
retention of water. Extreme fluid retention observed in individuals suffering from
kwashiorkor is a direct result of irregularities in the lymphatic system and an
indication of capillary exchange. The lymphatic system serves three major purposes:
fluid recovery, immunity, and lipid absorption. Victims of kwashiorkor commonly
exhibit reduced ability to recover fluids, immune system failure, and low lipid
absorption, all of which result from a state of severe undernourishment. Fluid
recovery in the lymphatic system is accomplished by re-absorption of water and
proteins which are then returned to the blood. Compromised fluid recovery results
in the characteristic belly distension observed in highly malnourished children.
Capillary exchange between the lymphatic system and the bloodstream is stunted
due to the inability of the body to effectively overcome the hydrostatic pressure
gradient. Proteins, mainly albumin, are responsible for creating the Colloid Osmotic
Pressure (COP) observed in the blood and tissue fluids. Due to the lack of proteins,
Self-Instructional
102 Material
no substantial pressure gradient can be established to draw fluids from the tissue Protein Metabolism

back into the blood stream. This results in the pooling of fluids, causing the swelling
and distention of the abdomen.
The low protein intake leads to some specific signs: oedema of the hands
NOTES
and feet, irritability, anorexia, a desquamative rash, hair discolouration, and a large
fatty liver. The typical swollen abdomen is due to two causes: ascites because of
hypoalbuminemia (low oncotic pressure), and enlarged fatty liver.
Marasmus
Marasmus is a disease caused by a severe deficiency of protein and calories that
affect infants and very young children, often resulting in weight loss and dehydration.
Marasmus can develop into starvation and cause fatality caused by a lack of
essential nutrients. People with marasmus appear bony with little muscle tissue.
Marasmus is a form of severe malnutrition characterized by energy deficiency.
It can occur in anyone with severe malnutrition but usually occurs in children. A
child with marasmus looks emaciated. Body weight is reduced to less than 62% of
the normal (expected) body weight for the age. Marasmus occurrence increases
prior to age 1, whereas kwashiorkor occurrence increases after 18 months. It can
be distinguished from kwashiorkor in that kwashiorkor is protein deficiency with
adequate energy intake whereas marasmus is inadequate energy intake in all forms,
including protein. This clear-cut separation of marasmus and kwashiorkor is
however not always clinically evident as kwashiorkor is often seen in a context of
insufficient caloric intake, and mixed clinical pictures, called marasmic kwashiorkor,
are possible. Protein wasting in kwashiorkor generally leads to oedema and ascites,
while muscular wasting and loss of subcutaneous fat are the main clinical signs of
marasmus.
Signs and Symptoms: Marasmus is commonly represented by a shrunken, wasted
appearance, loss of muscle mass and subcutaneous fat mass. Buttocks and upper
limb muscle groups are usually more affected than others. Oedema is not a sign of
marasmus and is present in only kwashiorkor and marasmic kwashiorkor. Other
symptoms of marasmus include unusual body temperature (hypothermia, pyrexia);
anaemia; dehydration (as characterized with consistent thirst and shrunken eyes);
hypovolemic shock (weak radial pulse; cold extremities; decreased consciousness);
tachypnea (pneumonia, heart failure); abdominal manifestations (distension,
decreased or metallic bowel sounds; large or small liver; blood or mucus in the
stools), ocular manifestations (corneal lesions associated with Vitamin A deficiency);
dermal manifestations (evidence of infection, purpura, and ear, nose, and throat
symptoms (otitis, rhinitis). Dry skin and brittle hair are also symptoms of marasmus.
Marasmus can also make children short-tempered and irritable.
Causes: Marasmus is caused by a severe deficiency of nearly all nutrients,
especially protein, carbohydrates and lipids, usually due to poverty and scarcity of
food. Viral, bacterial and parasitic infections can cause children to absorb few
Self-Instructional
Material 103
Protein Metabolism nutrients, even when consumption is adequate. Marasmus can develop in children
who have weakening conditions, such as chronic diarrhoea.
In dry climates, marasmus is the more frequent disease associated with
malnutrition. Another malnutrition syndrome includes cachexia, although it is often
NOTES
caused by underlying illnesses. These are important considerations in the treatment
of the patients. Common symptoms of both marasmus and kwashiorkor include
fatigue, irritability, diarrhoea, stunted growth and impairment of cognition and mental
health.
Impaired Mental Health
Long term protein inadequacy can influence your psychological well-being in
various ways. It can prompt mental hindrance (especially in youngsters) and
furthermore cause tension, surliness, sorrow and crankiness.
Oedema
Not getting enough protein can prompt oedema (liquid maintenance). This can
cause swelling in various zones of the body, for example, the feet, hands and
stomach. Aside from the swelling oedema can likewise cause hurting in the
appendages, stained skin, hypertension and solid joints.
Organ Failure
Protein is required for the growth and maintenance of various body functions.
Deficiency of protein can cause the improper function of different body organs.
Deficiencies of Protein C and Protein S
Deficiencies of Protein C and Protein S are inherited conditions that cause abnormal
blood clotting, according to Medline Plus. Deficiency of Protein C occurs in about
1 out of 300 people while the deficiency of Protein S affects 1 in 20,000 people.
Symptoms for these deficiencies include redness, pain, tenderness or swelling in
the affected area. People with these protein deficiencies need to be careful about
activities that increase risk of blood clots, such as prolonged sitting, bed rest, and
long-time travel in cars and airplanes.

4.4 INBORN ERRORS OF PROTEIN METABOLISM

The following are the major diseases caused due to errors in protein metabolism.
1. Albinism
x This condition appears in the total absence of tyrosinase inside the
melanocytes in the skin.
x The black pigment melanin is not formed in the skin, eyes and hair.
x This inherited condition occurs to a greater or less extent in all types of
organisms.
Self-Instructional
104 Material
x The diagnostic advice is for the prevention of exposure of sunlight and Protein Metabolism

protection of the eyes by wearing dark glasses.


2. Tyrosinosis
x This syndrome is due to the absence either of hepatic p- NOTES
hydroxyphenylpyruvate hydroxylase or of tyrosine transaminase activities.
x The patient excretes large quantities of tyrosine in the urine.
3. Tyrosinernia
A. Neonatal Tyrosinernia
x Neonatal tyrosinernia occurs in the new born.
x Tyrosine, p-hydroxphenylpyruvic acid, p-hydroxyphenyl lactic acid and p-
hydroxyphenyl acetic acid appear in the urine.
x A transient deficiency of p-hydro-xyphenyl-pyruvic acid oxidase causes
this condition and the condition lasts for a few weeks.
x Administration of ascorbic acid and reduction of protein intake brings about
the normal condition.
B. Hereditary Tyrosinernia
x This disorder is similar to neo-nataltyrosinernia, but the amount of p-
hydroxyphenyl lactic acid in the urine is greater.
x This disorder is not controlled by the administration of ascorbic acid.
x It is due to the inherited deficiency of p-hydroxyphenyl pyruvic acid oxidase.
x Liver failure and death can occur six months after birth and the infant has a
characteristic odour.
x Some patients develop hepatosplenomegaly, a nodular cirrhosis of the liver,
abnormalities of tyrosine and methionine metabolism, multiple defects in
renal tubular reabsorption, rickets, hyperphosphaturia, pro-teinuria and
amino aciduria.
4. Phenylketonuria
x This inherited disorder appears in the absence of phenylalanine hydroxylase
which is responsible for the conversion of phenylalanine to tyrosine. As a
result, alternative catabolizes of phenylalanine are produced. Much of the
phenyl-acetyl-glutamine is excreted in the urine.
x Mental retardation develops among infants and children.
x Patients with phenylketonuria tend to have a deficiency of serotonin. This
may be connected with the defect of myelin synthesis.
x The accumulation of phenylalanine also impairs melanin synthesis and children
with this defect tend to have fair skin and fair hair.
x Excess of phenylalanine in the blood leads to excretion of the amino acid
into the intestine. Here, it competes with the tryptophan for absorption and
tryptophan is subjected to the action of intestinal bacteria.
Self-Instructional
Material 105
Protein Metabolism x Early diagnosis (shortly after birth) and extreme restriction of phenylalanine
intake is effective in preventing this disorder.
5. Alkaptonuria
NOTES x This condition is characterized by the excretion of homogentisic acid (di-
hydroxyphenyl acetic acid) in the urine owing to the lack of homogentisic
acid oxidase.
x This abnormal condition is often found in infancy. Over 600 cases have
been reported. The incidence of alkaptonuria is 2-5 per million live births.
x The most clinical manifestation is the dark urine due to the oxidation of
homogentisic acid in air.
x In this disorder, several grams of homogentisic acid are excreted daily.
x This condition is present at birth and persists throughout life.
x In later life, accumulation of dark pigment in cartilages and tendons gives
rise to the condition known as ochronosis, which is accompanied by arthritic
changes.
6. Maple Syrup Urine Disease
x This syndrome is characterized by the absence of the enzymes required for
the oxidative decarboxylation of the keto acids derived from the branched
chain amino acids - valine, leucine and isoleucine. As a result, these keto
acids are accumulated in the blood and excreted in urine. The urinary
excretion of these keto acids produce an odour that is similar to maple
syrup or of burnt sugar.
x The infant is difficult to feed and may vomit. The patient suffers from a
significant degree of lethargy.
x Death may occur by the end of the first year of life without treatment.
7. Histidinemia
x It is an inherited disorder of histidine metabolism in which the amounts of
histidine in the blood and urine are increased. There is also increased
excretion of imidazole pyruvic acid.
x The metabolic block of histidine is due to the insufficient activity of liver
histidase which impairs the conversion of histidine to urocanic acid.
x Development of speech in this condition is retarded. Mental development
is also retarded.
x Histidine excretion is also increased during normal pregnancy but not in the
toxaemia of pregnancy. This increase is not due to metabolic defect.
8. Hypervalinemia
x In this abnormal condition, the infants suffer from stunted growth, muscle
wasting and vomiting.
x The valine content of blood and urine is very high.
Self-Instructional
106 Material
Protein Metabolism
4.5 ENTNER DOUDOROFF PATHWAY

The Entner–Doudoroff pathway (ED pathway) describes a pathway which includes


a series of enzyme-catalyzed chemical reactions that are active in bacterial primary NOTES
metabolism. It is a pathway that catabolizes glucose to pyruvic acid using enzymes
distinct either from those used in glycolysis or the pentose phosphate pathway.
The ED pathway was named so because it was first reported by Michael Doudoroff
and Nathan Entner from the Bacteriology Department at the University of California,
Berkeley in 1952. Recent work on the Entner–Duodoroff pathway has shown
that its use is not restricted to prokaryotes as it was previously thought.
Distinct features of the Entner–Doudoroff pathway are as follows:
x Uses 6-phosphogluconate dehydratase and 2-keto-3-
deoxyphosphogluconate aldolase to create pyruvate from glucose.
x Has a net yield of 1 ATP for every one glucose molecule processed, as well
as 1 NADH and 1 NADPH. In comparison, glycolysis has a net yield of 2
ATP molecules and 2 NADH molecules for every one glucose molecule
metabolised.
Figure 4.3 illustrates the Entner-Doudoroff pathway (KDPG: 2-keto-3-deoxy-6-
phosphogluconate).

Fig. 4.3 Entner-Doudoroff Pathway (KDPG: 2-keto-3-deoxy-6-phosphogluconate)


Self-Instructional
Material 107
Protein Metabolism Organisms that Use the Entner-Doudoroff Pathway
There are several bacteria that use the Entner–Doudoroff pathway for metabolism
of glucose and are unable to catabolize via glycolysis, for example therefore lacking
NOTES essential glycolytic enzymes, such as phosphofructokinase as seen in
Pseudomonas). Genera in which the pathway is prominent include Gram-Negative
and Gram-Positive bacteria, such as Enterococcus faecalis, as well as several in
the Archaea, the second distinct branch of the Prokaryotes. Most organisms that
use the pathway are Aerobes, due to the low ATP yield per glucose molecule
metabolised.
Examples of bacteria using the pathway include the following:
Pseudomonas - A genus of Gram-Negative Bacteria
Azotobacter - A genus of Gram-Negative Bacteria
Rhizobium - A plant genus of Gram-Negative Bacteria
Agrobacterium - A plant pathogen (oncogenic) genus of Gram-Negative Bacteria
Escherichia coli - A Gram-Negative Bacterium
Enterococcus faecalis - A Gram-Positive Bacterium
Zymomonas mobilis - A Gram-Negative Facultative Anaerobe
Xanthomonas campestris - A Gram-Negative Bacterium uses this pathway for
providing energy
Conversion of Glucose to Glucose-6-Phosphate
The first step in Entner–Doudoroff pathway is phosphorylation of glucose by a
family of enzymes called hexokinases to form Glucose-6-Phosphate (G6P). This
reaction consumes ATP, but it acts to keep the glucose concentration low, promoting
continuous transport of glucose into the cell through the plasma membrane
transporters. In addition, it blocks the glucose from leaking out – the cell lacks
transporters for G6P, and free diffusion out of the cell is prevented due to the
charged nature of G6P. Glucose may alternatively be formed from the
phosphorolysis or hydrolysis of intracellular starch or glycogen.
In animals, an isozyme of hexokinase called glucokinase is also used in the
liver, which has a much lower affinity for glucose (Km in the vicinity of normal
glycemia), and differs in regulatory properties. The different substrate affinity and
alternate regulation of this enzyme are a reflection of the role of the liver in maintaining
blood sugar levels.
Conversion of Glucose-6-Phosphate to 6-Phosphoglucanolactone
The G6P is then converted to 6-phosphoglucanolactone in the presence of enzyme
glucose-6-phosphate dehydrogenase( an oxido-reductase) with the presence of
co-enzyme Nicotinamide Adenine Dinucleotide Phosphate (NADP+) which will
be reduced to nicotineamide adenine dinucleotide phosphate hydrogen along with
a free hydrogen atom H+.
Self-Instructional
108 Material
Conversion of 6-Phosphoglucanolactone to 6-Phosphogluconic Acid Protein Metabolism

The 6PGL is converted into 6-phosphogluconic acid in the presence of enzyme


hydrolase.
Conversion of 6-Phosphogluconic Acid to 2-Keto-3-Deoxy-6- NOTES
Phosphoglucanate
The 6-phosphogluconic acid is converted to 2-Keto-3-Deoxy-6-
Phosphogluconate (KDPG) in the presence of enzyme 6-phosphogluconate
dehydratase in which water molecule is released to the surroundings.
Conversion of 2-Keto-3-Deoxy-6-Phosphoglucanate to Pyruvate and
Glyceraldehyde-3-Phosphate
The KDPG is then converted into pyruvate or glyceraldehyde-3-phosphate in the
presence of enzyme KDPG aldolase. When KDPG is converted into pyruvate,
the Entner–Doudoroff pathway for that pyruvate ends here and then the pyruvate
goes into further metabolic pathways, such as the TCA cycle, ETC cycle, etc. The
other product (Glyceraldehyde-3-Phosphate) is further converted by entering into
the glycolysis pathway and at last get converted into pyruvate for further metabolism.
Conversion of Glyceraldehyde-3-Phosphate to 1, 3-Bisphosphoglycerate
The G3P is converted to 1, 3-bisphosphoglycerate in the presence of enzyme
Glyceraldehyde-3-Phosphate Dehydrogenase (an oxido-recductase). The
aldehyde groups of the triose sugars are oxidised, and inorganic phosphate is
added to them, forming 1, 3-Bisphosphoglycerate. The hydrogen is used to reduce
two molecules of NAD+, a hydrogen carrier, to give NADH + H+ for each triose.
Conversion of 1, 3-Bisphosphoglycerate to 3-Phosphoglycerate
This step is the enzymatic transfer of a phosphate group from 1, 3-
Bisphosphoglycerate to ADP by phosphoglycerate kinase, forming ATP and 3-
Phosphoglycerate.
Conversion of 3-Phosphoglycerate to 2-Phosphoglycerate
Phosphoglycerate Mutase isomerises 3-Phosphoglycerate into 2-Phosphoglycerate.
Conversion of 2-Phosphoglycerate to Phosphoenol Pyruvate
Enolase then converts 2-Phosphoglycerate to Phosphoenolpyruvate. This reaction
is an elimination reaction.
Conversion of Phosphoenol Pyruvate to Pyruvate
A final substrate-level phosphorylation now forms a molecule of pyruvate and a
molecule of ATP by means of the enzyme pyruvate kinase. This serves as an
additional regulatory step, similar to the Phosphoglycerate Kinase step.
Self-Instructional
Material 109
Protein Metabolism

Check Your Progress


1. Distinguish between hydrogen bond and disulphide bonds.
NOTES 2. How many types of secondary structures of proteins are there?
3. What are the basis of classification of proteins?
4. Write the classification of proteins on the basis of functional properties.
5. Write the classification of proteins on the basis of solubility and physical
properties.
6. What is Albumen? Give its main properties.

4.6 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. Hydrogen bonds: These are weak, low energy, non covalent bonds sharing
single hydrogen by two electronegative atoms i.e. O and N. The H bonding
in secondary structure occurs regularly. This regularity allows the protein to
assume helical configuration or sheeted structure. Disulphide bonds: These
are formed between two cysteine residues. They are strong, high energy,
covalent bonds.
2. The secondary structures of proteins are of the following two types:
x Alpha-helix
x Beta-pleated sheet structure
3. Proteins are classified on the following basis:
x Shape and size
x Functional properties
x Solubility and physical properties
4. Classification of proteins on the basis of functional properties: The proteins
are differentiated with regard to the functions they perform as follows:
x Defense proteins: Involved in defense mechanisms, for example,
immunoglobulins
x Respiratory proteins: Involved in the function of respiratory, for
example, haemoglobin, myoglobulin, cytochromes
x Contractile proteins: Involved in muscle contractions and relaxation,
for example, protein of skeletal muscles.
x Hormones: A protein released by a cell or a gland in one part of the
body to sends out messages that affect cells in other parts of the
organism
x Enzymes: Proteins that catalyse the cellular reactions
Self-Instructional
110 Material
x Structural proteins: Involved in structural integrity of cells, for example, Protein Metabolism

proteins of skin, cartilage, nail


5. Classification of proteins on the basis of solubility and physical properties:
This is the most acceptable scheme of classification of proteins. According
NOTES
to this scheme, proteins are classified on the basis of their solubility and
physical properties and are divided into the following three classes:
x Simple proteins
x Conjugated proteins
x Derived proteins acids
6. Albumen is found in egg, blood and milk. Its main properties are as follows:
x It is heat coagulable.
x It is water soluble.
x It is soluble in weak salt solution.
x It is insoluble in strong salt solution.

4.7 SUMMARY

x In 1939, G. J. Muller investigated the substances found in milk and egg.


x J. J. Berzelius suggested that these substances should be called proteins.
x Proteins are defined as the high molecular weight mixed polymers of amino
acids joined by peptide linkages.
x Proteins are the essence of life processes. They are the fundamental
constituents of all protoplasm and are involved in the structural integrity and
functions of living cells.
x Proteins serve as regulators of these reactions both directly as components
of enzymes and indirectly in the form of chemical messengers known as
hormones as well as the receptors for these hormones.
x Proteins, such as rhodopsin in the retina of eye, acquire sensory information
that is processed through the action of nerve cell proteins.
x The proteins of immune system such as immunoglobulin form an essential
biological defense system in higher animals.
x Structural proteins are generally inert to biochemical reactions. They maintain
the natural form and position of the organs, the cell wall and primary fibrous
constituent of the cell’ have structural proteins.
x Collagen is the most abundant known protein in animals, forming a major
part of the skin, cartilage, ligament, tendon and bone.
x The properties of living cells depend upon the biological processes, which
are regulated by enzymes.
Self-Instructional
Material 111
Protein Metabolism x The major portion of an enzyme is made up of proteins. These proteins are
mostly spherical in shape and are termed globular proteins.
x Some enzymes are simple proteins containing only ammo acids, others are
NOTES complex proteins.
x Enzymes catalyse a variety of reactions in the living cells. Certain proteins,
especially in the animals are involved in the transport of many essential
biological factors to different parts of the organisms.
x Haemoglobin transporting O2 from one part of the body to the other is an
example of carrier protein.
x Hydrogen bonds are weak, low energy, non-covalent bonds sharing single
hydrogen by two electronegative atoms, i.e., O and N.
x The H bonding in secondary structure occurs regularly. This regularity allows
the protein to assume helical configuration or sheeted structure.
x Disulphide bonds are formed between two cysteine residues. They are
strong, high energy, covalent bonds.
x Tertiary structure of a protein is its three-dimensional folding of its arrangement
i.e. polypeptide chain with the secondary structure which may be further
folded forming many sizes.
x A protein with two or more identical subunits is called oligomer. A protomer
is the structural unit of an oligomeric protein.
x Since proteolytic enzymes can degrade proteins to polypeptides, it seemed
possible that these might be the substrates for protein synthesis.
x Indeed, in view of the similarity of the domains within the different proteins,
even those possessing different functions, the possibility was considered
that the primary structures of proteins might be modified by the exchange of
amino acids within existing polypeptide chains.
x Proteins that are destined to be exported from a cell are usually synthesized
in a precursor form with an additional peptide at the N-terminus.
x Many propeptides, including those in the blood coagulation cascade and
numerous viral proteins, are linked to the mature protein through two adjacent
basic amino acids.
x Protein is the building block of the muscles, skin, enzymes and hormones of
our body. It plays an essential role in the functioning of all body tissues and
organs.
x Deficiency of protein leads to various health problems, specifically the low
protein intake may cause changes in body composition that develop over a
long period of time, such as muscle wasting.
x The most severe form of protein deficiency is known as kwashiorkor. It
most often occurs in children in developing countries where famine and
Self-Instructional imbalanced diets are common.
112 Material
x Protein not only helps maintain muscle and bone mass, but it’s also essential Protein Metabolism

for body growth.


x Kwashiorkor is a disease caused by a severe deficiency of protein in diets
that contain calories mostly from carbohydrates, such as yams, rice and
NOTES
bananas.
x Kwashiorkor is a severe form of malnutrition, caused by a deficiency in
dietary protein.
x The extreme lack of protein causes an osmotic imbalance in the gastro-
intestinal system causing swelling of the gut diagnosed as an oedema or
retention of water.
x The low protein intake leads to some specific signs: oedema of the hands
and feet, irritability, anorexia, a desquamative rash, hair discolouration, and
a large fatty liver.
x The typical swollen abdomen is due to two causes: ascites because of
hypoalbuminemia (low oncotic pressure), and enlarged fatty liver.
x Marasmus is a disease caused by a severe deficiency of protein and calories
that affect infants and very young children, often resulting in weight loss and
dehydration.
x Marasmus can develop into starvation and cause fatality caused by a lack
of essential nutrients. People with marasmus appear bony with little muscle
tissue

4.8 KEY WORDS

x Oligomer: A protein with two or more identical subunits is called oligomer.


x Simple proteins: These are the proteins, which on complete hydrolysis
yield only amino acids.
x Nucleoproteins: The nucleoproteins are compounds made up of simple
basic proteins such as protamine or histone with nucleic acid as prosthetic
group.
x Derived proteins: The proteins formed from native protein by the action
of heat, physical forces or chemical factors.

4.9 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. Distinguish between secondary structure and tertiary structure of proteins.
2. Write a short note on alpha-helix.
Self-Instructional
Material 113
Protein Metabolism 3. Draw a well-labelled diagram to show beta-pleated sheet of amino acid
4. Briefly discuss about ionic or electrostatic interactions.
5. Give the classification of proteins on the basis of solubility and physical
NOTES properties
6. What are derived proteins
Long Answer Questions
1. Discuss in detail about protein metabolism.
2. Write the biological significance of proteins.
3. Give the structural organization of proteins.
4. Classify protein on various categories.
5. Discuss about the deficiency diseases of proteins.
6. What are the inborn errors of protein metabolism?

4.10 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
114 Material
Lipids
BLOCK - II
LIPIDS, VITAMINS AND MINERALS

NOTES
UNIT 5 LIPIDS
Structure
5.0 Introduction
5.1 Objectives
5.2 Classification of Lipids
5.3 Properties of Lipids
5.4 Classfication of Fatty Acids
5.4.1 Types of Fats
5.4.2 Fatty Acyls
5.5 Properties of Fatty Acids
5.5.1 Saturated Fatty Acids
5.5.2 Unsaturated Fatty Acids
5.6 Phospholipid
5.7 Cholesterol Synthesis in E Coli
5.8 Answers to Check Your Progress Questions
5.9 Summary
5.10 Key Words
5.11 Self Assessment Questions and Exercises
5.12 Further Readings

5.0 INTRODUCTION
The term ‘lipids’ applies to a group of naturally occurring substances which are
insoluble in water but soluble in non-polar solvents like chloroform, ether, benzene
and carbon disulphide. They occur widely in nature in either free form or often
associated with other compounds like proteins and carbohydrates. The role of
dietary fats and oils in human nutrition is an essential area of concern and
investigation in the field of nutritional science. The findings of these investigations
have wide-ranging implications for consumers, health-care providers and nutrition
educators, as well as food producers, processors and distributors. New evidence
concerning the advantages and risks in relation to certain characteristics of dietary
fat is constantly emerging in both scientific literature and the popular media. Fat
is a major source of energy aiding the body in absorbing vitamins. Fats are
significant for the growth, development and the overall health of an individual. In
addition to this fat provides taste to foods and satiates the body. Fats serve as a
significant source of calories and nutrients for infants and toddlers.
In this unit, you will Study about lipids and fats and what role they play in
our lifestyle.
Self-Instructional
Material 115
Lipids
5.1 OBJECTIVES
After going through this unit, you will be able to:
NOTES x Classify lipids
x Discuss properties of lipids and fatty acids
x Describe phospholipid
x Explain cholesterol synthesis in E. coli
5.2 CLASSIFICATION OF LIPIDS

Lipids are classified as follows:


x Simple Lipids
x Compound Lipids
x Terpenoids and Steroids
x Derived Lipids
Simple Lipids
These include oils and fats. These are esters of fatty acids and glycerol. It has
been observed that oils tend to remain liquid at 20°C whereas fats tend to remain
solid this temperature. A fatty acid is a long chain carboxylic acid
(CH3.(CH2)n.COOH. The fatty acids that are found in natural fats and oils have
even number of carbon atoms. Figure 5.1 displays the general structure of a fat
molecule.
C H2-O-OC- R1
C H-O-OC- R2
C H2-O-OC-R3
Fig. 5.1 General Structure of a Fat Molecule

Where R1, R2, R3 are fatty acid components.


There are also known as ‘neutral fats’ and are commonly found in adipose
tissue, butter, lard, fish oil, olive olive oil, corn oil, mustard oil and so on. The
following reaction shows the formation of triglycerides form fatty acid and glycerol:

Most fatty acids are linear but some branched chain and cyclic fatty acids are also
found.
Compound Lipids
Compound lipids include phospholipids, sphingolipids, glycolipids and sulpholipids.
Self-Instructional
116 Material
x Phospholipids: These are also known as phosphatides. They contain Lipids

phosphoric acid, nitrogenous base and glycerol and fatty acids (lecithin,
cepahiln, etc). Figure 5.2 displays the structure of phospholipids; X can be
choline, serine, glycerol, etc.
NOTES

Fig. 5.2 Structure of Phospholipids

x Sphingolipids: They contain sphingosine or dihydrosphingosine.


x Glycolipids: They contain carbohydrates along with fatty acids and glycerol.
x Sulpholipids: They contain sulphuric acid alon with fatty avids and glycerol.
Terpenoids and Steroids
x Terpenes: Found in essential oils, resin acids, rubber, plant pigments such
as caotenese and lycopenes, Vitamin A, and camphor. Large group of
compounds made up of repeating isoprene units; Vitamin A of nutritional
interest; fat soluble Vitamin E and K, which are also related chemically to
terpenes.
x Sterols:
R Cholesterol: Found in egg yolk, dairy products, and animal tissues.
A constituent of bile acids and a precursor of Vitamin D.
R Ergosterol: Found in plant tissues, yeast, and fungi. Converted to
Vitamin D2 on irradiation.
R 7-dehydrocholesterol: Found in animal tissues and underneath skin.
Converted to D3 on irradiation.
x Androgens and Estrogens: (Sex hormones) Found in ovaries and testes.
x Adrenal Corticolsteroids: adrenal cortex, blood.
Derived Lipids
Fatty Acids: occur in plant and animal foods; also exhibit in complex forms with
other substances. Obtained from hydrolysis of fats; usually contains an even number
of carbon atoms and are straight chain derivatives.

Check Your Progress


1. What are simple lipids?
2. Where is cholesterol found?
3. Where are androgens and estrogens?
Self-Instructional
Material 117
Lipids
5.3 PROPERTIES OF LIPIDS

Lipids are usually defined as those components that are soluble in organic solvents
NOTES (such as ether, hexane or chloroform), but are insoluble in water. This group of
substances includes triacylglycercols, diacylglycercols, rnonoacylglycercols, free
fatty acids, phospholipids, sterols, caretonoids and vitamins A and D.
The lipid fraction of a fatty food therefore contains a complex mixture
of different types of molecule. Triacylglycercols are the major component of
most foods, typically making up more than 95 to 99per cent of the total lipids
present.
Triacylglycerols are esters of three fatty acids and a glycerol molecule.
The fatty acids normally found in foods vary in chain length, degree of
unsaturation and position on the glycerol molecule. Consequently, the triacylglycerol
fraction itself consists of a complex mixture of different types of molecules.
Each type of fat has a different profile of lipids present which determines the
precise nature of its nutritional and physiochemical properties.
The terms fat, oil and lipid are often used interchangeably by food scientists.
Although sometimes the term that is used to describe those lipids that are solid at
the specified temperature, whereas the term oil is used to describe those lipids
that are liquid at the specified temperature.
Solid Fat Content
The Solid Fat Content (SFC) of a lipid influences many of its sensory and physical
properties, such as spreadability, firmness, mouthfeel, processing and stability.
Melting Point
In many situations, it is not necessary to know the SFC over the whole temperature
range; instead, only information about the temperature at which melting starts or
ends is required. A pure triacylglycerol has a single melting point that occurs at a
specific temperature. Nevertheless, foods lipids contain a wide variety of different
triacylglycerols, each with their own unique melting point, and so they melt over a
wide range of temperatures. Thus, the ‘melting point’ of a food lipid can be
defined in a number of different ways, each corresponding to a different amount of
solid fat remaining.
Clear Point
A small amount of fat is placed in a capillary tube and heated at a controlled rate.
The temperature at which the fat completely melts and becomes transparent is
called the ‘clear point’.
Slip Point
A small amount of fat is placed in a capillary tube and heated at a controlled rate.
The temperature at which the fat just starts to move downwards due to its weight
is called the ‘slip point’.
Self-Instructional
118 Material
Wiley Melting Point Lipids

A disc of fat is suspended in an alcohol-water mixture of similar density and is then


heated at a controlled rate. The temperature at which the disc changes shape to a
sphere is called the ‘wiley melting point’. NOTES
Cloud Point
This gives a measure of the temperature at which crystallization begins in liquid oil.
A fat sample is heated to a temperature where all the crystals are known to have
melted.
Smoke, Flash and Fire Points
These tests give a measure of the effect of heating on the physicochemical properties
of lipids. They are particularly important for selecting lipids that are going to be
used at high temperatures, for example during baking or frying. The tests reflect
the amount of volatile organic material in oils and fats such as free fatty acids.
Saponification
It is the process of breakdown of neutral fat into glycerol and fatty acids by their
treatment with alkali. The saponification number is a measure of the average
molecular weight of the triacylglycerols in a sample. The saponification number is
defined as the mg of KOH required to saponify one gram of fat. The smaller the
saponification number the larger is the average molecular weight of the
triacylglycerols present.
Rheology
The rheology of lipids is important in many food applications. Rheology is the
science concerned with the deformation and flow of matter. Most rheological tests
involve applying a force to a material and measuring its flow or change in shape.
Many of the textural properties that people perceive when they consume
foods are largely rheological in nature, for example, creaminess, juiciness,
smoothness, brittleness, tenderness, hardness, etc. The stability and appearance
of foods often depends on the rheological characteristics of their components.
The flow of foods through pipes or the ease at which they can be packed into
containers are also determined by their rheology. Liquid oils are usually characterized
in terms of their flow properties (viscosity), whereas viscoelastic or plastic ‘solids’
are characterized in terms of both their elastic (elastic modulus) and flow properties.
A wide variety of experimental techniques are available to characterize the
rheological properties of food materials.
One of the most important rheological characteristics of lipids is their
‘plasticity’, because this determines their ‘spreadability’.
The plasticity of a lipid is due to the fact that fat crystals can form a three-
dimensional network that gives the product some solid-like characteristics.
Self-Instructional
Material 119
Lipids Below a certain stress (known as the ‘yield stress’) the product behaves
like a solid with an elastic modulus because the crystal network is not disrupted,
but above this stress it flows like a liquid because the crystal network is continually
disrupted.
NOTES
Rheological techniques are therefore needed to measure the change in
deformation of a lipid when stresses are applied.
Table 5.1 Properties of Different Oils
No. Name of the Oil Analysis
Percent of Oil Specific Saponification Value Iodine
in Seed or Gravity value
Kernal

1 Ambadi Oil 30-40 ---- 189-195 93-107


2 Castor Seed Oil 45-50 0.945- 177-187 83-86
0.965/25°C OH-160
3 Cheru Seed Oil Kernal-60-62 0.856- 191-200 90-101
(Phulware Fat) 0.862/25°C
4 Chullu(Wild apricot) 45-50 0.915- 186-188 101-106
Seed Oil 0.918/25°C
5 Coconut Oil 63-65 0.917- 251-263 7.5-10.5
0.919/25°C
6 Corn (Maize) Oil Germ-48 0.915- 187-193 103-128
0.920/25°C
7 Cotton Seed Oil 18-25 0.915- 191-196 103-115
0.926/15°C
8 Dhupa Seed Oil 20-22 0.912/25°C 187-192 36-43
(Dhupa fat)
9 Groundnut Oil (Penut 45-55 0.910- 188-195 84-100
Oil) 0.915/25°C
10 Hemp Seed Oil 30-35 0.923- 190-193 140-175
0.925/25°C
11 Jojoba(Hohoba) Seed 45-50 0.8635- 92-167 82-89
Oil 0.8640/25°C
12 Kamala Seed Oil 30-35 0.9409/40°C 195 166
13 Kapok Oil 22-25 0.920- 189-197 86-110
0.933/15°C
14 Karanja Seed Oil 27-39 0.936- 180-190 81-90
0.937/15°C Acetyl 14.5
15 Kokum Oil 25-34 0.895/40°C 187-192 25-38
(Kokum Butter)

16 Kusum Oil Kernal-28-34 0.9099/30°C 220-234 Acetyl 4.0 50-60


17 Linseed Oil (Flax 40-44 0.931- 189-196 170-180
Seed Oil) 0.938/15°C
18 Mahua Seed Oil 35-42 0.856- 187-194 58-70
(Mohwa, Mahuda) 0.870/15°C
19 Maroti Oil 41-45 0.940- 198-204 92-103
0.960/25°C

Self-Instructional
120 Material
Lipids
20 Mustard Seed Oil (Rap Seed 33-40 0.906- 169-176 98-110
Oil) 0.910/25°C
21 Nahor Seed Oil 60-70 0.956/25°C 193-209 73-93

22 Neem Oil Kernal-40- 0.915- 175-200 65-80 NOTES


50 0.922/30°C
23 Niger Seed Oil 30-45 0.917- 189-193 125-135
0.920/30°C
24 Oiticica Oil (Cicoil) 50-60 0.977- 186-193 ----
0.988/20°C
25 Olive Oil 45-70 0.909- 187-196 79-90
0.919/25°C
26 Palash(Palas Seed) Oil 17-19 ---- 175-190 65-85

27 Palm Kernal Oil Kernal-44- 0.886- 245-255 14-22


65 0.873/99°C
28 Palm Oil 30-60 0921-0.925/15°C 196-205 48-58

29 Perilla Oil 35-40 0.930- 188-197 193-208


0.937/15°C
30 Pilu Oil (Khakan Fat) 38-42 0.867/25°C 240-250 12-20

31 Pisa Oil 48-50 0.925/25°C 255.5- 8.5-10.9


257
32 Poppyseed Oil 36-50 0.898/60°C 197.5 133.4
33 Ratanjyot Oil (Jatropha Seed 30-40 0.918- 191-202 82-98
Oil) 0.923/15°C OH 4-20
34 Rice Bran Oil 15-23 0.916- 181-188 90-108
0.921/25°C
35 Rubber Seed Oil Kernal-45- 0.924- 190-195 132-148
52 0.930/15°C OH 12-
32
36 Safflower Oil 25-37 0.919- 188-194 140-150
0.924/25°C
37 Sal Seed Oil (Sal Fat) 14-20 0.868- 188-192 35-39
0.870/30°C
38 Sesame Oil(Til Oil) 45-54 0.916- 188-195 103-116
0.921/25°C
39 Sorghum Oil 30-50 ---- 181-191 108-122

40 Soya Bean Oil 15-20 0.916- 189-195 128-143


0.922/25°C

Self-Instructional
Material 121
Lipids
41 Sunflower Seed Oil 25-35 0.915-0.919/25°C 188-194 125-140

42 Tea Seed Oil 58-60 ---- 193-196 83-89


43 Thumba(Tumba) Seed Oil 19-22 0.924-0.927/28°C 172-174 118-122
NOTES 44 Tobacco Seed Oil 30-43 0.923-0.925/15°C 186-197 125-154

45 Tung Oil 16-18 0.930/15°C 189-195 160-175


46 Undi Oil (Punna Oil) Kernal-50-60 0.942-0.945/15°C 191-202 82-98
47 Wheat Germ Oil 8-14 0.925-0.933/25°C 179-190 115-129

Check Your Progress


4. What is SFC?
5. What is the slip point?

5.4 CLASSFICATION OF FATTY ACIDS

Fats are mainly the ester of glycerol and one, two or three fatty acids. They are
soft, solid, or semisolid organic compounds, constituting the esters of glycerol and
fatty acids and their associated organic groups. The mixtures of such compounds,
occurring widely in organic tissue, especially in the adipose tissue of animals and in
the seeds, nuts, and fruits of plants are known as fats.
Fats are the solid form of triacyglycerol. In common terms, lipids are also
known as fats, however, there is a difference—all fats are lipids, but all lipids are
not fats. Fats are a type of simple lipid. There is a very small difference between
fats and oils too. They differ only in their physical existence, that is, fats are solid
and oils are liquid at room temperature.
Fats act as storage of energy. They give 9 cal / gm of energy on catabolism.
They are stored as adipose tissue in various parts of our body. This tissue acts as
an insulator against shock from outside. Fats molecules act as precursors for
synthesis of various lipids. They also help in absorption of fat soluble vitamins, that
is, vitamin A, D, E and K4.
5.4.1 Types of Fats
Fats are classified according to the presence of unsaturation, that is, presence of
double bonds. They are mainly of the following two types:
1. Saturated Fats: Saturated fats (Refer Figure 5.3) do not have double
bond in their fatty acids.

Self-Instructional Fig. 5.3 Saturated Fat


122 Material
2. Unsaturated Fats: Unsaturated fats (Refer Figure 5.4) contain double Lipids

bonds in their fatty acids. Since they are able to take up hydrogen in them,
they are called unsaturated fats.

NOTES

Fig. 5.4 Unsaturated Fat

The various classifications of unsaturated fats are as follows:


x Monounsaturated Fats: These fats contain only one double bond in their
fatty acids. The fatty acid present in them is known as monounsaturated
fatty acid (MUFA). Common monounsaturated fatty acids are palmitoleic
acid (16:1 n–7), cis-vaccenic acid (18:1 n–7) and oleic acid (18:1 n–9).
Palmitoleic acid has 16 carbon atoms with the first double bond occurring 7
carbon atoms away from the methyl group (and 9 carbons from the carboxyl
end). It can be lengthened to the 18-carbon cis-vaccenic acid. Oleic acid
has 18 carbon atoms with the first double bond occurring 9 carbon atoms
away from the carboxylic acid group.
x Polyunsaturated Fats: The fats, having more than one double bond present
in their fatty acids are called polyunsaturated fats. The fatty acid present in
them is known as PolyUnsaturated Fatty Acid (PUFA). The fatty acids can
exist in any of the following two types of confirmations, as a result of their
arrangement of the carbon atom around the double bonds:
– Trans Fats: In trans fats, the fatty acids have such an arrangement
that the carbon chains are present on the opposite side. This makes
the chain appear straight. Elaidic acid is an example of trans fat.
– Cis Fats: In cis fats, as the fatty acids have arrangement of chains in
the same side of the double bond, there is a kink or bend near the
double. An example of cis fats is oleic acid.
x Omega Fatty Acids: These fats have double bond from the methyl side,
that is, opposite side of the carboxyl functional group. These are of the
following three types:
– Omega-3 Fatty Acids: Omega-3 fatty acids (Refer Figure 5.5) have
final carbon–carbon double bond in the n–3 position, that is, the third
bond from the methyl end of the fatty acid. They are also called n–3
fatty acids. Nutritionally important n–3 fatty acids include
polyunsaturated EicosaPentaenoic Acid (EPA), A-Linolenic Acid
(ALA), (EPA) and docosahexaenoic acid (DHA).
Self-Instructional
Material 123
Lipids – Omega-6 Fatty Acid: Omega–6 fatty acids (Refer Figure 5.6) acids
have final carbon–carbon double bond in the n–6 position, that is, the
sixth bond, counting from the end opposite the carboxyl group. They
are also called n-6 fatty acids. Linoleic acids and arachidonic acids
NOTES are significant n-6 fatty acids.
– Omega-9 Fatty Acids: Omega-9 fatty acids (Refer Figure 4.9) are
also called n- 9 fatty acids because they contain carbon – carbon
double bond in n-9 position from methyl end. Oleic acid and Erucic
acid are significant n-9 fatty acids.

Fig. 5.5 Omega-3 Fatty Acid

Fig. 5.6 Omega-6 Fatty Acid

Fig. 5.7 Omega-9 Fatty Acid (Oleic Acid)

5.4.2 Fatty Acyls


Fatty acyls is a generic term used to describe fatty acids and their conjugates
and derivatives. It refers to a diverse group of molecules synthesized by chain-
elongation of an acetyl-CoA primer with malonyl-CoA or methylmalonyl-CoA
groups in a process called fatty acid synthesis. These molecules are made of a
Self-Instructional
124 Material
hydrocarbon chain that terminates with a carboxylic acid group. This arrangement Lipids

confers the molecule with a polar, hydrophilic end and a non-polar, hydrophobic
end that is insoluble in water. The fatty acid structure is one of the most fundamental
categories of biological lipids and is commonly used as a building block of more
structurally complex lipids. The carbon chain, typically between four to 24 carbons NOTES
long, may be saturated or unsaturated and may be attached to functional groups
containing oxygen, halogens, nitrogen and sulfur. Where a double bond exists,
there is the possibility of either a cis or trans geometric isomerism, which
significantly affects the molecule’s molecular configuration. Cis-double bonds cause
the fatty acid chain to bend, an effect that is more pronounced with more double
bonds in the chain. This, in turn, plays an important role in the structure and function
of cell membranes. Most naturally occurring fatty acids are of the cis configuration,
although the trans form does exist in some natural and partially hydrogenated
fats and oils.
Eicosanoids are examples of biologically important fatty acids, which are
primarily derived from arachidonic acid and eicosapentaenoic acid, which includes
prostaglandins, leukotrienes, and thromboxanes. Other major lipid classes in the
fatty acid category are the fatty esters and fatty amides. Fatty esters include important
biochemical intermediates, such as wax esters, fatty acid thioester coenzyme A
derivatives, fatty acid thioester ACP derivatives and fatty acid carnitines. The fatty
amides include N-acyl ethanolamines, such as the cannabinoid neurotransmitter
anandamide. The number of carbons in fatty acids is generally even, because fatty
acid biosynthesis occurs with sequential addition of two carbon units.

Check Your Progress


6. What are saturated fats?
7. What are polyunsaturated fats?
8. What are omega-9 fatty acids?

5.5 PROPERTIES OF FATTY ACIDS

The properties of saturated and unsaturated fatty acids are described in the following
section.
5.5.1 Saturated Fatty Acids
The straight-chain (contain only single bond between carbon atoms) or normal-
chain fatty acid components (that are normally even numbered) make up 10 to 40
per cent of the total fatty acids in most natural lipids. The most abundant saturated
fatty acids in animal and plant tissues are straight chain compounds with 14, 16
and 18 carbon atoms. All the possible odd and even numbered homologues are
found in esterified form in nature. They are named systematically with saturated
Self-Instructional
Material 125
Lipids hydrocarbon with the same number of carbon atoms, the final ‘e’ being changes to
‘oic’. Thus the fatty acid with 16 carbon atoms is systematically named as
hexadecanoic acid. The normal term used to describe hexadecanoic acid in nutrition
is palmitic acid. Figure 5.8 displays the structural formula of palmitic acid .
NOTES

Fig. 5.8 Structural Formula of Palmitic Acid


It can be also termed as C16 fatty acid or 16:0 with a greater precision. The
number before the colon specifies the number of carbon atoms and the number
after the colon species the position of the double bond.
Saturated fatty acids with more than 24 carbon atoms normally occur in
foods and waxes. The fatty acids present in milk fats contain 4 to 10 carbon
atoms while those of chain length of C12 to C24 occur in most animal and vegetable
fats.
Some saturated fatty acids and their molecular formulae are as follows:
Butyric: CH3(CH2)2COOH
Lauric (Dodecanoic Acid): CH3 (CH2)10COOH
Myristic (Tetradecanoic Acid): CH3 (CH2)12COOH
Palmitic (Hexadecanoic Acid): CH3 (CH2) 14COOH
Stearic: Stearic acid also called octadecanoic acid is one of the many useful types
of saturated fatty acids that come from many animal and vegetable fats and oils. It
is a waxy solid, and its chemical formula is CH(CH)COOH.
Arachidic: Arachidic acid also called eicosanoic acid is a saturated fatty acids
found in peanut oil. It melts at 75. 4°C and its chemical formula is CH(CH)COOH.
(Eicosanoic Acid): CH3 (CH2)18COOH
5.5.2 Unsaturated Fatty Acids
Unsaturated fatty acids contain double bonds between their carbon atoms. The
number of double bonds may vary from one to six. The hydrogen atom is removed
where the double bond is formed. The unsaturated fatty are more vulnerable to
rancidity. The unsaturated fatty acids are of two types, which are as follows:
x Monounsaturated Fatty Acids
x Polyunsaturated Fatty Acids

Self-Instructional
126 Material
Table 5.2 displays unsaturated fat food sources and their fat content. Lipids

Table 5.2 Unsaturated Fat Food Sources and their Fat Content

Unsaturated Fat Food Sources Monounsatu Polyunsaturat


rated Fat ed Fat Content NOTES
Content (grams/100 g
(grams/100 g food)
food)
Nuts
Nuts, macadamia nuts, dry roasted, with salt added 1.50 59.27
Nuts, hazelnuts or filberts 7.92 45.65
Nuts, pecans 21.62 40.80
Nuts, almonds 12.22 32.16
Nuts, mixed nuts, oil roasted, with peanuts, with salt added 13.30 31.70
Nuts, mixed nuts, dry roasted, with peanuts, with salt added 10.77 31.39
Nuts, cashew nuts, dry roasted, with salt added 7.84 27.32
Nuts, cashew nuts, oil roasted, with salt added 8.55 25.92
Nuts, brazilnuts, dried, unblanched 20.58 24.55
Nuts, pistachio nuts, dry roasted, with salt added 13.90 24.22
Nuts, pine nuts, dried 34.07 18.77
Nuts, pine nuts, dried 34.07 18.77
Nuts, walnuts 47.17 8.93
Nuts, coconut meat, dried (desiccated), sweetened, shredded 0.39 1.51
Nuts, coconut meat, raw 0.37 1.42
Nuts, chestnuts, roasted 0.87 0.76
Peanuts
Peanuts, all types, oil-roasted, with salt 15.27 25.94
Peanuts, all types, dry-roasted, with salt 15.69 24.64
Peanuts, all types, dry-roasted, without salt 15.69 24.64
Peanut butter, chunk style, with salt 14.80 24.56
Peanut butter, smooth style, with salt 13.87 23.71
Vegatable Oils
Oil, vegetable safflower, salad or cooking, oleic, over 70per 14.35 74.65
cent (primary safflower oil of commerce)
Oil, olive, salad or cooking 10.53 72.96
Oil, peanut, salad or cooking 32.00 46.20
Oil, soybean, salad or cooking, (hydrogenated) 37.60 43.00
Oil, sesame, salad or cooking 41.70 39.70
Oil, soybean, salad or cooking, (hydrogenated) and cottonseed 48.10 29.50
Oil, vegetable, corn, industrial and retail, all purpose salad or 54.68 27.57
cooking
Oil, vegetable, sunflower, linoleic, (approx. 65 per cent) 65.70 19.50

Self-Instructional
Material 127
Lipids Monounsaturated Fatty Acids (MUFA)
Monounsaturated fatty acids contain only one double bond between the carbon
atoms in a molecule of fatty acid. These include oleic acid, palmitoleic acid,
myristoleic acid,etc. Figure 5.9 displays the structure oleic acids.
NOTES

Fig. 5.9 Structure of Oleic Acid


Monounsaturated fatty acids have a higher melting temperature than
polyunsaturated fatty acids but lower than saturated fatty acids. They are liquid at
room temperature and may become semi-solid or solid when refrigerated.
Polyunsaturated Fatty Acids (PUFA) or Essential Fatty Acids (EFA)
Polyunsaturated fatty acids contain more than one double bond and are derived
from those containing one double bond. Figure 5.10 displays the structure of stearic
acid, oleic acid, linoleic acid, D-linoleic acid.

Fig. 5.10 Comparison of Structures of Stearic, Oleic, Linoleic and D-Linoleic Acid
Classification of PUFA
Linoleic Acid Family
x Linolenic Acid (C18:2)
x J-linolenic Acid (C18:3)
x Arachidonic Acid (C20:4)
Self-Instructional
128 Material
Linolenic Acid Family Lipids

x Linolenic Acid (C18:3)


x Eicosapentaenoic Acid (C20:5)
x Decosahaexoic Acid (C22:6) NOTES
Table 5.3 displays unsaturated fatty acids and their chemical structure.
Table 5.3 Unsaturated Fatty Acids and their Chemical Structure

Common Chemical Structure Δx C:D n–x


Name

Myristoleic acid CH3(CH2)3CH=CH(CH2)7COOH cis-Δ9 14:1 n–5

Palmitoleic acid CH3(CH2)5CH=CH(CH2)7COOH cis-Δ9 16:1 n–7


6
Sapienic acid cis-Δ 16:1 n–10

Oleic acid CH3(CH2)7CH=CH(CH2)7COOH cis-Δ9 18:1 n–9


9 12
Linoleic acid CH3(CH2)4CH=CHCH2CH=CH(CH2)7COOH cis,cis-Δ ,Δ 18:2 n–6

α-Linolenic acid CH3CH2CH=CHCH2CH=CHCH2CH=CH cis,cis,cis-Δ9,Δ12,Δ15 18:3 n–3


(CH2)7COOH

Arachidonic CH3(CH2)4CH=CHCH2CH=CHCH2CH=CHC cis,cis,cis,cis-Δ5Δ8,Δ11,Δ14 20:4 n–6


acid H2CH=CH(CH2)3COOH

Eicosapentaenoi CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2 cis,cis,cis,cis,cis-Δ5,Δ8,Δ11,Δ14,Δ17 20:5 n–3


c acid CH=CHCH2CH=CH(CH2)3COOH

Erucic acid CH3(CH2)7CH=CH(CH2)11COOH cis-Δ13 22:1 n–9


4 7 10 13 16 19
Docosahexaenoi CH3CH2CH=CHCH2CH=CHCH2CH=CHCH2 cis,cis,cis,cis,cis,cis-Δ ,Δ ,Δ ,Δ ,Δ ,Δ 22:6 n–3
c acid CH=CHCH2CH=CHCH2CH=CH(CH2)2COOH

List of n–3 Fatty Acids


Table 5.4 lists the chemical names for the most common n–3 fatty acids found in
nature.
Table 5.4 Chemical Names for Common N”3 Fatty Acids Found in Nature

Common name Lipid name Chemical Name


16:3 (n–3) all-cis-7,10,13-Hexadecatrienoic Acid
α-Linolenic Acid (ALA) 18:3 (n–3) all-cis-9,12,15-Octadecatrienoic Acid
Stearidonic Acid (STD) 18:4 (n–3) all-cis-6,9,12,15-Octadecatetraenoic Acid
Eicosatrienoic Acid (ETE) 20:3 (n–3) all-cis-11,14,17-Eicosatrienoic Acid
Eicosatetraenoic Acid (ETA) 20:4 (n–3) all-cis-8,11,14,17-Eicosatetraenoic Acid
Eicosapentaenoic Acid (EPA) 20:5 (n–3) all-cis-5,8,11,14,17-Eicosapentaenoic Acid
Docosapentaenoic Acid (DPA), 22:5 (n–3) all-cis-7,10,13,16,19-Docosapentaenoic Acid
Clupanodonic Acid
Docosahexaenoic Acid (DHA) 22:6 (n–3) all-cis-4,7,10,13,16,19-Docosahexaenoic Acid
Tetracosapentaenoic Acid 24:5 (n–3) all-cis-9,12,15,18,21-Docosahexaenoic Acid
Tetracosahexaenoic Acid (Nisinic acid) 24:6 (n–3) all-cis-6,9,12,15,18,21-Tetracosenoic Acid

Table 5.5 lists common fatty acids of animal and plant origin.

Self-Instructional
Material 129
Lipids Table 5.5 Common Fatty Acids of Animal and Plant Origin

Systematic name Trivial Name Shorthand


Saturated Fatty Acids
NOTES
Ethanoic Acetic 2:0
Butanoic Butyric 4:0
Hexanoic Caproic 6:0
Octanoic Caprylic 8:0
Decanoic Capric 10:0
Dodecanoic Lauric 12:0
Tetradecanoic Myristic 14:0
Hexadecanoic Palmitic 16:0
Octadecanoic Stearic 18:0
Eicosanoic Arachidic 20:0
Docosanoic Behenic 22:0
Monoenoic Fatty Acids
cis-9-Hexadecenoic Palmitoleic 16:1(n-7)
cis-6-Octadecenoic Petroselinic 18:1(n-12)
cis-9-Octadecenoic Oleic 18:1(n-9)
cis-11-Octadecenoic Cis-Vaccenic 18:1(n-7)
cis-13-Docosenoic Erucic 22:1(n-9)
cis-15-Tetracosenoic Nervonic 24:1(n-9)
Polyunsaturated Fatty Acids*
9,12-Octadecadienoic Linoleic 18:2(n-6)
6,9,12-Octadecatrienoic γ-Linolenic 18:3(n-6)
9,12,15-Octadecatrienoic α-Linolenic 18:3(n-3)
5,8,11,14-Eicosatetraenoic Arachidonic 20:4(n-6)
5,8,11,14,17-Eicosapentaenoic EPA 20:5(n-3)
4,7,10,13,16,19-Docosahexaenoic DHA 22:6(n-3)
* all the double bonds are of the cis configuration
Table 5.6 lists the positional distribution of fatty acid in seed oil.

Self-Instructional
130 Material
Table 5.6 Positional Distributions of Fatty Acids (Mol Percent) Lipids
in Triacyl-sn-Glycerols of Seed Oils

Oil Position Fatty Acid


16:0 18:0 18:1 18:2 18:3 C20-C24
NOTES
Peanut TG 9 3 58 23 7
1 14 5 59 19 4
2 2 tr 59 39 1
3 11 5 57 10 15
Rapeseed* TG 3 2 26 17 10 43
1 4 2 23 11 53
2 1 37 36 6 6
3 4 3 17 4 20 70
Soyabean TG 9 4 24 54 8
1 14 6 23 48 9
2 1 tr 22 70 7
3 13 6 28 45 9
Linseed TG 6 4 16 17 57
1 10 6 15 16 53
2 2 1 16 21 60
3 6 4 17 13 59
Maize (Corn) TG 11 2 29 57 1
1 1 3 28 50 1
2 18 tr 27 70 1
3 2 3 31 52 1
Olive TG 10 2 76 10 1
1 13 3 72 10 1
2 1 83 14 1
3 17 4 74 5
Cacao Butter TG 24 35 36 3 tr 1
1 34 50 12 1 1 1
2 2 2 87 9
3 37 53 9 tr 2
Palm TG 48 4 36 10
1 60 3 27 9
2 13 tr 68 18
3 72 8 14 3

tr = trace ( <0.5per cent ). * High erucic acid rapeseed oil. TG = intact triacylglycerols
The positional distribution of fatty acid in fat depots of animals is listed in table 5.7.

Self-Instructional
Material 131
Lipids Table 5.7 Positional Distributions of Fatty Acids (Mol Percent)
in Triacyl-sn-Glycerols of Animal Depot Fats

Species Position Fatty Acid


14:0 16:0 16:1 18:0 18:1 18:2 18:3
NOTES

Human TG 5 24 7 8 46 7 1
1 4 42 3 15 27 6 1
2 6 10 12 2 55 4 2
3 4 19 6 6 57 11 1
Cattle TG 5 27 6 17 33 5 1
1 4 41 6 17 20 4 1
2 9 17 6 9 41 5 1
3 1 22 6 24 37 5 1
Sheep TG 3 22 2 35 36 2
1 1 35 2 47 4 -
2 4 14 2 15 52 5
3 3 16 1 42 26 2
Pig TG 2 27 3 13 45 9
1 1 10 2 30 51 6
2 4 72 5 2 13 3
3 - tr 2 7 70 18
Rat TG 2 23 5 6 35 26 1
1 2 32 5 9 32 15 1
2 1 10 4 1 37 45 1
3 2 27 5 7 37 17 1
Rabbit TG 3 28 9 3 29 20 4
1 3 34 9 6 25 14 2
2 6 25 12 1 26 23 5
3 1 24 7 3 35 22 5
Chicken TG 1 30 6 6 45 11 1
1 1 47 7 8 31 5 1
2 tr 13 5 6 55 19 1
3 1 31 7 3 49 8 1

Results are listed for cis-18:1 isomers only; trans-18:1 was present in positions sn-1, sn-2 and
sn-3 as 5, 2 and 6 per cent, respectively.
Tr = trace (<0.5per cent). TG = intact triacylglycerols
The positional distribution of fatty acid in milk fats of animals is listed in table 5.8.
Self-Instructional
132 Material
Table 5.8 Composition of the Fatty Aids Esterified to Each Position of the Lipids
Triacyl-sn-Glycerols in the Milk Fats of Various Species
Species Position Fatty Acid
4:0 6:0 8:0 10:0 12:0 14:0 16:0 16:1 18:0 18:1 18:2 18:3
Cow TG 12 5 2 4 4 11 24 2 7 24 3 NOTES
1 - 1 1 2 5 10 34 2 10 30 2
2 - 1 1 3 6 18 32 4 10 19 4
3 35 13 4 6 1 6 5 1 1 23 2
Human TG tr 3 26 27 6 7 36 11 1
1 tr 1 3 16 4 15 46 11 tr
2 tr 2 7 58 5 3 13 7 1
3 1 6 7 6 8 2 50 15 1
Rat TG 6 19 14 12 21 2 3 13 10 1
1 3 10 10 10 20 2 5 24 14 1
2 6 20 16 18 29 2 1 3 5 1
3 10 26 15 9 13 2 2 12 12 1
Pig TG 4 32 9 5 39 10 1
1 2 22 7 7 50 11 1
2 7 58 11 1 15 8 1
3 4 15 10 6 52 12 2

tr = trace (< 0.5 per cent). TG = intact triacylglycerols.


Table 5.9 lists the positional distribution of fatty acid in animal tissues other than
milk fats and fat depots of the body.
Table 5.9 Positional Distributions of Fatty Acids (Mol Percent) in
Triacyl-sn-glycerols from Animal Tissues Other than Depot Fats and Milk

Position Fatty Acid


Source 16:0 16:1 18:0 18:1 18:2
Sheep Livera TG 26 3 16 43 5
1 47 2 17 21 1
2 14 3 18 55 8
3 16 3 13 54 5
Sheep Plasma TG 28 2 22 31 2
1 34 2 33 21 2
2 38 3 6 33 1
3 11 2 28 40 2
Sheep Adrenals TG 23 2 16 32 3
1 33 2 31 28 1
2 41 4 4 26 3
3 4 1 15 48 4
Chicken Plasma TG 29 5 5 50 11
1 74 6 4 13 1
2 4 3 2 60 31
3 8 5 8 76 2
Chicken Egg TG 29 5 7 49 10
1 71 5 4 17 2
2 4 3 3 63 26
3 12 6 14 67 1
Self-Instructional
Material 133
Lipids Sheep tissues contain appreciable amounts of minor components not listed here
including odd-and branched-chain, etc.
TG = intact triacylglycerols.

NOTES Table 5.10 displays the positional distribution of fatty acid in fish oils.
Table 5.10 Positional Distributions of Fatty Acids (Mol Percent) in
Triacyl-sn-Glycerols of Fish Oils

Species Position Fatty Acid


14:0 16:0 16:1 18:0 18:1 18:2 20:1 22:1 20:5 22:5 22:6
Herring TG 7 12 9 1 11 2 17 23 9 2 5
1 6 12 13 1 16 3 25 14 3 1 1
2 10 17 10 1 10 3 6 5 18 3 13
3 4 7 5 1 8 1 20 50 4 1 1
Mackerel TG 6 14 7 2 17 2 11 16 9 2 9
1 6 15 11 3 21 2 8 18 5 1 2
2 10 21 6 1 9 1 5 5 12 3 20
3 2 5 4 2 21 2 19 24 10 1 5
Skate TG 2 13 8 2 23 1 13 9 7 3 18
1 2 19 12 5 30 1 12 8 4 1 5
2 3 15 7 1 9 1 8 5 6 7 37
3 1 6 6 1 28 2 19 11 11 2 11
Cod TG 6 13 13 3 20 2 12 6 12 2 9
1 6 15 14 6 28 2 12 6 2 1 1
2 8 16 12 1 9 2 7 5 12 3 20
3 4 7 14 1 23 2 17 7 13 1 6

TG = intact triacylglycerols.

5.6 PHOSPHOLIPID

The steps involved in the elongation of the fatty acid chain are quite similar in
bacteria, fungi, plants and animals. The formation of acetyl-ACP and malonyl-
ACP initiate the elongation reactions. Acetyl-ACP and malonyl-ACP are formed
by acetyl transacylase (acetyl transferase) and malonyl transacylase (malonyl
transferase), respectively. While the acetyl transacylase enzyme is not highly specific
as it can transfer other acyl groups, such as the propionyl group, malonyl
transacylase is highly specific.
The acetyl group from ACP gets transferred to E-keto-acyl-ACP synthase
(KSase), also known as acyl-malonyl-ACP condensing enzyme by another
transacylase reaction. The first actual elongation reaction involves the condensation
of acetyl-ACP and malonyl-ACP by the E-ketoacyl-ACP synthase to form
acetoacetyl-ACP. The decarboxylation that accompanies the reaction with malonyl-
ACP drives the synthesis of acetoacetyl-ACP. ATP is responsible for the
Self-Instructional
134 Material
condensation reaction to form acetoacetyl-ACP. Malonyl-CoA can be viewed as Lipids

a form of stored energy for driving fatty acid synthesis and it can be established
that all the carbons of acetoacetyl-ACP are derived from acetate units of acetyl-
CoA.
The biosynthetic cycle finally results in the synthesis of a four-carbon unit, NOTES
a butyryl group, from two smaller building blocks. This butyryl-ACP condenses
with another malonyl-ACP, in the next cycle of the process, making a six-carbon
E-ketoacyl-ACP and CO2. A six-carbon saturated acyl-ACP is yielded by a
subsequent reduction to a E-alcohol, dehydration and another reduction. Until the
chain is 16 carbons long, this cycle continues with the net addition of a two-
carbon units in each turn. Hydrolysis of the C16-acyl-ACP yields a palmitic acid
and the free ACP.
In the end, seven malonyl-CoA molecules and one acetyl-CoA yield a
palmitate (shown here as palmitoyl-CoA):
Acetyl-CoA + 7 malonyl-CoA2 + 14 NADPH + 14 H+
o palmitoyl-CoA + 7 HCO32 + 14 NADP+ + 7 CoASH
The formation of seven malonyl-CoA molecules requires:
7 Acetyl-CoA + 7 HCO32 + 7 ATP42 o
7 malonyl-CoA2 + 7 ADP32 + 7 Pi22 + 7 H1
Thus, the overall reaction of acetyl-CoA to yield palmitic acid is:
palmitoyl-CoA + 14 NADP+ + 7 CoASH + 7 ADP32 + 7Pi22
Thus, it can be said that the fatty acid synthase complex contains activities of
seven different enzymes and an Acyl Carrier Protein (ACP). In summary, the
following reactions occur in FAS:
x The two carbons of acyl CoA are transferred to ACP of FAS complex by
the enzyme acetyl CoA –ACP transacylase. The acetyl group attached to
ACP is transferred to cysteine.
x The malonyl group of malonyl CoA is transferred to ACP by enzyme malonyl
CoA – ACP transacylase
x The enzyme E –ketoacyl ACP synthase transfers acetyl group from cysteine
to malonyl, attached to ACP with loss of one CO2. This results in the
synthesis of E-ketoacyl ACP.
x The E –ketoacyl ACP is converted to E-hydroxyacyl- ACP by reduction
reaction in the presence of E-ketoyacyl –ACP reductase. The reducing
equivalent is NADPH.
x A dehydration reaction of E-hydroxyacyl- ACP introduces a double between
D and E carbons. This reaction is catalysed by E-hydroxyacyl- ACP
dehydratase. This results in the formation of Trans e 2 –Enoyl CoA.
x The enzyme enoyl CoA reductase reduces Trans '2 –Enoyl CoA to Acyl –
ACP(butyryl-ACP). The reduction is done with the help of NADPH.
x The steps repeat till all the 16 carbons of palmitate are added.
Self-Instructional
Material 135
Lipids x The enzyme palmitoyl thioesterase separates palmitate from fatty acid
synthase.
Additional Elongation
NOTES Though the primary product of the fatty acid synthase is palmitate, cells synthesize
many other fatty acids. If the chain is released before reaching 16 carbons in
length, many shorter chains can be easily made. Special elongation reactions, taking
place both in the mitochondria and at the surface of the endoplasmic reticulum are
used to make longer chains. The ER reactions of adding two-carbon units at the
carboxyl end of the chain by means of oxidative decarboxylations involving malonyl-
CoA provide the thermodynamic driving force for the condensation reaction. The
mitochondrial reactions involve addition (and subsequent reduction) of acetyl units.
These reactions are essentially a reversal of fatty acid oxidation, with the exception
that NADPH is utilized in the saturation of the double bond, instead of FADH2.
In a newly synthesized fatty acid, both prokaryotes and eukaryotes can
introduce a single cis double bond. While eukaryotes need an O2-dependent
pathway to carry out this process, bacteria like E. coli can need an O2-independent
pathway. While the O2-independent reaction requires some other means to activate
the desired bond toward dehydrogenation, O2-dependent reaction can occur
anywhere in the fatty acid chain.
In E. coli, four normal cycles of elongation begin the biosynthesis of a
monounsaturated fatty acid to form a 10-carbon intermediate, E-hydroxydecanoyl-
ACP. At this point, E-hydroxydecanoyl thioester dehydrase forms a double bond
3,4 to the thioester and in the cis configuration. This is followed by three rounds of
the normal elongation reactions to form palmitoleoyl-ACP. Elongation may
terminate at this point or may be followed by additional biosynthetic events. cis-
vaccenic acid, the principal unsaturated fatty acid in E. coli, is formed by an
additional elongation step, using palmitoleoyl-ACP as a substrate.
It is only after the fatty acyl chain has reached its full length, does the addition
of double bonds to fatty acids in eukaryotes occur. Even if no useful functional
group exists on the chain to facilitate activation, dehydrogenation of stearoyl-CoA
occurs in the middle of the chain:
CH3O(CH2)16CO-SCoA o CH3O(CH2)7CH = CH(CH2)7CO-SCoA
stearoyl-CoA desaturase, a 53-kD enzyme containing a non-heme iron center
catalyzes this reaction. Other requirements include, NADH, and oxygen (O2) and
two other proteins; cytochrome b5 reductase (a 43-kD flavoprotein) and
cytochrome b5 (16.7 kD). A pair of electrons is transferred by cytochrome b5
reductase from NADH through FAD to cytochrome b5. Reduction of nonheme
Fe3+ to Fe2+ in the desaturase occurs with oxidation of reduced cytochrome b5.
The Fe3+ accepts a pair of electrons (one at a time in a cycle) from cytochrome b5
and creates a cis double bond at the 9, 10 position of the stearoyl-CoA substrate.
O2 is the terminal electron acceptor in this fatty acyl desaturation cycle. Animals
can synthesize fatty acids with double bonds at positions beyond C-9 only by this
method.
Self-Instructional
136 Material
This single desaturation reaction may lead to additional chain elongation. Lipids

Two carbons can be used to elongate the oleoyl-CoA produced to form a 20:1
cis-'11 fatty acyl-CoA. Reactions similar to the preceding scheme yield
palmitoleoyl-CoA, if the starting fatty acid is palmitate. This can subsequently be
elongated to yield cis-vaccenic acid. Similarly, C16 and C18 fatty acids can be NOTES
elongated to yield C22 and C24 fatty acids.
Differences occur in organisms with respect to formation, processing and
utilization of polyunsaturated fatty acids. For example, while Eukaryotes synthesize
different types of polyunsaturated fatty acids, E.Coli does not have any such acids.
While plants manufacture double bonds between the '9 and the methyl end of the
chain, mammals cannot. However, they can introduce double bonds between the
double bond at the 8- or 9-position and the carboxyl group. Additional double
bonds to unsaturated fatty acids can be added by mammals in their diets. They
can make arachidonic acid from linoleic acid. This fatty acid serves as the precursor
for prostaglandins and other biologically active derivatives like leukotrienes.
Synthesis involves formation of a linoleoyl ester of CoA from dietary linoleic acid
and introduction of a double bond at the 6-position. This is followed by elongation
of a triply unsaturated product by malonyl-CoA with a decarboxylation step so as
to yield a 20-carbon fatty acid with double bonds at the 8, 11, and 14-positions.
Finally, a 20-carbon fatty acid with double bonds at the 5-, 8-, 11-, and 14-
positions is liberated by a second desaturation reaction at the 5-position followed
by an acyl-CoA synthetase reaction. These steps are shown in Figure 5.11.

Fig. 5.11 Coupling of Fatty Acid Synthesis and Fatty Acid Oxidation
Self-Instructional
Material 137
Lipids Biosynthesis of Complex Lipids
Fatty acids are covalently bound to the backbone structures of complex lipids.
The major classes of lipids are glycerolipids and sphingolipids. For glycerolipids,
NOTES glycerol is the backbone and for sphingolipids sphingosine is the backbone. The
two major classes of glycerolipids are glycerophospholipids and triacylglycerols.
The phospholipids are precursors of hormones such as the eicosanoids (for
example, prostaglandins) and signal molecules. They consists of both
glycerophospholipids and sphingomyelins are important parts of the membrane
structure.
As different organisms have different complements of lipids, they invoke
different lipid biosynthetic pathways. For example, while bacteria have simple
lipid compositions, sphingolipids and triacylglycerols are produced only in
eukaryotes. 75 per cent of the phospholipids in E. coli is accounted by
posphatidylethanol-amine and the rest is taken care by phosphatidyl-glycerol and
cardiolipin. There are no phosphatidylcholine, phosphatidylinositol, sphingolipids,
or cholesterol in E. coli membranes.
Glycerolipid Biosynthesis
A common pathway that operates in nearly all organisms for the synthesis of
phosphatidic acid is glycerolipid biosynthesis. Glycerokinase catalyzes the
phosphorylation of glycerol to form glycerol-3-phosphate. It is then acylated at
both the 1- and 2-positions to yield phosphatidic acid. Glycerol-3-phosphate
acyltransferase catalyzes the first acylation at position 1.
Dihydroxyacetone phosphate can also be utilized by eukaryotic systems as
a begining point for synthesis of phosphatidic acid. The reduction of the backbone
keto group by acyldihydroxyacetone phosphate reductase, using NADPH as the
reductant occurs after a specific acyltransferase adds the first acyl chain.
Alternatively, glycerol-3-phosphate dehydrogenase can be used to reduce
dihydroxyacetone phosphate to glycerol-3-phosphate. Figure 5.12 shows the
synthesis of glycerolipids in eukaryotes.

Self-Instructional
138 Material
Lipids

NOTES

Fig. 5.12 Synthesis of Glycerolipids in Eukaryotes

In eukaryotes, phosphatidic acid is converted directly either to diacylglycerol or


to cytidine diphosphodiacylglycerol or CDP-diacylglycerol, from which other
glycerophospholipids are derived. Triacylglycerol, phosphatidylethanolamine and
phosphatidylcholine are necessary for synthesis of diacylglycerol. Out of these,
Triacylglycerol, which is synthesized primarily in adipose tissue, liver and
intestines, is the principal energy storage molecule in eukaryotes. While in liver
and adipose tissue, the biosynthesis of triacylglycerol occurs via diacylglycerol
acyltransferase, a different route is used in intestines. After triacylglycerols from
the diet are broken down by specific lipases to to 2-monoacylglycerols,
acyltransferases, acylate 2-monoacylglycerol to produce new triacylglycerols
as shown Figure 5.13.

Self-Instructional
Material 139
Lipids

NOTES

Fig. 5.13 Formation of Triacylglycerols

The phosphorylation of ethanol-amine to form phosphoethanolamine, initiates the


phosphatidylethanolamine synthesis. This is followed by transfer of a cytidylyl group
from CTP to form CDP-ethanolamine and pyrophosphate. This reaction is driven
forward by PPi hydrolysis. Phosphoethanolamine is thereby linked to the di-
acylglycerol backbone by a specific phosphoethanolamine transferase. Since
animals synthesize biosynthesis of phosphatidylcholine directly, its synthesis is entirely
Self-Instructional
140 Material
analogous. It is necessary for all the choline utilized in this pathway to be acquired Lipids

from the diet. However, yeast, certain bacteria and animal livers are able to convert
phosphatidylethanolamine to phosphatidylcholine by methylation reactions involving
S-adenosylmethionine.
NOTES
PhosphatidylSerine (PS) is synthesized by mammals in a calcium ion-
dependent reaction, which involves aminoalcohol exchange. The enzyme, which
catalyzes this reaction is linked with the endoplasmic reticulum and accepts
PhosphatidylEthanolamine (PE) and other phospholipid substrates. PS is
subsequently converted to PE by a mitochondrial PS decarboxylase. This is
illustrated in Figure 5.14.

Fig. 5.14 Inter-Conversion of Phosphatidylethanolamine and


Phosphatidylserine in Mammals

For some other important phospholipids, like Phosphatidyl-Inositol (PI),


PhosphatidylGlycerol (PG) and cardiolipin, eukaryotes use CDP-diacylglycerol
as a precursor. Though PI itself accounts for only about 2 to 8 per cent of the
lipids in most animal membranes, the breakdown products of PI like inositol-1,4,
5-trisphosphate and diacylglycerol serve as second messengers in many cellular
signaling processes. This is shown in Figure 5.15.

Self-Instructional
Material 141
Lipids

NOTES

Fig. 5.15 CDP-Diacylglycerol as a Precursor of Phosphatidylinositol,


Phosphatidylglycerol and Cardiolipin in Eukaryotes

At the 1-position, some glycerophospholipids, instead of an acyl ester group,


possess alkyl or alkenyl ether groups. These glyceroether phospholipids are
synthesized from DiHydroxyAcetone Phosphate((DHAP). After an exchange
reaction, in which the acyl group is removed as a carboxylic acid and a long-chain
alcohol is added to the 1-position, the acylation of the DHAP occurs. An acyl-
CoA reductase reaction, which involves oxidation of two molecules of NADH is
used to derive this long-chain alcohol. This reaction is obtained from the
corresponding acyl-CoA. The DHAP backbone of the 2-keto group is thereby
reduced to an alcohol, followed by acylation. Ether analogs of phosphatidylcholine,
Self-Instructional
142 Material
Lipids
phosphatidylethanolamine and so forth can be produced, if the resulting 1-alkyl-
2-acylglycero-3-phosphate reacts to phosphatidic acid in a similar manner. The
alkyl ether chains of these lipids can be desaturated by specific desaturase enzymes,
which are linked with the endoplasmic reticulum. Plasmalogens, which refer to the NOTES
products, which contain DE-unsaturated ether-linked chains at the C-1 position,
are abundantly found in the cardiac tissue and in the central nervous system. The
desaturases, catalyzing these reactions use cytochrome b5 as a cofactor, NADH
as a reductant and O2 as a terminal electron acceptor. This biosynthesis of
plasmogens in animals is shown in Figure 5.16.

Fig. 5.16 Biosynthesis of Plasmalogens in Animals

Platelet activating factor is an interesting ether phospholipid with unusual


physiological properties. As shown in Figure 5.17, it possesses an alkyl ether at
C-1 and an acetyl group at C-2. The very short chain at C-2 makes this molecule
much more water-soluble than typical glycerolipids. Platelet activating factor is
able to dilate blood vessels and aggregate platelets in animals.
Self-Instructional
Material 143
Lipids

NOTES

Fig. 5.17 Platelet Activating Factor

Sphingolipid Biosynthesis
Neural tissues contain high-levels of Sphingolipids, which are ubiquitous
components of eukaryotic cell membranes. The myelin sheath that insulates nerve
axons is particularly rich in sphingomyelin and other related lipids. Sphingolipids,
which are built upon sphingosine backbones, are normally not present in prokaryotic
organisms.
3-ketosphinganine synthase, which is a PLP-dependent enzyme, catalyses
the initial reaction, that involves condensation of serine and palmitoyl-CoA with
release of bicarbonate (Refer Figure 5.18). Reduction of the ketone product to
form sphinganine is catalyzed by 3-keto-sphinganine reductase, with NADPH as
a reactant. This is followed by acylation of sphinganine to form N-acyl sphinganine.
It is then desaturated to form ceramide. In this pathway, sphingosine itself does
not appear as an intermediate.

Self-Instructional
144 Material
Lipids

NOTES

Fig. 5.19 Biosynthesis of Sphingolipids in Animals

Ceramide is the building block for all other sphingolipids, for example,
sphingomyelin is produced by transferring phosphocholine from phosphatidylcholine.
About 15 per cent of the lipids of myelin sheath structures are made up by
cerebrosides, which are yielded by glycosylation of ceramide by sugar nucleotides.
Cerebrosides that contain one or more sialic acid (N-acetylneuraminic acid)
moieties are called gangliosides. Figure 5.20 illustrates the general form of the
biosynthetic pathway for ganglioside GM2. As shown in the figure, sugar units
from nucleotide derivatives, like UDP-N-acetylglucosamine, UDP-galactose, and
UDP-glucose are added to the developing ganglioside.
Self-Instructional
Material 145
Lipids

NOTES

Fig. 5.20 Synthesis of Glycosylceramides, Gangliosides, and


Sphingomyelins from Ceramide in Animals

Eicosanoid Biosynthesis
The ubiquitous breakdown products of phospholipids, derived from 20-carbon
fatty acids are known as Eicosanoids. Cells activate the breakdown of selected
phospholipids, in response to appropriate stimuli. Fatty acids are selectively cleaved
from the C-2 position of phospholipids by phospholipase A2. These acids are
mainly unsaturated fatty acids, one of them being arachidonic acid. The combined
actions of phospholipase C and diacylglycerol lipase also release arachidonic acid
from phospholipids. This is shown in Figure 5.21.
Self-Instructional
146 Material
Lipids

NOTES

Fig. 5.21 Eicosanoid Biosynthesis


Arachidonic acid and other polyunsaturated fatty acids are modified by
animal cells by processes like cyclization and oxygenation. This leads to production
of local hormones that act near their sites of synthesis, exerting their effects at very
low concentrations. These hormones include ProstaGlandins (PG), ThromboXanes
(Tx), leukotrienes and other hydroxyeicosanoic acids. Thromboxanes, discovered
in blood platelets (thrombocytes), are cyclic ethers with a hydroxyl group at
C-15.
All prostaglandins are cyclopentanoic acids, derived from arachidonic acid.
Prostaglandin endoperoxide synthase, which is an enzyme associated with the
endoplasmic reticulum, initiates the biosynthesis of prostaglandins. It catalyzes
oxidation and cyclization of arachidonic acid, simultaneously and is viewed to possess
two distinct activities, cyclooxygenase and peroxidase, as shown in Figure 5.22.

Fig. 5.22 Oxidation and Cyclization of Arachidonic Acid by


Prostaglandin Endoperoxide Synthase Self-Instructional
Material 147
Lipids Stimuli like histamine, epinephrine and bradykinin, proteases such as thrombin
and even serum albumin can initiate the release of arachidonate and the synthesis
or interconversion of eicosanoids. These can also be carried by tissue injury and
inflammation. Arachidonate release and eicosanoid synthesis also occur when special
NOTES inflammatory cells like monocytes and neutrophils invade injured tissues and interact
with the resident cells. Heart attack (myocardial infarction), rheumatoid arthritis
and ulcerative colitis are some examples of tissue injury in which eicosanoid
synthesis has been characterized.

Check Your Progress


9. What is hexadecanoic acid?
10. What are eicosanoids?

5.7 CHOLESTEROL SYNTHESIS IN E. COLI

Cholesterol (Refer Figure 5.23), which is the most commonly occuring steroid in
animal cells, is an important part of the cell membrane. It also serves as a precursor
to bile acids (for example, cholate, glycocholate, taurocholate) and steroid
hormones (for example, testosterone, estradiol, progesterone). Moreover, 7-
dehydrocholesterol, the immediate precursor of cholesterol, allows derivation of
vitamin D3. The primary site of cholesterol biosynthesis is liver.

Fig. 5.23 Structure of Cholesterol


Self-Instructional
148 Material
The synthesis of mevalonate from acetyl-CoA initiates the cholesterol Lipids

biosynthetic pathway in the cytosol. The first step involves formation of acetoacetyl-
CoA by the E-ketothi-olase-catalyzed Claisen condensation of two molecules of
acetyl-CoA. Thereby, 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) is formed
by union of acetyl-CoA and acetoacetyl-CoA. The next step involves catalyses of NOTES
Claisen condensationis by HMG-CoA synthase. This is followed by the rate-
limiting step, in which 3R-mevalonate is produced bythe HMG-CoA by undergoing
two NADPH-dependent reductions. Thereafter, HMG-CoA reductase, a 97-kD
glycoprotein that traverses the endoplasmic reticulum membrane with its active
site facing the cytosol,catalyses the reaction. Figure 5.24 shows the3R-mevlonate
from acetyl-CoA. Figure 5.25 shows a reaction mechanism for HMG-CoA
reductase.

Fig. 5.24 Biosynthesis of 3R-Mevalonate from Acetyl-CoA


Self-Instructional
Material 149
Lipids

NOTES

Fig. 5.25 A Reaction Mechanism for HMG-CoA Reductase

In the biosynthesis of squalene, mevalonate first gets converted to two key


5-carbon intermediates, isopentenyl pyrophosphate and dimethylallyl
pyrophosphate by four consecutive reactions. These two then join to yield farnesyl
pyrophosphate and squalene.
Pyrophosphomevalonate decarboxylase phosphorylates the 3-hydroxyl
group and the next step involves trans-elimination of the phosphate and carboxyl
groups to form the double bond in isopentenyl pyrophosphate. Isomerization of
the double bond yields the dimethylallyl pyrophosphate. Geranyl pyrophosphate
is produced by the condensation of the two 5-carbon intermediates and farnesyl
pyrophosphate is produced by addition of another 5-carbon isopentenyl group.
Pyrophosphate is released in both the steps of farnesyl pyrophosphate production.
The hydrolysis of this pyrophosphate drives these reactions forward. Also, the
isoprene units are linked in a head-to-tail fashion to form farnesyl pyrophosphate.
This is the general rule in biosynthesis of molecules involving isoprene linkages. An
exception to this rule occurs in the next step when two farnesyl pyrophosphates
Self-Instructional
150 Material
are joined to produce squalene in a tail-to-tail condensation. Figure 5.26 shows Lipids

the conversion of mevalonate to squalene.

NOTES

Fig. 5.26 Conversion of Mevalonate to Squalene

Squalene is converted to squalene-2, 3-epoxide by squalene


monooxygenase, an enzyme bound to the endoplasmic reticulum. This reaction
requires oxygen and a cytosolic protein called soluble protein activator. It also
employs FAD and NADPH as coenzymes. The second reaction, which involves
a succession of 1, 2 shifts of hydride ions and methyl groups, is catalyzed by a
second ER membrane enzyme, called 2, 3-oxidosqualene lanosterol cyclase.
Even though the structure of lanosterol and cholesterol looks similar, the
conversion of lanosterol to cholesterol requires 20 steps to be followed (Refer
Figure 5.27). The enzymes responsible for all this are associated with the
endoplasmic reticulum.7-dehydrocholesterol is the penultimate intermediate of
the primary pathway. An alternative pathway, also composed of many steps,
produces the intermediate desmosterol. Reduction of the double bond at C-24
yields cholesterol. Acyl-CoA:cholesterol AcylTransferases (ACAT ) synthesizes
cholesterol ester, a principal form of circulating cholesterol on the cytoplasmic
face of the endoplasmic reticulum.

Self-Instructional
Material 151
Lipids

NOTES

Fig. 5.27 Conversion of Squalene to Squalene-2, 3-epoxide and


Lanosterol to Cholesterol

Check Your Progress


11. What is cholesterol?
12. What is the primary site of cholesterol biosynthesis?
Self-Instructional
152 Material
Lipids
5.8 ANSWERS TO CHECK YOUR PROGRESS
QUESTIONS

1. Simple lipids include oils and fats. These are esters of fatty acids and glycerol. NOTES
2. Cholesterol is found in egg yolk, dairy products, and animal tissues. A
constituent of bile acids and a precursor of Vitamin D.
3. Sex hormones are found in ovaries and testes.
4. The Solid Fat Content (SFC) of a lipid influences many of its sensory and
physical properties, such as spreadability, firmness, mouthfeel, processing
and stability.
5. A small amount of fat is placed in a capillary tube and heated at a controlled
rate. The temperature at which the fat just starts to move downwards due
to its weight is called the ‘slip point’.
6. Saturated fats do not have double bond in their fatty acids.
7. The fats, having more than one double bond present in their fatty acids are
called polyunsaturated fats. The fatty acid present in them is known as
PolyUnsaturated Fatty Acid (PUFA).
8. Omega-9 fatty acids are also called n- 9 fatty acids because they contain
carbon – carbon double bond in n-9 position from methyl end. Oleic acid
and Erucic acid are significant n-9 fatty acids.
9. The fatty acid with 16 carbon atoms is systematically named as hexadecanoic
acid.
10. The ubiquitous breakdown products of phospholipids, derived from 20-
carbon fatty acids are known as Eicosanoids.
11. Cholesterol which is the most commonly occurring steroid in animal cells, is
an important part of the cell membrane. It also serves as a precursor to bile
acids (for example, cholate, glycocholate, taurocholate) and steroid
hormones (for example, testosterone, estradiol, progesterone).
12. Liver is the primary site of cholesterol biosynthesis.

5.9 SUMMARY

x Lipids are classified as follows: Simple lipids, Compound lipids, Terpenoids


and Steroids, Derived lipids.
x Lipids are usually defined as those components that are soluble in organic
solvents (such as ether, hexane or chloroform), but are insoluble in water.
This group of substances includes triacylglycercols, diacylglycercols,
rnonoacylglycercols, free fatty acids, phospholipids, sterols, caretonoids
and vitamins A and D.
Self-Instructional
Material 153
Lipids x Simple lipids include oils and fats. These are esters of fatty acids and glycerol.
x Compound lipids include phospholipids, sphingolipids, glycolipids and
sulpholipids.
NOTES x Each type of fat has a different profile of lipids present which determines
the precise nature of its nutritional and physiochemical properties.
x Fats are classified according to the presence of unsaturation, that is, presence
of double bonds. They are mainly of the following two types: saturated fats
and unsaturated fats.
x Saturated fats do not have double bond in their fatty acids.
x Unsaturated fats contain double bonds in their fatty acids. Since they are
able to take up hydrogen in them, they are called unsaturated fats.
x Omega fatty acids have double bond from the methyl side, that is, opposite
side of the carboxyl functional group. These are of the following three types:
Omega-3 fatty acids, Omega-6 fatty acids, and Omega-9 fatty acids.
x Fatty acyls is a generic term used to describe fatty acids and their conjugates
and derivatives.
x Monounsaturated fatty acids contain only one double bond between the
carbon atoms in a molecule of fatty acid. These include oleic acid, palmitoleic
acid, myristoleic acid, etc.
x Neural tissues contain high-levels of Sphingolipids, which are ubiquitous
components of eukaryotic cell membranes.
x The ubiquitous breakdown products of phospholipids, derived from 20-
carbon fatty acids are known as Eicosanoids.
x Cholesterol, which is the most commonly occurring steroid in animal cells,
is an important part of the cell membrane. It also serves as a precursor to
bile acids (for example, cholate, glycocholate, taurocholate) and steroid
hormones (for example, testosterone, estradiol, progesterone).

5.10 KEY WORDS

x Lipids: A lipid is a biomolecule that is soluble in nonpolar solvents.


x Non-polar solvents: Non-polar solvents are typically hydrocarbons used
to dissolve other naturally occurring hydrocarbon lipid molecules that do
not (or do not easily) dissolve in water, including fatty acids, waxes, sterols,
fat-soluble vitamins (such as vitamins A, D, E, and K).
x Fatty acids: A carboxylic acid consisting of a hydrocarbon chain and a
terminal carboxyl group, especially any of those occurring as esters in fats
and oils.
Self-Instructional
154 Material
x Saturated fat: A saturated fat is a type of fat in which the fatty acid chains Lipids

have all or predominantly single bonds. A fat is made of two kinds of smaller
molecules: glycerol and fatty acids. Fats are made of long chains of carbon
atoms.
NOTES
x Unsaturated fat: An unsaturated fat is a fat or fatty acid in which there is at
least one double bond within the fatty acid chain.
x Biosynthesis: The production of complex molecules within living organisms
or cells.

5.11 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. What are simple lipids?
2. Discuss briefly about compound lipids.
3. Write a short note on omega fatty acids.
4. What do you understand by biosynthesis of complex lipids?
5. Briefly explain sphingolipid biosynthesis.
6. What are eicosanoid biosynthesis?
Long Answer Questions

1. Explain properties of lipids.


2. Discuss the types of fats.
3. Describe fatty acyls in detail.
4. Discuss properties of saturated fatty acids.
5. Discuss the properties of unsaturated fatty acids.
6. Explain in detail about phospholipid.
7. Discuss cholesterol synthesis in E coli.

5.12 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Self-Instructional
Material 155
Lipids Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
NOTES
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
156 Material
Lipid Metabolism

UNIT 6 LIPID METABOLISM


Structure NOTES
6.0 Introduction
6.1 Objectives
6.2 Lipids
6.2.1 Lipid Metabolism
6.2.2 Lipid Peroxidation
6.3 Fatty Acid: Oxidation
6.4 Fatty Acid Metabolism
6.5 Answers to Check Your Progress Questions
6.6 Summary
6.7 Key Words
6.8 Self Assessment Questions and Exercises
6.9 Further Readings

6.0 INTRODUCTION

Metabolism is a chemical processes that occur within a living organism in order to


maintain life. Lipid metabolism is the synthesis and degradation of lipids in cells,
involving the break down or storage of fats for energy. These fats are obtained
from consuming food and absorbing them or they are synthesized by an animal's
liver. Lipogenesis is the process of synthesizing these fats. Fatty acid metabolism
consists of catabolic processes that generate energy, and anabolic processes that
create biologically important molecules. Fatty acids are a family of molecules
classified within the lipid macronutrient class. Lipid peroxidation is the oxidative
degradation of lipids. It is the process in which free radicals steal electrons from
the lipids in cell membranes, resulting in cell damage.
In this unit, you will study about the metabolism of lipids and fatty acids.

6.1 OBJECTIVES

After going through this unit, you will be able to:


x Discuss metabolism of lipids
x Discuss metabolism of fatty acids
x Understand D, E, Zoxidation of fatty acids
x Describe lipid peroxidation

Self-Instructional
Material 157
Lipid Metabolism
6.2 LIPIDS

Lipids containing polyunsaturated fatty acids are susceptible to free radical-initiated


NOTES oxidation and can participate in chain reactions that increase damage to
biomolecules.
Characteristically the lipids are divided into two major groups: apolar and
polar. Triglycerides (apolar), stored in various cells, but especially in adipose (fat)
tissue, are usually the main form of energy storage in mammals. Polar lipids are
structural components of cell membranes, where they participate in the formation
of the permeability barrier of cells and subcellular organelles in the form of a lipid
bilayer. The major lipid type defining this bilayer in almost all membranes is glycerol-
based phospholipid. The importance of the membrane lipid physical (phase) state
is evidenced by the fact that lipids may control the physiological state of a membrane
organelle by modifying its biophysical aspects, such as the polarity and permeability.
Lipids also have a key role in biology as signaling molecules.
Lipids comprise about 15 – 20 per cent of body weight of human beings, with
TriacylGlycerol or TriGlycerides (TAG or TG or Neutral fat or fat depot) constituting
about 85–90 per cent of the total proportion. This fat is stored in the body in the
form of adipose tissue. It serves as the prime form of energy, due to the following
reasons:
x It exists in concentrated form and provides cal /gm of energy, whereas
carbohydrate and protein gives only 4cal /gm.
x While triacylglycerols are non- polar and hydrophobic in nature,
carbohydrates and proteins are polar in nature. This means that while the
former are stored without water, the latter contain large amount of water.
x The triacylgycerol is cleaved by enzyme pancreatic lipase into Free Fatty
Acid (FFA) and glycerol. This is known as lipolysis(lysis or breakdown of
lipids).
The glycerol is converted to glycerol-3-phosphate, in presence of enzyme
glycerol kinase with utilization of one ATP molecule. The conversion of glycerol to
glycerol-3-phosphate occurs in liver and not in adipose tissue. Thus, the glycerol
phosphate formed can either be utilized for the synthesis of triacylglycerol or
phospholipids or it can form dihyroxyacetone phosphate in presence of enzyme
glycerol-3-phosphate ehydrogenase with release of one NADH + H+.
6.2.1 Lipid Metabolism
Lipid metabolism is the synthesis and degradation of lipids in cells, involving the
break down or storage of fats for energy. These fats are obtained from consuming
food and absorbing them or they are synthesized by an animal’s liver. Lipogenesis
is the process of synthesizing these fats. The majority of lipids found in the human
Self-Instructional
158 Material
body from ingesting food are triglycerides and cholesterol. Other types of lipids Lipid Metabolism

found in the body are fatty acids and membrane Lipids. Lipid metabolism is often
considered the digestion and absorption process of dietary fat; however, there are
two ways organisms can use fats to obtain energy: consumed dietary fats and
storage fat. Vertebrates and humans use both methods of fat usage as their sources NOTES
of energy for organs such as the heart to function. Since lipids are hydrophobic
molecules, they need to be solubilized before their metabolism begin. Lipid
metabolism often begins with hydrolysis, which occurs with the help of various
enzymes in the digestive system. Lipid metabolism does exist in plants, though the
processes differ in some ways when compared to animals. The second step after
the hydrolysis is the absorption of the fatty acids into the epithelial cells of the
intestinal wall. In the epithelial cells, fatty acids are packaged and transported to
the rest of the body.
Lipid Digestion
Digestion is the first step to lipid metabolism, and it is the process of breaking the
triglycerides down into smaller monoglyceride units with the help of lipase enzymes.
Digestion of fats begin in the mouth through chemical digestion by lingual lipase.
Ingested cholesterol is not broken down by the lipases and stays intact until it
enters the epithelium cells of small intestine. Lipids then continue to the stomach
where chemical digestion continues by gastric lipase and mechanical digestion
begins (Peristalsis). The majority of lipid digestion and absorption, however, occurs
once the fats reach the small intestines. Chemicals from the pancreas (pancreatic
lipase family and Bile salt-dependent lipase) are secreted into the small intestines
to help breakdown the triglycerides, along with further mechanical digestion, until
they are individual fatty acid units able to be absorbed into the small
intestine’s epithelial cells. It is the pancreatic lipase that is responsible for signaling
for the hydrolysis of the triglycerides into separate free fatty acids and glycerol
units.
Lipid Absorption
The second step in lipid metabolism is the absorption of fats. Absorption of fats
occurs only in the small intestines. Once the triglycerides are broken down into
individual fatty acids and glycerols, along with cholesterol, they will aggregate into
structures called micelles. Fatty acids and monoglycerides leave the micelles and
diffuse across the membrane to enter the intestinal epithelial cells. In the cytosol of
epithelial cells, fatty acids and monoglycerides are recombined back into
triglycerides. In the cytosol of epithelial cells, triglycerides and cholesterol are
packaged into bigger particles called chylomicrons which are amphipathic structures
that transport digested lipids. Chylomicrons will travel through the blood stream
to enter adipose and other tissues in the body.
Self-Instructional
Material 159
Lipid Metabolism Transporting Lipids
Due to the hydrophobic nature of membrane lipids, triglycerides and cholesterols,
they require special transport proteins known as lipoproteins. The amphipathic
NOTES structure of lipoproteins allows the tryglycerols and cholesterol to be transported
through the blood. Chilomicrons are one sub-group of lipoproteins which carry
the digested lipids from small intestine to the rest of the body. The varying densities
between the types of lipoproteins are characteristic to what type of fats they
transport. For example, Very-Low-Density-Lipoproteins (VLDL) carry the
synthesized triglycerides by our body and Low-Density-Lipoprotein (LDL)
transport cholesterol to our peripheral tissues. A number of these lipoproteins are
synthesized in the liver, but not all of them originate from this organ.
Lipid Catabolism

Once the chylomicrons (or other lipoproteins) travel through the tissues, these
particles will be broken down by lipoprotein lipase in the luminal surface of
endothelial cells in capillaries to release tryglycerides. Tryglycerides will get broken
down into fatty acids and glycerol before entering cells and remaining cholesterol
will again travel through the blood to the liver.

Fig. 6.1 Lipid Catabolism

In the cytosol of the cell (for example a muscle cell), the glycerol will be
converted to glyceraldehyde 3-phosphate, which is an intermediate in the glycolysis,
to get further oxidized and produce energy. However, the main steps of fatty
acids catabolism occur in the mitochondria. Long chain fatty acids (more than 14
carbon) need to be converted to Fatty acyl-CoA in order to pass across the
Self-Instructional
160 Material
mitochondria membrane. Fatty acid catabolism begins in the cytoplasm Lipid Metabolism

of cells as Acyl-CoA synthetase uses the energy from cleavage of an ATP to


catalyze the addition of Coenzyme A to the fatty acid. The resulting Acyl-CoA cross
the mitochondria membrane and enter the process of beta oxidation. The main
NOTES
products of the beta oxidation pathway are Acetyl-CoA (which is used in the Citric
acid cycle to produce energy), NADH and FADH. The process of beta oxidation
requires the following enzymes: Acyl CoA dehydrogenase, Enoyl-CoA
hydratase, 3-hydroxyacyl-CoA dehydrogenase, and 3-ketoacyl-CoA thiolase. The
diagram to the left shows how fatty acids are converted into Acetyl-CoA. The
overall net reaction, using palmitoyl CoA as a model substrate is:
7 FAD + 7 NAD+ + 7 CoASH + 7 H2O + H(CH2CH2)7CH2CO-SCoA
o 8 CH3CO-SCoA + 7 FADH2 + 7 NADH + 7 H+
Lipid Biosynthesis

In addition to dietary fats, storage lipids stored in the adipose tissues are one of
the main sources of energy for living organisms. Triacylglycerols, lipid membrane
and cholesterol can be synthesized by the organisms through various pathways.
Membrane Lipid Biosynthesis

There are two major classes of membrane lipids: glycerophospholipids and


sphingolipids. Although many different membrane lipids are synthesized in our body,
pathways share the same pattern. The first step is synthesizing the backbone
(sphingosine or glycerol), the second step is addition of fatty acids to the back
bone to make phosphatidic acid. Phosphatidic acid is further modified with the
attachment of different hydrophilic head groups to the back bone. Membrane lipid
biosynthesis occurs in the endoplasmic reticulum membrane.
Triglyceride Biosynthesis

The phosphatidic acid is also a precursor for triglyceride biosynthesis. Phosphatidic


acid phosphotase catalyzes the conversion of phosphatidic acid to diacylglyceride,
which will be converted to triacylglyceride by acyltransferase. Tryglyceride
biosynthesis occurs in the cytosol.
Fatty Acid Biosynthesis

The precursor for fatty acids is acetyl CoA and it occurs in the cytosol of the
cell. The overall net reaction, using palmitate as a model substrate is:
8 Acetyl-coA + 7 ATP + 14 NADPH + 6H+ o palmitate + 14 NADP+ +
6H2O + 7ADP + 7Pi

Self-Instructional
Material 161
Lipid Metabolism Cholesterol Biosynthesis

Cholesterol can be made from acetyl-CoA through a multiple-step pathway known


as Isoprenoid Pathway. Cholesterols are essential because they can be modified
NOTES to form different hormones in the body such as progesteron 70% of Choleserol
biosynthesis occurs in the cytosol of liver cells.
6.2.2 Lipid Peroxidation
Lipid peroxidation, which leads to lipid hydroperoxide formation often, occurs in
response to oxidative stress. Basically, the lipid peroxidation is considered as the
main molecular mechanisms involved in the oxidative damage to cell structures
and in the toxicity process that lead to cell death. First, lipid peroxidation was
studied for food scientists as a mechanism for the damage to alimentary oils and
fats, however other researchers considered that lipid peroxidation was the
consequence of toxic metabolites, for example CCl4, that produced highly reactive
species, disruption of the intracellular membranes and cellular damage. Lipid
peroxidation is a complex process known to occur in both plants and animals. It
involves the formation and propagation of lipid radicals, the uptake of oxygen, a
rearrangement of the double bonds in unsaturated lipids and the eventual destruction
of membrane lipids, with the production of a variety of breakdown products,
including alcohols, ketones, alkanes, aldehydes and ethers.
In pathological situations the reactive oxygen and nitrogen species are
generated at higher than normal rates, and as a consequence, lipid peroxidation
occurs with a-tocopherol deficiency. In addition to containing high concentrations
of polyunsaturated fatty acids and transition metals, biological membranes of cells
and organelles are constantly being subjected to various types of damage. The
mechanism of biological damage and the toxicity of these reactive species on
biological systems are currently explained by the sequential stages of reversible
oxidative stress and irreversible oxidative damage. Oxidative stress is understood
as an imbalance situation with increased oxidants or decreased antioxidants. The
concept implies the recognition of the physiological production of oxidants (oxidizing
free-radicals and related species) and the existence of operative antioxidant
defenses.
Lipid peroxidation is a chain reaction initiated by the hydrogen abstraction
or addition of an oxygen radical, resulting in the oxidative damage of
PolyUnsaturated fatty acids (PUFA). Since polyunsaturated fatty acids are more
sensitive than saturated ones, it is obvious that the activated methylene (RH) bridge
represents a critical target site. The presence of a double bond adjacent to a
methylene group makes the methylene C-H bond weaker and therefore the
hydrogen in more susceptible to abstraction. This leaves an unpaired electron on
Self-Instructional
162 Material
the carbon, forming a carbon-centered radical, which is stabilized by a molecular Lipid Metabolism

rearrangement of the double bonds to form a conjugated diene which then combines
with oxygen to form a peroxyl radical. The peroxyl radical is itself capable of
abstracting a hydrogen atom from another polyunsaturated fatty acid and so of NOTES
starting a chain reaction.

Fig. 6.2 Lipid Peroxidation Process

Check Your Progress


1. What are polar lipids?
2. What is lipolysis?
3. What is lipid metabolism?
4. What is lipid peroxidation?

6.3 FATTY ACID: OXIDATION

After lipolysis, free fatty acid is released into blood and transported in blood in a
bound form to albumin. Then, the free fatty acid enters the cell and is oxidized.
The fatty acids are oxidized mainly by E-oxidation . E-oxidation is the fatty acid
oxidation of E-carbon (second carbon from the function group) atom. In E-
oxidation, acetyl CoA is removed from fatty acid in series. Thus, the acetyl CoA
formed enters into TCA cycle for release of energy. The fatty acid oxidation has
the following three major stages:
1. Formation of Acyl CoA: This occurs in cytosol. The enzyme thiokinase
or acyl CoA synthetase activates fatty acids to acyl CoA. This reaction
requires ATP, coenzyme A and Mg2+. During this activation process, one
ATP is converted AMP and Inorganic Pyrophosphate (PPi). The PPi is
hydrolysed to release two Phosphate (Pi) in presence of enzyme
pyrophosphatase.
2. Transport of Acyl CoA into Mitochondria: The Acyl Coa cannot be
transported into mitochondria, because the mitochondrial membrane is
impermeable to fatty acid. For transportation of Acyl coA, a special carnitine
carrier is required. The acyl group of acyl CoA gets attached to carnitine
and reaction occurs in presence of enzyme carnitine Acyltransferase I. The
Self-Instructional
Material 163
Lipid Metabolism acyl-carnitine complex is then transported across mitochondrial membrane
to matrix by a carrier protein in the membrane. After entering the matrix, the
Acyl-carnitine complex is cleaved into acyl CoA and carnitine by enzyme
Carnitine–acyl transferase II.
NOTES
3. E -Oxidation: This occurs in matrix of mitochondria. Following are the
steps of E-oxidation:
x Formation of Double Bond between D and E-Carbon, that is, 2
and 3 Carbon: Acyl CoA dehydrogenase catalyse dehydrogenation
of AcylCoA in presence of FAD. In this reaction, a double bond is
formed in between D and E- carbon.
x Formation of E-Hydroxyacyl CoA: The double bond is hydrated
to E-hydroxyacyl CoA by enzyme Enoyl CoA hydratase.
x Formation of E -ketoacyl CoA: E-hydroxyacyl CoA undergoes
oxidation reaction and forms E-Ketoacyl CoA in presence of enzyme
E-hydroxyacyl CoA dehydrogenase.
x Formation of Acetyl CoA: This is the final reaction in which two
carbons are removed from Acyl CoA in the form of Acetyl CoA. This
occurs in presence of E–ketoacyl CoA thiolase. The remaining acyl
coA contain two carbons less than the original one. The oxidation
process continues till all acyl CoA is converted to Acyl CO.
Therefore, the net reaction is:
Cn Acyl CoA + FAD + NAD + + H2 O +CoASH o C(n-2) AcylCoA
+ Acetyl CoA + FADH2 + NADH + H+
E -Oxidation of Unsaturated Fatty Acids
E-oxidation of unsaturated fatty acids poses a problem since the location of a cis
bond can prevent the formation of a trans-'2 bond. Additional two enzymes are
used to solve this problem. E-oxidation normally occurs until the acyl CoA is not
an appropriate substrate for acyl CoA dehydrogenase or enoyl CoA hydratase
for any conformation of the hydrocarbon. The steps of E-oxidation, in this case
are as follows:
x Formation of Trans-'2 Bond from Cis-'3 bond: If the acyl CoA contains
a cis-'3 bond, then cis-'3- Enoyl CoA isomerase converts the bond to a
trans-'2 bond, which is a regular substrate.
x Formation of Cis-'3-Enoyl CoA from Cis-'4 Double Bond: If the
acyl CoA contains a cis-'4 double bond, then its dehydrogenation yields a
2, 4-dienoyl intermediate, which is not a substrate for enoyl CoA hydratase.
However, the enzyme 2,4 Dienoyl CoA reductase reduces the intermediate,
using NADPH, into cis-'3-enoyl CoA. As in the above case, this compound
is converted into a suitable intermediate by 3,2-Enoyl CoA isomerase.
Therefore, it could be concluded that odd-numbered double bonds are
Self-Instructional
164 Material
handled by the isomerase an even-numbered double bonds are handled by Lipid Metabolism

the reductase (which creates an odd numbered double bond) and the
isomerase.
E -Oxidation of Odd-Numbered Chains NOTES
Fatty acids, possessing an odd number of carbons are usually found in plant lipids
and certain marine organisms. Several ruminant animals form large quantities of 3-
carbon propionate. The oxidations of odd and even- numbered chains are similar,
with end products as propionyl-CoA and acetyl-CoA.
Propionyl-CoA is initially carboxylated with the help of a bicarbonate ion
into D-stereoisomer of methylmalonyl-CoA, in a reaction involving a biotin co-
factor, ATP and the enzyme propionyl-CoA carboxylase. The bicarbonate ion’s
carbon is added to the middle carbon of propionyl-CoA, to form a D-
methylmalonyl-CoA. However, the D conformation is enzymatically converted
into the L conformation by methylmalonyl-CoA epimerase. It then goes through
intra-molecular rearrangement, the catalysis of which is done by methylmalonyl-
CoA mutase (requires coenzyme-B12 as its coenzyme) to form succinyl-CoA.
The succinyl-CoA can then get into the citric acid cycle. Since it is not possible to
fully metabolize in the citric acid cycle, the products of its incomplete reaction
should be eliminated in a process known as cataplerosis. This permits regeneration
of the citric acid cycle intermediates, probably a vital process in some metabolic
diseases.
Oxidation in Peroxisomes
A Fatty acid gets oxidized in peroxisomes, when the fatty acid chains are too long
to be managed by the mitochondria. However, the oxidation terminates at octanyl
CoA. It is known that extremely long chain (greater than C-22) fatty acids go
through initial oxidation in peroxisomes followed by mitochondrial oxidation.
One important difference is that oxidation in peroxisomes is not combined
with ATP synthesis. On the other hand, the high-potential electrons are shifted to
O2, which produces H2O2. The enzyme catalase, present only in peroxisomes,
converts the hydrogen peroxide into water and oxygen.
Peroxisomal E-oxidation too needs enzymes specific to the peroxisome
and very long fatty acids. There are three main differences between the enzymes
used for mitochondrial and peroxisomal E-oxidation. These differences are as
follows:
x E-oxidation in the peroxisome needs the utilization of a peroxisomal carnitine
acyltransferase (instead of carnitine acyltransferase I and II used by the
mitochondria) for transporting the activated acyl group into the peroxisome.
x The initial oxidation step in the peroxisome undergoes catalysis with the
help of the enzyme, acyl CoA oxidase.
Self-Instructional
Material 165
Lipid Metabolism x The E-ketothiolase that helps in peroxisomal E-oxidation has altered
substrate specificity, which differs from the mitochondrial E-ketothiolase.
High fat diet and administration of hypolipidemic drugs, such as, clofibrate induce
peroxisomal oxidation.
NOTES
Alpha Oxidation (D
D-Oxidation)
Alpha oxidation (D-oxidation) is a process which enables the breaking down of
certain fatty acids by removing only one carbon from the carboxyl end. In humans,
alpha-oxidation is brought into peroxisomes to break down dietary phytanic acid,
which cannot undergo beta-oxidation because of its E-methyl branch, into pristanic
acid.
Omega Oxidation (Z Z-Oxidation)
Omega oxidation is an alternative pathway to beta oxidation. Fatty acid is
metabolized in some species of animals by this pathway. This pathway, instead of
E carbon, involves the oxidation of the Z carbon (the carbon most distant from
the carboxyl group of the fatty acid). This process is normally a minor catabolic
pathway for medium-chain fatty acids (10-12 carbon atoms). It becomes more
important when E oxidation is defective.
In vertebrates, the enzymes for Z-oxidation are situated in the endoplasmic
reticulum of liver and kidney cells, in place of in the mitochondria similar to E-
oxidation. The steps of the process are as follows:
x Hydroxylation: The first step initiates a hydroxyl group onto the Z carbon
in the existence of the enzyme mixed function oxidase. The oxygen for the
group is derived from molecular oxygen in a complex reaction involving
cytochrome P450 and the electron donor NADPH.
x Oxidation of the Hydroxyl Group: The next step is the oxidation of the
hydroxyl group to an aldehyde by NAD by enzyme alcohol dehydrogenase+.
x Oxidation of the Aldehyde Group: The third step is the oxidation of the
aldehyde group to a carboxylic acid by NAD+ by enzyme aldehyde
dehydrogenase. A fatty acid with a carboxyl group at each end is produced
as a result.
After the three steps, any end of the fatty acid can be attached to coenzyme A.
The molecule can get into the mitochondrion and undergo D oxidation. The end
products after successive oxidation include succinic acid that can get into the citric
acid cycle and adipic acid.

Check Your Progress


5. What is oxidation?
6. What is alpha oxidation?
Self-Instructional
166 Material
Lipid Metabolism
6.4 FATTY ACID METABOLISM

Fatty acid metabolism consists of catabolic processes that generate energy, and
anabolic processes that create biologically important molecules (triglycerides, NOTES
phospholipids, second messengers, local hormones and ketone bodies). Fatty
acids are a family of molecules classified within the lipid macronutrient class. One
role of fatty acids in animal metabolism is energy production, captured in the form
of Adenosine TriphosPhate (ATP). When compared to other macronutrient classes
(carbohydrates and protein), fatty acids yield the most ATP on an energy per gram
basis, when they are completely oxidized to CO2 and water by beta oxidation and
the citric acid cycle. Fatty acids (mainly in the form of triglycerides) are therefore
the foremost storage form of fuel in most animals, and to a lesser extent in plants.
In addition, fatty acids are important components of the phospholipids that form
the phospholipid bilayers out of which all the membranes of the cell are constructed
(the cell wall, and the membranes that enclose all the organelles within the cells,
such as the nucleus, the mitochondria, endoplasmic reticulum, and the Golgi
apparatus). Fatty acids can also be cleaved, or partially cleaved, from their chemical
attachments in the cell membrane to form second messengers within the cell, and
local hormones in the immediate vicinity of the cell. The prostaglandins made from
arachidonic acid stored in the cell membrane, are probably the most well-known
group of these local hormones.
Fatty Acid Catabolism
Fatty acids are released, between meals, from the fat depots in adipose tissue,
where they are stored as triglycerides, as follows:
x Lipolysis, the removal of the fatty acid chains from the glycerol to which
they are bound in their storage form as triglycerides (or fats), is carried out
by lipases. These lipases are activated by high epinephrine and glucagon
levels in the blood (or norepinephrine secreted by sympathetic nerves in
adipose tissue), caused by declining blood glucose levels after meals, which
simultaneously lowers the insulin level in the blood.
x Once freed from glycerol, the free fatty acids enter the blood, which
transports them, attached to plasma albumin, throughout the body.
x Long chain free fatty acids enter the metabolizing cells (i.e., most living cells
in the body except red blood cells and neurons in the central nervous system)
through specific transport proteins, such as the SLC27family fatty acid
transport protein. Red blood cells do not contain mitochondria and are
therefore incapable of metabolizing fatty acids; the tissues of the central
nervous system cannot use fatty acids, despite containing mitochondria,
because long chain fatty acids (as opposed to medium chain fatty acids
cannot cross the blood brain barrier into the interstitial fluids that bathe these
cells.
Self-Instructional
Material 167
Lipid Metabolism x Once inside the cell long-chain-fatty-acid—CoA ligase catalyzes the reaction
between a fatty acid molecule with ATP (which is broken down to AMP and
inorganic pyrophosphate) to give a fatty acyl-adenylate, which then reacts
with free coenzyme A to give a fatty acyl-CoA molecule.
NOTES
x In order for the acyl-CoA to enter the mitochondrion the carnitine shuttle is
used:
R Acyl-CoA is transferred to the hydroxyl group of carnitine by carnitine
palmitoyltransferase I, located on the cytosolic faces of
the outer and inner mitochondrial membranes.
R Acyl-carnitine is shuttled inside by a carnitine-acylcarnitine translocase,
as a carnitine is shuttled outside.
R Acyl-carnitine is converted back to acyl-CoA by carnitine
palmitoyltransferase II, located on the interior face of the inner
mitochondrial membrane. The liberated carnitine is shuttled back to
the cytosol, as an acyl-CoA is shuttled into the matrix.
x Beta oxidation, in the mitochondrial matrix, then cuts the long carbon chains
of the fatty acids (in the form of acyl-CoA molecules) into a series of two-
carbon (acetate) units, which, combined with co-enzyme A, form molecules
of acetyl CoA, which condense with oxaloacetate to form citrate at the
‘beginning’ of the citric acid cycle. It is convenient to think of this reaction
as marking the ‘starting point’ of the cycle, as this is when fuel - acetyl-CoA
- is added to the cycle, which will be dissipated as CO2 and H2O with the
release of a substantial quantity of energy captured in the form of ATP,
during the course of each turn of the cycle.
Briefly, the steps in beta oxidation (the initial breakdown of free fatty acids
into acetyl-CoA) are as follows:
R Dehydrogenation by acyl-CoA dehydrogenase, yielding 1 FADH2
R Hydration by enoyl-CoA hydratase
R Dehydrogenation by 3-hydroxyacyl-CoA dehydrogenase, yielding
1 NADH + H+
R Cleavage by thiolase, yielding 1 acetyl-CoA and a fatty acid that has
now been shortened by 2 carbons (forming a new, shortened acyl-
CoA)
This beta oxidation reaction is repeated until the fatty acid has been
completely reduced to acetyl-CoA or, in, the case of fatty acids with odd
numbers of carbon atoms, acetyl-CoA and 1 molecule of propionyl-
CoAper molecule of fatty acid. Each beta oxidative cut of the acyl-CoA
molecule yields 5 ATP molecules.
x The acetyl-CoA produced by beta oxidation enters the citric acid cycle in
the mitochondrion by combining with oxaloacetate to form citrate. This
Self-Instructional
168 Material
results in the complete combustion of the acetyl-CoA to CO2 and water. Lipid Metabolism

The energy released in this process is captured in the form of 1 GTP and
11 ATP molecules per acetyl-CoA molecule oxidized. This is the fate of
acetyl-CoA wherever beta oxidation of fatty acids occurs, except under
certain circumstances in the liver. NOTES
In the liver oxaloacetate can be wholly or partially diverted into
the gluconeogenic pathway during fasting, starvation, a low carbohydrate diet,
prolonged strenuous exercise, and in uncontrolled type 1 diabetes mellitus. Under
these circumstances oxaloacetate is hydrogenated to malate which is then removed
from the mitochondrion to be converted into glucose in the cytoplasm of the liver
cells, from where it is released into the blood. In the liver, therefore, oxaloacetate
is unavailable for condensation with acetyl-CoA when significant gluconeogenesis
has been stimulated by low (or absent) insulin and high glucagon concentrations in
the blood. Under these circumstances acetyl-CoA is diverted to the formation
of acetoacetate and beta-hydroxybutyrate. Acetoacetate, beta-hydroxybutyrate,
and their spontaneous breakdown product, acetone, are frequently, but confusingly,
known as ketone bodies (as they are not ‘bodies’ at all, but water-soluble chemical
substances). The ketones are released by the liver into the blood. All cells with
mitochondria can take ketones up from the blood and reconvert them into acetyl-
CoA, which can then be used as fuel in their citric acid cycles, as no other tissue
can divert its oxaloacetate into the gluconeogenic pathway in the way that this can
occur in the liver. Unlike free fatty acids, ketones can cross the blood-brain
barrier and are therefore available as fuel for the cells of the central nervous system,
acting as a substitute for glucose, on which these cells normally survive. The
occurrence of high levels of ketones in the blood during starvation, a low
carbohydrate diet, prolonged heavy exercise and uncontrolled type 1 diabetes
mellitus is known as ketosis, and, in its extreme form, in out-of-control type 1
diabetes mellitus, as ketoacidosis.
The glycerol released by lipase action is phosphorylated by glycerol kinase in
the liver (the only tissue in which this reaction can occur), and the resulting glycerol
3-phosphate is oxidized to dihydroxyacetone phosphate. The glycolytic
enzyme triose phosphate isomerase converts this compound to glyceraldehyde 3-
phosphate, which is oxidized via glycolysis, or converted to glucose
via gluconeogenesis.

Check Your Progress


7. What is the full form of ATP?
8. Which organ of the body releases ketones?

Self-Instructional
Material 169
Lipid Metabolism
6.5 ANSWERS TO CHECK YOUR PROGRESS
QUESTIONS

NOTES 1. Polar lipids are structural components of cell membranes, where they
participate in the formation of the permeability barrier of cells and subcellular
organelles in the form of a lipid bilayer.
2. The triacylgycerol is cleaved by enzyme pancreatic lipase into Free Fatty
Acid (FFA) and glycerol. This is known as lipolysis (lysis or breakdown of
lipids).
3. Lipid metabolism is the synthesis and degradation of lipids in cells, involving
the break down or storage of fats for energy.
4. The lipid peroxidation is considered as the main molecular mechanisms
involved in the oxidative damage to cell structures and in the toxicity process
that lead to cell death.
5. Oxidation is the fatty acid oxidation of -carbon (second carbon from the
function group) atom. In E-oxidation, acetyl CoA is removed from fatty
acid in series.
6. Alpha oxidation (D-oxidation) is a process which enables the breaking down
of certain fatty acids by removing only one carbon from the carboxyl end.
7. Adenosine triphosphate.
8. The ketones are released by the liver into the blood.

6.6 SUMMARY

x Lipids containing polyunsaturated fatty acids are susceptible to free radical-


initiated oxidation and can participate in chain reactions that increase damage
to biomolecules.
x Characteristically the lipids are divided into two major groups: apolar and
polar. Triglycerides (apolar), stored in various cells, but especially in adipose
(fat) tissue, are usually the main form of energy storage in mammals.
x Lipids comprise about 15 – 20 per cent of body weight of human beings,
with triacylglycerol or triglycerides (TAG or TG or Neutral fat or fat depot)
constituting about 85–90 per cent of the total proportion.
x Lipid metabolism is the synthesis and degradation of lipids in cells, involving
the break down or storage of fats for energy. These fats are obtained from
consuming food and absorbing them or they are synthesized by an animal’s
liver.
Self-Instructional
170 Material
x Digestion is the first step to lipid metabolism, and it is the process of breaking Lipid Metabolism

the triglycerides down into smaller monoglyceride units with the help of
lipase enzymes.
x Lipid peroxidation, which leads to lipid hydroperoxide formation often, NOTES
occurs in response to oxidative stress. Basically, the lipid peroxidation
is considered as the main molecular mechanisms involved in the oxidative
damage to cell structures and in the toxicity process that lead to cell
death.
x After lipolysis, free fatty acid is released into blood and transported in blood
in a bound form to albumin. Then, the free fatty acid enters the cell and is
oxidized. The fatty acids are oxidized mainly by -oxidation.
x A Fatty acid gets oxidized in peroxisomes, when the fatty acid chains are
too long to be managed by the mitochondria.
x Alpha oxidation (D-oxidation) is a process which enables the breaking
down of certain fatty acids by removing only one carbon from the carboxyl
end.
x Omega oxidation is an alternative pathway to beta oxidation. Fatty acid is
metabolized in some species of animals by this pathway. This pathway,
instead of carbon, involves the oxidation of the carbon (the carbon most
distant from the carboxyl group of the fatty acid).
x Fatty acid metabolism consists of catabolic processes that generate energy,
and anabolic processes that create biologically important molecules
(triglycerides, phospholipids, second messengers, local hormones and ketone
bodies).

6.7 KEY WORDS

x Lipids: A lipid is a biomolecule that is soluble in nonpolar solvents. Non-


polar solvents are typically hydrocarbons used to dissolve other naturally
occurring hydrocarbon lipid molecules that do not (or do not easily) dissolve
in water, including fatty acids, waxes, sterols, fat-soluble vitamins (such as
vitamins A, D, E, and K).
x Fatty acids: A carboxylic acid consisting of a hydrocarbon chain and a
terminal carboxyl group, especially any of those occurring as esters in fats
and oils.
x Oxidation: The process or result of oxidizing or being oxidized.
x Metabolism: The chemical processes that occur within a living organism
in order to maintain life.
Self-Instructional
Material 171
Lipid Metabolism
6.8 SELF ASSESSMENT QUESTIONS AND
EXERCISES

NOTES Short Answer Questions


1. Write a short note on lipid peroxidation.
2. Describe in brief the metabolism of fatty acids.
3. What is lipid catabolism?
4. Discuss fatty acid catabolism.
Long Answer Questions
1. Explain metabolism of lipids.
2. Discuss oxidation of fatty acids.
3. How is oxidation carried out in Peroxisomes.
4. Explain the metabolism of fatty acids.

6.9 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.
Self-Instructional
172 Material
Vitamins

UNIT 7 VITAMINS
Structure NOTES
7.0 Introduction
7.1 Objectives
7.2 Classification, Properties, and Functions of Vitamins
7.2.1 Fat-Soluble Vitamins
7.2.2 Water-Soluble Vitamins
7.3 Vitamins as Co-Factors and Co-Enzymes
7.4 Answers to Check Your Progress Questions
7.5 Summary
7.6 Key Words
7.7 Self Assessment Questions and Exercises
7.8 Further Readings

7.0 INTRODUCTION

Vitamin is an organic compound required as a nutrient in tiny amounts by an


organism. In other words, an organic chemical compound (or related set of
compounds) is called a vitamin when it cannot be synthesized in sufficient quantities
by an organism, and must be obtained from the diet.
Vitamins are classified by their biological and chemical activity, not their
structure. Thus, each vitamin refers to a number of vitamer compounds that all
show the biological activity associated with a particular vitamin. Such a set of
chemicals are grouped under an alphabetized vitamin generic descriptor title, such
as vitamin A, which includes the compounds retinal, retinol and four known
carotenoids. Thirteen vitamins are presently universally recognized.
In this unit, you will study about vitamins, their classification and role in
metabolism.

7.1 OBJECTIVES

After going through this unit, you will be able to:


x Analyse the significance of vitamins
x Identify various diseases occurring due to lack of vitamins
x Differentiate between fat-soluble and water-soluble vitamins
x Discuss vitamins as co-factors and co-enzymes

Self-Instructional
Material 173
Vitamins
7.2 CLASSIFICATION, PROPERTIES AND
FUNCTIONS OF VITAMINS

NOTES Vitamins are substances which are different from food. These substances are
essential for the growth of animals. Even though their requirement is in very minute
quantities, their absence causes specific deficiencies. The main sources of vitamins
are plants and bacteria.
The general features of vitamins are as follows:
x They are widespread in nature.
x All common food materials contain more than one type of vitamin.
x Mostly plants can synthesize the vitamins.
x Human body synthesizes some vitamin such as vitamin A and vitamin D.
Some members of vitamin B are synthesized by the microorganisms present
in the intestine.
x Nearly all the cells of body can store vitamins up to some extent.
x They are non-antigenic.
x They are required in very small quantities.
All vitamins can be classified into the following two groups:
x Fat-Soluble Vitamins
x Water Soluble Vitamins
7.2.1 Fat-Soluble Vitamins
The examples of fat-soluble vitamins are vitamin A, D, E and K. These vitamins
are not readily soluble in water and contain only carbon, hydrogen and oxygen. In
some animals, they play very specific roles, like vitamin Aplays a role in the formation
of visual compounds and Vitamin D absorbs calcium and phosphorus from the
vertebrate intestine. Vitamin A, E and K are terpenoids and vitamin D is steroid.
Fat-soluble vitamins can be stored in the body, for example vitamin A is
stored in liver for several months. Even though it is possible to store these vitamins,
excessive intake of these could lead to a toxic condition.
Vitamin A
Vitamin A was recognized as an essential nutritional factor by Elmer McCollumm
in 1915. After this, in 1917, Holmes isolated from fish liver oil and in 1947, Milas
it synthesized it for the first time.
Occurrence
The liver oil of fish is the best natural source of vitamin A. The shark and halibut
contain maximum amount of vitamin A and cod liver contains the lowest amount of
vitamin A. Polar bear’s liver is a concentrated source of vitamin A. The other
Self-Instructional
174 Material
major sources of vitamin A are butter, milk, eggs, carrot, pumpkins, cantaloupes, Vitamins

turnips, peppers, peas, sweet potato, papaya, tomato, apricots, peaches, plum,
cherries, mango etc. The infants have very low vitamin A content, which needs to
be supplemented by colostrums and breast milk. Vitamin A originates in the marine
algae and then passes up by the food chain to reach the larger carnivorous animals. NOTES
Figure 7.1 shows some sources of vitamin A.

Fig. 7.1 Sources of vitamin A

Structure of Vitamin A
Vitamin A is found in two forms, A1 and A2. The A1 form is given by carotenoids in
animals. This includes D, E, J- carotenoids and cryptoxanthin. A molecule of E-
carotene is made up of eight, 5 carcon isoprenoid units, linked to form a long chain
of 40 carbon atoms with an ionone ring at each end. This orange red hydrocarbon,
upon hydrolysis, gives two molecules of vitamin A1. One form ofvitamin A is a
complex primary alcohol, called retinol, whose empirical formula is C20H29OH.
The second form of vitamin A is present in fresh water fish and is called
vitamin A2. It is different from vitamin A1 asit has an additional conjugate double
bond between carbon atom 3 and 4 of the E-ionone ring. Figure 7.2 shows the
structure of vitamin A.

Fig. 7.2 Structure of Vitamin A

Properties of Vitamin A
The various properties of vitamin A are as follows:
x It is viscid, colourless oil that can be isolated by careful fractionation as pale
yellowish needles.
x It displays characteristic absorption band in the UV spectrum at 328 mµ.
x It is soluble in fat and fat solvents.
x It gets lost in very less amount when in foodstuffs are cooked, canned or
frozen.
x It is unstable in air unless protected by antioxidants including vitamin E.
Self-Instructional
Material 175
Vitamins Metabolism
In the intestine, the E-carotene is first reduced to retinal and then to retinol, which
in turn is converted to retinyl ester by way of a reaction of a fatty acid, for example
palmitic acid. This is absorbed through the lymphatic system and stored in liver.
NOTES
Since the vitamin A that circulates in the blood is in the form of reinol that bounds
to retinol binding protein (RBP), the retinyl ester is converted into retinol before it
mixes into blood.
Deficiency
Vitamin A plays several important roles in physiology and its deficiency can lead to
various disorders like nyctalopia (night blindness), xerophthalmia (scaly condition
of the membrane covering the eye), keratomalacia (softening of the cornea),
phrynoderma (toad skin, hard and horny skin) and stunted growth.
x Xerophthalmia (Xerosis): It is a major cause of blindness in children and is
characterized by drying of eyes. This happens because the lacrymal glands
become stratified and keratinized and produces tears. As a result of this, the
external surface becomes dry and dull. The bacteria do not get washed away
and the eyelids swell and become sticky. Due to this frequent exudation of
blood and severe infection of eyes happens. This results in untreated blindness.
· Keratomalacia: It is a corneal disease that occurs in the children of age 3-
4 years. In this case, the cornea loses its lusture, undergoes a necrosis and
develops a few pinpoint ulcers, which later on cause an ulcer. After the
perforation of cornea, release of aqueous humour or iris occurs. Sometimes,
the whole eyeball may shrink.
x Phrynoderma: It is a skin lesion that is characterized by follicular
hyperkeratosis. In this case, the outer part of forearm in the region of elbow
and thighs and buttocks becomes rough and spiky. In severe deficiency, the
eyes, kidney and the respiratory tract become infected.
x In the alimentary canal, the deficiency of vitamin A causes damage to the
intestinal mucosa, resulting diarrhea.
x Hypervitaminosis: Vitamin A is less toxic to animals and human beings if
taken in the large quantity. The Recommended Dietary Allowance (RDA) of
vitamin A is 5,000 IU. The children taking overdose of vitaminAshow swelling
over bones, limited motion and definite hyperosteoses. Adults taking overdose
of vitamin A show nosebleed, weakness, headache, anorexia and nausea.
Vitamin D
The existence of vitamin D was demonstrated by Elmer McCollum in 1922 for the
first time. He found that cod liver oil is effective in preventing rickets. Vitamin D is
also called sunshine vitamin or antirachitic factor because its provitamin form,
present in human skin, is easily converted into the active form by radiation with
UV. At least 10 different compounds are known to have antirachitic properties.
These are designated as D2 (Ergocalciferol), D3 (Cholecalciferol), etc.
Self-Instructional
176 Material
Occurrence Vitamins

The best source of vitamin D is the liver oil of many fish such as cod. Though egg
yolk is a very good natural source of vitamin D, milk and butter contain very less
amount of it. Cereals, vegetables and fruits also have very low amount of vitamin D.
NOTES
Structure
The transformation of ergosterol (C28H44O) to active vitamin D2 involves a series
of steps which are as follows:
Ergosterol Lumisterol Protachysterol Tachysterol Precalciferol

Toxisterol

Calciferol

SUPRASTEROL

Thus, the cholecalciferol (C22H44O) is produced from 7-dehydrocholsterol through


a series of steps, which are as follows.
7-dehydrocholsterol Lumisterol3 Tachysterol3 Precalciferol3 —
Cholecalciferol
Figure 7.3 illustrates the production process of vitamins D2 and D3.

Fig. 7.3 Production of Vitamins D2 And D3

Properties
The various properties of vitamin D are as follows:
x It is a white and odourless crystalline solid.
Self-Instructional
Material 177
Vitamins x It is soluble in fat.
x It is heat resistant.
x It is not affected by acids and alkalies.
NOTES Metabolism
The vitamin D3 is generally not needed as an external dose, because it can be
synthesized in the human body. It plays a very critical role in calcification of bones
and teeth. It also increases the absorption of calcium and phosphate into the blood.
The release of bound calcium is also stimulated by vitamin D, but only in the
parathyroid hormone.
Deficiency
The most common disease that occurs in children due to deficiency of vitamin D is
rickets. The deficiency of vitamin D in adults is known as osteomalacia.
x Rickets: It means to weaken or twist. In this disease, calcification fails to
occur in the child. The early signs of this disease are craniotabes,that is,
thickening of the outer table of the skull and thickening of the wrists and
ankles.
In case of this disease, longitudinal grooves develop posterior to rosary.
The chest protrudes in forward direction, giving it pigeon rest appearance
and the pelvic entrance is narrowed. The muscles poorly develop.
x Osteomalacia: It means softening of bones. In this disease, the bones
soften and the C\P ratio does not remain constant. The disease happens if
the body is not sufficiently exposed to sunlight. Overdose of calciferol to
children and adults leads to de-mineralization of bones. Serum concentrations
of calcium and phosphorus increase very much, which results in the metabolic
calcification of many soft tissues and the formation of renal calculi.
Vitamin E
Vitamin E was first demonstrated in vegetable oils by Evans and Mattill in 1920.
In 1936, the compound with vitamin E were isolated from wheat germ oil and
named D and E tocopherol. After that, five other tocopherols were discovered
from various cereals, wheat germ, corn oil, rice etc.
Occurrence
The tocopherols are found in many plants oil like wheat germ rice, corn, cottonseed,
soybean and peanuts. They are also present in meat, eggs, leafy plants and some
fruits, but in small amounts.
Structure
The tocopherols are the derivatives of 6-hydroxychroma containing an isoprenoid
side chain at carbon-2. The various tocopherols differ in substituent on carbon 5,
7 and 8. There are three methyl groups that are attached to the benzene ring,
which are essential for full activity. Figure 7.4 shows the structure of tocopherols.
Self-Instructional
178 Material
Vitamins

NOTES
Fig. 7.4 Structure of Tocopherols

Properties
The various properties of vitamin D are as follows:
x It is light yellow in colour.
x It can resist the heat up to 200OC.
x It can get slowly oxidized and destroyed by UV rays.
Metabolism
Tocopherols can prevent the oxidation of many other easily oxidized substances.
Vitamin A affects only the ectoderm and endoderm in the embryo, but vitamin E
affects mesoderm. Tocopherol protects the mitochondrial system from inactivation
by fat peroxides. Vitamin E deficient muscles consume high oxygen.
Deficiency
Because of deficiency of vitamin E, sterility develops in female rats and in males,
germinative epithelium of testes degenerate and the spermatozoa becomes
nonmotile. In herbivores its deficiency causes muscular dystrophy.
The minimum daily requirement of vitamin D in male is 30 IU and in female
it is 25 IU. In pregnant ladies and lactating mothers, the minimum requirement is
30 IU.
Vitamin K
In German, vitamin K means koagulations-vitamin, which denotes a group of
lipophili, hydrophobic vitamins that are needed for the posttranslational modification
of certain proteins.
In 1929, Henrick Dam investigated about this vitamin while working on
chickens. There are two naturally forms of vitamin K, K1 and K2, K1 was first
isolated in 1939 by the Dam et al from alfalfa and the K2from fish meal by Doisy
et al, also in 1939.
Occurrence
Vitamin K is mainly found in the green leafy vegetables such as alfalfa, spinach,
cabbage, swiss chard, brassica etc. Fruits and cereals are poor sources of vitamin
K. Putrefied fish meal is a rich source of vitamin K2.
Structure
The two forms of vitamin K are the derivatives of quinones and they differ in their
side chain, present at carbon 3 of the naphthoquinone ring. In vitamin K1 it is a
Self-Instructional
Material 179
Vitamins phytol redical and in vitamin K2 it is a difarnesyl radical. Vitamin K1 is found in
plants and has four isoprene units in its side chain. Vitamin K2 is found in animals
and has six isoprene subunits each with double bonds. Figure 7.5 shows the structure
of vitamin K1and K2.
NOTES

Vitamin K1

Vitamin K2

Fig. 7.5 Structure of Vitamin K1 and K2.

Properties
The properties of vitamin K are as follows:
x Vitamin K1 is yellow viscid oil and vitamin K2 is yellowish crystalline solid.
x It is sensitive to light.
x It is destroyed by the irradiation, strong acids, alkalies and oxidizing agents.
Metabolism
Vitamin K plays a very important role in blood clotting. The absorption of vitamin
K occurs in the small intestine in the form of bile.
Deficiency
The deficiency of vitamin K in the body causes loss of blood clotting. In man,
deficiency of vitamin K results in steatorrhea with decreased intestinal absorption
of lipids. It is very rare to find lack of vitamin K in human beings.
7.2.2 Water-Soluble Vitamins
Just like the fat-soluble vitamins, these vitamins contain carbon, hydrogen and
oxygen. Besides that they also contain nitrogen. These vitamins act as catalysts
and perform vital roles in many biochemical reactions.
Like the fat-soluble vitamins, the water soluble vitamins do not possess any
close chemical resemblance. The common water-soluble vitamins are vitamins B
complex (B1, B2, B3, B5, B6, B7, B9 and B12) vitamin C, Choline, inositol, p-
aminobenzoic acid, bioflavinoids and á-lipoic acid.
Self-Instructional
180 Material
Vitamin B Vitamins

These comprise nearly 13 components, B1, B2, B3 etc. All of them, except lipoic
acid are water soluble. They play a vital role in metabolism. Mostly they are
synthesized by the intestinal bacteria. NOTES
Vitamin B1 (Thiamine)
This vitamin was first isolated in 1949, by Jansen in Holland and Adolf Windaus in
Germany. Its common name is antiberiberi factor. It is also known as antineuritic
factor.
Occurrence
Thiamine is found in all plants, yeast and pork, cereals, heart, liver and kidney. It is
mainly found as a coenzyme, Thiamine PyroPhosphate (TPP) and is destroyed by
heat in neutral or alkaline media. Figure 7.6 shows some sources of vitamin B1.

Fig. 7.6 Some Sources of Vitamin B1.

Structure
The chemical structure of thiamine was determined in 1935 by Robert R. Williams
in USA. Its chemical formula is (C12H17N4OS). It is a 2,5-dimethyle1-6-
aminopyrimidine bond through a methylene linkage to 4-methyle1-5-hydroxyethyle-
thiazole. Figure 7.7 shows the chemical structure of thiamine. It is the only pyrimidine
containing an alkyl group at carbon 2 position and except penicillin it is the only
natural compound which contains a thiazole group.
Self-Instructional
Material 181
Vitamins

NOTES
Fig. 7.7 Chemical structure of Thiamine

Properties
The various properties of thiamine are as follows:
x It is a white crystalline substance.
x It is easily soluble in water and insoluble in ether and chloroform.
x It contains odour like yeast.
x It is partly lost in cooking and canning
x It is stable in acids.
Metabolism
The requirements of thiamine increase in case of fever, increased muscular activity,
pregnancy and lactation. Its absorption decreases in case of gastrointestinal or
liver diseases. The people who consume raw sea food show the symptoms of
thiamine deficiency. Thiamine is phosphorylated with ATP to form thiamine
PyroPhosphate (TPP).
Deficiency
The deficiency of vitamin B1 causes beriberi in humans and polyneuritis in animals.
x Beriberi: The initial symptoms of this disease are fatigue, apathy, irritability,
depression, drowsiness, anorexia, insomnia and abdominal discomfort. These
symptoms are generally followed by symptoms like burning paresthesis of
toes and feet, decreased tendon reflexes, loss of vibration sense, tenderness
and crampling of leg muscules, congestive heart failure etc.
Three major categories of beriberi according to symptoms are as follows:
R Dry Beriberi: In this type of beriberi, the child appears plump, but
pale, fabby, listless and dyspneic. The heart beat is rapid and the liver
is enlarged.
R Wet Beriberi: In this type of beriberi, the child is undernourished,
pale and edematous and has dyspnea, vomiting and trachycardia. The
skin appears waxy. There may be albumin and casts presents in the
urine.
R Acute Pernicious Beriberi: In this type of beriberi, lesions are found
in the heart, peripheral nerves, subcutaneous tissues and serous cavities.
The heart is enlarged and edema, serous effusions and venous
engorgement may be seen.
Often the symptoms of more than one type of beriberi appear and then it is
called mixed beriberi.
Self-Instructional
182 Material
The normal requirements of vitamin B1 for a man are 1.2-1.4 mg per day Vitamins

and for woman it is 1.0 mg per day. Pregnant and lactating women need 1.5 mg of
vitamin B1per day.
Vitamin B2 (Riboflavin) NOTES
This vitamin was first isolated from milk whey in 1879 and its chemical synthesis
was done by Richard Kuhn and Paul Karrer. It is also known as lactoflavin and is
called yellow enzyme due to its yellow colour.
Occurrence
Riboflavin is chiefly found in milk, cheese, eggs, liver, kidney, heart and brewer’s
yeast. Cow’s milk and leafy vegetables are also good sources of riboflavin. In
nature, it is a constituent of one of the two coenzymes, Flavin MonoNucleotides
(FMN) and Flavin Adenine Dinucleotide (FAD). During germination, the riboflavin
constituent increases to a great extent. Roasted or boiled meat retains nearly 75%
riboflavin in it. General cooking generally does not affect riboflavin concentration
in food. Figure 7.8 shows some sources of riboflavin.

Fig. 7.8 Some Sources of Riboflavin

Structure
The chemical formula of riboflavin is C17H20N4O6. A mol of thiamine consists of a
sugar alcohol, D-ribitol, attached to a chromogenic dimethyl isolloxazine ring at
position 9. Figure 7.9 shows the structure of riboflavin.

Fig. 7.9 Structure of Riboflavin


Self-Instructional
Material 183
Vitamins Properties
The various properties of riboflavin are as follows:
x It is a bright orange yellow crystalline powder.
NOTES x It is soluble in water and insoluble in ether and chloroform.
x It is stable in heat and acids.
x It is easily decomposed when exposed to alkalies and light.
Metabolism
Riboflavin is synthesized nearly by all green plants, bacteria, yeasts etc. It is essential
for growth and tissue respiration. In the presence of flavokinase, phosphorylation
enzymes of riboflavin convert it into FMN. FMN is further converted to FAD
upon reaction with ATP. These two coenzymes play very critical role in cell
metabolism.
Deficiency
Hepatitis patients and people using oral contraceptives or phenothiazine or
probenecid may have a deficiency of riboflavin. This deficiency leads to certain
diseases in human like chelosisare lead, glossitis, keratitis, conjunctivitis, lacrimation
and corneal vascularization etc.
While the children daily need 0.6 mg of riboflavin, adults need 1.7 mg.
During pregnancy and lactation, the daily need of riboflavin is 2.0 mg for women.
Vitamin B3
The vitamin B3 was first isolated from yeast liver in 1938 by Roger J. Williams.
He named it pentothenic acid. FritzA. Lipmann determined the structure of coenzyme
of this vitamin. This enzyme is also called filtrate factor or the yeast factor.
Occurrence
The richest source of vitamin B3 is yeast, liver and eggs. Potato, cabbage, cauliflower,
broccoli, tomatoes, peanuts, wheat bran, are some of the sources of vitamin B3.
Structure
Vitamin B3 is an amide of pantoic acids and â-alanine. Figure 7.10 shows the
structure of this vitamin.

Fig. 7.10 Structure of Vitamin B3

Properties
The various properties of Vitamin B3 are as follows:
x It is pale yellow viscous oil.
x It is soluble in water and ethyl acetate and insoluble in chloroform.
x It gets destroyed in an acidic and alkaline medium.
Self-Instructional
184 Material
Metabolism Vitamins

Green plants and several micro-organisms can synthesize pentothenic acids.


However, it cannot be synthesized by mammals. Pentothenic acids should be present
in the diet to serve as a starting point for coenzyme A. This coenzyme A should be
NOTES
in the form of acetyl co-enzyme A. The only known metabolic use of vitamin B3 is
its participation in the formation of the biologically important CoA.
Deficiency
The deficiency of vitamin B3 leads to achromatrichia (premature graying of the
hair).
Vitamin B5
Vitamin B5 is referred as nicotinic acid. An American physician named Joseph
Goldberger named it pellagra preventive factor. It is also called vitamin G in the
honour of Joseph Goldberger. In 1937, Conard Elvehjem and D. Wayne first time
recognized the role of nicotinic acid as vitamin. This was first isolated by Funk
(1911).
Occurrence
Nicotinic acid (Niacin) is widely found in plants and animals. It is found in the
yeast in a very good amount. Liver, pork, poultry and red meat are other rich
sources of this vitamin. A vegetarian diet lacks or poorly contains this vitamin.
Structure
Vitamin B5 is the simplest of all the vitamins. Figure 7.11 shows its structure.

Fig. 7.11 Nicotinic Acid

Properties
The various properties of vitamin B5are as follows:
x It is a white crystalline substance.
x It is soluble in water, ethyl alcohol and less soluble in ether and nicotinamide.
x It is heat stable.
x It is white needles of crystals when pure.
Metabolism
Nicotinamide is synthesized by the amidation of niacin acenine dincleotide and
subsequent degradation of NAD. In human, niacin is synthesized from the amino
Self-Instructional
Material 185
Vitamins acid tryptophan. A series of conversion takes place to from niacin which are shown
in Figure 7.12.

NOTES

Fig. 7.12 Metabolism of Niacin

Deficiency
Deficiency of vitamin B5 causes pellagra in man and blacktongue in dogs. The
pellagra is characterized by anorexia, lassitude, fatigue, burning sensation, numbness
and dizziness.
The blacktongue disease leads to complete loss of appetite. Pustules develop
in the inner lips and cheeks and intensive salivation and diarrhea may occur as
later symptoms.
The normal per day requirement of vitamin B5 is 8-15 mg in children, 15-
20 mg in men and 13-15 mg in women. For pregnant and lactating woman the
requirement is 20mg daily.
Self-Instructional
186 Material
Vitamin B6 Vitamins

While working on dermatitis in rats, Albert Szent-Gyorgyi found some substance


other than riboflavin and thiamine, which could cure the disease. He named the
substance vitamin B6 or antidermatitis factor or adermin. Vitamin B6 group includes NOTES
3 compounds; pyridoxine, pyridoxal and pyridoxamine.
Occurrence
Vitamin B6 are rich in rice, cereals, peas, turnip, Brussels sprouts, carrots, potatoes,
bananas, avocadoes, watermelons, egg yolk, salmon, chicken, fish, pork, beef
and yeasts.
Structure
Vitamin B6 are derivatives of pyridine and the three forms differ from each other
at position 4 of the ring. Figure 7.13 (a), (b) and (c) shows the structure of the
three compounds of vitamin B6.

(a) Pyridoxine (b) Pyridoxal (c) Pyidoxamine


Fig. 7.13 Structure of the Three Compounds of Vitamin B6

Properties
The various properties of vitamin B6are as follows:
x Pyridoxine is a white crystalline substance.
x It is soluble in water and alcohol.
x It is heat stable, but the two other allies (pyridoxal and pyridoxamine) get
destroyed in high temperature.
x It is sensitive to UV and light radiation.
Metabolism
All the three forms convert into pyridoxal 5 phosphate and act as a coenzyme in
many reactions. The phosphate of pyridoxal also acts as a carrier in the transport
of amino acid and in tryptophan. All the three forms can be detected in human
urine. It is a very important compound for nervous systems.
Deficiency
The deficiency of vitamin B6 in rats leads to development of acrodynia. In humans
its deficiency leads to convulsions, anemia, dermatitis, nausea and vomiting. The
deficiency can also occur in malabsorption such as celiac syndrome.
Self-Instructional
Material 187
Vitamins For children vitamin B6 daily requirement is .02-1.2 mg and for normal
adult it is 2.0 mg per day. But during pregnancy and lactation, per day requirement
of vitamin B6 is 2.5 mg.

NOTES Vitamin B7
Fritz Kogl in 1935 isolated a growth promoting factor from dried egg yolk and
named it biotin. Szent-Gyorgyi et al. proved that it can cure egg white injury in rats
and other animals and so called antiegg white injury factor. It is also called coenzyme
R since it is growth factor for rhizobium.
Occurrence
Biotin found in a wide range of foods sources. But there are few sources which
are rich in biotin such as egg yolk, liver and some vegetables, yeasts, milk and
molasses.
Structure
The structure of biotin is given by Vincent du Vigneaud in 1942. The formula of
biotin is C10H16O3N2S. There are two forms of biotin, that is, allobiotin and
epibiotin. Biotin also possesses optically active properties. Figure 7.14 shows the
structure of biotin.

Fig. 7.14 Biotin

Properties
The various properties of biotin are as follows:
x Its crystals are like needles.
x It is soluble in water and ethyl alcohol and insoluble in chloroform and
ether.
x It is stable in heat.
x Its melting point is 230°C.
Metabolism
The biotin sersves as a prosthetic group and carries out the carboxylation reaction.
This enzyme also brings about synthesis of fatty acids.
Deficiency
The deficiency of biotin causes dermatitis, edema and weight loss in man. The
deficiency is induced by sterilization of intestine and feeding with raw egg white. In
egg white, avidin protein is present which inactivates the biotin.
Self-Instructional
188 Material
Vitamins
Vitamin B9
In 1931, Lucy Wills identified folate as a nutrient for prevention of anemia. In
1941, the folate is was isolated from spinach leaves by Mitchell for the first time.
In 1943, Bob Stokstad isolated it in a pure crystalline form and described the NOTES
chemical nature of it.
Occurrence
The rich sources of vitamin B9are liver, kidney, tuna fish, salmon, yeast, wheat,
dates and spinach, asparagus and turnip greens, beans, peas, lentils, orange juice,
grapefruit juice, honeydew melon, banana, raspberry etc.
Structure
Folic acids contain three different units namely, glutamic acid, p-aminobenzoic
acid and pterin. Its molecular formula is C19H19O6N7. The various types of vitamin
B9 differ from each other in the number of glutamic acid group present. Figure
7.15 shows the structure of folic acid.

Fig. 7.15 Structure of Folic acid

Properties
The various properties of folic acid are as follows:
x It is a white crystalline substance.
x It is slightly soluble in water.
x It is stable to heat in alkalies and neutral substances.
Metabolism
Folic acid after reduction acts as coenzymes. TetraHydroFolic Acid (THFA) is
involved in the transfer of methyl group and in the formation of serine, methionine,
thymine, purines, choline and inosinic acid. Folic acid with ascorbic acid is related
to tyrosine metabolism.
Deficiency
The deficiency of vitamin B9 leads to anemia in chickens, achromotrichia in rats,
macrocystic anemia, leucopenia, diarrhea and edema in monkeys.
In man, folic acid deficiency leads to megaloblastic anemia, glossitis and
gastrointestinal disorders.
Self-Instructional
Material 189
Vitamins The daily requirement of this vitamin in infants is 0.1 mg, in children 0.2 mg,
in adult 0.4 mg and in pregnant and lactating woman 0.8mg.
Vitamin B12
NOTES In 1926, George Minot and George William Murphy discovered the importance
of liver in pernicious anemia and got Nobel Prize for this discovery. In 1948 Laster
Smith (England) and Edward Rickes and Karl Folkers (USA) isolated this anti
pernicious factor in crystalline form. It was named cyancobalamin or vitamin B12.
Occurrence
This vitamin can be occur in the liver, milk, meat, eggs, fish, oysters and clams. It
can be get from bacteria directly or indirectly. Cynocoalamin can only be synthesized
by the micro-organism especially anaerobic bacteria.
Structure
The structure of this vitamin is described by Dorothy Crowfoot Hodgkin. It is a
molecule having the molecular formula C63H88O14N14PCo. Cobalt is present in it
in trivalent state and centrally situated, surrounded by 4 pyrrole rings. Some other
natural compounds having the same activity as vitamin B12 have been isolated
such as hydroxocobalamin. Figure 7.16 shows the structure of vitamin B12.

Fig. 7.16 Structure of Vitamin B12

Properties
The various properties of vitamin B12are as follows:
x It is a deep red crystalline substance.
x It is soluble in water, alcohol and acetone.
x It is heat stable in neutral solution, but destroyed by heat in acidic or alkaline
solution.
Self-Instructional
190 Material
Metabolism Vitamins

Vitamin B12is converted to coenzyme B12 which is associated with many biological
reactions, such as 1, 2 shift of hydrogen atom, carrier of a methyl group,
isomerization of diacarboxylic acids etc.
NOTES
Deficiency
A deficiency of vitamin B12 occurs in the person who abstains from all animal
products. Pernicious anemia can occur the symptoms of which are irritability,
anorexia and listlessness. The adult pernicious anemia is a typical deficiency disease
in which failure of formation of RBCs occurs.
For children vitamin B12 is required 2-4 µg per day, for adult 5 µg per day
and for pregnant and lactating woman 8 µg per day.
Vitamin C (Ascorbic acid)
In 1750, vitamin C in the form of lemon was given to the British sailors to prevent
scurvy. In 1928, Albert G. Szent-Gyorgyi isolated vitamin C in crystalline form
and named it hexuronic acid. Later, it has been isolated from lemon by C. Glen
King and W.A. Waugh.
Occurrence
Vitamin C is present in all fresh fruits and vegetables. Citrus fruit such as orange,
lemon, lime etc. gooseberry, pineapple, guavas, tomatoes, melons, raw cabbage
and green pepper are the richest source of vitamin C. Vitamin C is absent in fishes,
fats and oils. Figure 7.17 shows the various sources of vitamin C.

Fig. 7.17 Some Sources of Vitamin C


Structure
Howorth describe the structure of ascorbic acid. It molecular formula is C6H8O6.
Chemically, it is 1-threo-2,4,5, 6-pentoxyhexane-2-carboxylic acid lactone. Figure
7.18 shows the structure of ascorbic acid.
Self-Instructional
Material 191
Vitamins

NOTES

Fig. 7.18 Structure of Ascorbic Acid

Properties
The various properties of ascorbic acid are as follows:
x It is a colourless, odourless, crystalline substance.
x It is slightly sour in taste.
x It is soluble in water and alcohol and insoluble in chloroform, ether and light
petroleum.
Metabolism
All mammals except man, higher plants, primates and guinea pig can synthesize
ascorbic acid. In the presence of metal ion the ascorbic acid can be oxidized to
dehydroascorbic acid. It is a more powerful electron donor than ascorbic acid.
Tissue collagen formation is the major function of ascorbic acid. It is involved in a
large number of enzymatic reactions. Its electron accepting property makes it
possible to be an anti aging substance and probably because of this property it is
deficient in old age.
Deficiency
The deficiency of vitamin C leads to scurvy, which may occur at any stage of life.
After a viable period of vitamin C deficiency, one can show symptoms like
irrtitability, tachypnea, digestive disturbance and loss of appetite. The major
symptoms which develop in later period include tenderizing of bones, edemaous
swellings, petechial hemorrhage, bleeding gums, scorbutic rosary, delayed wound
healing, sicca syndrome, anemia and pyrexia.
Water-Soluble Vitamins
Food Source Functions Deficiency Overconsumption
Symptoms Symptoms
Vitamin C (Ascorbic Acid)
Citrus fruits, Formation of collagen Bleeding gums; Nontoxic under normal
broccoli, (a component of wounds don't heal; conditions; rebound
strawberries, tissues), helps hold bruise easily; dry, scurvy when high doses
melon, green them together; wound rough skin; scurvy; discontinued; diarrhea,
pepper, healing; maintaining sore joints and bloating, cramps;
tomatoes, dark blood vessels, bones, bones; increased increased incidence of
green teeth; absorption of infections. kidney stones.
vegetables, iron, calcium, folacin;
potatoes. production of brain
hormones, immune
Self-Instructional factors; antioxidant.
192 Material Thiamin (Vitamin B )
factors; antioxidant.
Thiamin (Vitamin B1 ) Vitamins
Pork, liver, Helps release energy Mental confusion; None known.
whole grains, from foods; promotes muscle weakness,
enriched grain normal appetite; wasting; edema;
products, peas, important in function impaired growth;
meat, legumes. of nervous system. beriberi. NOTES
Riboflavin (Vitamin B2)
Liver, milk, Helps release energy Cracks at corners None known.
dark green from foods; promotes of mouth;
vegetables, good vision, healthy dermatitis around
whole and skin. nose and lips; eyes
enriched grain sensitive to light.
products, eggs.
Niacin (Nicotinamide, Nicotinic Acid)
Liver, fish, Energy production Skin disorders; Abnormal liver
poultry, meat, from foods; aids diarrhea; function; cramps;
peanuts, whole digestion, promotes weakness; mental nausea; irritability.
and enriched normal appetite; confusion;
grain products. promotes healthy skin, irritability.
nerves.
Vitamin B6 (Pyridoxine, Pyridoxal, Pyridoxamine)
Pork, meats, Aids in protein Skin disorders, None known.
whole grains metabolism, dermatitis, cracks
and cereals, absorption; aids in red at corners of
legumes, green, blood cell formation; mouth; irritability;
leafy vegetables. helps body use fats. anemia; kidney
stones; nausea;
smooth tongue.
Folacin (Folic Acid)
Liver, kidney, Aids in protein Anemia; smooth May mask vitamin B12
dark green leafy metabolism; promotes tongue; diarrhea. deficiency (pernicious
vegetables, red blood cell anemia).
meats, fish, formation; prevents
whole grains, birth defects of spine,
fortified grains brain; lowers
and cereals, homocystein levels
legumes, citrus and thus coronary
fruits. heart disease risk.
Vitamin B12
Found only in Aids in building of Pernicious anemia, None known.
animal foods: genetic material; aids anemia;
meats, liver, in development of neurological
kidney, fish, normal red blood disorders;
eggs, milk and cells; maintenance of degeneration of
milk products, nervous system. peripheral nerves
oysters, that may cause
shellfish. numbness, tingling
in fingers and toes.
Pantothenic Acid

Self-Instructional
Material 193
Vitamins Pantothenic Acid
Liver, kidney, Involved in energy Uncommon due to None known.
meats, egg yolk, production; aids in availability in most
whole grains, formation of hormones. foods; fatigue; nausea,
legumes; also abdominal cramps;
NOTES made by difficulty sleeping.
intestinal
bacteria.
Biotin
Liver, kidney, Helps release energy Uncommon under None known.
egg yolk, milk, from carbohydrates; normal circumstances;
most fresh aids in fat synthesis. fatigue; loss of
vegetables, also appetite, nausea,
made by vomiting; depression;
intestinal muscle pains; anemia.
bacteria.

Fat-Soluble Vitamins

Fat-Soluble Vitamin
Vitamin Source Physiological Deficiency Over
Functions consumption
A (retinol) Vitamin A: Helps to form skin Mild: night Mild: nausea,
(provitamin liver, vitamin and mucous blindness, irritability,
A, such as A fortified membranes and diarrhea, blurred vision.
beta milk and keep them healthy, intestinal Severe: growth
carotene) dairy thus increasing infections, retardation,
products, resistance to impaired enlargement of
butter, whole infections; essential vision. liver and spleen,
milk, cheese, for night vision; Severe: loss of hair,
egg yolk. promotes bones and inflammation bone pain,
Provitamin A: tooth development. of eyes, increased
carrots, leafy Beta carotene is an keratinization pressure in skull,
green antioxidant and may of skin and skin changes.
vegetables, protect against eyes. Blindness
sweet cancer. in children.
potatoes,
pumpkins,
winter
squash,
apricots,
cantaloupe.

Self-Instructional
194 Material
D Vitamin D-fortified Promotes Severe: rickets in Mild: nausea, Vitamins
dairy products, hardening of children; weight loss,
fortified margarine, bones and teeth, osteomalacia in irritability.
fish oils, egg yolk. increases the adults. Severe: mental and
Synthesized by absorption of physical growth
sunlight action on calcium. retardation, kidney NOTES
skin. damage, movement
of calcium from
bones into soft
tissues.
E Vegetable oil, Protects vitamins Almost impossible Nontoxic under
margarine, butter, A and C and fatty to produce without normal conditions.
shortening, green acids; prevents starvation; possible Severe: nausea,
and leafy vegetables, damage to cell anemia in low digestive tract
wheat germ, whole membranes. birth-weight disorders.
grain products, nuts, Antioxidant. infants.
egg yolk, liver.
K Dark green leafy Helps blood to Excessive None reported.
vegetables, liver; clot. bleeding.
also made by
bacteria in the
intestine.

Check Your Progress


1. What are the main sources of vitamins?
2. When was vitamin A recognized as an essential nutritional factor?
3. Which is the most common disease that occurs in children due to deficiency
of vitamin D?
4. Name some sources of vitamin K.
5. When was thiamine isolated for the first time?
6. Name a disease that occurs due to the deficiency of vitamin B5 in man.

7.3 VITAMINS AS CO-FACTORS AND CO-


ENZYMES

Coenzymes and Cofactors


Some enzymes need assistance so that the catalytic process goes smoothly.
Molecules, which can provide this assistance, are either cofactors or coenzymes.
Function of Coenzymes
Coenzymes are organic carrier molecules. They are non-protein components of
an enzyme that are required for the catalytic process to occur smoothly. They bind
to the active sites of enzymes when the substrate molecules bind, and although
they are not substrate molecules they participate in the catalysis process.
Self-Instructional
Material 195
Vitamins They can be carriers or electrons or groups of atoms, which enables the
catalytic reaction to occur.
Some examples include:
NOTES x NADH which is an electron carrying coenzyme.
x CoA (coenzyme A) is an acyl group (-COR) carrying coenzyme.
Function of cofactors
They are either loosely or tightly bond to the enzyme, which is often denatured
when the cofactor is removed. These participate directly in the catalytic process,
unlike coenzymes. Cofactors stabilize the enzyme or the substrate and assist directly
in the reaction process.
In summary, they are inorganic stabilisers that assist directly in the catalytic
reaction.
Some examples include:
x Mg2+ ions are cofactors in the DNA replication process that stabilizes the
negatively charged DNA molecules.
Vitamins and Minerals
Vitamins are often precursors to many organic coenzymes and minerals are
inorganic cofactors.
Examples Include:
1. Vitamins – some are coenzymes
x B3 – ‘Niacin’ is a precursor of NAD
x B5 – Is a precursor of CoA
2. Minerals – some are cofactors
x Mg2+ - Cofactor involved in DNA replication
x Ca2+ - Not a cofactor because it is actually involved in the structure of teeth
and bones
Vitamins are organic molecules that are needed in small amounts in the diets
of some higher animals.
Vitamins can be grouped according to whether they are soluble in water or
in nonpolar solvents.
Water-Soluble Vitamins
Examples include ascorbic acid (vitamin C) and a series known as the vitamin B
complex.
x Ascorbate, the ionized form of ascorbic acid, serves as a reducing agent
(an antioxidant).
Self-Instructional
196 Material
x The vitamin B series consists of parts of coenzymes. It is worth noting that Vitamins

except for Vit C, all vitamins must be modified before they can serve their
function.
Fat-Soluble Vitamins NOTES
Not all vitamins function as coenzymes. Fat-soluble vitamins have a variety of
functions. Examples include:
x Vit K is needed in the blood clotting process
x Vitamin D is a hormone that regulates the metabolism of calcium and
phosphorus. A deficiency in vitamin D impairs bone formation in growing
animals.
x Vitamin E reacts with and neutralizes reactive oxygen species before they
can oxidize unsaturated membrane lipids, damaging cell structures.

Check Your Progress


7. What are co-enzymes?
8. What are co-factors?

7.4 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. The main sources of vitamins are plants and bacteria.


2. Vitamin A was recognized as an essential nutritional factor by Elmer
McCollumm in 1915.
3. The most common disease that occurs in children due to deficiency of vitamin
D is rickets.
4. Vitamin K is mainly found in the green leafy vegetables such as alfalfa,
spinach, cabbage, swiss chard, brassica, etc.
5. Thiamine was first isolated in 1949, by Jansen in Holland and Adolf Windaus
in Germany.
6. The deficiency of vitamin B5 causes a disease called pellagra in man.
7. Coenzymes are organic carrier molecules. They are non-protein
components of an enzyme that are required for the catalytic process to
occur smoothly.
8. They are either loosely or tightly bond to the enzyme, which is often denatured
when the cofactor is removed. These participate directly in the catalytic
process, unlike coenzymes.

Self-Instructional
Material 197
Vitamins
7.5 SUMMARY

x Vitamins are substances which are different from food. These substances
NOTES are essential for the growth of animals. Even though their requirement is in
very minute quantities, their absence causes specific deficiencies.
x The examples of fat-soluble vitamins are vitamin A, D, E and K. These
vitamins are not readily soluble in water and contain only carbon, hydrogen
and oxygen.
x In some animals, they play very specific roles, like vitamin A plays a role in
the formation of visual compounds and Vitamin D absorbs calcium and
phosphorus from the vertebrate intestine.
x Vitamin A, E and K are terpenoids and vitamin D is steroid.
x Fat-soluble vitamins can be stored in the body, for example vitamin A is
stored in liver for several months. Even though it is possible to store these
vitamins, excessive intake of these could lead to a toxic condition.
x Vitamin A was recognized as an essential nutritional factor by Elmer
McCollumm in 1915. After this, in 1917, Holmes isolated from fish liver oil
and in 1947, Milas it synthesized it for the first time.
x The liver oil of fish is the best natural source of vitamin A. The shark and
halibut contain maximum amount of vitamin A and cod liver contains the
lowest amount of vitamin A. Polar bear’s liver is a concentrated source of
vitamin A.
x Vitamin A is found in two forms, A1 and A2. The A1 form is given by
carotenoids in animals.
x A molecule of beta-carotene is made up of eight, 5 carcon isoprenoid units,
linked to form a long chain of 40 carbon atoms with an ionone ring at each
end.
x In the intestine, the beta-carotene is first reduced to retinal and then to
retinol, which in turn is converted to retinyl ester by way of a reaction of a
fatty acid, for example palmitic acid. This is absorbed through the lymphatic
system and stored in liver.
x Vitamin A plays several important roles in physiology and its deficiency can
lead to various disorders like nyctalopia (night blindness), xerophthalmia
(scaly condition of the membrane covering the eye), keratomalacia (softening
of the cornea), phrynoderma (toad skin, hard and horny skin) and stunted
growth.
x Keratomalacia is a corneal disease that occurs in the children of age 3-4
years. In this case, the cornea loses its lusture, undergoes a necrosis and
develops a few pinpoint ulcers, which later on cause an ulcer.
Self-Instructional
198 Material
x Phrynoderma is a skin lesion that is characterized by follicular hyperkeratosis. Vitamins

In this case, the outer part of forearm in the region of elbow and thighs and
buttocks becomes rough and spiky.
x The existence of vitamin D was demonstrated by Elmer McCollum in 1922
NOTES
for the first time. He found that cod liver oil is effective in preventing rickets.
x Vitamin D is also called sunshine vitamin or antirachitic factor because its
provitamin form, present in human skin, is easily converted into the active
form by radiation with UV. At least 10 different compounds are known to
have antirachitic properties. These are designated as D2 (Ergocalciferol),
D3 (Cholecalciferol), etc.
x The best source of vitamin D is the liver oil of many fish such as cod. Though
egg yolk is a very good natural source of vitamin D, milk and butter contain
very less amount of it. Cereals, vegetables and fruits also have very low
amount of vitamin D.
x The most common disease that occurs in children due to deficiency of vitamin
D is rickets. The deficiency of vitamin D in adults is known as osteomalacia.
x Rickets means to weaken or twist. In this disease, calcification fails to occur
in the child. The early signs of this disease are craniotabes, that is, thickening
of the outer table of the skull and thickening of the wrists and ankles.
x Tocopherols can prevent the oxidation of many other easily oxidized
substances. Vitamin A affects only the ectoderm and endoderm in the embryo,
but vitamin E affects mesoderm.
x In German, vitamin K means koagulations-vitamin, which denotes a group
of lipophili, hydrophobic vitamins that are needed for the posttranslational
modification of certain proteins.
x In 1929, Henrick Dam investigated about this vitamin while working on
chickens. There are two naturally forms of vitamin K, K1 and K2, K1 was
first isolated in 1939 by the Dam et al from alfalfa and the K2 from fish meal
by Doisy et al, also in 1939.
x Vitamin K is mainly found in the green leafy vegetables such as alfalfa,
spinach, cabbage, swiss chard, brassica, etc. Fruits and cereals are poor
sources of vitamin K. Putrefied fish meal is a rich source of vitamin K2.
x The two forms of vitamin K are the derivatives of quinones and they differ
in their side chain, present at carbon 3 of the naphthoquinone ring. In vitamin
K1 it is aphytol redical and in vitamin K2 it is a difarnesyl radical.
x Vitamin K1 is found in plants and has four isoprene units in its side chain.
Vitamin K2 is found in animals and has six isoprene subunits each with
double bonds.
x Vitamin K plays a very important role in blood clotting. The absorption of
vitamin K occurs in the small intestine in the form of bile.
Self-Instructional
Material 199
Vitamins x The deficiency of vitamin K in the body causes loss of blood clotting. In
man, deficiency of vitamin K results in steatorrhea with decreased intestinal
absorption of lipids. It is very rare to find lack of vitamin K in human beings.

NOTES x Vitamin B1 (Thiamine) was first isolated in 1949, by Jansen in Holland and
Adolf Windaus in Germany. Its common name is antiberiberi factor. It is
also known as antineuritic factor.
x Thiamine is found in all plants, yeast and pork, cereals, heart, liver and
kidney. It is mainly found as a coenzyme, Thiamine Pyrophosphate (TPP)
and is destroyed by heat in neutral or alkaline media.
x Vitamin B2 (Riboflavin) was first isolated from milk whey in 1879 and its
chemical synthesis was done by Richard Kuhn and Paul Karrer. It is also
known as lactoflavin and is called yellow enzyme due to its yellow colour.
x Riboflavin is chiefly found in milk, cheese, eggs, liver, kidney, heart and
brewer’s yeast. Cow’s milk and leafy vegetables are also good sources of
riboflavin.
x Riboflavin is synthesized nearly by all green plants, bacteria, yeasts etc. It is
essential for growth and tissue respiration. In the presence of flavokinase,
phosphorylation enzymes of riboflavin convert it into FMN.
x Hepatitis patients and people using oral contraceptives or phenothiazine or
probenecid may have a deficiency of riboflavin. This deficiency leads to
certain diseases in human like chelosisare lead, glossitis, keratitis,
conjunctivitis, lacrimation and corneal vascularization, etc.
x The vitamin B3 was first isolated from yeast liver in 1938 by Roger J.
Williams. He named it pentothenic acid. Fritz A. Lipmann determined the
structure of coenzyme of this vitamin. This enzyme is also called filtrate
factor or the yeast factor.
x The richest source of vitamin B3 is yeast, liver and eggs. Potato, cabbage,
cauliflower, broccoli, tomatoes, peanuts, wheat bran, are some of the
sources of vitamin B3.
x The deficiency of vitamin B3 leads to achromatrichia (premature graying of
the hair).
x Vitamin B5 is referred as nicotinic acid. An American physician named Joseph
Goldberger named it pellagra preventive factor. It is also called vitamin G in
the honour of Joseph Goldberger.
x In 1937, Conard Elvehjem and D. Wayne first time recognized the role of
nicotinic acid as vitamin. This was first isolated by Funk (1911).
x Deficiency of vitamin B5 causes pellagra in man and blacktongue in dogs.
The pellagra is characterized by anorexia, lassitude, fatigue, burning
sensation, numbness and dizziness.
Self-Instructional
200 Material
x Vitamin B6 are rich in rice, cereals, peas, turnip, Brussels sprouts, carrots, Vitamins

potatoes, bananas, avocadoes, watermelons, egg yolk, salmon, chicken,


fish, pork, beef and yeasts.
x The deficiency of vitamin B6 in rats leads to development of acrodynia. In NOTES
humans its deficiency leads to convulsions, anemia, dermatitis, nausea and
vomiting. The deficiency can also occur in malabsorption such as celiac
syndrome
x The rich sources of vitamin B9 are liver, kidney, tuna fish, salmon, yeast,
wheat, dates and spinach, asparagus and turnip greens, beans, peas, lentils,
orange juice, grapefruit juice, honeydew melon, banana, raspberry etc.
x Folic acids contain three different units namely, glutamic acid, p-aminobenzoic
acid and pterin. Its molecular formula is C19H19O6N7. The various types
of vitamin B9 differ from each other in the number of glutamic acid group
present.
x Folic acid after reduction acts as coenzymes. TetraHydroFolic acid (THFA)
is involved in the transfer of methyl group and in the formation of serine,
methionine, thymine, purines, choline and inosinic acid. Folic acid with
ascorbic acid is related to tyrosine metabolism.
x A deficiency of vitamin B12 occurs in the person who abstains from all
animal products. Pernicious anemia can occur the symptoms of which are
irritability, anorexia and listlessness. The adult pernicious anemia is a typical
deficiency disease in which failure of formation of RBCs occurs.
x Coenzymes are organic carrier molecules. They are non-protein components
of an enzyme that are required for the catalytic process to occur smoothly.
They bind to the active sites of enzymes when the substrate molecules bind,
and although they are not substrate molecules they participate in the catalysis
process.

7.6 KEY WORDS

x Ascorbic acid: It refers to a water-soluble sugar acid with antioxidant


properties whose appearance is white to light-yellow crystals or
powder.
x Fat-soluble vitamins: These refer to vitamins that dissolve in dietary and
body fat (A, D, E, and K).
x Metabolism: It refers to the set of chemical reactions that happen in living
organisms to maintain life.

Self-Instructional
Material 201
Vitamins
7.7 SELF ASSESSMENT QUESTIONS AND
EXERCISES

NOTES Short Answer Questions


1. What are the general features of Vitamins?
2. What are the different sources of Vitamin A?
3. List some properties of Vitamin D.
4. Which diseases occur due to deficiency of Vitamin E?
5. What are the major categories of Beriberi?
Long Answer Questions
1. Explain the structures of Vitamins A, D, E and K.
2. Discuss the metabolism of Vitamin B1, B2, B3and B5.
3. Describe some diseases occurring due to deficiency of Vitamin A.
4. Compare and contrast the structure of Vitamin B1 and B2.

7.8 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.
Self-Instructional
202 Material
Minerals

UNIT 8 MINERALS
Structure NOTES
8.0 Introduction
8.1 Objectives
8.2 Minerals in Food
8.3 Classification of Minerals
8.3.1 Major Elements
8.3.2 Trace Elements
8.4 Bioavailability and Deficiency of Minerals
8.4.1 Calcium
8.4.2 Iron
8.4.3 Iodine
8.4.4 Fluorine
8.4.5 Sodium
8.4.6 Potassium
8.5 Minerals in Food Processing
8.6 Answers to Check Your Progress Questions
8.7 Summary
8.8 Key Words
8.9 Self Assessment Questions and Exercises
8.10 Further Readings

8.0 INTRODUCTION

A mineral is, a solid chemical compound that occurs naturally in pure form.
A rock may consist of a single mineral, or may be an aggregate of two or more
different minerals, specially segregated into distinct phases. Compounds that occur
only in living beings are usually excluded, but some minerals are often biogenic (such
as calcite) and/or are organic compounds in the sense of chemistry (such as mellite).
Moreover, living beings often synthesize inorganic minerals (such as hydroxylapatite)
that also occur in rocks.
In geology and mineralogy, the term ‘mineral’ is usually reserved for mineral
species: crystalline compounds with a fairly well-defined chemical composition and
a specific crystal structure. Minerals without a definite crystalline structure, such
as opal or obsidian, are then more properly called mineraloids. If a chemical
compound may occur naturally with different crystal structures, each structure is
considered different mineral species. Thus, for example, quartz and stishovite are
two different minerals consisting of the same compound, silicon dioxide.
The International Mineralogical Association (IMA) is the world’s premier
standard body for the definition and nomenclature of mineral species. As of
November 2018, the IMA recognizes 5,413 official mineral species. Out of more
than 5,500 proposed or traditional ones. The chemical composition of a named
Self-Instructional
Material 203
Minerals mineral species may vary somewhat by the inclusion of small amounts of impurities.
Specific varieties of a species sometimes have conventional or official names of
their own.
Besides the essential chemical composition and crystal structure, the
NOTES
description of a mineral species usually includes its common physical properties
such as habit, hardness, lustre, diaphaneity, colour, streak, tenacity, cleavage,
fracture, parting, specific gravity, magnetism, fluorescence, radioactivity, as well
as its taste or smell and its reaction to acid.
Minerals are classified by key chemical constituents; the two dominant
systems are the Dana classification and the Strunz classification. Silicate minerals
comprise approximately 90% of the Earth’s crust. Other important mineral groups
include the native elements, sulfides, oxides, halides, carbonates, sulfates,
and phosphates.
In this unit, you will study about minerals - types, absorption and role of
minerals in metabolism, minerals deficiency diseases in detail.

8.1 OBJECTIVES

After going through this unit, you will be able to:


x List the minerals contained in foods
x Classify minerals
x Analyse the bioavailability and deficiency of minerals
x Discuss the minerals in food processing

8.2 MINERALS IN FOOD

A seed, after falling from a plant to the earth, immediately begins absorbing minerals
and nutrients from the soil. Minerals are essential in the process transformation
from seed to plant. Different minerals function in specific ways within plant, animal,
and human bodies. Our bodies need minerals from food for the bodily processes
to take place in a healthy manner. Our bodies ‘mine’ minerals from the plants we
consume and thus we are supplied with some of the integral elements that constitute
our bodies and keep them functioning effectively. Our own life blood is to an
extent manufactured from minerals in the Earth. Iron, absorbed by a plant from
the soil, is used for the creation of chlorophyll that is the life blood of a plant. We
consume the plant and our bodies transform the iron found in the plant into
haemoglobin — an essential element constituting our blood.
Minerals are solid inorganic substances of natural occurrence. Minerals
perform various functions. For instance, minerals such as calcium, phosphorus,
and magnesium contribute to the bone structure. Your body continually forms and
destroys bone cells by the process of bone remodelling that allows growth and
Self-Instructional
204 Material
maintenance. Sodium, potassium, and chloride are electrolytes that help conduct Minerals

electrical currents in your nervous system and maintain normal blood pressure.
Iron is a major constituent of haemoglobin and myoglobin, which are oxygen-
carrying compounds of red blood cells. Copper, zinc, manganese, and molybdenum
work with B-vitamins and enzymes to decompose carbohydrates, fats, and proteins NOTES
into digestible forms and convert them into energy.
Importance of Minerals: Minerals are important in human nutrition because of
the roles played by them in human body and they are:
x All nutrients such as vitamins, proteins, enzymes, amino acids, carbohydrates,
fats, sugars, oils, etc. require minerals for effective cellular function. All bodily
functions are dependent on the action and presence of minerals. Minerals
are more important to nutrition compared to vitamins. Vitamins are required
for every bodily biochemical process but cannot function in the absence of
minerals.
x Minerals are helpful in the process of healing. Tissue rebuilding occurs more
rapidly when the body has access to the necessary minerals, which is why
soaking in water quickly heals wounds.
x Minerals are difficult to absorb into the body. Calcium, for instance, must
be taken with vitamins D and C, essential fatty acids and in the proper ratio
to magnesium, for the function of digestion to take place appropriately. One
of the reasons why women have a tendency of being anaemic is for improper
digestion of iron. Iron is present in every food we eat, according to the late
nutritionist, Adelle Davis. However, as iron is difficult to digest, most iron
ingested passes through the body unassimilated. Iron is best absorbed in
combination with additional vitamin C.
x Many mineral supplements are not easily assimilated by the body. It is
essential that mineral supplements be water soluble, not in rock form, and
that the elements be in an oxygenated state, supplying more oxygen to the
blood cells and thereby leading to the release of toxins from the body.
Sources: Most of the minerals in the human diet come directly derived from plants,
such as fruits and vegetables, or indirectly from animal sources. For example,
dairy products are a strong source of calcium, organ meats, fishes, and cashews
are strong sources of copper, whole grain products and leafy green products are
a good source of magnesium and so on. In addition, a deficiency of minerals as
result of lack of sources leads to various types of diseases. For example, in high-
altitude regions, the intake of sodium is less and thus the inhabitants are often
affected by cold extremities, weakness, etc.

8.3 CLASSIFICATION OF MINERALS

Minerals are classified as macro- or major minerals and micro- or trace minerals.
Macro-minerals are present in amounts greater than 0.005 per cent of the body
Self-Instructional
Material 205
Minerals weight whereas micro-minerals are present in less than 0.005 per cent of the body
weight. Macrominerals are required in greater quantity by the body, as compared
to microminerals.
Microminerals perform one or more functions in the body in their optimal
NOTES
concentration. A high concentration may lead to toxicity and death. Similarly, a
very low concentration may lead to severe deficiency diseases that may be fatal if
not cured. It has been found that consumption of one divalent cation (like Ca++,
Mg++, Fe++) may inhibit the absorption of another cation. On the contrary, the
deficiency of one cation enhances the absorption of the other cation.
About 4 to 6 per cent of body weight constitutes mineral elements. Their
largest concentration is found in bones and teeth. They are also found in soft
tissues such as nerves and muscles as well as in blood and other body fluids.
They help in regulating various body functions that include the following:
x Maintenance of the acid-base balance
x Control of water balance
x Muscle contraction
x Stimulation of the nervous system
x Blood coagulation
8.3.1 Major Elements
Major elements are commonly referred to as macrominerals. Macrominerals are
present at larger levels in the animal body or are required in larger amounts in the
diet. Macrominerals constitute the following:

8.3.2 Trace Elements


Microminerals were initially termed as ‘trace’ as they were not easily quantified by
the earlier analytical methods. Iron is considered to be the mineral that divides the
macrominerals from microminerals. Trace elements (as defined by some scientists)
are the minerals that are needed by the body in concentration lower to or equal to
that of iron. According to some scientists trace elements are required by the body
in amounts less than 100 mg per day.
Trace elements are also considered to be essential as their deficiency affects
the biological functions of the body. The dietary deficiency caused due to these
minerals is preventable and reversible if the required amounts of minerals are
administered. The major features of microminerals are as follows:
x They are components of living tissues.
x The concentration of microminerals is almost fairly constant from one species
to another.
Self-Instructional
206 Material
x Their deficiency results in some physiological and structural abnormalities Minerals

as well as biochemical changes in the body.


x Their supplementation reduces or prevents the abnormalities or biochemical
changes caused by their deficiency.
NOTES
Microminerals (trace elements) include the following:

8.4 BIOAVAILABILITY AND DEFICIENCY OF


MINERALS

Let us now discuss the bioavailability and deficiency of some important minerals
that are necessary for human consumption:
8.4.1 Calcium

Bioavailability
Calcium is the most abundant mineral found in the body. It is a divalent cation and
accounts for about 1.5 per cent of the total body weight. The maximum amount of
calcium is found in bones and teeth (99 per cent). The remaining amount (1 per
cent) is found in both intracellular fluid and extra cellular fluid.
Milk, milk products , sesame seeds, tofu, turnips, small dried fish, ragi, leafy
vegetables, broccoli, legumes, kale, etc., are the chief sources of calcium. meat,
poultry, fish, grains and nuts are poor sources of calcium.
Milk and milk products are considered to be the best sources of calcium
because calcium present in milk is very easily assimilated. This assimilation is
facilitated by vitamin D that is also present in milk. Table 8.1 lists various sources
of calcium.
Calcium is present in the form of insoluble salts in foods. It is first converted
into its ionic form (Ca++) and then absorbed in slightly acidic pH. At higher
phosphorus (intestines), it forms complexes with other minerals and thus its
bioavailability is reduced.
The non-osseous calcium (calcium not associated with bones) is found within
mitochondriaendoplasmic reticulum, nucleus and vesicles.
Deficiency
A calcium deficiency may result from the following:
x Inadequate intake in diet
x Poor absorption of calcium
x Excessive losses in urine and faeces.
Self-Instructional
Material 207
Minerals All of the above reasons may lead to inadequate mineralization of bones. In
children, rickets is observed due to deficient amounts of calcium accretion in the
bones.
Hypocalcemia may result in tetany. Tetany is the intermittent contractions of
NOTES
the muscles. Muscle pain, muscle spasm and parasthesias are common signs of
tetany.
Calcium deficiency in adults leads to osteoporosis, which is the loss of bone
mass. Bone mass includes protein matrix and minerals. Hypertension and colon
cancer are observed in case of deficiency of calcium over a long period of time.
Persons consuming high-protein diets and high-fibre diets are also found
deficient in calcium. Fat malabsorption also leads to calcium deficiency. Apart
from this, loss of calcium from bones due to immobilization, decreased gastro-
intestinal transit time and prolonged use of thiazide drugs leads to calcium deficiency.
Postmenopausal women, Amenorrheic women, and people who are lactose
intolerant need extra calcium.
Table 8.1 Various Sources of Calcium

8.4.2 Iron
Now, we shall discuss the bioavailability and effect of deficiency of iron.
Bioavailability
Iron, one of the most abundant metals on Earth, is essential to most life forms and
to normal human physiology. Iron is an integral part of many proteins and enzymes
that are required to maintain good health. In humans, iron is an essential component
of proteins involved in oxygen transport. It is also essential for the regulation of
cell growth and differentiation.
Our body contains about 2 to 4 gm of iron. Iron is a vital component of
haemoglobin, which transports oxygen to various tissues of the body. Almost, all
living organisms require iron as an essential element for growth and multiplication.
Iron deficiency is the most common nutritional problem in the world. Free ionic
iron hardly exists in the body. It is found in the following forms:
x 65 per cent of iron is found in haemoglobin.
x 10 per cent of iron is found as myoglobin.
Self-Instructional
208 Material
x 1 per cent to 5 per cent is found as a component of enzymes. Minerals

x The remaining body iron is found in blood or storage organs.


It has been found that the total body iron is related to body weight, age,
gender, pregnancy and state of growth of an individual. NOTES
There are two forms of dietary iron:
x Heme
x Non-Heme
Heme iron is derived from haemoglobin, the protein in red blood cells that
delivers oxygen to cells. Heme iron is found in animal foods that originally contained
haemoglobin, such as red meat, fish, and poultry. Iron in plant foods such as lentils
and beans is arranged in a chemical structure called nonheme iron. This is the form
of iron added to iron-enriched and iron-fortified foods.
Deficiency
The World Health Organization considers iron deficiency as the number one
nutritional disorder in the world. It has been observed that 80 per cent of the
world’s population may be iron deficient, while 30 per cent may have iron deficiency
anaemia.
Iron deficiency develops gradually and usually begins with a negative iron
balance, when iron intake does not meet the daily need for dietary iron. This
negative balance initially depletes the storage form of iron while the blood
haemoglobin level, a marker of iron status, remains normal. Iron deficiency anaemia
is an advanced stage of iron depletion. It occurs when storage sites of iron are
deficient and blood levels of iron cannot meet daily needs. Blood haemoglobin
levels are below normal with iron deficiency anaemia.
Iron deficiency anaemia can be associated with low dietary intake of iron,
inadequate absorption of iron, or excessive blood loss. Women of childbearing
age, pregnant women, preterm and low birth weight infants, older infants and
toddlers, and teenage girls are at greatest risk of developing iron deficiency anaemia
because they have the greatest need for iron. Women with heavy menstrual losses
can lose a significant amount of iron and are at considerable risk of iron deficiency.
Adult men and post-menopausal women lose very little iron, and have a low risk
of iron deficiency.
Individuals with kidney failure, especially those being treated with dialysis,
are at high risk for developing iron deficiency anaemia. This is because their kidneys
cannot create enough erythropoietin, a hormone needed to make red blood cells.
Both iron and erythropoietin can be lost during kidney dialysis. Individuals who
receive routine dialysis treatments usually need extra iron and synthetic erythropoietin
to prevent iron deficiency.
Vitamin A helps mobilize iron from its storage sites, so a deficiency of vitamin
A limits the body’s ability to use stored iron. This results in an apparent iron
deficiency because haemoglobin levels are low even though the body can maintain
Self-Instructional
Material 209
Minerals normal amounts of stored iron. This problem is seen in developing countries where
vitamin A deficiency often occurs.
Chronic malabsorption can contribute to iron depletion and deficiency by
limiting dietary iron absorption or by contributing to intestinal blood loss. Most
NOTES
iron is absorbed in the small intestines. Gastrointestinal disorders that result in
inflammation of the small intestine may result in diarrhoea, poor absorption of
dietary iron, and iron depletion.
Eating nonnutritive substances such as dirt and clay, often referred to as
pica or geophagia, is sometimes seen in persons with iron deficiency.
People with chronic infectious, inflammatory, or malignant disorders such
as arthritis and cancer may become anaemic. Three groups of people are most
likely to benefit from iron supplements: people with a greater need for iron,
individuals who tend to lose more iron, and people who do not absorb iron normally.
These individuals include the following:
x Pregnant women.
x Preterm and low birth weight infants.
x Older infants and toddlers.
x Teenage girls.
x Women of childbearing age, especially those with heavy menstrual losses.
x People with renal failure, especially those undergoing routine dialysis.
x People with gastrointestinal disorders who do not absorb iron normally.
Foods rich in Iron

8.4.3 Iodine
We shall discuss this mineral with regard to: (i) bioavailability and (ii) deficiency.
Bioavailability
Iodine is found in dairy products, seafood, kelp, eggs, some vegetables, and iodized
salt. Iodine is important for essential hormone development in the human body.
Iodine is found in food in the form of iodide. Iodide is required in the formation
of thyroid hormones.
Iodized salt is the main food source of iodine. Seafood is naturally rich in
iodine. Cod, sea bass, haddock, and perch are good sources (Refer Table 8.2).
Self-Instructional
210 Material
Kelp is the most common vegetable seafood that is a rich source of iodine. Dairy Minerals

products also contain iodine. Other good sources of iodine include plants grown
in iodine rich soil.
Iodine is also found in seawater so any type of seafood is a rich source of
NOTES
iodine, particularly seaweed (kelp). Since an adult only requires around one
teaspoonful of iodine over a lifetime, eating fish once a week is likely to be enough
to fulfill the average iodine requirement. Although it comes from the ocean, sea salt
is not a good source of iodine.
Deficiency
Iodine is needed for the normal metabolism of cells. Metabolism is the process of
converting food into energy. Humans need iodine for normal thyroid function, and
for the production of thyroid hormones.
In developing foetus, babies, and young children, the effects of iodine
deficiency are serious. They include stunted growth, diminished intelligence, and
retardation. Lack of iodine is a major problem in developing countries. Iodine is
the world’s number one cause of preventable intellectual disability in children
(cretinism). There is evidence that some levels of iodine deficiency may be too
mild to cause goitre but may still retard brain development. Iodine is a trace mineral
and an essential nutrient found naturally in the body.
In the case of adults, an enlarged thyroid gland, or goitre, is not the only side
effect of iodine deficiency. If the deficiency is long term, it results in hypothyroidism.
Symptoms include dry skin, hair loss, fatigue and slowed reflexes.
Goitre can also increase the risk of thyroid cancer. Goitre can be associated
with hyperthyroidism, a condition in which too much thyroid hormone is produced.
Pregnant women need higher levels of iodine, as lack of this nutrient can
retard normal development in babies.
Vegetarians may be at risk of iodine deficiency if they do not eat seafood.
Vegetarians can get iodine from iodized salt or seaweed. Lack of enough iodine
(deficiency) may occur in places that have iodine-poor soil. Many months of iodine
deficiency in a person’s diet may cause goitre or hypothyroidism.
Table 8.2 Sources of Iron

Self-Instructional
Material 211
Minerals 8.4.4 Fluorine
Now, we shall discuss fluorine bioavailability and results of deficiency of fluorine.
Bioavailability
NOTES
Fluorine is naturally present in the body as calcium fluoride. Calcium fluoride is
mostly found in bones and teeth.
Fluoridated water, and food prepared in fluoridated water, contains fluoride.
Natural sodium fluoride is in the ocean, so most seafood contains fluoride. Tea
and gelatin also contain fluoride. The ready-to- eat infant formula is made of water
containing fluoride.
Small amounts of fluoride help reduce tooth decay. Fluoridation of tap water
helps reduce cavities in children by 50 – 60 per cent. Fluorides also help maintain
bone structure. Low doses of fluoride salts may be used to treat conditions that
cause faster-than-normal bone loss, such as menopause.
Deficiency
Fluoride deficiency may appear in the form of increased cavities and weak bones
and teeth. Fluoride supplementation is necessary to prevent cavities, especially in
children.

Some Sources of Fluorine

8.4.5 Sodium
Sodium is an important mineral from the point of view of human health. We shall
focus on its bioavailability and deficiency.
Bioavailability
Sodium constitutes about 90 per cent of the cations of the extracellular compartment
(vascular and interstitial fluid). It constitutes about 142 mEq/l. The osmotic pressure
of ECF is basically due to the presence of sodium. About 30 per cent of the total
sodium is located on the surface of bone crystals from where they are released
into the blood in the case of hyponatremia.
Sodium occurs naturally in most foods. The most common form of sodium
is sodium chloride, which is table salt. Milk, beets, and celery also naturally contain
sodium. Drinking water also contains varying amounts of sodium.
Self-Instructional
212 Material
Sodium is added to various food products. Some of these added forms are Minerals

monosodium glutamate, sodium nitrite, sodium saccharin, baking soda (sodium


bicarbonate), and sodium benzoate. These are ingredients in condiments and
seasonings such as, soy sauce, onion salt, garlic salt, and bouillon cubes.
NOTES
Deficiency
Hyponatraemia is usually defined as serum sodium below 125 mEq per litre, and
may be without symptoms. The symptoms occur due to overhydration of cells of
the central nervous system. Some symptoms of sodium deficiency include:
x Weakness
x Giddiness
x Anorexia
x Cramps in Calf Muscles
x Low Blood Pressure
x Water Deficiency
Causes of deficiency include inadequate intake of sodium, excessive fluids
(dilutionalhyponatraemia), and effect of drugs.

Some Sources of Sodium

8.4.6 Potassium
Potassium is a mineral involved in electrical and cellular body functions. In the
body, potassium is classified as an electrolyte.
Bioavailability
About 95 per cent of potassium is present in the intracellular component of the
body. Due to this potassium is the main intracellular cation of the body. It influences
the contractibility of the skeletal and cardiac muscles. It also affects the excitability
of the nervous system. It maintains the electrolyte and pH balance of the body.
Many foods contain potassium. All meats (red meat and chicken) and fish
such as salmon, cod, flounder, and sardines are good sources of potassium. Soy
products and veggie burgers are also good sources of potassium.
Vegetables including broccoli, peas, lima beans, tomatoes, potatoes
(especially their skins), sweet potatoes, and winter squashes are all good sources
of potassium.
Self-Instructional
Material 213
Minerals Fruits that contain significant sources of potassium include citrus fruits,
cantaloupe, bananas, kiwi, prunes, and apricots. Dried apricots contain more
potassium than fresh apricots.
Milk and yogurt, as well as nuts, are also excellent sources of potassium.
NOTES
People on dialysis for kidney failure should avoid consuming potassium-
rich foods. They require specialized diets to avoid excessive potassium in the
blood.
Deficiency
Excessive or little potassium in the body can have serious consequences. As many
foods contain potassium, potassium deficiency is rarely caused by inadequate
diet. However, even a moderate reduction in the body’s potassium levels can lead
to salt sensitivity and high blood pressure.
A deficiency of potassium (hypokalaemia) can occur in people with certain
diseases or as a result of diuretics (water pills) for the treatment of high blood
pressure or heart failure. Additionally, many medications, such as diuretics, laxatives,
and steroids, can cause a loss of potassium, which occasionally may be severe.
A variety of conditions can cause potassium loss from the body. The most
common are vomiting and diarrhoea. Several rare kidney and adrenal gland
disorders may also cause low potassium levels.
Hypokalaemia is a lower than normal amount of potassium in the blood
caused by sweating, vomiting, breakdown of muscle fibres and so on.
Potassium is needed for the proper functioning of cells, especially nerve
and muscle cells. The kidneys remove excess potassium in the urine to keep a
proper balance of the mineral in the body.

Some Sources of Potassium

8.5 MINERALS IN FOOD PROCESSING

Minerals such as iron and iodine play a very important role in food processing.
They are discussed as follows:
Iron compounds used in food fortification are commonly classified according to
their solubility. Selection of an appropriate iron fortificant for any given application
is based on the following criteria:
x Organoleptic considerations
Self-Instructional
214 Material
x Bioavailability Minerals

x Cost
x Safety
The most important factor in fortifying light-coloured foods is the colour of NOTES
the iron compounds. For the enrichment of rice, white iron and ferric orthophosphate
are used as fortificants. The use of more soluble iron compounds often leads to the
development of off-colours and off-flavours due to reactions with other components
of the food material.
Ferrous sulphates often turn infant cereals grey or green. Off-flavours can
be the result of lipid oxidation catalysed by iron. The iron compounds may also
give metallic flavour to the food.
Some of these undesirable interactions with the food matrix can be avoided
by coating the fortificant with hydrogenated oils or ethyl cellulose. Bioavailability
of iron compounds is normally stated relative to a ferrous sulphate standard. Highly
water soluble iron compounds have superior bioavailability. By reducing the particle
size the bioavailability of an insoluble or very poorly soluble iron compounds can
be improved. This may also lead to increased reactivity in deteriorative processes.
The most commonly used compounds in the iodization of foods include the
iodides and iodates of sodium and potassium. These are the additives allowed
by Codex Alimentarius in the iodization of salt.
The iodide compounds are superior to iodates because of the following
reasons:
x Iodides are cheaper.
x Iodides are more soluble.
x Iodides have a higher iodine content.
Iodates are more stable under conditions of high moisture, high ambient
temperature, sunlight, aeration and the presence of impurities.
Potassium iodide is used in the following conditions:
x Salt is dry.
x Salt is free from impurities.
x Salt has a slightly alkaline pH.
If the salt is not alkaline, the iodide may be oxidized to molecular iodine and
lost through evaporation. If excess water is present the iodide may be separated
from the salt in the water film (FAO/WHO, 1991). Loss of iodide can be reduced
through the addition of stabilizers such as 0.1 per cent sodium thiosulphate and
0.1 per cent calcium hydroxide combined or 0.04 per cent dextrose and 0.006
per cent sodium bicarbonate. The presence of calcium salts has been reported to
produce off-flavour due to calcium ions (Kuhajek and Fiedelman, 1973).
Self-Instructional
Material 215
Minerals

Check Your Progress


1. Give the importance of minerals.
NOTES 2. Name some of the major elements.
3. What does calcium deficiency result into?
4. What are heme iron?
5. List some of the symptoms of sodium deficiency.
6. When is potassium iodide used?

8.6 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. Minerals are important in human nutrition because of the roles played by


them in human body and they are:
x All nutrients such as vitamins, proteins, enzymes, amino acids,
carbohydrates, fats, sugars, oils, etc. require minerals for effective cellular
function. All bodily functions are dependent on the action and presence
of minerals. Minerals are more important to nutrition compared to
vitamins. Vitamins are required for every bodily biochemical process
but cannot function in the absence of minerals.
x Minerals are helpful in the process of healing. Tissue rebuilding occurs
more rapidly when the body has access to the necessary minerals, which
is why soaking in water quickly heals wounds.
x Minerals are difficult to absorb into the body. Calcium, for instance,
must be taken with vitamins D and C, essential fatty acids and in the
proper ratio to magnesium, for the function of digestion to take place
appropriately. One of the reasons why women have a tendency of being
anaemic is for improper digestion of iron. Iron is present in every food
we eat, according to the late nutritionist, Adelle Davis. However, as iron
is difficult to digest, most iron ingested passes through the body
unassimilated. Iron is best absorbed in combination with additional
vitamin C.
x Many mineral supplements are not easily assimilated by the body. It is
essential that mineral supplements be water soluble, not in rock form,
and that the elements be in an oxygenated state, supplying more oxygen
to the blood cells and thereby leading to the release of toxins from the
body.
2. Microminerals (trace elements) include the following:
Iron, Flurine, Zink, Copper, Iodine, Chromium, Cobalt, Selenium.
Self-Instructional
216 Material
3. A calcium deficiency may result from the following: Minerals

x Inadequate intake in diet.


x Poor absorption of calcium.
x Excessive losses in urine and faeces. NOTES
4. Heme iron is derived from haemoglobin, the protein in red blood cells that
delivers oxygen to cells. Heme iron is found in animal foods that originally
contained haemoglobin, such as red meat, fish, and poultry. Iron in plant
foods such as lentils and beans is arranged in a chemical structure called
nonheme iron. This is the form of iron added to iron-enriched and iron-
fortified foods.
5. Some symptoms of sodium deficiency include:
x Weakness
x Giddiness
x Anorexia
x Cramps in calf muscles
x Low blood pressure
x Water deficiency
6. Potassium iodide is used in the following conditions:
x Salt is dry.
x Salt is free from impurities.
x Salt has a slightly alkaline pH.

8.7 SUMMARY

x The mineral elements that are recognized as essential for body functions
include calcium, phosphorus, sodium, molybdenum, chlorine, magnesium,
iron, selenium, iodine, manganese, copper, cobalt, zinc, fluorine and
chromium.
x The prominence of each mineral element in body tissues is closely related
to its functional role.
x As constituents of bones and teeth, minerals provide strength and rigidity to
skeletal structures.
x Minerals are classified as macro minerals and micro minerals. Macrominerals
are present in amounts greater than 0.005 per cent of the body weight
whereas micro minerals are present in less than 0.005 per cent of the body
weight.
x Macrominerals include calcium, phosphorus, potassium, chlorine, sodium,
magnesium and sulphur.
Self-Instructional
Material 217
Minerals x Microminerals were initially termed as ‘trace’ as they were not easily
quantified by the earlier analytical methods.
x Iron is considered to be the mineral that divides the macrominerals from
microminerals.
NOTES
x Trace elements (as defined by some scientists) are the minerals that are
needed by the body in concentration lower to or equal to that of iron.
x Microminerals perform one or more functions in the body in their optimal
concentration.
x Microminerals include iron, fluorine, zinc, copper, iodine, chromium, cobalt
and selenium.
x Calcium is the most abundant mineral found in the body. It is a divalent
cation and accounts for about 1.5 per cent of the total body weight.
x Milk and milk products such as curd, sesame seeds, tofu, turnips, small
dried fish, leafy vegetables, broccoli, legumes, kale, etc., are the chief sources
of calcium.
x Phosphorus is second only to calcium in terms of its abundance in our body.
x The best food sources of phosphorus include meat, poultry, fish, eggs, milk
and milk products, nuts, legumes, cereal grains and chocolates.
x Magnesium stands fourth, amongst all the minerals in terms of its overall
abundance in the body.
x Magnesium is found widely in food and beverages. Magnesium is rich in
tea, coffee and cocoa. It is also found in green leafy vegetables, nuts, legumes,
whole grain cereals, spices, chocolates, molasses, corn, peas, carrots, brown
rice and sea foods.
x Sodium constitutes about 90 per cent of the cation of the extracellular
compartment (vascular and interstitial fluid).
x The most common form of sodium is sodium chloride, which is table salt.
Milk, beets, and celery also naturally contain sodium.
x Potassium is a mineral involved in electrical and cellular body functions. In
the body, potassium is classified as an electrolyte.
x All meats (red meat and chicken) and fish such as salmon, cod, flounder,
and sardines are good sources of potassium.
x Chloride is the main anion of the ECF and of the total anion content of ECF
chloride constitutes 103 mEq per litre.
x Chloride can be found in table salt or sea salt in form of sodium chloride. It
is also found in many vegetables.
x Sulphur is a mineral found naturally in many different forms in all foods. It is
also used in the form of sulphates and sulphites as food additives in some
processed foods.
Self-Instructional
218 Material
Minerals
x Iron is an integral part of many proteins and enzymes that are required to
maintain good health.
x Heme iron is found in animal foods that originally contained haemoglobin,
such as red meat, fish, and poultry. Iron in plant foods such as lentils and NOTES
beans is arranged in a chemical structure called nonheme iron.
x Zinc is an essential mineral that is naturally present in some foods, added to
others, and available as a dietary supplement.
x Copper is a mineral that occurs naturally in many foods, including vegetables,
legumes, nuts, grains, and fruits, as well as shellfish, avocado, and beef
(organs such as liver).
x Selenium is a trace mineral found in soil, water, and certain foods. It is an
essential element in several metabolic pathways.
x Specific dietary sources of selenium include brewer’s yeast, wheat germ,
butter, garlic, grains, sunflower seeds, Brazil nuts, walnuts, raisins, liver,
kidney, shellfish (lobster, oyster, shrimp, scallops), and fresh water and salt
water fish (red snapper, salmon, swordfish, tuna, mackerel, halibut, flounder,
herring, smelts).
x Chromium is an essential mineral that is not made by the body and must be
obtained from the diet.
x The best source of chromium is brewer’s yeast.
x Iodine is found in dairy products, seafood, kelp, eggs, some vegetables and
iodized salt. Iodine is important for essential hormone development in the
human body.
x Manganese is a trace element found in a variety of foods. These include
bread, nuts, cereals and green vegetables (such as peas and runner beans).
x Molybdenum does not exist naturally in metallic state, but occurs in
association with other elements. The predominant form of molybdenum
occurring in soil and natural waters is the molybdate anion, MoO4–2.
x Fluoride occurs naturally in the body as calcium fluoride. Calcium fluoride
is mostly found in bones and teeth.
x Fluoridated water, and food prepared in fluoridated water, contains fluoride.

8.8 KEY WORDS

x Bioavailability: The proportion of a drug or other substance that enters


the circulation when introduced into the body.
x Haemoglobin: A haemoprotein composed of globin and heme that gives
red blood cells their characteristic colour; function primarily to transport
oxygen from the lungs to the body tissues.
x Anaemic: Deficiency in the number or quality of red blood cells.
Self-Instructional
Material 219
Minerals
8.9 SELF ASSESSMENT QUESTIONS AND
EXERCISES

NOTES Short Answer Questions


1. What are minerals?
2. How many types of minerals are there? Explain them briefly.
3. Write a short note on deficiency of iron.
4. Briefly discuss about bioavailability and deficiency of minerals.
Long Answer Questions
1. Define minerals.
2. Discuss the importance of minerals.
3. Write a note on the sources of minerals.
4. Classify minerals with examples.
5. Elaborate on the bioavailability and deficiency results associated with
minerals.
6. Explain the role of minerals in food processing.

8.10 FURTHER READINGS


Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.
Self-Instructional
220 Material
Nucleic Acids
BLOCK - III
NUCLEIC ACIDS AND ENZYMES
NOTES
UNIT 9 NUCLEIC ACIDS
Structure
9.0 Introduction
9.1 Objectives
9.2 Nucleic Acids: Structure
9.2.1 Nitrogenous Bases (Pyrimidine and Purine)
9.2.2 Nucleosides and Nucleotides
9.3 Ribonucleic Acid (RNA) and Deoxyribonucleic Acid (DNA)
9.3.1 Structure and Properties of DNA
9.3.2 Structure and Properties of RNA
9.3.3 DNA is Genetic Material
9.4 Synthesis and Degradation of Purines and Pyrimidines
9.4.1 Purine Nucleotide Synthesis
9.4.2 Pyrimidine Synthesis
9.4.3 Degradation of Purines and Pyrimidines
9.5 Answers to Check Your Progress Questions
9.6 Summary
9.7 Key Words
9.8 Self Assessment Questions and Exercises
9.9 Further Readings

9.0 INTRODUCTION

Nucleic acids are the biopolymers, or small biomolecules, essential to all known
forms of life. The term nucleic acid is the overall name for DNA and RNA. They
are composed of nucleotides, which are the monomers made of three components:
a 5-carbon sugar, a phosphate group and a nitrogenous base. If the sugar is a
compound ribose, the polymer is RNA (RiboNucleic Acid); if the sugar is derived
from ribose as deoxyribose, the polymer is DNA (DeoxyriboNucleic Acid).
Nucleic acids are the most important of all biomolecules. They are found in
abundance in all living things, where they function to create and encode and then
store information in the nucleus of every living cell of every life-form organism on
Earth. In turn, they function to transmit and express that information inside and
outside the cell nucleus—to the interior operations of the cell and ultimately to the
next generation of each living organism. The encoded information is contained and
conveyed via the nucleic acid sequence, which provides the 'ladder-step' ordering
of nucleotides within the molecules of RNA and DNA.
In this unit you will study about nucleic acids–DNA and RNA, structure,
function, metabolism and genetic disorders in detail.
Self-Instructional
Material 221
Nucleic Acids
9.1 OBJECTIVES

After going through this unit, you will be able to:


NOTES x Describe structure of nucleic acids
x Understand synthesis of purines and pyrimidines
x Discuss about the degradation of purines and pyrimidines

9.2 NUCLEIC ACIDS: STRUCTURE

The origins of nucleic acids can be traced back to 1869, when Friedrich Miescher,
a Swiss chemist isolated nuclei from pus cells and found that they contain a
phosphate rich substance. He named this substance nuclein. In 1874, he isolated
pure nucleic acid from salmon sperm nuclei. In 1899, Altman gave it the name
nucleic acid because it possesses acidic properties. In 1880s, Fischer identified
purine and pyrimidine bases in nucleic acid. In 1881, Zacharis identified nucleic
acid with chromatin. In 1882, Sachs discovered that nucleic acid of sperm and
egg are different.
In 1894, Geheimrat Albrecht Kossel, recognized that histones ad protamins
are associated with nucleic acid and the histones are the basic proteins. In 1910
he got Nobel Prize for the discovery of two purines and pyrimidine bases in nucleic
acids. In 1914, Robert Feulgen discovered the method of colour test of DNA,
which is called Feulgen test. In 1931, P.A. Levine defined that there are two types
of nucleic acids; DNA and RNA. In 1941, Caspersson and Brachet independently
stated that nucleic acid is directly related to protein synthesis.
In 1944, Oswald T. Avery, Colin M. Macleod and Maclyn McCarty stated
that DNA is directly related to inheritance. In 1953, James D. Watson and Francis
H.C. Crick constructed the double helical model for the DNA molecule.
Before going on to studying the types of nucleic acides, you need to know
what is gene and genome.
A gene is a unit of heredity in a living organism. It normally resides on a
stretch of DNA that codes for a type of protein or for an RNA chain that has a
function in the organism. All living things depend on genes. This is because they
specify all proteins and functional RNA chains. Genes contain the information on
which to build and maintain an organism’s cells and pass genetic traits to offspring,
although some organelles are self-replicating and not coded for by the organism’s
DNA. Most organisms have several genes corresponding to several different
biological traits. Some of these traits are visible instantly, for example, the colour
of the eye and the number of limbs. Some are not visible immediately, such as
blood type or increased risk for specific diseases, or the thousands of basic
biochemical processes comprising life.
Self-Instructional
222 Material
The word ‘genome’ is derived from the Greek word genome, which means Nucleic Acids

‘I become, I am born, to come into being’. Some suggest it to be a mix of the


gene and chromosome. In modern molecular biology and genetics, the genome
is the hereditary information of an organism in totality. It is encoded either in DNA
or for several types of viruses in RNA. The genome includes both the genes and NOTES
the non-coding sequences of the DNA.
Types of Nucleic Acids
There are two types of nucleic acids; DeoxyriboNucleic Acid (DNA) and
RiboNucleic Acid (RNA) . These two types of nucleic acids are present in all
types of plants and animals. The viruses also contain DNA or RNA, but not both.
DNA is present in the cell nucleus and nearly 90 per cent RNA is present in the
cytoplasm and 10 per cent in the nucleolus. Both of these types of nucleic acid
possess three components; phosphoric acid, pentose sugar and nitrogenous bases.
Phosphoric Acid
The molecular formula of phosphoric acid is H3PO4. It attaches three monovalent
hydrogen atoms and three divalent oxygen atoms to the pentavalent phosphorus
atom. Figure 9.1 shows the molecular structure of phosphoric acid.

Fig. 9.1 Phosphoric Acid

Pentose Sugar
Both the categories of nucleic acid are differentiated on the bases of pentose sugar
present in them. There are two types of pentose sugar; D-2-deoxyribose sugar
present in the deoxyribonucleic acid and the D-ribose sugar present in the D-
ribonucleic acid. These two sugars are present in the furanose form. Figure 9.2(a)
and (b) shows the molecular structure of D-ribose and D-2-deoxyribose,
respectively.

(a) D-Ribose (b) D-2-Deoxyribose

Fig. 9.2 Molecular Structures


Self-Instructional
Material 223
Nucleic Acids The main difference between these two structures is that only
D-2-deoxyribose sugar replaces OH atom at position 2 by an H atom. These two
sugars are also differentiated by means of colour reaction.

NOTES 9.2.1 Nitrogenous Bases (Pyrimidine and Purine)


There are two types of nitrogenous bases present in all nucleic acids. These are
pyrimidine and purine.
Pyrimidine: These are heterocyclic aromatic compounds similar to pyridine and
benzene. They possess six membered ring and two N atoms and three double
bonds. Its melting point is 220C. Figure 9.3 shows the molecular structure of
pyrimidine.

Fig. 9.3 Pyrimidine

In nucleic acids, pyrimidine, are of the following three types:


x Thymine (C5H6O2N2): It is named thymine as it has being isolated from
thymus. It is found in DNA and its molecular weight is 126.13 daltons.
Figure 9.4 shows the molecular structure of thymine.

Fig. 9.4 Thymine

x Cytosine (C5H5ON3): It is found both in RNA and DNA. It is a white


crystalline substance and its molecular weight is 111.12 daltons. Figure 9.5
shows the molecular structure of cytosine.

Fig. 9.5 Cytosine


Self-Instructional
224 Material
x Uracil (C4H4O2N2): It is found only in RNA. It is a white crystalline Nucleic Acids

substance and its molecular weight is 111.10 daltons. Figure 9.6 shows the
molecular structure of uracil.

NOTES

Fig. 9.6 Uracil

Purine: In 1884, Purine name was coined by the German chemist Emil Fischer.
Purine is a heterocyclic aromatic organic compound, in which a 6 membered
pyrimidine ring is fused with an imidazole ring. Purine has a melting point of 2160C.
Figure 9.7 shows the molecular structure of purine. There are two different types
of purines; adenine and guanine.

Fig. 9.7 Purine

x Adenine (C5H5N5): This is found in DNA as well as in RNA. It is a white


crystalline substance and its molecular weight is 135.15 daltons. Its melting
point is 360-3650C. Figure 9.8 shows the molecular structure of adenine.

Fig. 9.8 Adenine

x Guanine (C5H5ON5): This is also present in DNA as well as in RNA. It is


a white colourless, insoluble substance and its molecular weight is 151.15
daltons. Figure 9.9 shows the molecular structure of guanine.

Fig. 9.9 Guanine


Self-Instructional
Material 225
Nucleic Acids 9.2.2 Nucleosides and Nucleotides
Following are the characteristic features of Nucleosides and Nucleotides.
Nucleosides
NOTES
Nucleosides are the compounds in which nitrogenous bases are conjugated to
the pentose sugars by a E-Glycosidic linkage. The E-Glycosidic linkage involves
C-1 molecule of sugar and hydrogen of N-1 (in Pyrimidine), N-9 (in Purine). If
there is a presence of ribose sugar in the nucleoside, then it is named
ribonucleosides and if the nucleoside has deoxyribose sugar, then it is named
deoxyribonucleosides. The molecule without the phosphate group is called
nucleosides.

Fig. 9.10 Nucleosides

Nucleotides
Nucleotides are the phosphoric esters of nucleosides. When phosphate attaches
itself to the nucleosides, then it is called nucleotides. In ribose moiety, the
esterification can happen at three different positions, namely at C2, C3 and C5.
In deoxyribose moiety, the esterification can happen at two different positions,
namely at C3 and C5 position because C2 position does not have a hydroxyl
group.
A nucleotide refers to the monomer structural units that form the two primary
nucleic acids; RiboNucleic Acid (RNA) and DeoxyriboNucleic Acid (DNA) and
several other lesser nucleic acids. It comprises a heterocyclic nucleobase, a pentose
sugar (ribose or deoxyribose) and a phosphate or polyphosphate group.
The function of nucleotides is to code for proteins and enzymes. They also
determine the genetic structure of life, transport and transform cellular energy and
regulate enzymes. Most nucleotides are Adenine (A), Guanine (G), Thymine (T),
Cytosine (C) and Urasil (U). When nucleotides group together to constitute RNA
or DNA, only A and T group together and C groups with G. T will never pair with
U. Thymine is found only in DNA, while Urasil is found only in RNA.
It has three components: a nitrogenous base, a pentose sugar and a
phosphate.
Self-Instructional
226 Material
Nucleic Acids

NOTES

Fig. 9.11 Nucleotides


Structure
The nucleotide units themselves consist of many smaller components. Each
nucleotide has a phosphate unit, a sugar unit and a heterocyclic base unit.
x Phosphate Unit: The phosphate unit is represented either as a phosphate
ion or as a phosphoric acid molecule.
x Sugar Unit: The sugar unit contains a five-carbon atom sugar in its ring
form. It is either ribose in RNA or deoxyribose in DNA.
x Heterocyclic Bases: Nucleotides consist of different bases. These bases
are heterocyclic bases. They are also termed as nitrogenous bases since
they have nitrogen within the rings. Pyrimidine bases have one ring and are
similar in structure to the compound pyrimidine. They consist of cytosine,
thymine and uracil. Cytosine is found in both DNA and RNA. Thymine is
found only in DNA. Uracil is found only in RNA. Purines refer to heterocyclic
bases that contain two rings like the compound purine. They include adenine
and guanine, present in both DNA and RNA.
Functions
Nucleotides have a big role in metabolism. Besides forming the structural units of
RNA and DNA, they are the main sources of chemical energy, such as Adenosine
TriphosPhate (ATP) and guanosine triphosphate. They play an important role in
cellular signalling. They are incorporated into important cofactors in enzymatic
reactions.

Check Your Progress


1. Define gene.
2. What is genome?
3. Give the molecular formula of Phosphoric acid.
4. What is the melting point of Pyrimidine?
5. What is sugar unit?
Self-Instructional
Material 227
Nucleic Acids
9.3 RIBONUCLEIC ACID (RNA) AND
DEOXYRIBONUCLEIC ACID (DNA)

NOTES RNA and DNA are polymers constituted of monomers called mononucleotide
units. These mononucleotide units are united together by intermolecular dehydration
reactions that make phosphate ester bonds. Specialized enzymes catalyse such
reactions.
9.3.1 Structure and Properties of DNA
In the early 1950s, four scientists, James Watson and Francis Crick at
Cambridge University and Maurice Wilkins and Rosalind Franklin at King’s
College, determined the true structure of DNA from data and X-ray pictures
of a molecule taken by Franklin. In 1953, Watson and Crick published a
paper in the scientific journal Nature describing their research. They showed
that DNA molecule is not only double-stranded, but also has two strands
attached around each other forming a coil, or helix. A double helix represents
the exact structure of DNA.
Except viruses, in most living organisms, genetic information is stored in
DNA. DNA resides in the nucleus of living cells. Four different nucleotide bases
occur in DNA, namely Adenine (A), Cytosine (C), Guanine (G) and Thymine (T).
The nucleotide bases of the DNA molecule form complementary pairs
with the nucleotides hydrogen bond to another nucleotide base in a strand of
DNA, opposite to the original. This bonding is specific, with adenine always
pairing with thymine (and vice versa) and guanine always pairing with cytosine
(and vice versa).
A unique feature of DNA molecule is that it is able to make exact copies of
itself, or is self-replicate. When an organism requires more DNA, such as during
reproduction or cell growth, a break in the hydrogen bonds between the nucleotide
bases takes place, leading to the separation of two single strands of DNA. New
complementary bases are brought in by the cell and paired up with each of the
two separate strands, thus forming two new, identical, double-stranded DNA
molecules.
DNA is present in cells in the long structures in the nucleus, also known
as chromosomes. These chromosomes duplicate before the cells divide and
this process is known as DNA replication. Chromosomes are condensed
thread-like structures of DNA. DNA is present in the eukaryotes inside the
cell nucleus.
A small amount of DNA is also available in cell organelles like mitochondria
or chloroplasts. On the other hand, in prokaryotes like bacteria and archaea,
Self-Instructional
228 Material
DNA is present in the cytoplasm. Chromatin proteins, such as histones, compact Nucleic Acids

and organize DNA into chromosomes. A DNA molecule consists of two long
polymers, which face in the opposite direction and are known as ‘anti-parallel’.
This means that in a double helix the direction of the nucleotides in one strand is
opposite to the direction of the other strand. NOTES
Every sugar moiety is connected to one of the four types of molecules known
as bases. Each stand has polarity, that is, a top and a bottom that imparts 3' (three
prime) and 5' (five prime) ends. Information is encoded within the sequence of
these bases along the backbone. The 5' end has a terminal phosphate group
while whereas the 3' end has a terminal hydroxyl group. Hydrogen bonds that
bind the bases attached to the two strands help in stabilizing the DNA double
helix. Figure 9.12 shows the DNA molecular topography.

Fig. 9.12 Molecular Topography of DNA

Chemically, DNA is a long polymer that is comprised of nucleotide units.


The DNA chain is 22–26 Å (Ångströms) wide and each nucleotide unit is 3.3 Å
long. DNA polymers are large molecules that may have millions of nucleotides.
DNA exists as paired molecules in living beings. The two polymers, or
strands, interweave like vines. This gives it the characteristic double helical shape.
A nitrogenous base bonded to a sugar is known as a nucleoside and a base bonded
to a sugar and one or more phosphate groups is known as a nucleotide. If multiple
nucleotides are bonded together, like in DNA, this polymer is called a
polynucleotide.
Nitrogenous Base + Sugar = Nucleoside

Nitrogenous Base + Sugar + Phosphate Group = Nucleotide

Multiple Nucleotides Linked Together = Polynucleotide

Figure 9.13 shows the chemical structure of DNA and the hydrogen bonds
are shown as dotted lines.
Self-Instructional
Material 229
Nucleic Acids

NOTES

Fig. 9.13 Chemical Structure of DNA

The backbone of the DNA strand consists of alternating phosphate and


sugar residues. The sugar in DNA is known as 2-deoxyribose, a pentose (five-
carbon) sugar. Pentose can be differentiated from ribose as shown in Figure 9.14.
In RNA ribose sugar is present in place of deoxyribose. The sugars join together
forming phosphodiester bonds between third and fifth carbon atoms of adjacent
sugar rings, as shown as Figure 9.15.

Fig. 9.14 Difference between 2-Deoxyribose Sugar and Ribose Sugar

Self-Instructional
230 Material
Nucleic Acids

NOTES

Fig. 9.15 Formation of Phosphodiester Bond and Polynucleotide

Bases in DNA
The bases in DNA are classified into two types:
x Purines: Examples of purines include adenine and guanine (fused five- and
six-membered heterocyclic compounds).
x Pyrimidines: Examples of pyrimidines include cytosine and thymine (six-
membered rings).
Uracil (U), a fifth pyrimidine base, usually replaces thymine in RNA and
differs from thymine because it lacks a methyl group on its ring.
Grooves
A twisted DNA molecule shows two types of grooves that are different based on
size. The major groove is 22 Å wide while the minor groove is 12 Å wide. Because
it is narrow, the edges of the bases are less accessible in the minor groove in
comparison to major groove that leads to lesser interaction. Proteins such as
transcription factors that can bind to specific sequences in double-stranded DNA
therefore make contacts to the sides of the bases exposed in the major groove
normally.
Base Pairing
Complementary base pairing is when each type of base on one strand establishes a
link with exactly one type of base on the other strand. Purines form hydrogen bonds
with pyrimidines. The arrangement of two nucleotides binding together across the
double helix is known as a base pair.
Self-Instructional
Material 231
Nucleic Acids The double helical DNA strands can be pulled apart like a zipper either by
mechanical force or by applying high temperature. The concept of ‘complementarity’
causes duplication of all information in the double-stranded sequence of a DNA
helix, which is essential in DNA replication. This reversible, specific interaction
NOTES between complementary base pairs is crucial for all DNA functions in living organisms
(Refer Figure 9.16). In this figure, non-covalent hydrogen bonds between the pairs
are shown as dashed lines.

(a) GC base pair with three hydrogen bonds

(b) AT base pair with two hydrogen bonds


Figure 9.16 Base Pairing

The two types of base pairs are able to form various numbers of hydrogen
bonds. The AT base pair forms two hydrogen bonds while the GC base pair forms
three hydrogen bonds. DNA that has high GC content is more stable than DNA that
has low GC content. The DNA double helix that separates easily tends to have a
high AT content. The strength of the interaction can be calculated by finding the
temperature that is needed to break hydrogen bonds. This is known as the melting
temperature or Tm value. When all the base pairs in a DNA double helix melt, the
strands separate and are there in solution as two completely independent molecules.
Sense and Anti-Sense
A DNA sequence is referred to as ‘Sense’ if its sequence is the same as that of a
messenger RNA copy that is translated into protein. Its opposite is known as the
‘Anti-Sense’ when the sequence is on the opposite strand. Some DNA sequences
in prokaryotes and eukaryotes, and more in plasmids and viruses, distort the
difference between sense and anti-sense strands by having overlapping genes.
Self-Instructional
232 Material
Supercoiling Nucleic Acids

The process of DNA supercoiling involves twisting DNA into a rope-like structure.
If the DNA is twisted in the direction of the helix, it is known as positive
supercoiling. The bases are held more tightly together but if they are twisted in the NOTES
opposite direction, it is known as negative supercoiling. This way the bases
come apart more easily. Most DNA have slightly negative supercoiling introduced
by enzymes called topoisomerases.
Alternate DNA Structures
The many conformations in which DNA exists include A-DNA, B-DNA and Z-
DNA (Refer Figure 9.17). The conformation adopted by DNA is dependent on
the hydration level, DNA sequence, amount and direction of supercoiling, chemical
modifications of bases, type and concentration of metal ions and the presence of
polyamines in solution.

Fig. 9.17 Structures of A, Z and B DNA (from Left to Right)

As compared with B-DNA, A-DNA forms a wider, right-handed spiral


with a shallow, wide minor groove and a narrower, deeper major groove. The A
form occurs under non-physiological conditions in partially dehydrated samples of
DNA. DNA segments, where the bases have been chemically modified by
methylation, may undergo a larger change in conformation and adopt the Z form.
Here, the strands turn about the helical axis in a left-handed spiral, the opposite of
the more common B form.
Base Modifications
It is supposed that base modifications are involved in packaging. Regions that
have low or no gene expression usually have high methylation levels of cytosine
bases (Refer Figure 9.18). Despite the significance of 5-methylcytosine, it can
deaminate to leave a thymine base. Methylated cytosines are therefore particularly
prone to mutations. Other base modifications include adenine methylation in bacteria
and the presence of 5-hydroxymethylcytosine in the brain. Deamination converts
5-methylcytosine into thymine.
Self-Instructional
Material 233
Nucleic Acids

NOTES
Cytosine 5-Methylcytosine Thymine

Fig. 9.18 Structure of Cytosine, 5-Methylcytosin

9.3.2 Structure and Properties of RNA


RNA has derived its name from the sugar group present in its backbone, that is,
ribose. Some of the features of DNA and RNA are common. However, there are
also features that differentiate DNA from RNA. Both have a sugar-phosphate
backbone with nucleotide bases attached to it. Adenine, cytosine and guanine are
common in both. However, RNA lacks thymine. It contains uracil in place of
thymine. Further, DNA is a double-stranded molecule, whereas RNA is a single-
stranded molecule.
RNA is the key genetic material used in microorganisms, such as viruses. It
also plays an important role in the production of proteins in other living organisms.
Due to its ablity to move around the cells of living organisms, it functions as a
genetic messenger, transmitting the information contained in the cell’s DNA to
other parts of the cell. This transmission helps in protein synthesis.
Ribonucleic Acid (RNA) is a biologically important molecule that consists
of a long chain of nucleotide units. Each nucleotide has a nitrogenous base, a
ribose sugar and a phosphate. RNA is transcribed from DNA by enzymes known
as RNA polymerases and is usually processed further by other enzymes. RNA is
considered very important in the synthesis of proteins. Messenger RNA (mRNA),
a type of RNA, carries information from DNA to ribosomes, made of proteins
and ribosomal RNAs that bind together to form a molecular machine that can
read mRNAs and translate the information they carry into proteins. Other RNAs
with diverse roles exist – in particular, these regulate which genes are expressed
and are also the genomes of most viruses.
Each nucleotide in RNA comprises ribose sugar, with carbons numbered
from 1' to 5'. A base is attached to the 1' position, generally Adenine (A), Cytosine
(C), Guanine (G) or Uracil (U). Adenine and Guanine are Purines while Cytosine
and Uracil are Pyrimidines.
A phosphate group is attached to the 3' position of one ribose and the
5' position of the next ribose. Phosphate groups have a negative charge at
physiological pH, making RNA a charged molecule. The bases may form hydrogen
bonds between Cytosine and Guanine, between Adenine and Uracil and between
Guanine and Uracil (Refer Figure 9.19).

Self-Instructional
234 Material
Nucleic Acids

NOTES

Fig. 9.19 RNA Nucleotide with Ribose Sugar, Phosphate and Base

The functional form of single-stranded RNA molecules frequently asks for a


certain tertiary structure. The basis for this structure is given by secondary structural
elements that are hydrogen bonds within the molecule. This causes many
recognizable secondary structure ‘domains’, such as hairpin loops, bulges and
internal loops. Since RNA is charged, metal ions, such as Mg2+ are required to
stabilize many secondary and tertiary structures.
Comparison with DNA
RNA and DNA are both nucleic acids, but differ in the following ways:
DNA RNA
1. Long polymer with a deoxyribose and 1. Polymer with a ribose and phosphate
phosphate backbone backbone
2. Four different bases: adenine, guanine, 2. Four different bases: adenine,
cytosine and thymine guanine, cytosine, and uracil
3. It is a nucleic acid containing the genetic 3. The main job of RNA is to transfer the
instructions used in the development and genetic code required creating proteins
functioning of living organisms from the nucleus to the ribosome. This
4. Stands for deoxyribonucleic acid process prevents the DNA from
5. A double- stranded molecule with a long leaving the nucleus, so that it stays
chain of nucleotides safe. Without RNA, proteins could
never be made.
6. Deoxyribose sugar in DNA is less
reactive because of C-H bonds. 4. Stands for ribonucleic acid.
7. Stable in alkaline conditions 5. A single-stranded molecule in most of
its biological roles and has a shorter
8. Has smaller grooves where the damaging
chain of nucleotides
enzyme can attach which makes it harder
for the enzyme to attack DNA. 6. Ribose sugar is more reactive because
of C-OH (hydroxyl) bonds.
9. The helix geometry of DNA is of B-
Form. 7. Not stable in alkaline conditions.
10. Can be damaged by exposure to Ultra- 8. has larger grooves which makes it
violet rays easier to be attacked by enzymes.
9. The helix geometry of RNA is of A-
Form.
10. RNA is more resistant to damage by
Ultra-violet rays.
Self-Instructional
Material 235
Nucleic Acids Most biologically active RNAs, such as mRNA, tRNA, rRNA, snRNAs
and other non-coding RNAs, have self-complementary sequences that enable
parts of the RNA to fold and pair with itself. This causes double helices to form.
Structural analysis of these RNAs shows that they are highly structured and their
NOTES structures, unlike DNA, do not consist of long double helices but rather collections
of short helices packed together into structures similar to that of proteins. Thus,
RNAs can attain chemical catalysis like enzymes do.
Types of RNA
There are three types of RNA, which are as follows:
Ribosomal RNA (rRNA) (Insoluble RNA): This form of RNA has the highest
molecular weight. It is the most abundant RNA of all types of RNAs. There are
two different types of RNA in the prokaryotic and in eukaryotic cells. In prokaryotes
the ribosome possesses two different subunits, large and small. Large subunits
(50 s) contains two types of rRNA;. 23 s and 5 s named upon the sedimentation
behaviour. The smaller subunit (30 s) contains 16 s types.
The ribosome of eukaryotes also possesses three different types of rRNAs
named on the sedimentation behaviour. Larger subunit contains 5 s and 28 s types.
The smaller subunit contains 18 s types.
The mammalian rRNA possesses 5 s, 5.8 s and 28 s types in larger subunit
and in smaller subunit it contains only one 18 s types. Figure 9.20 shows the
structure of rRNA.

Fig. 9.20 Structure of rRNA

Messenger RNA (mRNA) (Template RNA): Messenger RNA is the most


heterogeneous in size and stability. Its amount is 5 per cent of total RNA of the
cell. It is synthesized on the surface of the DNA template. This carries the genetic
information to assemble the amino acids from DNA to ribosomes, hence it is
called messenger RNA. In prokaryotes the mRNA is unstable rather than in
eukaryotes. The mRNA is complementary to the DNA template strand. In
Self-Instructional
236 Material
Nucleic Acids
eukaryotes there are specific 5’ cap, which is the recognition site for ribosome
subunit, whereas in prokaryotes there is no specific 5’ recognition site. As a result,
there can be many ribosomal binding sites in prokaryotes, each one giving rise to
a different type of protein.
NOTES
In mammals the 5’ cap is methylated that means a cap of 7-methyle guanosine
triphosphate is linked to the 5’ end. The 3’ end of mRNA possess 20 -250
nucleotides of adenylate residue, which is called poly A tail. This tail probably
works as a stabilizer in the cell. Figure 9.21 shows the structure of mRNA.

Fig. 9.21 Structure of mRNA

Transfer RNA (tRNA) (Soluble RNA): It is the smallest polymeric form of


RNA. It is nearly 15 per cent of total RNA of the cellular RNA. The most important
function of tRNA is to act as a specific carrier of activated amino acids to specific
site on the protein synthesis. Now, nearly 50 sequences are known. Robert W.
Holley presented the clover leaf model for the tRNA. Figure 9.22 shows the
structure of tRNA.
The common structural features of tRNA are as follows:
x All tRNAs have a common design, consisting of three folds. This gives
them a shape of a clover leaf.
x The molecular weight of all tRNA molecules range from 24,000 to 31,000.
x tRNAs contain 7-15 unusual bases. Many of them are methylate of
unmethylated derivatives of A, U, G, C.
x The 5’ end is phosphorylated and terminal residue is usually guanylate.
x The base sequence of 3’ end of all tRNAs is CCA.
x Nearly 60 per cent of the nucleotides in tRNAs bases are paired to form
double helices.
x There are 5 groups of bases which are not base paired. These forms loops
which are as follows:
o 3’ CCA Terminal Region
o The Ribothymine-Pseudouracil-Cytosine (TOC) Loop
o The Extra Arm or Little Loop
o The DiyHdrouracil Loop (DHU)
o Anticodon Loop
x The unique feature of all the tRNAs is that distance between CCA to
anticodon is constant.
Self-Instructional
Material 237
Nucleic Acids

NOTES

Fig. 9.22 Structure of tRNA

9.3.3 DNA is Genetic Material


There is direct evidence that DNA is genetic material. According to Frederick
Griffith (1928), there are two types of strains of dipococcus pneumonia:
x Smooth (Virulent Strain)
x Rough (Avirulent Strain)
Smooth strain has a specific polysaccharide capsule and the rough strain
lacks this specific capsule. Both these S and R strains are of many types designated
as SI, SII, SIII. and RI, RII, RIII , and so on. In his experiment, Griffith injected
laboratory mice with live RII strain and the mice suffered no illness. When he
injected the SIII strain the mice became ill and died. But when he injected heat-
killed SIII, the mice did not fall ill. When he injected a mixture of RII and heat-
killed SIII, the mice became ill and died, which he found strange. By conducting
a postmortem on the dead mice, he found that the heart of the mice contained both
RII and SIII strains.
On the basis of the fact that S form can mutate to R form but R form cannot
mutate to S form, he concluded that bacteria must have a transformation of the
living RII strain and restoration of the capsule in the presence of heat-killed SIII
strain.
Self-Instructional
238 Material
Nucleic Acids

NOTES

Fig. 9.23 Bacterial Transfomation

However, he could not understand the cause of bacterial transfomation. In


1944, Oswald Avery, Colin Macleod and Maclyn McCary demonstrated that it
was DNA.
Semi-Conservative Nature of DNA: The Watson and Crick’s model of DNA
structure and replication suggested that the replication is semi conservative.
According to this method, the two original polynucleotide strands of the duplex
will unwind and each will serve as a template for the new strand. Both duplexes
that result from replication should be hybrid in nature, each containing an old strand
derived from the original strand and a new strand which has been formed during
the replication process.
Since each of the two double helices conserves only one of the parent
polynucleotide strand, the process is said to be semiconservative.
Genetic Code: The DNA molecule is composed of three kinds of moieties
phosphoic acids, deoxyribose sugar and nitrogen base. The poly sugar phosphate
backbone is always the same but the nitrogenous bases differ from one segment
to another. Therefore, the information might depend on their sequence. The
sequences of nitrogen bases of a given segment of DNA molecule, have been
found to be identical to the linear sequence of amino acids sequence in protein
molecule.

Fig. 9.24 Genetic Code


Self-Instructional
Material 239
Nucleic Acids The properties of the genetic code are as follows:
x The code is a triplet codon: The nucleotides of mRNA are arranged as a
linear sequence of codons, each consisting of three successive nirogenous
bases.
NOTES
x The codon is non-overlapping: In translating mRNA molecules, the codons
do not overlap but read sequentially.
x The code is commaless: It means that after one amino acid is coded, the
second amino acid will be coded automatically by the next three letters.
x The code is non- ambiguous: Non-ambiguous code means that a particular
codon will always code for the same amino acids.
x The code has polarity: The code is always read in a fixed direction, i.e, 5'
3'direction.
x The code is degenerate: More than one codon may specify the same amino
acid, which is called degeneracy of code.
x Some codes act as start codons: In most organisms, the AUG codon act as
the start codon. In rare cases, GUG also serves as a start codon.
x Some codes act as stop codons: Three codon UAG, UAA ad UGA are the
chain stop or termination codon.
x The code is universal: Same genetic code is found valid for all organisms
ranging from bacteria to man.

Check Your Progress


6. What are the nucleic bases that occur in DNA?
7. What is base pair?
8. What is sense?
9. Name the two types of strains of dipococcus pneumonia.

9.4 SYNTHESIS AND DEGRADATION OF


PURINES AND PYRIMIDINES

Nucleotides perform a diversity of important functions in all living cells. The key
role of nucleotide is to act as building blocks of DNA and RNA biomolecule.
Two classes of nucleotides (Purines and Pyrimidines) are found in the cells.
Nucleotides are vital carriers of chemical energy—a role primarily of Adenosine
Triphosphate (ATP) and to some extent Guanine Triphosphate (GTP). Cofactors
such as NAD, FAD, S-Adenosyl Methionine, and Coenzyme A, as well as of
activated biosynthetic intermediates, such as UDP-Glucose and CDP-
Diacylglycerol are components of nucleotides. Some, such as cAMP and cGMP,
acts as cellular second messengers for signal transduction in different biochemical
Self-Instructional
pathways.
240 Material
Two types of pathways lead to formation of nucleotides (Refer Figure 9.25): Nucleic Acids

x The De Novo Pathways


x The Salvage Pathways.
The de novo pathways are the biochemical pathway where nucleotides are NOTES
synthesized new from simple precursor molecules. Salvage pathways used to
recover bases and nucleosides formed during the degradation of DNA and RNA
biomolecules. Both types of pathways are important in cellular metabolism and
are described in this unit.
Purine and Pyrimidine biosynthesis through de novo synthesis appear to be
nearly similar in almost all living organisms. The Purine ring structure is built up one
or a few atoms at a time, attached to ribose throughout the process. The Pyrimidine
ring is synthesized as orotate, attached to ribose phosphate, and then converted to
the common pyrimidine nucleotides required in nucleic acid synthesis.
In both pathways, PhosphoRibosyl PyroPhosphate (PRPP) is important.
An amino acid is an important precursor in each type of pathway: Glycine for
Purines and Aspartate for Pyrimidines. Glutamine is the most important source of
amino groups in five different steps in the de novo pathways. Aspartate also serves
as the source of an amino group in the Purine pathways, in two peculiar steps.

Fig. 9.25 Pathways of Nucleotide Metabolism

9.4.1 Purine Nucleotide Synthesis


Adenosine 5’-MonoPhosphate (AMP; adenylate) and Guanosine 5´-
MonoPhosphate (GMP; guanylate are the two parent purine nucleotides of nucleic
acids which contains the purine bases namely, adenine and guanine. The origin of
the carbon and nitrogen atoms of the purine ring system is demonstrated in
Figure 9.26 which was identified using isotopic tracer testing in birds by John
Buchanan. In the year 1950s, Purine biosynthesis pathway in details was studied
out mainly by Buchanan and G. Robert Greenberg.
Self-Instructional
Material 241
Nucleic Acids

NOTES

Fig. 9.26 Origin of Ring Atoms of Purine

De Novo Synthesis of Purine


The purine (adenine and guanine) synthesis starts with Inosine -5’-MonoPhosphate
(IMP). IMP is synthesized in 11 steps from the simple precursors.
Step 1: Activation of Ribose -5-Phosphate Activation and PRPP Formation
Ribose 5-phosphate which is formed during the carbohydrate metabolism through
Hexose Monophosphate Shunt pathway, acts as preliminary material for Purine
nucleotide synthesis. PhosophoRibosyl PyroPhosphate (PRPP) is formed when
Ribose 5-phosphate reacts with ATP molecule in the presence of PRPP Synthetase
enzyme.

Step 2: Acquirement of N9 Atom of Purine


The amide nitrogen present in the Glutamine get transferred to PRPP which in
result 5- phosphoribosylamine is formed by replacing pyrophosphate. PRPP
glutamyl amidotransferase enzyme activity is monitored by feedback inhibition of
nucleotides (AMP, GMP and IMP).

Self-Instructional
242 Material
Nucleic Acids

NOTES

Step 3: Acquirement of C4, C5 and N7 Atom of Purine


GlycinAmide Ribotide (GAR) or Glycinamide ribosyl 5- phosphate is produced
in the presence of ATP when phosphoribosylamine reacts with glycine .

Step 4: Acquirement of C8 Atom of Purine


FormylGlycinAmide Ribosyl 5-phosphate (FGAR) is produced when N5, N10
formyl tetrahydrofolate donates the formyl group in the presence of Formyl
Transferase enzyme.

Self-Instructional
Material 243
Nucleic Acids Step 5: Acquisition of N3 Atom of Purine
FormylGlycinAmideine Ribosyl 5- Phosphate (FGAM) is formed from FGAR
when Glutamine shifts the second amido amino group.
NOTES

Step 6: Formation of Purine Imidazole Ring


5-AminoImidazole Ribosyl 5-phosphate (AIR) is formed through an ATP
dependent reaction.

Step 7: Acquirement of C6 Atom of Purine


Aminoimidazole carboxylate ribosyl 5-phosphate is produced through carboxylation
reaction (Incorporation of CO2). The majority of the carboxylation reaction requires
the vitamin biotin and /or ATP but this reaction does not require any additional
cofactors/coenzyme.

Step 8: Acquisition of N1 Atom of Purine


Aminoimidazole 4-succinyl carboxamide ribosyl 5- phosphate is produced when
Aspartate condenses with aminoimidazole carboxylate ribosyl 5-phosphate.

Self-Instructional
244 Material
Nucleic Acids

NOTES

Step 9: Removal of Fumarate


Aminoimidazole 4-carboxamide ribosyl 5-phosphate is formed when
Adenosuccinase cut off to fumarte and only the amino group of aspartate is
preserved.

Step 10: Acquirement of C2 Atom of Purine


Formimidoimidazole 4-carboxamide ribosyl 5-phosphate is produced when N10
formyl tetrahydrofolate donates a one-carbon moiety. All the nitrogen and carbon
atoms of Purine ring are donated by the particular sources within this reaction.

Self-Instructional
Material 245
Nucleic Acids Step 11: Cyclization to form IMP
IMP cyclohydrolase catalyzed the final reaction by the removal of water molecule
leading to the formation of ring closure structure of Inosine MonoPhosphate (IMP)
NOTES from formimidoimidazole ribosyl-5-P.

IMP is transformed into Guanine as GMP and adenine as AMP. AMP varies
from IMP in substitute of its 6-keto group by an amino group. IMP is the instant
precursor for the production of AMP and GMP. The amino group of aspartate is
enzymatically connected to IMP (C6 of Purine) with the assistance of GTP
hydrolysis to form adenylosuccinate. Adenyl succinate is enzymatically transformed
to AMP by the elimination of Fumarate group. For the production of GMP, IMP
undertakes NAD+ dependent dehydrogenation to produce Xanthosine
monophosphate (XMP). Glutamine then transports amide nitrogen to XMP to
form GMP (Refer Figure 9.27).

Fig. 9.27 Biosynthesis of AMP and GMP from IMP


Self-Instructional
246 Material
Salvage Pathway of Purine Synthesis Nucleic Acids

The formation of Purine in the cells occurs through salvage pathways. The turnover
of nucleic acids (particularly RNA) in the majority of the cells liberates adenine,
guanine and hypoxanthine. These free purines are retransformed to their consequent NOTES
nucleotides through salvage pathways. These pathways are varied in numerous
organisms in contrast to the de novo purine nucleotide synthesis pathway which is
analogous in approximately all the organisms.
In mammals, Adenine PhosphoRibosylTranferase (APRT) and
Hypoxanthine–Guanine PhosphoRibosylTranferase (HGPRT) are the two different
enzymes which are involved in Purines salvage pathway. The formation of AMP is
mediated through APRT using PRPP. The similar reaction for both hypoxanthine
and guanine is catalyzed by HGPRT enzyme.

Nucleoside Monophosphates are Converted to Nucleoside Triphosphates


The Nucleotides formed through salvage pathway are usually transformed to
nucleoside triphosphates. The conversion pathways are mostly common to all
cells. The enzyme adenylate kinase gets involved in the phosphorylation of AMP
to ADP as in the reversible reaction illustrated below:

By the glycolytic enzymes or through oxidative phosphorylation, ADP so


formed is phosphorylated to ATP. By the action of a class of enzymes called
nucleoside monophosphate kinases, ATP also bring about the production of other
nucleoside diphosphates. Nucleoside monophosphate kinases are normally specific
for a particular base but are nonspecific for the (ribose or deoxyribose) sugar.

Self-Instructional
Material 247
Nucleic Acids The well-organized cellular systems for rephosphorylating ADP to ATP tend
to pull this reaction in the direction of products. By the action of a ubiquitous
enzyme, nucleoside diphosphate kinase, nucleoside diphosphates are converted
into nucleoside triphosphates.
NOTES

Regulation of Purine Nucleotide Biosynthesis


To meet the cellular demands, the synthesis of purine nucleotide is well coordinated
in living system. The regulation of purine synthesis in large extent is maintained by
intracellular concentration of PRPP. The synthesis of purine is also dependent on
the availability of the enzyme PRPP Synthetase and ribose 5 phosphate.
PRPP glutamyl amidotransferse is maintained by a feedback mechanism by
purine nucleotides. If AMP and GMP are presented in sufficient quantity to meet
up the cellular necessities, their production is turned off at the amidotransferase
reaction. Another significant phase of regulation is in the conversion of IMP to
AMP & GMP. GMP inhibits IMP dehydrogenase while AMP inhibit
adenylosuccinate synthetase. Therefore, GMP and AMP control their particular
synthesis from IMP by a feedback mechanism. The regulation of interconversion
of IMP to AMP and GMP in represented in Figure 9.28. The solid line represents
the metabolic flow and dash line shows the positive and negative feedback
regulation.

Fig. 9.28 Regulation of Interconversion of IMP to AMP and GMP

9.4.2 Pyrimidine Synthesis


The biosynthesis of pyrimidine is simple than that of purine. Figure 9.29 shows the
source of different atoms in a pyrimidine skeleton which were identified by radio
labeling studies. The N1, C6, C5 and C4 atoms are derived from aspartate, N3
from glutamine and C2 from bicarbonate (HCO3–). In pyrimidine nucleotide
synthesis, the ring is completed before being linked to ribose-5-phosphate.
Self-Instructional
248 Material
Nucleic Acids

NOTES

Fig. 9.29 Origin of Ring Atoms of Pyrimidine

De Novo Synthesis of Uridine Monophosphate (UMP)


Uridine MonoPhosphate (UMP) is also act as the precursor of CMP. UMP is
synthesized in 6 steps.
Step 1: Formation of Carbomyl Phosphate
Carbomyl phosphate is formed by the hydrolysis of two molecules of ATPs,
bicarbonate and amide nitrogen of glutamine. The reaction is catalysed by
CPS –II.

Step 2: Synthesis of Carbomyl Aspartate


Carbomyl phosphate condenses with aspartate to from carbomylaspartate, cataylsed
by aspartatetranscarbomylase.

Self-Instructional
Material 249
Nucleic Acids Step 3: Ring Closure and Dihydroorolate Formation
By the elimination (condensation) reaction, the carbamoyl aspartate is converted
to a ring compound dihydroorotate. The reaction is catalyzed by dihydroorotase.
NOTES

Step 4: Oxidation of Dihydroorotate


Removal of hydrogen atoms from C5 and C6, by dihydroorotate dehydrogenase
and orotate is formed.

Step 5: Acquisition of the Ribo Phosphate Moiety


Orotate reacts with PRPP to produce Orotidine -5’-MonoPhosphate (OMP).
The reaction is catalysed by orotate phosphoribosyl transferase.

Step 6: Decarboxylation of UMP


OMP is decarboxylated forming UMP. UMP is the first true pyrimidine
Ribonucleotide

Self-Instructional
250 Material
Nucleic Acids

NOTES

Synthesis of UTP
Phosphorylation of UMP forms UDP and UTP, with the help of ATP.

Fig. 9.30 Synthesis of UTP

Synthesis of CTP
CTP is synthesized by the amination of UTP by the enzyme CTP synthase. In
animals, amino group is donated by glutamine and in bacteria; amino group is
donated by ammonia.
Synthesis of Thymine
Thymine is a methylated uracil and is synthesized as dTTP from UMP by
methylation. First dUTP is hydrolyzed to dUMP and PPi by the enzyme dUTP
diphosphohydrolase (dUTPase).

dUMP is then methylated to form dTMP. dTMP is then phosphorylated


with ATP rounds to form dTTP.
Self-Instructional
Material 251
Nucleic Acids Salvage Pathway of Pyrimidine Nucleotide Synthesis
Similar to purines, pyrimidines are also recovered from the derivative intermediates
of nucleic acids such as DNA and RNA. The recoveries of pyrimidines are
NOTES catalysed by the enzyme pyrimidine phosphoribosyltranferase. PRPP is utilized as
a source of ribose -5-phosphate by the enzyme pyrimidine phosphoribosyltranferase

Regulation of Pyrimidine Synthesis


CPSII, aspartate transcarbomylase and dihydrooratase are present as multienzyme
complex. Orotate phosphoribosyl transferase and OMP decarboxylase are present
as single functional enzyme.Due to cluster of those enzymes, the synthesis is well
coordinated. Dihydroorotate dehydrogenase is mitochondrial enzyme (CPSII and
aspartate transcarbomylase) and (OPRTransferase and OMP-decarboxylase) are
sensitive to allosteric regulation. CPSII is main regulatory enzyme in mammalian
cells. CPS II – is inhibited by UTP and activated by PRPP. Aspartate
transcarbomylase is the main regulatory enzyme in prokaryotes which is inhibited
by CTP and activated by ATP. The requirement of ATP for CTP synthesis and
stimulatory effect of GTP on CTP synthase ensures a balanced synthesis of purines
and pyrimidines.
9.4.3 Degradation of Purines and Pyrimidines
Purine nucleotides are degraded by a pathway in which they lose their phosphate
through the action of 5’- nucleotidase (Refer Figure 9.31).Adenylate yields
nucleoside, which is deaminated to inosine by adenosine deaminase, and inosine
is hydrolyzed to hypoxanthine (its purine base) and D-ribose. Hypoxanthine is
change to xanthine then into uric acid by xanthine oxidase enzyme, a flavoenzyme
with an atom of molybdenum and four iron-sulfur centers in its prosthetic group.
Molecular oxygen is the electron acceptor during this complicated reaction.
GMP destructive metabolism conjointly yields uric acid as end result. GMP
is initial hydrolyzed to nucleoside, which is then cleaved to free guanine. Guanine
undergoes hydrolytic removal of its amino to yield organic compound, which is
converted to uric acid by xanthine oxidase. Uric acid is that the excreted end
result of purine destructive metabolism in primates, birds, and some other animals.
Self-Instructional
252 Material
A healthy adult human excretes uric acid at a rate of about 0.6 g/24 h; the excreted Nucleic Acids

product arises in part from ingested purines and in part from turnover of the Purine
nucleotides of nucleic acids. In most mammals and plenty of different vertebrates,
uric acid is more degraded to allantoin by the action of urate oxidase enzyme.
NOTES

Fig. 9.31 Catabolism of Purine Nucleotides

Degradation of Pyrimidines
The pathways for degradation of pyrimidines generally lead to NH4+ production
and thus to urea synthesis. Thymine, for example, is degraded to
methylmalonylsemialdehyde (Refer Figure 9.32), an intermediate of valine
catabolism. It is further degraded through propionyl-CoA and methylmalonyl-CoA
to succinyl- CoA.
Self-Instructional
Material 253
Nucleic Acids

NOTES

Fig. 9.32 Catabolism of a Pyrimidine

Genetic aberrations in human purine metabolism have been found, some


with serious consequences. For example, Adenosine Deaminase (ADA) deficiency
leads to severe immunodeficiency disease in which T Lymphocytes and B
lymphocytes do not develop properly. Lack of ADA leads to a 100-fold increase
in the cellular concentration of dATP, a strong inhibitor of ribonucleotide reductase.
High levels of dATP produce a general deficiency of other dNTPs in T
Lymphocytes. The basis for B-Lymphocyte toxicity is less clear. Individuals with
ADA deficiency lack an effective immune system and do not survive unless isolated
in a sterile “bubble” environment. ADA deficiency is one of the first targets of
human gene therapy trials.
Self-Instructional
254 Material
Inhibitors of Nucleotide Synthesis Nucleic Acids

The structural analogs of folic acid (e.g.: methotrexate) inhibit the synthesis of
purine nucleotides and thus nucleic acids. These inhibitors also affect the proliferation
of normally growing cells. This causes many side-effects including anemia, baldness, NOTES
scaly skin, etc. They are widely used to control cancer. Sulfa drugs such as
Sulfonamides are the structural analogs of paraaminobenzoic acid (PABA)
indirectly reduce the synthesis of purines and therefore, the nucleic acids (DNA
and RNA). These can be used to inhibit the synthesis of folic acid by microgranisms.
Sulfonamides have no influence on humans, since folic acid is not synthesized and
is supplied through diet.

Check Your Progress


10. What does UMP stand for?
11. Which disease gets severe by the deficiency of simple Adenosine
DeAminase (ADA)?

9.5 ANSWERS TO CHECK YOUR RPROGRESS


QUESTIONS

1. A gene is a unit of heredity in a living organism. It normally resides on a


stretch of DNA that codes for a type of protein or for an RNA chain that
has a function in the organism. All living things depend on genes.
2. Genome is the hereditary information of an organism in totality. It is encoded
either in DNA or for several types of viruses in RNA. The genome includes
both the genes and the non-coding sequences of the DNA.
3. H3PO4.
4. 22oC.
5. The sugar unit contains a five-carbon atom sugar in its ring form. It is either
ribose in RNA or deoxyribose in DNA.
6. Four different nucleotide bases occur in DNA, namelyAdenine (A), Cytosine
(C), Guanine (G) and Thymine (T).
7. Complementary base pairing is when each type of base on one strand
establishes a link with exactly one type of base on the other strand. Purines
form hydrogen bonds with pyrimidines. The arrangement of two nucleotides
binding together across the double helix is known as a base pair.
8. A DNA sequence is referred to as ‘Sense’ if its sequence is the same as that
of a messenger RNA copy that is translated into protein. Its opposite is
known as the ‘Anti-Sense’.
9. (i) Smoth (virulent strain) (ii) Rough (avirulent strain).
Self-Instructional
Material 255
Nucleic Acids 10. Uridine Monophosphate.
11. Simple Adenosine DeAminase (ADA) deficiency leads to severe
immunodeficiency disease in which T Lymphocytes and B lymphocytes do
not develop properly.
NOTES

9.6 SUMMARY

x The origins of nucleic acids can be traced back to 1869, when Friedrich
Miescher, a Swiss chemist isolated nuclei from pus cells and found that they
contain a phosphate rich substance. He named this substance nuclein. In
1874, he isolated pure nucleic acid from salmon sperm nuclei. In 1899,
Altman gave it the name nucleic acid because it possesses acidic properties.
x A gene is a unit of heredity in a living organism.
x Genome is the hereditary information of an organism in totality. It is encoded
either in DNA or for several types of viruses in RNA.
x The genome includes both the genes and the non-coding sequences of the
DNA.
x There are two types of nucleic acids; DeoxyriboNucleic Acid (DNA) and
RiboNucleic Acid (RNA). These two types of nucleic acids are present in
all types of plants and animals.
x The viruses also contain DNA or RNA, but not both.
x Pyrimidines are heterocyclic aromatic compounds similar to pyridine and
benzene. They possess six membered ring and two N atoms and three
double bonds.
x Nucleosides are the compounds in which nitrogenous bases are conjugated
to the pentose sugars by DE-Glycosidic linkage.
x Nucleotides are the phosphoric esters of nucleosides.
x RNA and DNA are polymers constituted of monomers called
mononucleotide units.
x Two types of pathways lead to formation of nucleotides– the de novo
pathways and the salvage pathways.
x The de novo pathways are the biochemical pathway where nucleotides are
synthesized new from simple precursor molecules.
x Salvage pathways used to recover bases and nucleosides formed during
the degradation of DNA and RNA biomolecules. Both types of pathways
are important in cellular metabolism

Self-Instructional
256 Material
Nucleic Acids
9.8 KEY WORDS

x Nucleic acid: A complex organic substance present in living cells, especially


DNA or RNA, whose molecules consist of many nucleotides linked in a NOTES
long chain.
x RNA: Ribonucleic acid, a nucleic acid present in all living cells. Its principal
role is to act as a messenger carrying instructions from DNA for controlling
the synthesis of proteins, although in some viruses RNA rather than DNA
carries the genetic information.
x DNA: Deoxyribonucleic acid, a self-replicating material which is present in
nearly all living organisms as the main constituent of chromosomes. It is the
carrier of genetic information.
x Purine: A colourless crystalline compound with basic properties, forming
uric acid on oxidation.
x Pyrimidine: Any of a class of organic compounds of the heterocyclic series
characterized by a ring structure composed of four carbon atoms and two
nitrogen atoms.

9.9 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. Write a short note on pentose sugar.
2. What is the molecular structure of Uracil and Adenine?
3. Explain the process of DNA supercoiling.
4. Differentiate DNA and RNA.
5. Explain the types of RNA.
6. What is DNA as a genetic material?
Long Answer Questions
1. Explain the types of nucleic acids.
2. What are the types of nitrogenous bases present in nucleic acids? Explain.
3. What are nucleotides? Discuss their structure.
4. Discuss structure and properties of DNA.
5. Discuss structure and properties of RNA.
6. Explain synthesis of purines.
Self-Instructional
Material 257
Nucleic Acids 7. Explain synthesis of pyrimidines.
8. Explain degradation of purines and pyrimidines.

NOTES 9.10 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
258 Material
Enzymes

UNIT 10 ENZYMES
Structure NOTES
10.0 Introduction
10.1 Objectives
10.2 Classification, Chemical Nature and Properties of Enzymes
10.3 Factors Affecting Enzyme Activity
10.3.1 Factors Affecting the Rate of Enzyme Activity
10.4 Active Site of Enzyme
10.4.1 Structure of Active Sites
10.4.2 Models of Active Sites
10.4.3 Overview of Ligands
10.5 Answers to Check Your Progress Questions
10.6 Summary
10.7 Key Words
10.8 Self Assessment Questions and Exercises
10.9 Further Readings

10.0 INTRODUCTION

Enzymes are biological catalysts used in biochemical reactions. They are usually
proteins that provide alternative reaction pathways to give a boost to reactions.
These pathways have a lower level of activation energy. Although enzymes are
involved in the reaction, they do not undergo any changes and remain so at the end
of the reaction. This means, they can only alter the rate of reaction. In various
enzymatic reactions, substrates are the molecules at the beginning of the process,
which are then converted by the enzymes into different molecules. These are known
as products. Almost all chemical processes require enzymes to act as catalysts.
Almost all enzymes have protein and non-protein (called the co-factor) elements.
The protein part is usually globular.
In this unit, you will study about enzymes, classification and nomenclature of
mechanism of enzyme action, enzyme specificity and applications of enzymes in
classical diagnosis.

10.1 OBJECTIVES

After going through this unit, you will be able to:


x Get a comprehensive overview of enzymes
x Discuss how the enzymes are classified
x Explain the factors affecting enzymes activity
x Understand active sites of enzymes
Self-Instructional
Material 259
Enzymes
10.2 CLASSIFICATION, CHEMICAL NATURE AND
PROPERTIES OF ENZYMES

NOTES Enzymes can be named in various ways. For instance, in older days, enzymes
were named after people who discovered them. The names of enzymes like
pepsin, trypsin and cymotrypsin do not contain any information of the function
they are designed to carry out. The suffix ‘–ase’ has been added to the substrate
to name the enzyme; lipase acts on lipids, maltase acts on maltose, and so on.
These are known as the trivial names of enzymes that are not complete in
themselves.
After that, the International Union of Biochemistry (IUB) was established,
which appointed a commission on enzymes in 1961. This commission suggested
some principles for the classification and nomenclature of enzymes. The IUB system
is in use since 1964. They have divided enzymes into six major classes that
represent the general type reactions about the enzyme of that class. The following
are the major classes of enzymes:
x Oxidoreductase: Enzymes involved in oxidation reduction reaction, such
as, alcohol dehydrogenase.
x Transferase: Enzymes that catalyse the transfer of functional groups, such
as, hexokinase.
x Hydrolases: Enzymes that bring about hydrolysis of various compounds,
such as, lipase.
x Lyases: Enzymes that are specialized in the addition or removal of water,
ammonia, carbon dioxide, such as, aldolase.
x Isomerases: Enzymes that are involved in isomerization reactions, such
as, phosphohexose isomerase.
x Ligases: Enzymes that catalyse the synthetic reaction where two molecules
are joined by using ATP, such as, succinate thiokinase.
Each class is then further subdivided into many sub-classes. A four-digit Enzyme
Commission (EC) number is given to each enzyme representing the main class
(first digit), sub-class (second digit), sub-sub class (third digit) and the individual
enzyme (fourth digit). Each enzyme is given a specific name indicating the substrate,
coenzyme (if any) and the type of the reaction catalysed by the enzyme. Though
the IUB names are very hard to remember, trivial names and EC numbers are
often used.
Chemical Nature and Properties
All enzymes are proteinaceous in nature. However, now few RNA have been
observed to act as enzymes (ribozymes). Like proteins, enzymes have primary,
secondary and tertiary structures. They are also soluble in polar solvent and
insoluble in non-polar organic solvent. They are acidic or basic in nature
Self-Instructional depending on the ratio of base: acid. If this ratio is more than 1, the enzyme is
260 Material
basic; if the ratio is less than 1, it is acidic. They act as Zwitterions in their pI Enzymes

values. In their zwitterionic state or dipolar state, they are insoluble, precipitate
and have less mobility. Each enzyme has a specific tertiary structure which is
very important for its proper functioning. The holoenzyme is the functional unit
of enzymes, which is made of a protein part, apoenzyme, and a non-protein NOTES
organic part, known as coenzyme.
Holoenzyme Apoenzyme + Coenzyme

(Active Enzyme) (Protein Part) (Non-Protein Part)


When the non-protein part tightly binds to apoenzyme, it is known the asproathetic
group. The non–protein part can again be of two types – coenzyme and prosthetic
group. The coenzyme binds loosely to the protein part, whereas the prosthetic
group binds tightly to the apoenzyme. On dialysis, the coenzyme can be removed
from the apoenzyme, but the prosthetic group cannot be separated. Vitamins and
their derivatives act as coenzyme and act as second substrate.
Enzymes are of two types, depending on the number of polypeptides. They
are listed as follows:
x Monomeric Enzymes: A monomeric enzyme is made of one polypeptide
unit. Example: ribonuclease and trypsin.
x Oligomeric Enzymes: An oligomeric enzyme contains more than one
polypeptide chain. Examples: lactate dehydrogenase, aspartate
ranscarbamylase, and so on. Some enzymes are multi-enzyme complex.
They have specific sites to catalyse different reactions in a sequence. The
single specific site cannot complete its function. Only when they are in
complex, they can run the reaction in series to ultimately obtain the product.
The best examples of these types of enzymes are pyruvate dehyrogenase
and fatty acid synthase. The pyruvate dehyrogenase is a 3-enzyme complex,
whereas synthase is a 14- enzyme complex.
Sometimes enzymes require co–factors to complete their reactions. Most
of the inorganic minerals act as cofactors. Table 10.1 lists some important
ions and enzymes containing these ions.
Table 10.1 Ions and Enzymes
Ion Examples of Enzymes Containing This Ion
Cupric Cytochrome Oxidase
Ferrous or Ferric Catalase
Cytochrome (via Heme)
Nitrogenase
Hydrogenase
Magnesium Glucose 6-Phosphatase
Hexokinase
Manganese Arginase
Molybdenum Nitrate Reductase
Nickel Urease
Selenium Glutathione Peroxidase
Zinc Alcohol Dehydrogenase
Carbonic Anhydrase Self-Instructional
DNA Polymerase Material 261
Enzymes Functioning of Enzymes
The reactions taking place in biological systems require specific environments for
their occurrence. Enzymes provide this specific environment for their occurrence.
NOTES The salient feature of enzymes is catalysed reaction, meaning the reaction occurs
in a pocket on the enzyme known as active site. The molecule, that is, the substrate,
which is attached to the active site, is acted upon by the enzyme in this site. The
surface of the active site is lined with amino acid residues that have substitute
groups which bind the substrate, and chemical transformation is catalysed. The
active site separates substrates from the outer environment till the completion of
reaction. After that it is released in the solution.
Enzymes affect the rate of reaction and not the equilibrium of the reaction.
They decrease the activation energy to enhance reaction rates. Not only the enzymes
increase the reaction, but also organize and control energy released to be recovered
in other chemical forms and for other cell activities.

Check Your Progress


1. Define lyases.
2. What are ligases?
3. What are the types of enzymes, depending on the number of polypeptides?

10.3 FACTORS AFFECTING ENZYME ACTIVITY

Enzyme kinetics is the study of enzyme-catalysed chemical reactions, where the


reaction rate is calculated and the impact of varying the conditions of the reaction
examined. This process can reveal the catalytic mechanism of enzymes, their role
in metabolism and how their activity can be controlled. Kinetic studies aim to
determine the affinity with which the enzyme binds the substrate and the turnover
rate. Enzyme kinetics can also show the sequence in which substrates bind and the
sequence in which products are released.
It is important to examine the concept of enzyme kinetics for two reasons:
x It helps explain how enzymes work.
x It helps predict how enzymes behave in living organisms.
Enzymes are catalysts that do not change the chemical equilibrium of reactions.
The only way to utilize all substrate is to remove products after reaction. Enzymatic
catalysis is shown by the following equation:
E + S U ES U E + P
Where,
E: Enzyme
S: Substrate
Self-Instructional
262 Material
ES: Enzyme–Substrate Complex Enzymes

P: Product
The second part of the reaction is irreversible at the beginning of the reaction,
when the concentration of product is significantly lower than substrate concentration. NOTES
10.3.1 Factors Affecting the Rate of Enzyme Activity
The collision theory of chemical kinetics talks about the rate at which two molecules
will react when they approach within bond forming distance, or collide. Thus, it
could be said that the factors affecting the reaction rate should influence the frequency
or energy of collision between substrates.
Following are the important factors affecting the rate of enzyme-catalysed reactions:
x Concentration of Enzyme: If the concentration of enzyme is increased,
the rate of reaction also increases.
x Concentration of Substrate: The velocity of enzyme reaction increases
with an increase in the substrate concentration within the limiting range of
substrate levels. When a graph is plotted for velocity of enzyme-catalysed
reaction against the substrate concentration, a rectangular hyperbola graph
is obtained.
x Effect of Temperature: Velocity of enzymes increases with temperature
up to certain limits, and after that it declines. When a graph is plotted for
enzyme velocity against temperature, a bell-shaped curve is obtained. Q10
or temperature coefficient is a mathematical expression to describe the effect
of temperature on the velocity of enzyme catalysed reaction. Q10 is defined
as an increase in enzyme velocity when the temperature is increased by
10°C. When the temperature is increased, the activation energy of the
reactions are increased. This results in increased enzyme substrate interacts
and the reaction becomes faster. The optimum temperature of most enzymes
is between 40°C and 45°C. However, when temperature exceeds 50°C,
the enzyme gets denatured. As a result, it loses its activity.
x Effect of pH: Enzyme activity is affected by hydrogen ion concentration,
i.e., pH, and a similar bell-shaped curve as an effect of temperature is
obtained. Each enzyme has a specific optimum pH at which the rate of
reaction is maximum. Optimum pH is between pH 6 and 8 for maximum
enzyme activity. If pH is more than or less than optimum, the velocity of
reaction decreases, and at extreme condition the enzyme gets inactivated
because the extreme pH alters the ionic charges on the amino acid
(particularly at the active site), substrate and enzyme–substrate complex.
However, there are some exceptions, for example, pepsin (optimum pH
1–2), acid phosphatase (optimum pH 4–5) and alkaline phosphatase
(optimum pH 10–11).
x Effect of Product Concentration: Product concentration affects the
velocity of enzymes. This reduces the rate of reaction because the product
Self-Instructional
Material 263
Enzymes binds to the enzyme and enzyme activity is inhibited. However, this problem
is not seen in living systems because the product is continuously removed
after formation.

NOTES Check Your Progress


4. What is enzyme kinetics?
5. What is the effect of concentration of enzymes on rate of reaction?

10.4 ACTIVE SITE OF ENZYME

The active site is that area of an enzyme that has a big cleft surrounded by amino
acids and other side chains at the surface of the enzyme. This contains amino acid
residues and is the reason for substrate specificity and catalytic residues that act
as proton donors or acceptors or bind cofactors like Pyridoxal Pyrophosphate
(PLP), Thiamine Pyrophosphate (TPP) or Nicotinamide Adenine Diphosphate
(NAD). The active site is also the site of inhibition for enzymes. This site contains
the catalytic and binding sites and the recognition and binding of substrates is due
to the structural and chemical properties of the active site.
The interaction between enzymes and substrates promotes the formation
of the transition state structure by forming and breaking the bond. Enzymes help
a reaction to occur by stabilizing the transition state intermediate. This is done
by lowering the energy barrier or activation energy—the energy that is required
to promote the formation of transition state intermediate. The three-dimensional
cleft is formed by the groups that come from different part of the amino acid
sequences.
The active site is only a small part of the total enzyme volume. It enhances
the enzyme to bind to substrates and catalyse by many different weak interactions
because of its non-polar microenvironment. The weak interactions are the van
der Waals forces, hydrogen bonding and electrostatic interactions. The
arrangement of atoms in the active site is important for binding specificity. The
overall result is the speeding of the reaction process and increase in the rate of
reaction.
The enzyme active site is:
x The binding site for catalytic and inhibition reactions of enzyme and substrate.
x Structure of the active site.
x Its chemical characteristic are specific for the binding of a particular substrate.
The binding of substrates to enzyme causes changes in the chemical bonds
of the substrate and cause reactions that lead to the formation of products. The
products are released from the enzyme surface to regenerate the enzymes for
another reaction cycle. Thus, the enzymes remain unchanged and that is why they
are known as biocatalysts.
Self-Instructional
264 Material
10.4.1 Structure of Active Sites Enzymes

Active sites are in the shape of three-dimensional clefts that are composed of
amino acids from different residues of the primary amino acid sequence. Amino
acids that play a significant role in the binding specificity of the active site are NOTES
usually not adjacent to each other in the primary structure, but come near to each
other in order to form the active site because of the three-dimensional folding in
creating the tertiary structure.
The active site region is relatively small compared with the rest of the enzyme.
Similar to a ligand-binding site, majority of enzymes (non-binding amino acid
residues) exist primarily to serve as framework to support the structure of the
active site by providing correct orientation. The unique amino acids contained in
an active site promote specific interactions that are necessary for proper binding
and resulting catalysis. Enzyme specificity depends on the arrangement of atoms in
the active site.
Complementary shapes between enzyme and substrate(s) allow a greater
amount of weak non-covalent interactions which are electrostatic forces, van der
Waals forces, hydrogen bonding and hydrophobic interactions. Specific amino
acids also allow the formation of hydrogen bonds that shows the uniqueness of the
microenvironment for the active site.
10.4.2 Models of Active Sites
There are three different models that represent enzyme–substrate binding:
x Lock-and-Key Model
x Induced Fit Model
x Substrate Strain Model
Lock-and-Key Model: The Lock-and-Key Model was proposed by Emil
Fischer in 1890. This model presumes that there is a perfect fit between the
substrate and the active site—two molecules are complementary in shape. Lock-
and-Key is such a model that the active site of enzyme is a good fit for the substrate
that does not require change of enzyme structure after the enzyme binds substrate.
Thus, the active site has a pre-shaped template where only a specific substrate
can bind. This model failed because it could not explain many aspects of enzymatic
reactions and even could not explain the allosteric modulation. The substrate and
enzyme active site have complementary shapes.

Fig. 10.1 Lock-and-Key Model


Self-Instructional
Material 265
Enzymes Induced Fit Model: The Induced Fit Model involves the changing the
conformation of the active site to fit the substrate after binding. Also, in this model,
it was stated that there are amino acids that aid the correct substrate to bind to the
active site. This leads to lending shaping of the active site to the complementary
NOTES shape. The Induced Fit Model is such that the structure of the active site of enzyme
can be easily changed after binding of enzyme and substrate.
According to this model, the binding or active site is rigid as opposed to the
Lock-and-Key Model. The original or nascent active site has a different
confirmation. However, on interaction with the substrate, the enzyme active site
changes and substrate binds to enzyme very strongly. Thus, the substrate binding
to enzyme induces a better enzyme substrate complex. Now it is known that
amino acids of inactive sites are reoriented after interaction with enzymes and help
in activating catalytic activity of the enzyme. The binding in the active site involves
hydrogen bonding, hydrophobic interactions and temporary covalent bonds. The
active site will then stabilize the transition state intermediate to decrease the
activation energy. However, the intermediate is most likely unstable, allowing the
enzyme to release the substrate and return to the unbound state. The enzyme
active site forms a complementary shape to the substrate after binding.

Fig. 10.2 Induced-Fit Model


Substrate Strain Model: According to this model, the substrate is strained due
to induced change in the confirmation of the enzyme. When a Substrate binds to
a preformed active site, the enzyme induces a Strain to the Substrate. This Strained
Substrate helps in the formation of a new product. Nowadays, combinations of
both Induced Fit Model and Substrate Strain Models are used to explain the
mechanism of enzyme action.
10.4.3 Overview of Ligands
A binding site is a position on a protein that binds to an incoming molecule which
is smaller in size comparatively called a ligand. In proteins, binding sites are small
clefts on the tertiary structure where ligands bind to it using weak forces. There
are few amino acid residues that participate in binding the ligand. The remaining
residues in the protein act as a framework to provide correct conformation and
Self-Instructional
266 Material
orientation. Most binding sites have a concave shape, but convex and flat shapes Enzymes

are also sometimes found.


A Ligand-Binding site is a place of chemical specificity and affinity on protein
that binds or forms chemical bonds with other molecules and ions or protein ligands.
NOTES
The affinity of the binding of protein and ligand is a chemically attractive force
between the protein and ligand. As such, there can be competition between different
ligands for the same binding site of proteins, and the chemical reaction will result in
an equilibrium state between bonding and non-bonding ligands. The saturation of
the binding site is defined as the total number of binding sites that are occupied by
ligands per unit time.
The most common model of enzymatic binding sites is the induced fit model.
It differs from the more simple Lock-and-Key school of thought because the
induced fit model states that the substrate of an enzyme does not fit perfectly into
the binding site. With the lock and key model, it is assumed that the substrate is a
relatively rigid model that does not change its conformation and just states that the
substrate binds to the active site perfectly.
According to the induced fit model, the binding site of an enzyme is
complimentary to the transition state of the substrate in question, not the normal
substrate state. The enzyme stabilizes this transition state by having its NH3+ residues
stabilize the negative charge of the transition state substrate. This results in a dramatic
decrease in the activation energy required to bring forth the intended reaction. The
substrate is then converted to its product(s) by having the reaction go to equilibrium
quicker.
Properties Affecting Binding
The properties that affect binding are listed as follows:
x Complementarity: Molecular recognition depends on the tertiary structure
of enzymes which creates unique microenvironments in the active/binding
sites. These specialized microenvironments contribute to binding site
catalysis.
x Flexibility: Tertiary structure allows proteins to adapt to their ligands
(induced fit) and is essential for the vast diversity of biochemical functions.
x Surfaces: Binding sites can be concave, convex or flat; for small ligands, it
is clefts, pockets or cavities. Catalytic sites are often at domain and subunit
interfaces.
x Non-Covalent Forces: Non-covalent forces are also characteristic
properties of binding sites. Such characteristics are higher than average
amounts of exposed hydrophobic surface (small molecules—partly concave
and hydrophobic) and displacement of water can drive binding events.
x Affinity: Binding ability of the enzyme to the substrate (can be graphed as
partial pressure increases of the substrate against the affinity increases
Self-Instructional
Material 267
Enzymes (0–1.0); affinity of binding of protein and ligand is chemical attractive force
between the protein and ligand.

Check Your Progress


NOTES
6. What is an active site of an enzyme?
7. What is a binding site?

10.5 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. Enzymes that are specialized in the addition or removal of water, ammonia,


carbon dioxide, such as, aldolase.
2. Enzymes that catalyze the synthetic reaction where two molecules are joined
by using ATP, such as, succinate thiokinase.
3. Enzymes are of two types, depending on the number of polypeptides. They
are Monomeric enzymes and Oligomeric enzymes.
4. Enzyme kinetics is the study of enzyme-catalysed chemical reactions, where
the reaction rate is calculated and the impact of varying the conditions of
the reaction examined.
5. If the concentration of enzyme is increased, the rate of reaction also increases.
6. The active site is that area of an enzyme that has a big cleft surrounded by
amino acids and other side chains at the surface of the enzyme.
7. A binding site is a position on a protein that binds to an incoming molecule
which is smaller in size comparatively called a ligand.

10.6 SUMMARY

x Enzymes can be named in various ways. For instance, in older days, enzymes
were named after people who discovered them. The names of enzymes like
pepsin, trypsin and cymotrypsin do not contain any information of the function
they are designed to carry out.
x Oxidoreductase, transferase, hydrolases, lyases, isomerases and ligases are
the major classes of enzymes.
x All enzymes are proteinaceous in nature. However, now few RNA have
been observed to act as enzymes (ribozymes).
x Enzymes are of two types, depending on the number of polypeptides. They
are Monomeric enzymes and Oligomeric enzymes.
x Enzymes affect the rate of reaction and not the equilibrium of the reaction.
They decrease the activation energy to enhance reaction rates. Not only do
Self-Instructional
268 Material
enzymes increase the reaction, but also organize and control energy released Enzymes

to be recovered in other chemical forms and for other cell activities.


x Enzyme kinetics is the study of enzyme-catalysed chemical reactions, where
the reaction rate is calculated and the impact of varying the conditions of the
NOTES
reaction examined.
x Enzyme kinetics helps explain how enzymes work and predict how enzymes
behave in living organisms.
x The active site is that area of an enzyme that has a big cleft surrounded by
amino acids and other side chains at the surface of the enzyme.
x The active site is also the site of inhibition for enzymes. This site contains the
catalytic and binding sites and the recognition and binding of substrates is
due to the structural and chemical properties of the active site.
x A binding site is a position on a protein that binds to an incoming molecule
which is smaller in size comparatively called a ligand. In proteins, binding
sites are small clefts on the tertiary structure where ligands bind to it using
weak forces. There are few amino acid residues that participate in binding
the ligand.

10.7 KEY WORDS

x Active site: It is that area of an enzyme that has a big cleft surrounded by
amino acids and other side chains at the surface of the enzyme.
x Catalysts: They can be defined as substances that increase the velocity or
rate of a chemical reaction without themselves undergoing any change in
the overall process.
x Enzyme kinetics: It is the study of enzyme-catalyzed chemical reactions,
where the reaction rate is calculated and the impact of varying the conditions
of the reaction examined.
x Enzymes: They are biological catalysts produced by cells and responsible
for the high rate and specificity of one or more intracellular and extracellular
biochemical reactions.

10.8 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. What are oligomeric enzymes?
2. How do enzymes function?
3. How enzyme activity is get affected by pH?
Self-Instructional
Material 269
Enzymes 4. What is the effect of temperature on enzyme activity?
5. Explain about Lock-and-Key Model.
6. What is Induced Fit Model?
NOTES Long Answer Questions
1. Describe the major classes of enzymes.
2. Discuss chemical nature and properties of enzymes.
3. Explain the factors affecting the rate of enzyme activity.
4. Discuss different models that represent enzyme-substrate binding.
5. Give an overview of ligands.
6. Write the properties that affect binding.

10.9 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
270 Material
Enzyme Activity

UNIT 11 ENZYME ACTIVITY


Structure NOTES
11.0 Introduction
11.1 Objectives
11.2 Enzyme Activity
11.2.1 Factors Affecting Enzyme Activity
11.2.2 Michaelis-Menten Hypothesis
11.2.3 Models of Enzyme Action
11.3 Co-Enzymes and Co-Factors
11.4 Answers to Check Your Progress Questions
11.5 Summary
11.6 Key Words
11.7 Self Assessment Questions and Exercises
11.8 Further Readings

11.0 INTRODUCTION

Enzymes are macromolecular biological catalysts. Enzymes accelerate chemical


reactions. The molecules upon which enzymes may act are called substrates and
the enzyme converts the substrates into different molecules known as products.
Almost all metabolic processes in the cell need enzyme catalysis in order to occur
at rates fast enough to sustain life. Metabolic pathways depend upon enzymes to
catalyze individual steps. The study of enzymes is called enzymology and a new
field of pseudoenzyme analysis has recently grown up, recognising that during
evolution, some enzymes have lost the ability to carry out biological catalysis,
which is often reflected in their amino acid sequences and unusual ‘pseudocatalytic’
properties.
Enzymes are known to catalyze more than 5,000 biochemical reaction types.
Most enzymes are proteins, although a few are catalytic RNA molecules. The
latter are called ribozymes. Enzymes’ specificity comes from their unique three-
dimensional structures.
Like all catalysts, enzymes increase the reaction rate by lowering its activation
energy. Some enzymes can make their conversion of substrate to product occur
many millions of times faster. An extreme example is orotidine 5'-phosphate
decarboxylase, which allows a reaction that would otherwise take millions of years
to occur in milliseconds. Chemically, enzymes are like any catalyst and are not
consumed in chemical reactions, nor do they alter the equilibrium of a reaction.
Enzymes differ from most other catalysts by being much more specific. Enzyme
activity can be affected by other molecules: inhibitors are molecules that decrease
enzyme activity, and activators are molecules that increase activity. Many
Self-Instructional
Material 271
Enzyme Activity therapeutic drugs and poisons are enzyme inhibitors. An enzyme’s activity decreases
markedly outside its optimal temperature and pH, and many enzymes are
(permanently) denatured when exposed to excessive heat, losing their structure
and catalytic properties.
NOTES
Some enzymes are used commercially, for example, in the synthesis
of antibiotics. Some household products use enzymes to speed up chemical
reactions: enzymes in biological washing powders break down protein, starch
or fat stains on clothes, and enzymes in meat tenderizer break down proteins into
smaller molecules, making the meat easier to chew.
In this unit, you will study about enzyme activity, factors affecting enzyme
activity, co- enzymes and co-factors.

11.1 OBJECTIVES

After going through this unit, you will be able to:


x Understand what enzyme activity is
x Discuss the factors affecting enzyme activity
x Explain co- enzymes and co-factors

11.2 ENZYME ACTIVITY

Enzymes are macromolecular biological catalysts. Enzymes accelerate chemical


reactions. The molecules upon which enzymes may act are called substrates and
the enzyme converts the substrates into different molecules known as products.
Enzyme Activity = Moles of Substrate Converted Per Unit Time = Rate ×
Reaction Volume.
Enzyme activity is a measure of the quantity of active enzyme present and is
thus dependent on conditions, which should be specified. The SI unit is the katal,
1 katal = 1 mol s–1, but this is an excessively large unit. A more practical and
commonly used value is enzyme unit (U) = 1 µmol min–1. 1 U corresponds to
16.67 nanokatals.
Enzyme activity as given in katal generally refers to that of the assumed
natural target substrate of the enzyme. Enzyme activity can also be given as that of
certain standardized substrates, such as gelatin, then measured in Gelatin Digesting
Units (GDU), or milk proteins, then measured in Milk Clotting Units (MCU). The
units GDU and MCU are based on how fast one gram of the enzyme will digest
gelatin or milk proteins, respectively. 1 GDU equals approximately 1.5 MCU.
An increased amount of substrate will increase the rate of reaction with
enzymes, however once past a certain point, the rate of reaction will level out
because the amount of active sites available has stayed constant.
Self-Instructional
272 Material
Specific Activity Enzyme Activity

The specific activity of an enzyme is another common unit. This is the activity of an
enzyme per milligram of total protein (expressed in ìmol min–1 mg–1). Specific activity
gives a measurement of enzyme purity in the mixture. It is the micro moles of NOTES
product formed by an enzyme in a given amount of time (minutes) under given
conditions per milligram of total proteins. Specific activity is equal to the rate of
reaction multiplied by the volume of reaction divided by the mass of total protein.
The SI unit is katal/kg, but a more practical unit is ìmol/mgmin.
Specific activity is a measure of enzyme processivity (the capability of enzyme
to be processed), at a specific (usually saturating) substrate concentration, and is
usually constant for a pure enzyme.
An active site titration process can be done for the elimination of errors
arising from differences in cultivation batches and/or misfolded enzyme and similar
issues. This is a measure of the amount of active enzyme, calculated by for example,
titrating the amount of active sites present by employing an irreversible inhibitor.
The specific activity should then be expressed as µmol min–1 mg–1 active enzyme.
If the molecular weight of the enzyme is known, the turnover number, or ìmol
product per second per ìmol of active enzyme, can be calculated from the specific
activity. The turnover number can be visualized as the number of times each enzyme
molecule carries out its catalytic cycle per second.
Related Terminology
The rate of a reaction is the concentration of substrate disappearing (or product
produced) per unit time (mol L–1 s–1).
The % purity is 100% × (specific activity of enzyme sample / specific activity
of pure enzyme). The impure sample has lower specific activity because some of
the mass is not actually enzyme. If the specific activity of 100% pure enzyme is
known, then an impure sample will have a lower specific activity, allowing purity to
be calculated.
11.2.1 Factors Affecting Enzyme Activity
The function of enzyme is to increase the rate of reaction in a biological system to
sustain life. Consider the following reaction:
E+S ES EP E+P
Where E, S and P represent Enzyme, Substrate and Product, respectively. ES
and EP represent the complex of enzyme with substrate and product, respectively.
Figure 11.1 depicts the change in activation energy in a reaction taking
place without enzyme.

Self-Instructional
Material 273
Enzyme Activity

NOTES

Fig. 11.1 Process of a Reaction without an Enzyme

Symbols
In the Figure 11.1,
S = Free Energy of Substrate
P = Free Energy of the Product
TS = Transition State
G+= Activation Energy for the Two Reactions
G = Overall Standard Free Energy Change in Moving From S-P
According to the reaction coordinate in Figure 11.2, the free energy of P is
lower than that of S. Therefore, free energy change is negative for this reaction
that favours P. But there exists an energy barrier between the S and P, which is
required for the reaction to proceed in either direction. This energy barrier is
called the transition state. For the reaction to proceed, the reactant must reach this
energy barrier. The difference between the ground state and transition state is
known as activation energy.
Therefore, activation energy and rate of reaction are inversely proportional
to each other (i.e., if the activation energy is higher, the rate of reaction is slower
and vice versa). To overcome this problem, a catalyst is used to enhance the rate
of a chemical reaction or, in natural reactions, a catalyst is used by the system itself
in the form of enzymes (Refer Figure 11.2). Therefore, a catalyst enhances the
rate of a reaction by lowering the activation energy.

Self-Instructional
274 Material
Enzyme Activity

NOTES

Fig. 11.2 Process of a Reaction with an Enzyme

Symbols
In the figure 11.2,
E = Enzymes
ES = Enzyme and Substrate Complex
EP = Enzyme and Product Complex
The enzymes do not get used up in the reaction, and the equilibrium point
remains unaffected. They only enhance the rate of a reaction. When there are
more steps in the reaction, the overall rate is determined by the step with great
activation energy, which is known as the rate-limiting step. In Figure 11.2, the ES
and EP are intermediates and occupy the valleys in the curve.
11.2.2 Michaelis-Menten Hypothesis
Leonor Michaelis was a German-born American biochemist and physician famous
for his work in enzyme kinetics and Michaelis-Menten kinetics. Maud Leonora
Menten was a Canadian medical scientist who made significant contributions to
enzyme kinetics and histochemistry. They proposed a theory based on the following
assumptions:
x Only single substrate and single product are involved.
x The reaction proceeds essentially to completion.
x The concentration of substrate is much greater than that of the enzyme.
x An enzyme–substrate complex is formed as an intermediate step.
x The rate of decomposition of the substrate is proportional to the concentration
of the enzyme–substrate complex.
Consider the following reaction:
E+S ES E+P
Self-Instructional
Material 275
Enzyme Activity It is practically difficult to measure the quantity of ES or even S at any point
of the reaction. Therefore, Michaelis and Menten replaced these immeasurable
quantities with practically measurable quantities.

NOTES The following symbols are used to derive Michaelis-Menten equation:


(Et ) = Total Concentration of Enzyme
(S) = Total Concentration of Substrate
(ES) = Concentration of Enzyme Substrate Complex
(Et ) – (ES) = Concentration of Free Enzyme
The rate of reaction is proportional to the concentration (V) of the enzyme–
substrate complex.
V = k(ES) ...(1)
When total enzymes (Et) bound to the substrate, maximum rate of reaction
occurs. At this point the maximum concentration of ES (Vm) is equal to the total
concentration of enzyme:
Vm = k(Et) ...(2)
By dividing Equation (1) by Equation (2), Equation (3) is obtained:
V/Vm = (ES)/(Et) ...(3)
For the reversible reaction E + S ES, the equilibrium constant
for dissociation (km) of ES is as follows: (Et) – (ES) × (S)
(E t )–(ES)×(S)
Km = ...(4)
(ES)
(ES) × Km = (Et) × (S) – (ES) × (S)
(ES) × Km + (ES) × (S) = (Et) × (S)
(ES) × [Km + (S)] = (Et) × (S)
(ES)/(Et) = (S)/ Km + (S) ...(5)
Now, substitute the value of Equation (3) in Equation (5):
V/Vm = (S)/Km + (S)
V = Vm [(S)/Km + (S)] ...(6)
Km = (S) [Vm/V – 1] ...(7)
Where Km = Michaelis-Menten Constant
Equation (6) is the Michaelis-Menten Equation, which can be used to
calculate Km after experimentally determining the rate of reaction at various substrate
concentrations.
Michaelis-Menten constant is the measurement of the affinity of an enzyme
for its substrate.
Self-Instructional
276 Material
Km is numerically equal to the substrate concentrations or Km is equal to the Enzyme Activity

concentration of the substrate, which gives half the maximum velocity, Vm/2
(Refer Figure 11.3).

NOTES

Fig. 11.3 Rate of Reaction versus Substrate Concentration when V = ½ Vm

Lineweaver–Burk Methods
The determination of Km is a complex process; therefore, simpler methods were
devised by Lineweaver and Burk. Two such methods are discussed here:
First Method: This method involves plotting the kinetic data as the reciprocals of
V and (S). This double reciprocal plot was developed in 1934 by the Hans
Lineweaver and Dean Burk. The following equation has been derived from the
reciprocal of Michaelis-Menten equation:
1/V = Km + (S)/Vm × (S)
1/V = Km / Vm × 1/(S) + 1/Vm
After plotting the graph of 1/V versus 1/(S), a straight line is obtained
(Refer Figure 11.4). The slope of this curve corresponds to Km / Vm.

Fig. 11.4 The Graph of 1/V versus 1/(S)

Second Method: In this method, the Lineweaver-Burk equation is multiplied by


(S) on both sides to get the following equation:
(S)/V = Km/Vm + (S)/Vm
Self-Instructional
Material 277
Enzyme Activity The plot of (S)/V versus (S) gives a straight line (Refer Figure 11.5). The
intercept of the line on (S)/V axis is Km/Vm and the slope is 1/Vm.

NOTES

(S)/V Slope = 1/Vm

- Km

Km/Vm

Fig. 11.5 The Plot of (S)/V versus (S)

Active Site
As the substrate molecules are comparatively smaller than the enzyme molecules,
there are some specific regions on the enzymes for binding of substrate molecule.
These regions are known as active/catalytic/substrate sites.
The active site possesses some common features listed as follows:
x The active site is a small portion of the enzyme molecule.
x The active site is three dimensional.
x The arrangement of atoms in active site is well defined.
x The bond between the substrate molecule and the active site involves relative
weak forces.
x The active sites in the enzymes are grooves, which are devoid of water
content. In these grooves, the polar residues acquire special properties,
which are essential for catalysis and make the microenvironment in the
enzyme.
11.2.3 Models of Enzyme Action
The following models have been proposed to explain the mode of interaction of
enzymes and substrates:
x Fischer’s Lock and Key Model
x Koshland’s Induced Fit Model
Fischer’s Lock and Key Model
This model was proposed by Emil Fischer in 1898. According to this model, the
enzyme and the substrate unite in the same manner as a key in the lock. This
results in the formation of an enzyme–substrate complex (Refer Figure 11.6). The
enzyme-substrate complex is highly unstable and immediately decomposes to
produce the end products of the reaction and regenerate the free enzyme.
Self-Instructional
278 Material
Enzyme Activity

NOTES

Fig. 11.6 Fischer’s Lock and Key Model

Koshland’s Induced Fit Model


This model was proposed by the Koshland in 1958. He presumed that the enzyme
molecule does not retain its original shape and structure when it catalyses a reaction.
The substrate induces some configurational or geometrical changes in the active
site of the enzyme molecule (Refer Figure 11.7). Consequently, the enzyme molecule
fits the substrate molecule completely.

Fig. 11.7 Koshland’s Induced Fit Model

Isozyme, Ribozyme and Abzyme

Isozyme
The enzymes that occur in a number of differ-ent forms and differ from each other
chemically, immunologically and electrophoretically are called ‘Isoenzymes’ or
‘Isozymes’. Isozymes are enzymes that differ in amino acid sequence but catalyze
the same chemical reaction. These enzymes usually display different kinetic
parameters for example different KM values, or different regulatory properties.
The existence of isozymes permits the fine-tuning of metabolism to meet the particular
needs of a given tissue or developmental stage are isoforms (closely related variants)
of enzymes.
Isozymes are present in the serum and tissues of mammals, amphibians,
birds, insects, plants and unicellular organisms.
Examples
Isozymes of numerous dehydro-genases, and several oxidases, transaminases,
phosphatases, transphosphorylases, proteolytic en-zymes, aldolases.
Self-Instructional
Material 279
Enzyme Activity Ribozyme
A ribozyme is a RiboNucleic Acid (RNA) enzyme that catalyzes a chemical
reaction. The ribozyme catalyses specific reactions in a similar way to that of
NOTES protein enzymes.
Also called catalytic RNA, ribozymes are found in the ribosome where
they join amino acids together to form protein chains. Ribozymes also play a role
in other vital reactions such as RNA splicing, transfer RNA biosynthesis, and viral
replication.
The first ribozyme was discovered in the early 1980s and led to researchers
demonstrating that RNA functions both as a genetic material and as a biological
catalyst. This contributed to the worldwide hypothesis that RNA may have played
a crucial role in the evolution of self-replicating systems. This is referred to as the
RNA World Hypothesis and today, many scientists believe that ribozymes are
remnants of an ancient world that existed before the evolution of proteins. It is
thought that RNAs used to catalyse functions such as cleavage, replication and
RNA molecule ligation before proteins evolved and took over these catalytic
functions, which they could perform in a more efficient and versatile way.
Ribozymes have been extensively investigated by researchers to try and
determine their exact structure and function. Scientists have developed synthetic
ribozymes in the laboratory that are able to catalyze their own synthesis under
specific conditions. One example is the RNA polymerase ribozyme. Using
mutagenesis and selection, scientists have managed to develop and improve variants
of the Round-18 polymerase ribozyme from 2001. The best variant so far is called
B6.61, which can add up to 20 nucleotides to a primer template over a period of
24 hours. After 24 hours, the hydrolysis of its phosphodiester bonds causes the
ribozyme to decompose.
Ribozymes may also play an important role in therapeutic areas, acting as
molecules that can tailor specific RNA sequences, serving as biosensors and
providing a useful tool in areas such as gene research and functional genomics.
For example, strands of circular ribozymes celled viriods have been discovered
and these can have a devastating effect on plants. The viriods replicate by making
copies of themselves based on their own genome and their catalytic properties
enable them to undergo self-cleavage and send fragments off to colonize and
harm areas of a plant by proliferating and using the genetic material that the plant
itself needs. Researchers have now identified a site in these viriods that enables
them to self-cleave. The site is less than 30 nucleotides in length and has three
stems that form a central loop which is referred to as a ‘hammerhead.’ This structure
cleaves very specific RNA sequences to release viable RNA daughter strands.
Now, hammerheads of just 19 nucleotides in length have been synthesized that act
Self-Instructional
280 Material
Enzyme Activity
as highly specific catalysts. Similar ribozymes are also being made that could be
used to break up RNA viruses and RNA that is required for the transcription and
translation of DNA that contains mutations.
Such detailed studies of RNAs have led to rules being established regarding NOTES
how they achieve target recognition and based on those rules, scientists have
managed to adjust ribozymes so that they target and cleave new RNA molecule
targets that would not usually undergo cleavage by ribozymes. This raises the
exciting possibility that artificial ribozymes could be used as a therapeutic agents
to target RNA molecules that cause diseases such as HIV. In models of such
diseases, ribozymes have been successful at achieving this and a ribozyme that has
been shown to target and break up the RNA that makes up the HIV virus has
already been approved for testing in patients with HIV. In the future, ribozymes
may also be used as therapeutic agents in the correction of genetic disorders.
They could be used to eliminate abnormal proteins before they even exist by
attacking and breaking up the molecules of RNA that are needed for their translation
and transcription.
Abzyme
An abzyme is an antibody that expresses catalytic activity. A single molecule of an
antibody-enzyme, or abzyme, is capable of catalyzing the destruction of thousands
of target molecules. The efficiency of abzyme technology could permit treatments
with smaller doses of medicines at lower costs than are possible today. An abzyme
is used to lower the activation energy of a reaction allowing for the transition state
to be possible and the product to be formed. Abzymes are typically artificially
made by having the immune system make antibodies that bind to a molecule that
resembles the transition state (Transition State Analogue) of the catalytic process
that the researchers want to emulate. Therefore by creating this antibody, now
becoming a catalytic antibody allows for this antibody to act as an abzyme reducing
the activation energy of the reaction and allowing for the transition state to occur.
Abzymes however do occur naturally in the human body.
Uses in Medicine
Abzyme are currently being researched for the possible use against HIV infection.
The abzymes could target a specific site on the HIV infected cells that do not
mutate and then make the virus inert. This is an ongoing research project by the
University of Texas Medical School. By exploiting the highly specific antigen
binding properties of antibodies, experimental strategies have been made to
produce antibodies to catalyze that chemical reactions. These abzymes are chosen
from monoclonal antibodies which are created by immunizing mice with haptens
which mimic the transition states of enzyme-catalyzed reactions. The rate of this
reaction is promoted by enzyme catalysts that stabilize the transition state of this
Self-Instructional
Material 281
Enzyme Activity reaction, thereby decreasing the activation energy and allowing for more rapid
conversions of substrate product. To successfully create abzymes that are
complementary in structure to this transition state, mice were immunized with an
aminophosphonic acid hapten. The study of catalytic antibodies as a whole has
NOTES vastly increased current understanding of the mechanisms of enzyme catalysis and
represents another step forward in the attempts to create artificially engineered
biological enzymes.

Check Your Progress


1. What is the function of enzyme?
2. On what assumptions did Michaelis-Menten proposed a theory?
3. What is active site?
4. What are the common features of active site?

11.3 CO-ENZYMES AND CO-FACTORS

A co-enzyme is a substance that works with an enzyme to initiate or aid the function
of the enzyme. It may be considered a helper molecule for a biochemical reaction.
Co-enzymes are small, nonproteinaceous molecules that provide a transfer site
for a functioning enzyme. They are intermediate carriers of an atom or group of
atoms, allowing a reaction to occur. co-enzymes are not considered part of an
enzyme’s structure. They are sometimes referred to as cosubstrates. co-enzymes
cannot function on their own and do require the presence of an enzyme. Some
enzymes require several co-enzymes and co-factors. The B vitamins serve as co-
enzymes essential for enzymes to form fats, carbohydrates and proteins. An
example of a non-vitamin co-enzyme is S-adenosyl methionine, which transfers a
methyl group in bacteria as well as in eukaryotes and archaea.
Role of Co-Enzymes
Co-enzymes play a role in the functions of cells. Reactions within the cells work to
either break down nutrients or combine molecules for cellular activities that keep
the cells alive. Enzymes speed up these reactions. Without enzymes, these reactions
may not occur. Co-enzymes, in turn, support the functions of enzymes. They loosely
bind to enzymes to help them complete their activities. Co-enzymes are nonprotein,
organic molecules that facilitate the catalysis, or reaction, of its enzyme.
Co-enzymes are Co-Factors
Co-enzymes are one of two types of co-factors used by enzymes in these enzymatic
reactions. The other types of co-factors are inorganic ions. Magnesium, calcium
and potassium ions are commonly used with enzymes to speed up these reactions.
Self-Instructional
282 Material
Function of Co-Enzymes Enzyme Activity

Co-enzymes work by binding to the active side of the enzymes, the side that
works in the reaction. Since enzymes and co-enzymes are nonmetal organic
molecules, they bind together by forming covalent bonds. The co-enzymes share NOTES
electrons with the enzymes, rather than loss or gain electrons. When they form this
bond, they only help the reaction to occur by carrying and transferring electrons
through the reaction. Co-enzymes do not become integral parts of the enzymatic
reaction. Instead, the covalent bonds are broken at the end of the reaction, and
the co-enzyme returns back to free circulation within the cell until it is used again.
Enzyme Inhibitors
Enzyme inhibitors are molecules or compounds that bind to enzymes and result in
a decrease in their activity. An inhibitor can bind to an enzyme and stop a substrate
from entering the enzyme’s active site and/or prevent the enzyme from catalysing a
chemical reaction. The following are the categories of inhibitors:
x Reversible Inhibitors
x Irreversible Inhibitors
x Competitive Inhibitors
x Non-Competitive Inhibitors
x Un-Competitive Inhibitors
Reversible Inhibitors: These inhibitors are present naturally and can be
involved in metabolism regulation. For example, negative feedback caused by
inhibitors can help maintain homeostasis in cells. Other cellular enzyme inhibitors
include proteins that specifically bind to and inhibit enzyme targets. These inhibitors
are useful in eliminating harmful enzymes such as proteases and nucleases.
Reversible inhibitors bind non-covalently to enzymes, and many different types
of inhibition can occur depending on what the inhibitors bind to. The non-covalent
interactions between the inhibitors and enzymes include hydrogen bonds, hydrophobic
interactions and ionic bonds. Many of these weak bonds combine to produce strong
and specific binding. In contrast to substrates and irreversible inhibitors, reversible
inhibitors generally do not undergo chemical reactions when bound to the enzyme
and can be easily removed by dilution or dialysis (Refer Figure 11.8).

Fig 11.8 Reversible Inhibitors

Most reversible inhibitors follow the classic Michaelis–Menten scheme,


where an Enzyme (E) binds to its Substrate (S) to form an Enzyme–Substrate
Self-Instructional
Material 283
Enzyme Activity (ES) complex. Km is the Michaelis constant that corresponds to the concentration
of the substrate when the velocity is half the maximum. Vmax is the maximum velocity
of the enzyme.
Figure 11.9 shows the reaction between inhibitors and enzymes.
NOTES

Fig. 11.9 Interactions between Inhibitors and Enzymes

Irreversible Inhibitors: These inhibitors covalently bind to an enzyme,


cause chemical changes to the active sites of enzymes and cannot be reversed.
The main role of irreversible inhibitors includes modifying key amino acid residues
needed for enzymatic activity. They often contain reactive functional groups such
as aldehydes, alkenes or phenyl sulphonates. These electrophilic groups are
able to react with amino acid side chains to form covalent adducts. The amino
acid components are residues containing nucleophilic side chains such as hydroxyl
or sulfhydryl groups such as amino acids serine, cysteine, threonine or tyrosine
(Refer Figure 11.10).

Fig. 11.10 Irreversible Inhibitors

Irreversible inhibitors form a reversible non-covalent complex with the


enzyme (EI or ESI). Then, this complex reacts to produce the covalently modified
irreversible EI. The rate at which EI is formed is called the inactivation rate or
kinact. Binding of irreversible inhibitors can be prevented by competition with
either substrate or a second, reversible inhibitor since formation of EI may compete
with ES.
Self-Instructional
284 Material
Some reversible inhibitors can form irreversible products by binding very Enzyme Activity

tightly to their target enzyme. These tightly binding inhibitors show kinetics similar
to covalent irreversible inhibitors. The kinetic behavior is called slow-binding. Slow-
binding often involves a confirmational change as the enzyme ‘clams down’ around
the inhibitor molecule. Some examples of these slow-binding inhibitors include NOTES
drugs such as methotrexate and allopurinol.
Competitive Inhibitors: As the name suggests, these inhibitors compete
with substrates to bind to the enzyme at the same time. These inhibitors have an
affinity for the active site of an enzyme where the substrate also binds to and can
be overcome by increasing the concentrations of substrate out-competing the
inhibitor. Competitive inhibitors are often similar in structure to the real substrate.
Competitive inhibitors bind to active sites of enzymes and decrease the
amount of binding of substrate or ligand to enzyme, such that Km is increased and
Vmax not changed. The chemical reaction can be reversed by increasing concentration
of substrate. Competitive inhibitors can only bind to E and not to ES. They increase
Km by interfering with the binding of the substrate, but they do not affect Vmax
because the inhibitor does not change the catalysis in ES because it cannot bind to
ES (Refer Figure 11.11).

Fig. 11.11 Competitive Inhibitors

Uncompetitive Inhibitors: These inhibitors bind to the enzyme at the same


time as the enzyme’s substrate. However, the binding of the inhibitor affects the
binding of the substrate and vice versa. This type of inhibition cannot be overcome,
but can be reduced by increasing the concentrations of substrate. The inhibitor
usually follows an allosteric effect where it binds to a different site on the enzyme
than the substrate. This binding to an allosteric site changes the conformation of
the enzyme so that the affinity of the substrate for the active site is reduced.
Uncompetitive inhibitors bind to the enzyme–substrate complex to stop the
enzyme from reacting with the substrate to form a product. This works well at
higher substrate and enzyme concentrations that substrates are bonded to enzymes;
the binding results in decreasing concentration of substrate binding to enzyme, Km,
and Vmax, and increasing binding affinity of enzyme to substrate. These inhibitors
are able to bind to both E and ES, but their affinities for these two forms of the
enzyme are different. Therefore, these inhibitors increase Km and decrease Vmax
because they interfere with substrate binding and hamper catalysis in the ES
complex (Refer Figure 11.12).
Self-Instructional
Material 285
Enzyme Activity

NOTES

Fig. 11.12 Uncompetitive Inhibitors

Non-Competitive Inhibitors: These inhibitors bind to the active site and


reduce activity but do not affect the binding of the substrate. Therefore, the extent
of inhibition depends on the concentration of the substrate. These inhibitors bind
to other sites that are not active sites of enzymes that change the structure of
enzymes (Refer Figure 11.13). Non-competitive inhibitors have identical affinities
for E and ES. They do not change Km, but decrease Vmax.

Fig. 11.13 Noncompetitive Inhibitors

Enzyme Specificity
Enzyme activity is extremely specific in nature. There are three types of enzyme
activities and they are discussed as follows:
Stereospecificity or Optical Specificity: Stereoisomers compounds have
the same molecular formula but a different structural configuration. All enzymes
can work on only on single type of stereisomers. For example, trypsin only
hydrolyses polypeptides made of L-amino acid; hexiokinase and glucokinase only
metabolize D-glucose unit; amylase can hydrolyse D-Glycosidic bonds; cellulose
can hydrolyse E-Glycosidic bonds and so on.
Substrate specificity: The substrate binds to enzymes by weak non-
covalent bonds, for example, Van der Waals forces, electrostatic forces, hydrogen
bonding and hydrophobic interactions. Substrate-binding site or active sites have
a complementary shape that allows the substrate to bind. Substrate specificity is
of three types:
x Absolute Substrate Specificity: This type of enzymes can act exclusively
on a single type of substrates, for example, Enzyme Urease that cleaves
Urea to Ammonia and Carbon Dioxide andGlucokinase which converts
Glucose to Glucose-6-Phosphate.
Self-Instructional
286 Material
x Relative Substrate Specificity: Some enzymes have the ability to act on Enzyme Activity

structurally related substances but are specific towards groups or bonds


present on the structure.
x Group Specificity: Trypsin acts on Peptide Linkage having positively charged
NOTES
bonds, such as Arginine, Lysine, but not Proline. Chymotrypsin Hydrolyses
Peptide bonds involving bulky Hydrophobic residues, such as Phenylalanine,
Trypsin, tyrosine, but not Praline. Pepsin acts on Peptide bonds containing
Leucine, Phenylalanine, Tryptophan, Tyrosine, but not Proline.
Broad Specificity: They are specific for closely related substrates, for
example, Hexokinase, Glucose, Fructose, Mannose and Glucosamine.
Regulation of Enzyme Activity in Living Systems
Enzymes can be regulated by the following ways:
Allosteric Regulation: Some enzymes have sites other than active site.
Such sites are known as allosteric sites and such enzymes are known as allosteric
enzymes. Allosteric modulators/effectors/modifiers are substances that bind to the
allosteric site and act as regulators of enzyme activity to which they bind.
When a positive allosteric effector binds at the allosteric site, enzyme activity
is increased. Such allosteric sites are known as activator sites. On the other hand,
when a negative allosteric effector binds to the allosteric site, the activity of the
enzyme is reduced. Such sites are known as inhibitor site. Enzyme activity is
increased or reduced because of the change in the confirmation of the active site
of the enzyme by the non- covalent reversible binding of the effector molecule at
the allosteric site. Feedback inhibition or end product inhibition is a type of allosteric
inhibition where the end product inhibits the enzyme in a series of enzyme-catalysed
reactions of a metabolic pathway.
Activation of Latent Enzymes: Certain enzymes are produced in the
inactive or latent form. Such inactive enzymes are known as zymogen or
proenzymes. These are irreversibly activated by the breakdown of one or more
peptide bonds. Examples of such proenzymes are plasminogen, pepsinogen,
trypsinogen, procarboxypeptidases.
Some enzymes can exist in both active and inactive forms. These are
interconvertible, depending on the requirements of the body. The conversion from
active to inactive and from inactive to active occurs by reversible modification such
as phosphorylatoion, dephosphorylation and oxidation and reduction of disulfide
bonds. Examples of such enzymes are glycogen phosphorylase, which exists in two
forms Phosphorylase B (inactive form) and phosphorylase A (active form).
Compartmentalization of Metabolic Pathways: Usually, the anabolic
and catabolic pathways are operative in different cellular organelles to achieve
profit.
Control of Enzyme Synthesis: Rate-limiting enzymes are present in very
low concentration and have short half-lives. This helps in effective regulation of the
Self-Instructional
Material 287
Enzyme Activity enzyme level. There are two types of enzymes depending on their concentration
level:
x Constitutive enzymes (Housekeeping Enzymes): This enzyme level is
NOTES fairly constant and its level is not controlled.
x Adaptive Enzymes: This enzyme concentration can increase or decrease
depending on the body requirement and their level is regulated. Synthesis
of adaptive enzymes is regulated by the genes expressing them. There are
two terms used for genetic expression of protein synthesis and they are as
follows:
x Induction: Increased synthesis increases the concentration of enzymes
and is known as induction. For example, the insulin hormone increases
the level of glucokinase, phosphofructokinase.
x Repression: Decreased synthesis of enzyme because of a stopping
expression of the gene specific for an enzyme is known as repression.
Decrease in synthesis decreases the level of enzyme. For example,
pyruvate carboxylase is repressed by glucose.
Enzyme Degradation: Enzymes are degraded as are all proteins. Rate-
limiting enzymes are degraded rapidly as well as synthesized whenever required.
Barring some, all enzymes with long half-lives are slow in their activity.
Isoenzymes: Multiple forms of the same enzyme are known as isoenzymes.
These enzymes are generally tissue specific. Isoenzymes act on the same type of
substrate but they differ in Km, Vmax or both. This includes enzymes such as
glucokinase and hexokinase.

Check Your Progress


5. Define co-enzyme.
6. What is the role of co-enzymes?
7. What are enzyme inhibitors?
8. List the categories of inhibitors.

11.4 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. The function of enzyme is to increase the rate of reaction in a biological


system to sustain life.
2. Michaelis-Menten proposed a theory based on the following assumptions:
x Only single substrate and single product are involved.
Self-Instructional
288 Material
x The reaction proceeds essentially to completion. Enzyme Activity

x The concentration of substrate is much greater than that of the enzyme.


x An enzyme–substrate complex is formed as an intermediate step.
x The rate of decomposition of the substrate is proportional to the NOTES
concentration of the enzyme–substrate complex.
3. As the substrate molecules are comparatively smaller than the enzyme
molecules, there are some specific regions on the enzymes for binding of
substrate molecule. These regions are known as active/catalytic/substrate
sites.
4. The active site possesses some common features listed as follows:
x The active site is a small portion of the enzyme molecule.
x The active site is three dimensional.
x The arrangement of atoms in active site is well defined.
x The bond between the substrate molecule and the active site involves
relative weak forces.
x The active sites in the enzymes are grooves, which are devoid of water
content. In these grooves, the polar residues acquire special properties,
which are essent
5. A co-enzyme is a substance that works with an enzyme to initiate or aid the
function of the enzyme.
6. Co-enzymes play a role in the functions of cells. Reactions within the cells
work to either break down nutrients or combine molecules for cellular
activities that keep the cells alive. Enzymes speed up these reactions. Without
enzymes, these reactions may not occur. Co-enzymes, in turn, support the
functions of enzymes. They loosely bind to enzymes to help them complete
their activities. Co-enzymes are non-protein, organic molecules that facilitate
the catalysis, or reaction, of its enzyme.
7. Enzyme inhibitors are molecules or compounds that bind to enzymes and
result in a decrease in their activity.
8. The following are the categories of inhibitors:
x Reversible Inhibitors
x Irreversible Inhibitors
x Competitive Inhibitors
x Non-Competitive Inhibitors
x Un-Competitive Inhibitors

Self-Instructional
Material 289
Enzyme Activity
11.5 SUMMARY

x Enzymes are macromolecular biological catalysts.


NOTES x Enzymes accelerate chemical reactions.
x The molecules upon which enzymes may act are called substrates.
x The enzyme converts the substrates into different molecules known
as products.
x Enzyme Activity = Moles of Substrate Converted Per Unit Time = Rate ×
Reaction Volume.
x Enzyme activity is a measure of the quantity of active enzyme present and is
thus dependent on conditions, which should be specified.
x Enzyme activity as given in katal generally refers to that of the assumed
natural target substrate of the enzyme.
x An increased amount of substrate will increase the rate of reaction with
enzymes, however once past a certain point, the rate of reaction will level
out because the amount of active sites available has stayed constant.
x Specific activity is equal to the rate of reaction multiplied by the volume of
reaction divided by the mass of total protein.
x Specific activity is a measure of enzyme processivity (the capability of
enzyme to be processed), at a specific (usually saturating) substrate
concentration, and is usually constant for a pure enzyme.
x An active site titration process can be done for the elimination of errors
arising from differences in cultivation batches and/or misfolded enzyme and
similar issues.
x If the molecular weight of the enzyme is known, the turnover number, or
ìmol product per second per ìmol of active enzyme, can be calculated from
the specific activity.
x The function of enzyme is to increase the rate of reaction in a biological
system to sustain life.
x The enzymes do not get used up in the reaction, and the equilibrium point
remains unaffected. They only enhance the rate of a reaction.
x Leonor Michaelis was a German-born American biochemist and physician
famous for his work in enzyme kinetics and Michaelis-Menten kinetics.
x Maud Leonora Menten was a Canadian medical scientist who made
significant contributions to enzyme kinetics and histochemistry.
x Michaelis-Menten constant is the measurement of the affinity of an enzyme
for its substrate.
Self-Instructional
290 Material
x The determination of Km is a complex process; therefore, simpler methods Enzyme Activity

were devised by Lineweaver and Burk.


x The active sites in the enzymes are grooves, which are devoid of water
content. In these grooves, the polar residues acquire special properties, NOTES
which are essential for catalysis and make the microenvironment in the
enzyme.
x Fischer’s Lock and Key Model was proposed by Emil Fischer in 1898.
According to this model, the enzyme and the substrate unite in the same
manner as a key in the lock.
x Koshland’s Induced Fit Model was proposed by the Koshland in 1958.
He presumed that the enzyme molecule does not retain its original shape
and structure when it catalyses a reaction.
x The enzymes that occur in a number of different forms and differ from each
other chemically, immunologically and electrophoretically are called
‘Isoenzymes’ or ‘Isozymes’.
x Isozymes are enzymes that differ in amino acid sequence but catalyze the
same chemical reaction.
x The existence of isozymes permits the fine-tuning of metabolism to meet the
particular needs of a given tissue or developmental stage are isoforms (closely
related variants) of enzymes.
x Isozymes are present in the serum and tissues of mammals, amphibians,
birds, insects, plants and unicellular organisms.
x A ribozyme is a RiboNucleic Acid (RNA) enzyme that catalyzes a chemical
reaction. The ribozyme catalyses specific reactions in a similar way to that
of protein enzymes.
x The first ribozyme was discovered in the early 1980s and led to researchers
demonstrating that RNA functions both as a genetic material and as a
biological catalyst.
x Ribozymes have been extensively investigated by researchers to try and
determine their exact structure and function.
x Ribozymes may also play an important role in therapeutic areas, acting as
molecules that can tailor specific RNA sequences, serving as biosensors
and providing a useful tool in areas such as gene research and functional
genomics.
x An abzyme is an antibody that expresses catalytic activity. A single molecule
of an antibody-enzyme, or abzyme, is capable of catalyzing the destruction
of thousands of target molecules. The efficiency of abzyme technology could
permit treatments with smaller doses of medicines at lower costs than are
possible today.
Self-Instructional
Material 291
Enzyme Activity x An abzyme is used to lower the activation energy of a reaction allowing for
the transition state to be possible and the product to be formed.
x A co-enzyme is a substance that works with an enzyme to initiate or aid the
NOTES function of the enzyme. It may be considered a helper molecule for a
biochemical reaction.
x Co-enzymes are small, non-proteinaceous molecules that provide a transfer
site for a functioning enzyme. They are intermediate carriers of an atom or
group of atoms, allowing a reaction to occur.
x Co-enzymes are not considered part of an enzyme’s structure. They are
sometimes referred to as cosubstrates.
x Co-enzymes play a role in the functions of cells. Reactions within the cells
work to either break down nutrients or combine molecules for cellular
activities that keep the cells alive.
x Enzyme inhibitors are molecules or compounds that bind to enzymes and
result in a decrease in their activity.
x An inhibitor can bind to an enzyme and stop a substrate from entering the
enzyme’s active site and/or prevent the enzyme from catalysing a chemical
reaction
x The non-covalent interactions between the inhibitors and enzymes include
hydrogen bonds, hydrophobic interactions and ionic bonds. Many of these
weak bonds combine to produce strong and specific binding.
x Competitive inhibitors compete with substrates to bind to the enzyme at the
same time. These inhibitors have an affinity for the active site of an enzyme
where the substrate also binds to and can be overcome by increasing the
concentrations of substrate out-competing the inhibitor.
x Non-competitive inhibitors bind to the active site and reduce activity but do
not affect the binding of the substrate. Therefore, the extent of inhibition
depends on the concentration of the substrate.
x Stereoisomers compounds have the same molecular formula but a different
structural configuration.
x Absolute substrate specificity enzymes can act exclusively on a single type
of substrates, for example enzyme urease that cleaves urea to ammonia and
carbon dioxide and glucokinase which converts glucose to glucose-6-
phosphate.
x Some enzymes have sites other than active site. Such sites are known as
allosteric sites and such enzymes are known as allosteric enzymes.
x Allosteric modulators/effectors/modifiers are substances that bind to the
allosteric site and act as regulators of enzyme activity to which they bind.
Self-Instructional
292 Material
x Enzyme activity is increased or reduced because of the change in the Enzyme Activity

confirmation of the active site of the enzyme by the non- covalent reversible
binding of the effector molecule at the allosteric site.
x In adaptive enzymes the concentration can increase or decrease depending
NOTES
on the body requirement and their level is regulated.
x Increased synthesis increases the concentration of enzymes and is known
as induction. For example, the insulin hormone increases the level of
glucokinase, phosphofructokinase.
x Decreased synthesis of enzyme because of a stopping expression of the
gene specific for an enzyme is known as repression. Decrease in synthesis
decreases the level of enzyme. For example, pyruvate carboxylase is
repressed by glucose.
x Enzymes are degraded as are all proteins. Rate-limiting enzymes are
degraded rapidly as well as synthesized whenever required. Barring some,
all enzymes with long half-lives are slow in their activity.

11.6 KEY WORDS

x Enzymes: Enzymes are macromolecular biological catalysts and accelerate


chemical reactions.
x Substrates: The molecules upon which enzymes may act are called
substrates.
x Products: The enzyme converts the substrates into different molecules
known as products.
x Isozymes: The enzymes that occur in a number of different forms and
differ from each other chemically, immunologically and electrophoretically
are called ‘Isoenzymes’ or ‘Isozymes’.
x Abzyme: An abzyme is an antibody that expresses catalytic activity.
x Enzyme inhibitors: Enzyme inhibitors are molecules or compounds that
bind to enzymes and result in a decrease in their activity.
x Allosteric sites: Some enzymes have sites other than active site. Such
sites are known as allosteric sites.

11.7 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. Define the term enzymes.
2. What are the factors that affect enzyme activity?
Self-Instructional
Material 293
Enzyme Activity 3. What is active site?
4. How is abzyme helpful in field of medicines?
5. Give some of the roles of co-enzymes.
NOTES 6. Draw a well-labelled diagram of reversible inhibitor.
Long Answer Questions
1. What is enzyme activity? Explain the factors that affect enzyme activity.
2. Write a note on Michaelis-Menten Hypothesis.
3. Discuss about Lineweaver-Burk methods.
4. Explain about the methods of enzymes action.
5. Elaborate a note on co-enzymes and co-factors.
6. Discuss in detail about enzyme inhibitors.

11.8 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
294 Material
Hormones
BLOCK - IV
HORMONES, BUFFERS AND ELECTROLYTES
NOTES
UNIT 12 HORMONES
Structure
12.0 Introduction
12.1 Unit Objectives
12.2 Introduction of Hormones
12.2.1 Structure and Function of Hormones
12.3 General Classification of Hormones
12.4 Steroid Hormone Receptors
12.5 Peptide Hormones Receptor
12.6 Hormone Receptors and Intracellular Messengers
12.7 Insulin
12.8 Role of Glucagon
12.9 Epinephrine and their Mechanism for Various Endocrine
12.10 Regulatory Systems Mediated by Cyclic AMP
12.11 Answers To Check Your Progress Questions
12.12 Summary
12.13 Key Terms
12.14 Self Assessment Questions And Exercises
12.15 Further Readings

12.0 INTRODUCTION

Most glands in the human body deliver their secretions by the means of ducts
called exocrine glands. There are some other glands that produce chemical
substances that are directly secreted into the blood stream for transmission to
various target tissues. These are ductless or endocrine glands. The secretion of
endocrine glands is called as hormones. It is a chemical substance, which is
produced in one part of the body, enters the circulation and is carried to distant
target organs and tissues to modify their structures and functions. Hormones are
stimulating substances and act as body catalysts. The hormones catalyze and control
diverse metabolic processes. Despite their varying actions and different specificity’s
depending on the target organ, hormones act as body catalysts.
In this unit, you will study about the insulin, glucagons, epinephrine and
their mechanism, various endocrine and regulatory systems mediated by cyclic
AMP.

Self-Instructional
Material 295
Hormones
12.1 UNIT OBJECTIVES

After going through this unit, you will be able to:


NOTES x Describe the biochemical aspects of hormone
x Understand functions of harmones
x Explain the classification of harmones
x Understand the hormone receptors and intracellular messengers
x Identify protein kinase and phosphodiesterase
x Understand the role and mechanism of insulin, glucagons and epinephrine
x Describe the various endocrine and regulatory systems mediated by cyclic
AMP

12.2 INTRODUCTION OF HORMONES

The homeostatic adaptations an organism makes to a constantly changing


environment are in large part accomplished through alterations of the activity and
amount of proteins. Hormones provide a major means of facilitating these changes.
A hormone-receptor interaction results in generation of an intracellular signal that
can either regulate the activity of a select set of genes, thereby altering the amount
of certain proteins in the target cell, or affect the activity of specific proteins, including
enzymes and transporter or channel proteins. The signal can influence the location
of proteins in the cell and can affect general processes such as protein synthesis,
cell growth and replication, perhaps through effects on gene expression. Other
signaling molecules include cytokines, interleukins, growth factors and metabolites.
Excessive, deficient or inappropriate production and release of hormones and of
other regulatory molecules are major causes of disease.
All the steroid hormones are derived from cholesterol except Vitamin D.
They all consist of the same cyclopentanophenanthrene ring and atomic numbering
system as cholesterol does. Conversion of C27 cholesterol to the 18, 19 and 21
carbon steroid hormones consists of the rate-limiting, irreversible cleavage of a 6-
carbon residue from cholesterol, producing pregnenolone (C21) and
isocaproaldehyde. Popular names of steroid hormones are quite popular, but the
systematic nomenclature is just now gaining acceptance and familiarity with the
nomenclatures. Steroids that have 21 carbon atoms are systematically known as
pregnanes, whereas those steroids that contain 19 and 18 carbon atoms are termed
as androstanes and estranes, respectively.
12.2.1 Structure and Function of Hormones
Integration of body functions in humans and other higher organisms is carried out
through the nervous system, the immune system and the endocrine system. The
Self-Instructional
296 Material
endocrine system consists of a number of tissues that secrete their products in the Hormones

circulatory system and from there they get disseminated throughout the body,
regulating the function of distant tissues and maintaining the process of homeostasis.
In a separate but related system, exocrine tissues secrete their products in the
ducts and then outside the body or towards the intestinal tract. Basically, the NOTES
endocrine hormones are derived from the amino acids, peptides or sterols and to
act at sites distant from their tissue of origin. However, the latter definition has
started to blur as it is found that some secreted substances act at a distance (classical
endocrines), closer towards the cells, which secrete them (paracrines) or directly
on the cell that secreted them (autocrines). Insulin like Growth Factor -1(IGF-1)
that behaves as an endocrine, paracrine and autocrine, acts as a prime example of
this difficulty.
Hormones are usually present in the plasma and interstitial tissue at
concentrations in the range of 10–7M to 10–10M. Due to these low physiological
concentrations, sensitive protein receptors have come up in target tissues for sensing
the presence of weak signals. In addition, the systemic feedback mechanisms
have also evolved to regulate the endocrine hormone production.
Once a hormone gets secreted by an endocrine tissue, it usually binds with
a specific plasma protein carrier, with the complex being disseminated to distant
tissues. Plasma carrier proteins exist for all the classes of endocrine hormones.
Carrier proteins for peptide hormones prevent hormone destruction by plasma
proteases. Carriers for steroid and thyroid hormones permit these very hydrophobic
substances to be present in the plasma at concentrations several hundred-fold
greater than their solubility in water might permit. Carriers for small, hydrophilic
amino acid-derived hormones prevent their filtration through the renal glomerulus,
prolonging their circulating half-life greatly.
Those tissues that are capable of responding to endocrines have two
properties in common. These properties are as follows:
x They posses a receptor having very high affinity for hormone.
x The receptor is coupled to a process that regulates metabolism of the target
cells.
Receptors for most of the amino acid-derived hormones and all peptide
hormones are usually located on the plasma membrane. Activation of these
receptors by hormones (the first messenger) leads towards the intracellular
production of a second messenger, such as cAMP. It initiates the intracellular
biological response. Steroid and thyroid hormone are hydrophobic and diffuse
from their binding proteins in the plasma, across the plasma membrane to
intracellularly localize the receptors. The resultant complex of steroid and receptor
bind to response elements of nuclear DNA, regulating the production of mRNA
for specific proteins.
Self-Instructional
Material 297
Hormones
12.3 GENERAL CLASSIFICATION OF HORMONES

The following three categories of classification of hormones:


NOTES x According to Chemical Nature
x According to Origin
x According to Nature of Action
According to Chemical Nature: The hormones are classified into following
main classes according to the chemical structures:
x Steroid Hormones: Steroid hormones are the hormones which are the
derivative of cholesterol which includes sex hormones and hormones of the
adrenal cortex. They are comprised of three groups which include
glucocorticoids, mineralcorticoids and sex hormones (Testosterone,
Estrogen, and Progesterone).
Steroids also play roles in inflammatory responses, stress responses, bone
metabolism, cardiovascular fitness, behavior, cognition, and mood.
x Amine Hormones : Amino acid these are hormones derived from amino
acids. Many of the amino acid hormones are neurotransmitters. The
hormones derived from amino acids are thyroid hormones (T3, T4) and the
hormones of the adrenal medulla (epinephrine, norepinephrine).
x Peptide Hormones: Protein Hormones or peptide hormones are prepared
from polymers of amino acids. Most of these hormones encourage other
glands to create hormones. They are also significant in regulation of
metabolism, for exmaple, Oxytocin and vasopressin.
According to Origin: Mostly reproductive hormones are primarily derived from
four major organs or system (Hypothalamus, Anterior and posterior lobe of pituitary
gland, Gonads (testis and ovary including their interstitial tissues and corpus luteum),
Placenta and Uterus.
According to Nature of Action
x General Hormones: Growth hormone influence nearly all the body tissues,
similar is the case with Thyroid and Insulin hormones, hence they fall in
general category.
x Specific Hormones: these hormones affect functions of specific organs,
for example, FSH and androgens.
x Local Hormones: Prostaglandins, Acetyl cholin, Histamine act locally to
their site of production.
Chemical Classification of Hormones
The hormones are classified into following main classes according to the chemical
structures:
Self-Instructional
298 Material
Pitutary Hormones Hormones

x Oxytocin: The basic functions of oxytocin are as follows:


o It causes uterine contraction.
o It causes milk ejection in lactating females. NOTES
o It responds to suckling reflex and estradiol.
o It lowers steroid synthesis in testes.
x Vasopressin (AntiDiuretic Hormone, ADH): The major functions of
vasopression are as follows:
o It responds to osmoreceptor, which senses extracellular [Na+].
o It regulates blood pressure.
o It increases H2O re-absorption from distal tubules in kidney.
x Melanocyte-Stimulating Hormones (MSH): The major function of
melanocyte is pigmentation.
x Corticotropin (Adrenocorticotropin, ACTH)L: The major functions of
corticotrophin are as follows:
o It stimulates cells of adrenal gland.
o It increases the steroid synthesis and secretion.
x Lipotropin (LPH): Lipotropin basically increases the fatty acid release
from the adipocytes.
x Thyrotropin (Thyroid-Stimulating Hormone, TSH): It acts on thyroid
follicle cells to stimulate thyroid hormone synthesis.
x Growth Hormone (GH): The major functions of the growth hormone are
as follows:
o It acts as a general anabolic stimulant.
o It increases the release of Insulin-like Growth Factor-I (IGF-I).
o It leads to cell growth and bone sulfation.
x Prolactin (PRL): The major functions of prolactin are as follows:
o It stimulates the differentiation of secretory cells of mammary gland.
o It stimulates the milk synthesis.
x Luteinizing Hormone (LH): The major functions of luteinizing hormones
are as follows:
o It increases the Ovarian Progesterone synthesis.
o It acts on Leydig cells of testes to increase Testosterone synthesis.
o It releases and increases the interstitial cell development.
x Follicle-Stimulating Hormone (FSH): The major functions of Follicle
Stimulating Hormone are as follows:
Self-Instructional
Material 299
Hormones o It assists in the ovarian follicle development and ovulation.
o It increases the estrogen production.
o It acts on Sertoli cells of semi-ferous tubule to increase spermatogenesis.
NOTES Hypothalamic Hormones
x Somatostatin (SIF): It inhibits GH and TSH secretion.
Thyroid Hormones
x Thyroxine and Triiodothyronine: It responds to TSH and stimulates
oxidations in many cells.
x Calcitonin: It is produced in parafollicular C cells of the thyroid and it
regulates Ca2+ and Pi metabolism.
x Calcitonin Gene-Related Peptide (CGRP): It acts as a vasodilator.
Parathyroid Hormone
x Parathyroid Hormone (PTH): The basic functions of parathyroid hormone
are as follows:
o It regulates Ca2+ and Pi metabolism.
o It stimulates bone resorption.
o It increasesng serum [Ca2+].
o It stimulates Pi secretion through the kidneys.
Adipose Tissue Hormones
x Leptin: The major functions of leptin are as follows:
o It regulates the overall body weight by limiting the food intake
o It increases the energy expenditure.
o It regulates the neuroendocrine axis, inflammatory responses, blood
pressure and bone mass.
x Adiponectin: This hormone assists in increasing the major biological actions
of insulin sensitivity and fatty acid oxidation.
x Resistin: It induces the insulin resistance.
Hormones and Peptides of the Gut
x Glucagon-like Peptide 1 (GLP-1): The major functions of Glucagon are
as follows:
o It potentiates the glucose-dependent insulin secretion.
o It inhibits glucagon secretion.
o It inhibits gastric emptying.
x Glucose-Dependent Insulinotropic Polypeptide (GIP): It inhibits
secretion of gastric acid and it enhances the insulin secretion.
Self-Instructional
300 Material
x Ghrelin: The major functions of ghrelin are as follows: Hormones

o It assists in appetite stimulation.


o It stimulates the NPY release.
o It regulates the energy homeostasis, glucose metabolism, gastric secretion NOTES
and emptying.
o It helps in insulin secretion.
x Obestatin: It acts in the opposition to ghrelin action on appetite.
x Gastrin: It is produced by stomach antrum and it stimulates acid and pepsin
secretion and it also stimulates the pancreatic secretions.
x Secretin: It is secreted from duodenum at pH values below 12.5 and it
stimulates pancreatic acinar cells to release bicarbonate and H2O.
x CholeCystoKinin (CCK): It stimulates gallbladder contraction and bile
flow and it increases the secretion of digestive enzymes from pancreas.
x Motilin: It controls gastrointestinal muscles.
x Vasoactive Intestinal Peptide (VIP): It is produced by hypothalamus
and GI tract, and it relaxes the GI. It even inhibits the acid and pepsin
secretion and acts as a neurotransmitter in peripheral autonomic nervous
system. It increases the secretion of H2O and electrolytes from the pancreas
and gut.
x Somatostatin: It inhibits the release and action of numerous gut peptides
and it also inhibits the insulin and glucagon secretion from the pancreas.
x Peptide Tyrosine (PYY): It inhibits gastric motility by inhibiting cholinergic
neurotransmission and it inhibits the gastric acid secretion.
x Neuro-Peptide Tyrosine (NPY): It effects the hypothalamic function of
appetite and it controls the feeding behaviour and energy homeostasis.
Pancreatic Hormones
x Insulin: It is produced by E-cells of the pancreas and it increases the
glucose uptake and utilization and lipogenesis.
x Glucagon: It is produced by D-cells of the pancreas and it increases lipid
mobilization and glycogenolysis in order to increase the blood glucose levels.
x Pancreatic Polypeptide: It increases glycogenolysis and it regulates the
gastrointestinal activity.
x Somatostatin: It assists in the inhibition of glucagon and somatotropin
release.
Placental Hormones
x Estrogens: It helps in the maintenance of pregnancy.
x Progestins: It mimics the action of progesterone.
Self-Instructional
Material 301
Hormones x Chorionic Gonadotropin: Its activities are similar to luteinizing hormone.
x Placental Lactogen: It acts like prolactin and GH.
x Relaxin: It is produced in ovarian corpus luteum and it inhibits myometrial
NOTES contractions.
Gonadal Hormones
x Androgens (Testicular): It helps in the maturation and function of male
secondary sex organs.
x Inhibins A and B: It inhibits FSH secretion.
Adrenal Cortical Hormones
x Glucocorticoids: It has diverse effects on inflammation and protein synthesis.
x Mineralocorticoids: It helps in maintaining salt balance.
Adrenal Medullary Hormones
x Epinephrine (Adrenalin): It increases glycogenolysis, lipid mobilization,
smooth muscle contraction, cardiac function.
x Norepinephrine (Noradrenalin): It assists in lipid mobilization, arteriole
contraction and it also acts as neurotransmitter in the CNS.
Liver Hormones
x Angiotensin II: It is responsible for the essential hypertension through
stimulated synthesis.
Kidney Hormones
x Calcitriol [1,25-(OH)2-Vitamin D3]: It is responsible for maintenance of
calcium and phosphorous homeostasis and it increases the intestinal Ca2+
uptake and regulates the bone mineralization.
Cardiac Hormones
x Atrial Natriuretic Peptide (ANP): It is released from heart atria in response
to hypovolemia and it also acts on the outer adrenal cells to decrease the
aldosterone production.
Pineal Hormones
x Melatonin: It regulates the circadian rhythms.
General Considerations of Hormones
The hormones generally act as a body catalyst like enzymes. However, they differ
from the enzymes in the following ways:
x They are not always protein in nature.
Self-Instructional
302 Material
x The hormones are secreted in the blood stream prior to use, because, Hormones

circulating levels can indicate, the activity of endocrine gland and the exposure
of target organs.
Some other features of hormones are as follows:
NOTES
x Action in Low Concentration: Hormones act in a very low concentration
like vitamins.
x Storage Destruction and Excretion: Hormones are not ordinarily stored,
except in the gland, of origin. They do not have any cumulative action,
because they are destroyed and excreted as soon as their functions are
over. Some hormones work quickly and are destroyed quickly like thyroxine.
Mode of Action Role of C-AMP and Hormone Action
3'–5' c-AMP plays a unique role in the action of many protein hormones. Its level
may be decreased or increased by hormonal action as the effect varies depending
on the tissue. The hormones such as glucagon, catecholarnines, PTH, etc. act by
influencing a change in the intracellular c-AMP concentration through the adenylate
cyclase- c-AMP system. Different types of membrane receptors remain associated
with either Gs or Gi type of GTP dependent trimeric nucleotide regulatory complexes
of the membrane. Both Gs and Gi are made up of three subunits: Gs contains as 3y
while Gi contains a 3y. Formation of the receptor-hormone complex promotes the
binding of GTP with the subunit of either Gs or Gi. When a GTP is released, it binds
the adenylate cyclase located on the cytoplasmic surface of the membrane and
changes its conformation to activate it. However, in some cells calmodulin 4 Ca++
is also required for activation. Adenylate cyclase catalyzes the conversion of ATP
to c-AMP thus increasing the intracellular concentration of the latter.
On the other hand, aI-GTP inhibits adenylate cyclase by binding it. This
lowers the intracellular concentration of c-AMP.
Role of Calcium in Hormone Action
The action of most of the protein hormones are inhibited in the absence of calcium
even though ability to increase or decrease c-AMP is comparatively unimpaired.
Thus, calcium may have more terminal signal for hormone action than c-AMP. It is
suggested that the ionized calcium of the cytosol is the important signal. The source
of this calcium may be extracellular fluid or it may arise from mobilization of
intracellular tissue bound calcium. The hormone receptor binding may directly
inhibit the ‘Ca++- ATPase.’ It may also directly open up voltage-independent Ca++
channels in the membrane to increase the diffusion of Ca++ in the cell down its
inward concentration, which then acts as a second messenger to affect the cellular
activities. The receptor-hormone complex may produce ITP, which in turn can
increase cytosolic Ca++ concentration by enhancing the mobilization of Ca from
mitochondrial and endoplasmic reticular pools. Calcium is involved in the regulation
of several enzymes. All these enzymes have special biochemical metabolic roles.
Self-Instructional
Material 303
Hormones Ca++ also changes membrane permeability. Many of its effects are mediated through
its binding to -Ca++-dependent regulatory proteins like calmodulin and troponin.
Regulation of Hormone Secretion
NOTES Hormone secretion is strictly under control of several mechanisms.
Neuroendocrinal Control Mechanism
Nerve impulses control some endocrine secretions. Cholinergic sympathetic fibers
stimulate catecholamine secretion from adrenal medulla. Centers in the midbrain,
brainstem, hippocampus, etc. can send nerve impulses. At the terminations of
these neurons they release acetylcholine and biogenic amines to regulate the
secretions of hypophysiotropic peptide hormones from hypothalamic peptidergic
neurons. Some of the endocrine releases are controlled by either stimulatory or
inhibitory hormones from a controlling gland, for example, corticosteroids are
controlled by corticotropin and thyroid hormones are controlled by thyrotopin
from anterior pituitary. The tropins are further regulated by hypothalamic releasing
hormones.
Feedback Control Mechanism
It is mainly due to negative feedback that such control is brought about. When
there is a high blood level of target gland hormones, it may inhibit the secretion of
the tropic hormones stimulating that gland. Adrenal cortex secretes a hormone
called cortisol, which brings about the inhibition of secretion of corticotropin from
the anterior pituitary and corticotropin releasing hormone from the hypothalamus
by a long-loop feedback. This leads to the reduction in cortisol secretion.

Check Your Progress


1. How is the integration of body functions in humans and other higher
organisms carried out?
2. What is the basic function of oxytocin?
3. Name some of the mechanisms that control hormone secretion.

12.4 STEROID HORMONE RECEPTORS

Steroid hormones are bind to ligand-activated proteins, which regulates to the


transcription of selected genes. They are found in nucleus and cytosol and they
belong to the steroid and thyroid hormone receptor super-family of proteins, which
includes not just the receptors for steroid hormones, but also for Thyroid Hormone
(TR), vitamin D, Retinoic Acid (RAR) and GlucocoRticoids (GR). This large class
of receptors is termed as the nuclear receptor.
When these receptors bind the ligand they undergo a conformational change,
which renders them activated to recognize and bind to particular nucleotide
Self-Instructional
304 Material
sequences. These particular nucleotide sequences in the DNA are termed as Hormones

Hormone-Response Elements (HREs). When the ligand-receptor complexes


interact with the DNA they alter the transcriptional level of the associated gene.
Thus, the steroid-thyroid family of receptors all has three distinct domains as follows:
NOTES
x A Ligand-Binding Domain
x A DNA-Binding Domain
x A Transcriptional Regulatory Domain
Although there is the commonly observed effect of altered transcriptional
activity in response to hormone-receptor interaction, there are family member-
specific effects with ligand-receptor interaction. Binding of thyroid hormone to its
receptor, results in the release of the receptor from the DNA. Several receptors
are induced for interacting with other transcriptional mediators in response to the
ligand binding. Binding of glucocorticoid leads to the translocation of the ligand-
receptor complex from the cytosol to the nucleus.
The receptors for the retinoids are recognized as RARs and they exist in at
least three subtypes, namely, RARD, RARE and RARJ. In addition, there is another
family of nuclear receptors known as the retinoid X Receptors (RXRs), which
represent a second class of retinoid-responsive transcription factors. The RXRs
enhance the DNA-binding activity of RARs and the Thyroid Hormone Receptors
(TRs). The RXRs depict a class of receptors, which bind the retinoid 9-cis-retinoic
acid. They serve as obligatory heterodimeric partners for numerous members of
the nuclear receptor family. RXRD is expressed with highest levels like liver, kidney,
spleen, placenta and skin. The critical role for RXRD in development is explained
by the fact that null mice are embryonic lethals. RXRE is crucial for spermatogenesis
and RXRJ has a limited expression in both brain and muscle. The major difference
between the RARs and RXRs is that the former exhibits highest affinity for all-
trans-retinoic acid (all-trans-RA) and the latter for 9-cis-RA.
Additional super-family members are the Peroxisome Proliferator-Activated
Receptors (PPARs). The PPAR family consists of PPARD, PPARE/G and PPARJ.
All these receptors form a heterodimer with the RXRs. The first family member
identified was PPARD and it was found by the virtue of it binding with the fibrate
class of anti-hyperlipidemic drugs or peroxisome proliferators. Consequently, it
was shown that PPARD is the endogenous receptor for polyunsaturated fatty
acids. Expression of PPARD is also seen in macrophage foam cells and vascular
endothelium. Its role in these cells is thought to be the activation of anti-inflammatory
and anti-atherogenic effects. PPARJ is a master regulator of adipogenesis and is
most abundantly expressed in the adipose tissue. Low levels of expression are
also observed in liver and skeletal muscle. PPARJ was identified as the target of
the ThiaZoliDinedione (TZD) class of insulin sensitizing drug. The mechanism of
action of the TZDs is a function of the activation of PPARJ activity and the
consequent activation of adipocytes leading to increased fat storage and secretion
of insulin-sensitizing adipocytokines such asadiponectin. PPARG is expressed in
Self-Instructional
Material 305
Hormones most tissues and is involved in the promotion of mitochondrial fatty acid oxidation,
energy consumption and thermogenesis. PPARG serves as the receptor for
polyunsaturated fatty acids and VLDLs. Current pharmacologic targeting of PPARG
is aimed at increasing HDL levels in humans since experiments in animals have
NOTES shown that increased PPARG levels result in increased HDL and reduced levels of
serum triglycerides.
Mutations in the gene for PPARJ have been correlated with the insulin
resistance. It is unclear as to how the impaired PPARJ signaling can affect the
sensitivity of the body to insulin or indeed if the observed mutations are a direct or
indirect reasons for the symptoms of insulin resistance.

Check Your Progress


4. Define what steroid hormones are.
5. What is the major difference between the RARs and RXRs?

12.5 PEPTIDE HORMONES RECEPTOR

With the exception of the thyroid hormone receptor, the receptors for amino acid-
derived and peptide hormones are located in the plasma membrane. Receptor
structure is varied: some receptors consist of a single polypeptide chain with a
domain on either side of the membrane, connected with a membrane-spanning
domain. Some of the receptors comprise of a single polypeptide chain, which is
passed back and forth in serpentine fashion across the membrane, giving multiple
intracellular, transmembrane and extracellular domains. Other receptors consist of
multiple polypeptides.
Following the hormone binding, a signal is transduced to the interior of the
cell, where the second messengers and phosphorylated proteins generate the
appropriate metabolic responses. The main second messengers are cAMP, Ca2+,
Inositol TriPhosphate (IP3) and DiAcylGlycerol (DAG). Adenylate cyclase then
convert the ATP to cAMP and the subsequent increase in cAMP leads to the
activation of cAMP-dependent protein kinase (PKA) as shown in Figure 12.1.
GPCRs also couple with G-protein activation of PhosphoLipase C-J (PLCJ).
Activated PLCJ hydrolyzes membrane phospholipids result in the increased levels
of IP3 and DAG. Downstream signaling proteins are phosphorylated on serine
and threonine by PKA and DAG-activated Protein Kinase C (PKC) leading to
alterations in their activities.
The hormone-binding signal of most plasma membrane receptors is
transduced to the interior of cells by the binding of the receptor-ligand complexes
to a series of membrane-localized GDP/GTP binding proteins termed as G-proteins.
When G-proteins bind to the receptors, GTP exchanges with GDP bound to the
D subunit of the G-protein. The GD-GTP complex binds adenylate cyclase,
Self-Instructional
306 Material
activating the enzyme. The activation of the adenylate cyclase leads to c-AMP Hormones

production in the cytosol and to the activation of PKA, followed by the regulatory
phosphorylation of a lot of enzymes. Stimulatory G-proteins are designated Gs
and the inhibitory G-proteins are designated Gi.
NOTES

Fig. 12.1 Activation of cAMP-Dependent Protein Kinase

12.6 HORMONE RECEPTORS AND


INTRACELLULAR MESSENGERS

A hormone receptor is a receptor protein on the cell’s surface or in its interior that
binds a specific hormone. It causes many changes in the cell whereas the intracellular
messengers assert their actions primarily through the activation of protein kinases.
Hormones Transduce Signals to Affect Homeostatic Mechanisms
The stimulus can be a challenge or a threat to the organism, to an organ, or to the
integrity of a single cell within that organism. Recognition of the stimulus is the first
step in the adaptive response. At the organismic level, this generally involves the
nervous system and the special senses (sight, hearing, pain, smell, touch). At the
organismic or cellular level, recognition involves physicochemical factors such as
pH, O2 tension, temperature, nutrient supply, noxious metabolites and osmolarity.
Appropriate recognition results in the release of one or more hormones that will
govern generation of the necessary adaptive response. The hormones are
categorized by their specific cellular receptors and the type of signal generated.
Group I hormones interact with an intercellular receptor and group II hormones
with the receptor recognition sites located on the extracellular surface of the plasma
membrane of target cells.
Self-Instructional
Material 307
Hormones Hormones have Membrane Receptors and Use Intracellular
Messengers Group II (Peptide and Catecholamine)
Many hormones are water-soluble, have no transport proteins (and therefore have
NOTES a short plasma half-life) and initiate a response by binding a receptor located in the
plasma membrane (Refer Table 12.1). The mechanism of action of this group of
hormones can be discussed in terms of the intracellular signals they generate. Many
of these second messengers affect gene transcription but they also influence other
biological processes.
G Protein-Coupled Receptors
Many of the group II hormones bind the receptors that joined to effectors through
a GTP-binding protein intermediary. These receptors have seven hydrophobic
plasma membrane-spanning domains. This is illustrated by the seven
interconnected cylinders extending through the lipid bilayer. Receptors of this class,
which signal through guanine nucleotide-bound protein intermediates, are known
as G protein-coupled receptors, or GPCRs. Till today, over 130 G protein-linked
receptor genes have been cloned from various mammalian species. A wide variety
of responses are mediated by the GPCRs.
Adenylyl Cyclase
Different peptide hormones can either stimulate (s) or inhibit (i) the production of
cAMP from adenylyl cyclase.
Two parallel systems, a stimulatory (s) one and an inhibitory (i) one, converge
upon a single catalytic molecule (C). Each consists of a receptor, Rs, or Ri, and a
regulatory complex, G, and Gs. Gs and Gi are each times composed of D, E, and
J subunits. Because the D subunit in Gs, differs from that in Gi, the proteins, which
are distinct gene products, are designated Ds and Di. The D subunits bind guanine
nucleotides. The E, and J subunits are always associated (EJ) and appear to
function as a heterodimer. The binding of a hormone to Rs or R; results in a
receptor-mediated activation of G, which entails the exchange of GDP by GTP on
a and the concomitant dissociation of E, and J from D.
The D protein has intrinsic GTPase activity. The active form, D GTP, is
inactivated upon hydrolysis of the GTP to GDP; the trimeric G, complex is then
reformed and is ready for another cycle of activation. Cholera and pertussis toxins
catalyze the ADP fibosylation of Ds, and Di, respect tively. In the case of a, this
modification disrupts the intrinsic GTP-ase activity; thus, a, cannot reassociate
with 13y and is therefore irreversibly activated. ADP ribosylation of Di-2 prevents
the dissociation of Di2 from fry, and free Di-2 thus cannot be formed, Ds activity
in such cells is therefore unopposed.
There is a large family of G proteins, and these are part of the superfamily of
GTPases. The G protein family is classified according to sequence homology into
four subfamilies. There are 21 D, 5 E and 8 J subunit genes. Various combinations
of these subunits provide a large number of possible of DEJ and cyclase complexes.
Self-Instructional
308 Material
The D, subunits and the fry complex have actions independent of those on Hormones

adenylyl cyclase. Some forms of a, stimulate K+ channels and inhibit Ca+ channels,
and some a, molecules have the opposite effects. Members of the Gq family activate
the phospholipase C group of enzymes. The fry complexes have been associated
with K+ channel stimulation and phospholipase C activation. G proteins are involved NOTES
in many important biologic processes in addition to hormone action.
Table 12.1 DNA Sequences of Several Hormone Response Elements (HREs)
Hormone or effector HRE DNA sequence
Glucocorticoids GRE
Progestins PRE
GGTACA NNN TGTTCT
Mineralocorticoids MRE
Androgens ARE
Estrogens ERE AGGTCA --TGA/TCCT
Thyroid Hormone THE
Retinoic Acid RARE AGGTCA N3,4,5,AGGTCA
Vitamin D VDRE
cAMP CRE TGACGTCA

Letters indicate nucleotide; N means any one of the four can be used in that
position. The arrows pointing in the opposite directions illustrate the slightly
imperfect inverted palindromes present in many HREs; in some cases these are
called ‘half binding sites’ because each binds one monomer of the receptor. The
GRE, PRE, MRE and ARE consist of the same DNA sequence. Specificity may
be conferred by the intracellular concentration of the ligand or hormone receptor,
by flanking DNA sequences not included in the consensus, or by other accessory
elements. A second group of HREs includes those for thyroid hormones, estrogens,
retinoic acid and vitamin D. These HREs are similar except for the orientation and
spacing between the half palindromes. Spacing determines the hormone specificity.
VDRE (N=3), THE (N=4) and RARE (N=5) bind to direct repeats rather than
to inverted repeats. Another member of the steroid receptor superfamily, the retinoid
X receptor (RXR), forms heterodimers with VDR, TR and RARE, and these
constitute the transacting factors, cAMP affects gene transcription through the
CRE.
Protein Kinase
In prokaryotic cells, cAMP binds to a specific protein called Catabolite Regulatory
Protein (CRP) that binds directly to DNA and influences gene expression. In
eukaryotic cells, cAMP binds to a protein kinase called protein kinaseA (PKA)
that is a heterotetrameric molecule consisting of two regulatory subunits (R) and
two Catalytic subunits (C). cAMP binding results in the lowing reaction:
4 CAMP+R2C2 === R2 .( 4 CAMP)+2C
The R2C2 complex has no enzymatic activity but the binding of cAMP by R
dissociates R from C thereby activating the latter. The active C subunit catalyzes
the transfer of the y phosphate of the ATP to a serine or threonine residue in a
variety of proteins. The consensus phosphorylation sites are Arg/Lys-X-Ser/Thr
Self-Instructional
Material 309
Hormones and -Arg-Lys-X X-Ser-, who can be any amino acid. Protein kinase activities
were originally describing cAMP-dependent or cAMP-independent. This
classification has changed, as protein phosphorylation is now recognized as being
a major regulatory mechanism. Several hundred protein kinases have now been
NOTES described. The kinases are related in sequence and structure within the catalytic
domain, but each is a unique molecule with considerable variability with respect to
subunit composition, molecular weight, aurophosphorylation, Km for ATP and
substrate specificity.
Phosphodiesterases
Actions caused by hormones that increase cAMP concentration can be terminated
in a number of ways, including the hydrolysis of cAMP to 5'-AMP by
phosphodiesterases. The presence of these hydrolytic enzymes ensures a rapid
turnover of the signal (cAMP) and hence a rapid termination of the biologic process
once the hormonal stimulus is removed. There are at least eleven known members
of the phosphodiesterase family of enzymes. These are subject to regulation by
their substrates, cAMP and cGMP; by hormones; and by intracellular messengers
such as calcium, probably acting through calmodulin. Inhibitors of
phosphodiesterase, most notably methylated xanthine derivatives such as caffeine,
increase intracellular cAMP and mimic or prolong the actions of hormones through
this signal.

Check Your Progress


6. What is a hormone receptor?
7. What do different peptide hormones do?

12.7 INSULIN

Insulin is a hormone that is central to regulating fat and steroids metabolism in the
body. It causes cells in the liver, muscle and fat tissue to take up glucose from the
blood, storing it as glycogen in the liver and muscle.
Insulin Secretion
The insulin is packed into granules in the golgi apparatus. The insulin molecules
associate into a hexamer with two zinc ions and one calcium ion. The contents of
granules are released by exocytosis; both insulin and C-peptide are released into
circulation. Glucose is the major stimulant of insulin secretion. The E cells have
GluT 2 receptors, through which glucose is absorbed. This is oxidized, so that
more ATP is made available. ATP opens calcium channels. Increased intracellular
calcium activates adenyl cyclase, which produces cyclic AMP. This cAMP along
with calcium causes the insulin secretion. Insulin secretion is enhanced by GTP,
secretin, pancreozymin, gastrin and glucagon. Other stimulants of insulin secretion
Self-Instructional
310 Material
are proteins, amino acids especially leucine and arginine, parasympathetic stimulation Hormones
and 3-adrenergic stimulation. Inhibition of secretion occurs following D-adrenergic
stimulation and vagotomy. The drug tolbutamide will stimulate insulin secretion.
Biosynthesis of Insulin
NOTES
The insulin is synthesized as a larger precursor polypeptide chain, the pre-pro-
insulin. It has 109 amino acids. It is rapidly convene topro-insulin in the endoplasmic
reticulum by removal of leader sequence of 23 amino acid residues. The proinsulin
with 86 amino acids is transported to golgi apparatus where it is cleaved by a
protease. The site specific cleavage is done by enzymes named as ‘prohormone
convertase 1 and 2.’ Thus, C-peptideor connecting peptide with 33 amino acids
is removed. Now, insulin has 53 amino acids; the extra two amino acids are removed
by carboxy peptidase H and insulin with 51 amino acids is thus formed.
Actions of Insulin
Insulin acts by binding to a plasma membrane receptor on the target cells. Human
insulin receptor gene is located on chromosome 19. In obesity, the number of
receptors is decreased and the target tissue becomes less sensitive to insulin. Insulin
receptor is a glycoprotein with four subunits; two alpha and two beta subunits.
The alpha units (135 kD) are located on the extracellular side, to which insulin
binds. The E subunits (95 kD) traverse the membrane and are exposed on the
cytoplasmic side. Beta subunit has tyrosine kinase activity. Figure 12.2 shows the
insulin signalling to glycogen metabolism.

Fig. 12.2 Insulin Signalling to Glycogen Metabolism

Signal Transduction
Binding of insulin causes dimerization of the receptor. It is then internalized, so that
the signal is transmitted. Then the tyrosine kinase phosphorylates tyrosine residues
on the cytoplasmic side of insulin receptor: This event, in turn, phosphorylates
Self-Instructional
Material 311
Hormones Insulin Receptor Substrates (IRS). IRS-1 and 2 are characterized. The message
is later transmitted in a series of serine/threonine kinases. A summary of this cascade
is given in Figure 12.3. The final effect may be any one of the following:
x Gene Transcription
NOTES
x Activation of Enzymes
x NA Synthesis

Fig. 12.3 Signal Transduction

Degradation of Insulin
Insulin is rapidly degraded by the liver. Plasma half-life is less than five minutes. An
insulin specific protease (insulinase) and a hepatic glutathione- insulin-
transhydrogenase, which cleaves the disulphide linkages are involved in the
degradation of insulin. Normal insulin level in blood is 5–15 micro units/ml. Proinsulin
contributes5 to 10% of the total insulin measured in plasma. Proinsulin has about
one-third biological activity as of that insulin. Approximately 50 units of insulin are
secreted per day. Sometimes, measurement of endogenous insulin may be difficult
because of the presence of antibodies against insulin in the circulation. Then,
measurement of C-peptide is useful, Insulin and C-peptide are synthesized and
secrete in equimolar quantities. But half-life of C-peptide is 35 minutes (insulin has
only five minutes); so plasma level of C-pptide is five to ten-fold higher than insulin.
Physiological Actions of Insulin
Insulin plays a central role in the regulation of the metabolism of carbohydrates,
lipids and proteins.
Self-Instructional
312 Material
Uptake of Glucose by Tissues Hormones

Insulin facilitates the membrane transport of glucose Facilitated diffusion of glucose


in muscle is enhanced by insulin. In diabetes mellitus, the transporter, GluT4
reduced. About 80% of glucose uptake is independent of insulin and occurs in
NOTES
splanchnic tissue are brains. At basal rates, brain utilizes 60% of sugar oxidized.
Glucose uptake in liver (by GluT2) is independent of insulin. Hepatic glucokinase
is specifically activated by insulin and is not under the influence of insulin.
Utilization of Glucose
Insulin favours the utilization and storage of glucose. About 50% of the ingested
glucose is utilized for yielding energy by glycolysis. Another 40% is converted to fat
and the rest 10% is stored as glycogen. Glycolysis and lipogenesis are stimulated by
insulin. The activity and amount of several enzymes are increased by insulin. These
include key glycolytic enzymes (glucokinase, phosphofructokinase and pyruvate
kinase) as well as glycogen synthase.
Hypoglycemic Effect
Insulin lowers the blood glucose level by promoting utilization and storage.
Gluconeogenesis is inhibited by insulin by repressing the key enzymes. Insulin also
inhibits glycogenolysis by favouring the inactivation of glycogen phosphorylase
and inhibiting the glucose-6-phosphatase. The net effect is a lowering of blood
glucose level.
Anti-Ketogenic Effect
Insulin depresses HMG CoA synthase and so ketogenesis is decreased. Moreover,
in presence of insulin, acetyl CoA is completely utilized in the citric acid cycle,
because oxaloacetate generated from glucose is available in plenty. Insulin also
favours fatty acid synthesis from acetyl CoA. All these factors reduce the availability
of acetyl CoA.
Other general Effects
Protein synthesis is promoted and degradation is retarded. The uptake of amino
acids is favoured. It is ananabolic hormone. Insulin stimulates replication of cells in
culture. In fact, insulin is an essential growth factor for all mammalian cells.

12.8 ROLE OF GLUCAGON

It is a polypeptide hormone with 29 amino acids. It is secreted by the D cell of


pancreas. Enteroglucagon is a peptide hormone secreted by duodenal mucosa
having sequence similarity. Glucagon has 50% homology to secretin, another
intestinal hormone. Glucagon is secreted by the D cells as a big proglucagon
precursor. Glucagon has a half life in plasma for about five minutes. It is inactivated
in the liver. The major regulator of secretion of glucagon is glucose. An increase in
blood glucose level inhibits secretion of glucagons (Refer Figure 12.4).
Self-Instructional
Material 313
Hormones

NOTES

Fig. 12.4 Glucagon and Blood Relation

Physiologic Effects of Glucagon


Glucagon is anti-insulin in nature. Therefore, the net effect is decided by the insulin
glucagon ratio. It is mainly glycogenolytic. The active form of glycogen phosphorylase
is formed under the influence of glucagon. Liver is the primary target for the
glycogenolytic effect of glucagon. Gluconeogenesis is favoured by glucagon by
inducing enzymes like PEPCK, glucose 6-phosphatase and fructose-1,6-
bisphosphatase. Net effect is hyperglycemia.
The effect of glucagon on adipose tissue is to increase plasma FFA level. %-
oxidation is favoured, since carnitine acyltransferase is activated by glucagon. The
mitochondrial acetyl CoA level increases and ketogenesis is favoured. In a normal
person excessive ketogenesis is prevented between the effects of insulin and
glucagon.
Control of Glucagon Secretion
Hormone combines with a membrane bound receptor. This activates the GDP
bound G-protein, by converting it into GTP form. The D subunit of G-protein now
dissociates from the E and J subunits and D subunit binds to GTP. The GTP-G
protein, in turn, activates adenyl cyclase. Thus, ATP is converted to cAMP. This
combines with the regulatory subunit of the protein kinase. So, the catalytic subunit
is free to act. The active protein kinase will phosphorylate enzyme, so that glycogen
phosphorylase is activated.
Regulation of carbohydrate metabolism in general depends on anti-insulin
hormones; glucocorticoids, glucagon and growth hormone. The major action of
glucagon is glycogenolysis. Glucocorticoids acts mainly by gluconeogenesis. But
growth hormones antagonize insulin in many key metabolic reactions.
Diabetes Mellitus
Diabetes mellitus is a metabolic disease due to absolute or relative insulin deficiency.
It causes loss of weight as if the body mass is passed through the urine. Diabetes
mellitus is a disease known from very ancient times. In Western literature, Thomas
Willis in 1670 noticed the sweet taste of diabetic urine. In 1776, Mathew Dobson
showed that this sweetness of urine is due to sugar. In 1838, Bouchardt and Peligot
Self-Instructional
314 Material
proved that the sugar of diabetic urine is glucose. Qualitative test for urine sugar Hormones

was perfected by Hermann Fehling (1848) and semi-quantitative test by Francis


Benedict (1908).
Diabetes mellitus is a common clinical condition. About 10% of the total
NOTES
population and about 1/5th of persons above the age of 50 suffer from this disease.
It is a major cause for morbidity and mortality. The disease may be classified as
follows (WHO recommendation, 1999):
x Type I Diabetes Mellitus.
x Type 2 Diabetes Mellitus.
x Gestational Diabetes.

12.9 EPINEPHRINE AND THEIR MECHANISM FOR


VARIOUS ENDOCRINE

The adrenal gland is composed of an outer cortex and inner medulla. The cortex is
responsible for secretion of steroid hormones. The medulla is responsible for the
secretion of epinephrine.
The adrenal cortex hormones are of two types:
x Glucocorticoids
x Mineralocorticoids
Glucocorticoids
They primarily affect metabolism of carbohydrate, proteins and lipids and
relatively have minor affects on electrolytes and water metabolism like cortisol,
cortisone and corticosterone.
Metabolic Functions of Mineralocorticoids
In general, glucocorticoids have anti-insulin effect. They are catabolic to peripheral
tissues and anabolic to liver. Over-all effect increases blood glucose level.
The mineralocorticoids (aldosterone) stimulate renal absorption of sodium
hydrogen, ammonium and magnesium ions. Due to its pronounced sodium retention
property, it increases water absorption.
Adrenal Medullary Hormones
Two biologically active compounds have been isolated from the adrenal medulla
and synthesized. They are as follows:
x Epinephrine.
x Norepinephrine (nor Adrenaline).
The naturally occurring forms are laevorotatory, the synthetic are racemic,
the former being almost twice as active as the latter. The above two hormones are
called catecholamines and are closely related to tyrosine and synthesized in body
Self-Instructional
Material 315
Hormones from tyrosine. Epinephrine is primarily synthesized and stored in adrenal medulla.
Norepinephrine is primarily synthesized–in sympathetic nervous system and acts
locally as a ‘neurotransmitter’ at the post synaptic cell. Norepinephrine is also
synthesized and stored in adrenal medulla,
NOTES
Metabolic Functions of Catecholamines
Epinephrine stimulates rapid breakdown of glycogen of liver (glycogenolysis)
producing hyperglycaemia. In muscles, epinephrine also cause breakdown of
glycogen (glycogenolysis) by increasing cyclic AMP level (13-effect), but in this
tissue it is more active than glucagon. Glucagon has very little effect or no effect
due to lack of specific receptors. Increases in cyclic AMP after epinephrine
administration is seen in two to four seconds, the effect of epinephrine on cardiac
output (ionotropic effect) is seen shortly afterwards, whereas activation of
phosphorylase is not detectable, in vivo, actually epinephrine action can result in
an increase in heart glycogen. Both epinephrine and norepinephrine increases
breakdown of Triacyl glycerol in adipose tissue by increasing cyclic-AMP level
(31 effects). Net effect of lipolysis is rapid release of FFA and glycerol from adipose
tissue to blood. Epinephrine increases hepatic gluconeogenesis (132 effects).
Norepinephrine and epinephrine are almost equally potent in their calorigenic
action. They produce a prompt rise in the metabolic rate, which is independent of
the liver, and a smaller delayed rise, which is abolished by hepatectomy.

12.10 REGULATORY SYSTEMS MEDIATED BY


CYCLIC AMP

The discovery of messenger RNA was an unequivocal breakthrough in


understanding cell biochemistry and biochemical genetics.
Sutherland has worked on the interaction between hormone adrenalin and
blood sugar (glucose) in the circulatory system. This reaction generally takes place
in the grip of emotions such as anger, pain or fear and it provides an animal with
the energy either to fight or flee. Glycogen is stored in the liver cells as a polymeric
storage form of glucose. There are several steps, which transform glycogen into
glucose, and it finally enters the blood stream. Sutherland found that only the first
step of conversion of glycogen to glucose 1-phosphate was actually controlled -
by epinephrine and is catalysed by an enzyme phosphorylase. He further found
that the enzyme exists in two forms:
x Phosphorylase-D, which is active.
x Phosphorylase-E, which is rather inactive.
Cyclic AMP is an important second messenger. It forms, as shown, when
the membrane enzyme adenylyl cyclase is activated (as indicated, by the alpha
subunit of a G protein) (Refer Figure 12.5).
Self-Instructional
316 Material
Hormones

NOTES

Fig. 12.5 Protein Kinesis

The cyclic AMP then goes on to activate specific proteins. Some ion channels,
for example, are gated by cyclic AMP. But an especially important protein activated
by cyclic AMP is protein kinase A, which goes on the phosphorylate certain cellular
proteins.
A Metabolic Regulator and Second Messenger
The process of cyclic AMP in Escherichia coli was established in 1965 by
Sutherland and it was known to regulate a large number of biochemical processes.
By that time, Robert Perlman and Ira Pastan from the National Institute of Health
happened to discover an important regulatory role of cAMP in bacteria and it was
equated with the role of messenger RNA. Glucose repressed the formation of
several enzymes leading to the biosynthesis of various sugars in E. coli. Perlman
and Pastman studied the glucose effect and found that if cAMP was present in E.
coli cultures, then enzyme expression does not take place within these cells. It
then could metabolize most of the sugar including maltose, lactose and arabinose.
It is already clear that synthesis of enzyme with the help of genetic information
stored in RNA. They discovered a mutant strain in which the basis for the enzyme
for the formation of cAMP was missing. The mutant had the normal gene for the
metabolism of lactose and glatose and thus was able to transcribe mRNA for
metabolizing both the sugars. When cAMP was supplied to such mutant, they
synthesized excess of mRNA for both the sugars without altering the rate of
transcription of other genes. Similar results were obtained in cell free preparation.
Therefore, it was almost clear that cyclic AMP act as a messenger for increasing
the transcription process and consequently, regulate the cellular metabolism. Cyclic
AMP has been called the second messenger since it transmits the primary hormonal
message from the cell surface to the cell interior. A large amplification of the normal
signal is achieved by using cAMP as a second messenger.
Self-Instructional
Material 317
Hormones Functional Control of Various Biological Process
Cyclic AMP has a regulatory role to play in bacteria fungi, algae, lower plants,
higher plant and animal system including mammals. The diversity is rather a reflection
NOTES of the commonality of genetic information operational in any system. In bacteria a
large number of chemical reactions under the regulation of cycle AMP. Phenomenon
like differentiation and development are also very well studied in cellular smile
mould which like the myxomycetes, exist for a part of their life cycle as amoeba
like organism.
The amoebas from streams, all following towards the center of the growing
aggregate in the series of waves and sticking together. The amoeba at the centre,
the pacemaker, is the one that emits pulses of cAMp at the fastest rate. After a
period of migration, the cells in the mass begin to differentiate and some form–the
stalk cells. It has been further demonstrated that isolated amoebas, subjected to a
high concentration of cAMP, differentiate into stalk cells. In higher plants also, the
seed germination requires certain growth hormones such as gibberellin. It should
be noted that the growth hormones are triggered by cAMP. Spore germination in
mosses has been shown to be under the control of cAMP.
Molecular Structure
Adenosine TriPhosphate (ATP) has three phosphate groups joined, in an order to
the 5'- phosphate cleavage, that is, removal of two terminal phosphate groups
together; the third phosphate group is still attached to the 5'-carbon of the ribose.
Figure 12.6 shows the molecular structure of cAMP.

Fig. 12.6 Molecular Structure of cAMP

The cAMP differs from AMP in possessing the phosphate group in a cyclic
manner and involving both 3'-and- 5'-carbon of the ribose sugar. The adenine
moiety is, however, same in AMP or cAMP. In a technical- sense, cAMP is 3'-5'-
cyclic adenylic acid, or adenosine 3'- 5'- cyclic phosphoric acid.
AMP Adenosine5'-monophosphate
ADP Adenosine 5'-diphosphate
ATP Adenosine 5'-triphosphate
cAMP Adenosine 3'-; 5’cyclic phosphoric acid
Self-Instructional
318 Material
Biosynthesis and Degradation Hormones

Cyclic AMP is formed in the plasma membrane of a variety of cells by the action
of an enzyme called adenyl cyclase.
NOTES
Mg2+
ATP ——o cAMP + PPi adenyl cyclase
The formation of cAMP is accompanied by the release of pyrophosphate.
The reaction is endothermic and has a G° value of about 2 Kcal/mol. Further, the
degradation of cAMP is an exothermic reaction (G°= –12 Kcal/mol) and requires
a specific enzyme called phosphodiesterase.
Cyclic AMP is stable in the absence of phosphodicstcrase, and the
intercellular concentration of cAMP reflects the relative activities of adenyl cyclase
and phosphodiesterase. Normally, the concentration of cAMp is regulated primarily
by the alteration of adenyl cyclase activity, but the concentration also may be
manipulated artificially by inhibiting phosphodiesterase with methylaxanthines, such
as theophylline and caffeine. Adenyl cyclase in mammalian cells apparently consists
of regulatory and catalytic components that are responsible for specific hormone
recognition and cAMP synthesis, respectively. These complexes are locaiized in
the plasma membrane where they can interact with a variety of hormones in the
first stage of hormonal regulation of the target-cell metabolism. Polypeptide and
amino acid-derived hormones and the prostaglandins apparently bind themselves
to the regulatory component and alter the activity of the associated adenyl cyclase,
thereby altering the intracellular concentration of cAMP. The majority of the
hormones increase the activity of adenyl cyclase, whereas a few; such as insulin
and some prostaglandins, decrease its activity. Adenyl cyclase is synthesized in
response to hormonal action in the receptors or target cells in the membrane. The
enzyme has been found to be membrane-bound.
Adenyl cyclase activity localized in the cell plasma membrane represents
the site of interaction between the intercellular regulatory device represented by
the hormones and the intracellular communication channel represented by CAMP.
This system has the effect of being a receptor of the hormone signal-as well as a
sort of chemical amplification device, since the direct action of the hormone is
on an enzyme. This means that the hormone can lead to the production of cAMP
on a much more than one-to-one stoichiometry. The circulating concentration of
cAMP hormones is of the order of 10-10 M, whereas the concentration of cAMP
in a stimulated target cell is about 10-0 M. Thus, the synthesis if cAMP by
activated enzyme-adenyl cyclase leads to a 104-fold amplification of the hormonal
signal. It should be added that the activity of adenyl cyclase is influenced by large
number of other hormones and that its product, cAMP, regulates the activity of
a number of other metabolic processes. The effect of both the hormones and
Self-Instructional
Material 319
Hormones cAMP differs greatly in different tissues, so that a great deal of specificity of action
is possible.
Hunger Signal
NOTES
How does a bacterium know how to synthesize the enzymes required for
metabolizing sugar? In other words, a hunger signal initiates the synthesis of
enzymes, which can satisfy the hunger of a bacterium by utilizing various sugars. In
the presence of glucose, the bacterium does not feel hungry and, therefore, no
enzymes are synthesized. As soon as the glucose concentration is lowered, the
cyclic AMP increases and raises a hunger signal to synthesize the enzymes
responsible for using various other sugars. A similar role has been assigned to
cAMP in the mammalian cells. However, the synthesis of new enzymes is not
directly due to the increase in transcription, but is primarily because of the activation
of protein kinases which, thereby, are able to govern a variety of cellular processes.
Different target cells respond differently because they contain different substrates
for protein kinases. Kinases have been found tophosphorylate histones, ribosomal
proteins as well as specific enzymes.
Glycogen Metabolism
Glycogen is a branched polymer of glucose with a high molecular weight. Glucose
residues are linked by D-1 ‘4-glycosidic bonds and the branches are created by
D -1-6-glycosidic bonds. The glycogen can be obtained in multiple ways in the
intermediary metabolism.
One important feature about glycogen metabolism is that entirely different
pathways are operative for the synthesis and breakdown of glycogen. Although
the control of these two processes is mediated via different enzymes, yet, with a
coordinated mechanism and when synthesis is inhibited, breakdown is stimulated
and vice versa. The coordinated control of glycogen synthesis and breakdown is
under the regulation of cyclic AMP.
Two important enzymes involved are as follows:
The actual synthesis of the polymer glycogen occurs with the transfer of
glucose from UDP glucose molecule to the end of the glucose polymer chain.
In vertebrates, UDP-glucose gives maximum activity in, this reaction, ADP-
glucose is much less effective. However, in the lower organism ADP-glucose is
more effective.
Two hormones including glycogen (synthesized by the pancreas) and
epinephrine (synthesized in the adrenal medulla) influence the biosynthesis
degradation of glycogen. Both epinephrine and glucagon bind themselves to the
plasma membrane and stimulate the enzyme adenyl cyclase, which forms cAMP,
which, in turn, activate a protein kinase. This protein kinase now coordinately
Self-Instructional
320 Material
phosphorylates both glycogen phosphorylase and glycogen synthetase. Since, the Hormones

phosphorylation is coordinated, the synthesis and breakdown of glycogen are


also linked.
Thus, glycogen metabolism is regulated by cAMP and involves the following steps: NOTES
x Switching off phosphorylase and switching on synthetase
x Switching on phosphorylase and switching off synthetase
Control of Transcription
Cyclic AMP-plays a major role in controlling transcription particularly during
induction. A well known case, which has already been mentioned is that of the
synthesis of (D-galactosidase. It has been found that externally supplied cAMP
could overcome the inhibitory effect of E-galactosidase on synthesis. Cyclic AMP
binds itself to a catabolite gene activator protein to form a complex cAMP-CAP,
which binds itself, the promoter site of the lac operon. The initiation of transcription
is stimulated in response to this binding. Cyclic AMP is also considered to be
involved in the regulating of transcription during the cell division/cycle. Recently, it
has been observed that there are two distinct stages Gr and G2 during cell division
in eukaryotes and an intermediary stage S’ when maximum DNA synthesis occurs.
It has been observed that cAMP decreases in concentration in fast growing cells.
cAMP is apparently exerting transcriptional control over cell division inhibition
and differentiation. It is further suggested that, just as in prokaryotes, cAMP bind
itself to receptor protein while controlling transcription during the cell cycle, cAMP
has also been shown to regulate the synaptic transmission in the nervous system as
well as the inflammatory and immune reactions of tissues.
It must be clearly understood that cyclic AMP functions only as an allosteric
effector, by increasing or decreasing the rate of reaction. However, cAMP has not
been found to act as a substrate or product barring its own synthesis or degradation.
The concentration of cAMP is primarily controlled by extra cellular stimuli,
particularly hormones.

Check Your Progress


8. Name the various types of adrenal cortex hormones.
9. Define what glycogen is.

12.11 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. Integration of body functions in humans and other higher organisms is carried


out through the nervous system, the immune system and the endocrine system.
2. The basic function of oxytocin is to cause uterine contraction.
Self-Instructional
Material 321
Hormones 3. Hormone secretion is controlled through the following mechanisms:
x Neuroendocrinal control mechanism
x Feedback control mechanism
NOTES 4. Steroid hormones are bind to ligand-activated proteins, which regulate the
transcription of selected genes.
5. The major difference between the RARs and RXRs is that the former exhibits
highest affinity for all-trans-retinoic acid (all-trans-RA) and the latter for 9-
cis-RA.
6. A hormone receptor is a receptor protein on the cell’s surface or in its
interior that binds a specific hormone.
7. Different peptide hormones either stimulate (s) or inhibit (i) the production
of cAMP from adenylyl cyclase.
8. The various types of adrenal cortex hormones are as follows:
x Glucocorticoids
x Mineralocorticoids
9. Glycogen is a branched polymer of glucose with a high molecular weight.

12.12 SUMMARY
x Hormones, cytokines, interleukins and growth factor use a variety of signaling
mechanisms to facilitate cellular adaptive responses.
x The ligand–receptor complex serves as the initial signal for members of the
nuclear receptor family.
x Class II (peptide and catecholamine) hormones, which bind to cell surface
receptor, generate a variety of intracellular signals. These include CAMP,
cGMP, Cat, phosphatidylinositides and protein kinase cascades.
x Many hormone responses are accomplished through alterations in the rate
of transcription of specific genes.
x The nuclear receptor super family of proteins plays a central role in the
regulation of gene transcription.
x These receptors, which may have hormones, metabolites or drugs as ligands,
bind to specific DNA elements as homodimers or as heterodimers with
RXR.
x Some—orphan receptors—have no known ligand but bind DNA and
influence transcription.
x A large family of coregulator proteins remodels chromatin, modifies other
transcription factors and bridges the nuclear receptors to the basal
transcription apparatus.
Self-Instructional
322 Material
Hormones
12.13 KEY TERMS

x Hormone: It is a chemical released by a cell or a gland in one part of the


body that sends out messages that affect cells. NOTES
x Cholesterol: It is a waxy steroid metabolite found in the cell membranes.
x Enzyme: It is a protein that catalyzes chemical reactions.
x Peptide: It is a short polymer of amino acids.

12.14 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. What are the basic functions of hormones?
2. What do you mean by peptide hormone receptor?
3. How is insulin secreted?
4. How is glucagons secretion controlled?
5. Write briefly about role of Calcium in hormones section.
Long Answer Questions
1. Describe how hormones are chemically classified.
2. Discuss hormone receptors and intracellular messengers.
3. Discuss the usage of insulin in today’s world.
4. Discuss about the regulatory system mediated by Cycle AMP.

12.15 FURTHER READINGS

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Self-Instructional
Material 323
Hormones Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
NOTES
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
324 Material
Acid-Base Balance

UNIT 13 ACID-BASE BALANCE


Structure NOTES
13.0 Introduction
13.1 Objectives
13.2 Henderson–Hasselbalch Equation
13.3 Regulation of Acid–Base Balance
13.3.1 Blood Buffer System
13.3.2 Respiratory and Renal Regulation
13.4 Acid–Base Disorders
13.5 Anion Gap
13.6 Assessment of Acid–Base Homeostasis
13.7 Answers to Check Your Progress Questions
13.8 Summary
13.9 Key Words
13.10 Self Assessment Questions and Exercises
13.11 Further Readings

13.0 INTRODUCTION

The chemical balance or homeostasis, as it is called, of body fluids can be maintained


by regulating their pH (which gives a measure of how acidic or basic a substance
is). As you must be aware, substances releasing hydrogen ions (H+) in solution are
referred to as acids. Acids which release most of their ions are called strong acids
(for example, hydrochloric acid) and those which release only some of the hydrogen
ions are called weak acids (for example, carbonic acid). On the other hand, bases
or alkalis generally have a low hydrogen ion concentration and can accept hydrogen
ions in solution. The pH reflects the hydrogen ion concentration of the solution: the
higher the hydrogen ion concentration (and the more acidic the solution), the lower
will be the pH. Water has a pH of 7 and is neutral; that is, it is neither acidic nor
alkaline in nature. Solutions with a pH less than 7 are acidic while those with a pH
more than 7 are alkaline. The pH scale is logarithmic, i.e., a solution with a pH of
5 is 10 times more acidic than one with a pH of 6.
In this unit, you will study about the acid–base balance and pH regulation
inside the human body.

13.1 OBJECTIVES

After studying this unit, you should be able to:


x Derive the Henderson–Hasselbalch equation and identify its significance in
maintaining acid–base equilibrium
x Analyse the complete mechanism of regulation of acid–base balance and
also describe the various buffer systems present within the body Self-Instructional
Material 325
Acid-Base Balance x Identify the different types of acid–base disorders
x Elaborate upon the concept of anion gap
x Detail out the mechanism of acid–base homeostasis
NOTES
13.2 HENDERSON–HASSELBALCH EQUATION

We all know that our body is very sensitive to its pH level, as a result of which
strong mechanisms exist inside the body to maintain it. The negative impact that an
acid–base imbalance can have on your body is enormous. Outside the acceptable
range of pH, proteins are denatured and digested, enzymes lose their ability to
function and death may occur.
The Henderson–Hasselbalch equation aids in studying the nature of the
titration curve of a weak acid, which is crucial for understanding the buffer action
and the acid–base equilibrium in the blood and tissues of vertebrates. This equation
helps in restating the expression for the dissociation constant of an acid. For the
dissociation of a weak acid HA into H+ and A-, the Henderson–Hasselbalch
equation is derived as given below:
ª¬H º¼ ª¬ A  º¼
Ka
HA
First solve for [H+]:
HA
[H 1 K,
A
After that you consider the negative logarithm of both sides:
HA
 log ª¬H º¼  logk a  log
Ac
Then you replace pH for -log [H+] and pKa for -log Ka:
HA
pH pK a  log
ª¬ A  º¼

Ultimately you invert -log [HA]/[A-], which also involves changing its sign,
so that the Henderson–Hasselbalch equation can be derived:
ª¬ A  º¼
pH pK a  log
HA
In more general terms,
proton acceptor
pH pK a  log
proton donor
This equation is applicable for the titration curve of all weak acids and it
helps in deriving many important quantitative relationships. For instance, it clarifies
the reason why the pKa of a weak acid is equal to the pH of the solution when
Self-Instructional
titration is only through midway. At that point, [HA] equals [A–], and
326 Material
pH = pKa + log 1 = pKa + 0 = pka Acid-Base Balance

Through the Henderson–Hasselbalch equation you can calculate the


following:
x pKa, when pH and the molar ratio of proton donor and acceptor are NOTES
given.
x pH, when pKa and the molar ratio of proton donor and acceptor are
given.
x Molar ratio of proton donor and acceptor, when pH and pKa are given.

13.3 REGULATION OF ACID–BASE BALANCE

Your body needs to maintain the acid–base balance as it is very essential. A


discussion of acid–base balance requires a review of several basic concepts: acid,
base, buffer, pH and pK—and also the principles of equilibrium and the law of
mass action. As mentioned earlier, an acid is a substance that can yield a hydrogen
ion (H+) or hydronium ion when dissolved in water. A base is a substance that can
yield hydroxyl ions (OH-). The relative strengths of acids and bases, and their
ability to dissociate in water are described by their dissociation constant (also
known as ionization constant or K value). The pK, defined as the negative log of
the ionization constant, is also the pH in which the protonated and unprotonated
forms are present in equal concentrations. Strong acids have pK values of less
than 3.0, whereas strong bases have pK values greater than 9.0. For acids, raising
the pH above the pK will cause the acid to dissociate and yield an H+. For bases,
lowering the pH below the pK will cause the base to release OH-. Many species
have more than one pK, meaning they can accept or donate more than one H-.
A buffer, which is the combination of a weak acid or weak base and its salt,
is a system that resists changes in pH. The effectiveness of a buffer depends on the
pK of the buffering system and the pH of the environment in which it is placed. In
plasma, the bicarbonate–carbonic acid system, having a pK of 6.1, is one of the
principal buffers. The reference value for blood plasma pH is 7.40. Weisberg
cited an example to demonstrate the effectiveness of the blood buffers.
If the pH of 100 mL of distilled water is 7.35 and one drop of 0.05 mol/L
HCl is added, the pH will change to 7.00. To change 100 mL of normal blood
from a pH of 7.35 to a pH of 7.00, approximately 25 mL of 0.05 mol/L HCl is
needed. With 5.5 L of blood in the average body, more than 1300 mL of HCl
would be required to make this same change in pH.
Body fluids are maintained within a narrow range that is slightly alkaline.
The normal pH of arterial blood is between 7.35 and 7.45 (Refer Figure 13.1).
Acids are continually produced during metabolism. Several body systems, including
buffers, the respiratory system and the renal system, are actively involved in
maintaining the narrow pH range necessary for optimal function.
Self-Instructional
Material 327
Acid-Base Balance

NOTES

Figure 13.1 Range of pH Values from Acidic to Alkaline

Buffers help maintain acid–base balance by neutralizing excess acids or


bases. The lungs and the kidneys help maintain a normal pH by either excreting or
retaining acids and bases.
Maintenance of H+: The normal concentration of H+ in the extracellular body
fluid ranges from 36 to 44 nmol/L (pH 7.34–7.44); however, through metabolism,
the body produces much greater quantities of H+. Through exquisite mechanisms
that involve the lungs and kidneys, the body controls and excretes H+ in order to
maintain pH homeostasis. Any H+ value outside this range will cause alterations in
the rates of chemical reactions within the cell and affect the many metabolic
processes of the body, and can lead to alterations in consciousness, neuromuscular
irritability, tetany (Tetany or tetany seizure is a medical sign consisting of the
involuntary contraction of muscles, which may be caused by disease or other
conditions), coma and death. The reference value for arterial blood pH is 7.40
and is equivalent to an H+ concentration of 40 nmol/L.
13.3.1 Blood Buffer System
For the purpose of maintaining a specific pH, the molecule which has an inclination
of either binding or releasing hydrogen ions is called a buffer. For instance, as has
been mentioned earlier, human blood needs to maintain a pH of 7.4. For elaborating
this, let us consider the bicarbonate ion that you are already familiar with. You
have already seen that the bicarbonate ion forms carbonic acid as well as the
carbonate ion, both of which are reversible processes.
H2CO 3 U HCO3  H U CO3  2H

The bicarbonate ion is neither a strong acid nor a strong base. The direction
of the reaction is governed by the type of solution in which the ion is present.
As per the above explanation, because the buffers will accept and donate
hydrogen ions in acidic and basic solutions, respectively, there ought to exist some
pH value at which point an equilibrium will be obtained so that there is no more
gaining or donating of hydrogen ions. This equilibrium is the pH that the buffer
always strives to maintain. The equilibrium pH for the bicarbonate buffer, which is
very important in blood, is 7.4.
Self-Instructional
328 Material
The human body consists of three important buffer systems which are as follows: Acid-Base Balance

x Bicarbonate Buffer System


x Phosphate Buffer System
x Protein Buffer System NOTES
Bicarbonate Buffer System: In this system, carbon dioxide is converted into
bicarbonate ions in the blood, as a result of which the bicarbonate ions float around
in the blood, and maintain the blood pH of 7.4 (Refer Figure 13.2).

Carbon Carbonic Bicarbonate Carbonate


Dioxide Acid Ion Ion

Figure 13.2 The Carbonate–Bicarbonate Buffer System

Phosphate Buffer System: Phosphoric acid is produced as a result of the


breaking down of nucleic acid and phosphoprotein. Phosphoric acid is denoted
as H3PO4 (Refer Figure 13.3). H3PO4 converts very quickly to dihydrogen
phosphate (H2PO4-), which acts as an excellent buffer, as it can either gain a
hydrogen ion and produce phosphoric acid, or release another hydrogen ion and
produce monohydrogen phosphate or HPO42-. The following Figure 6.3 reveals
that under extreme basic conditions, monohydrogen phosphate can donate the
remaining hydrogen ion.
In case of an acidic solution, the above mentioned reactions move towards
the left, and in case of a basic solution, they proceed towards the right. Hence, it
is evident that the phosphate buffer system is capable of accepting or donating
hydrogen ions depending on the solution it is in.
pKa = 2.1 6.9 12.8

H3PO 4 l H2PO41  H l HPO42  H l PO43  H

Figure 13.3 The Phosphate Buffer System

Protein Buffer System: You are well aware of the fact that proteins themselves
act as buffers, because when bicarbonate ions are formed, the hydrogen ions,
which also result from bicarbonate production, are taken in by the blood proteins.
Proteins are composed of amino acids, which consist of a central carbon
with four groups off of it (Refer Figure 13.4):
x A Carboxyl Group (COOH)
x An Amino Group (NH2)
Self-Instructional
Material 329
Acid-Base Balance x A Hydrogen Atom
x An R Group

NOTES

Figure 13.4 Chemical Structure of an Amino Acid

It is because of these carboxyl and amino groups that the proteins can
themselves act as buffers.
Buffers prevent excessive changes in pH by removing or releasing hydrogen
ions. If excess hydrogen ion is present in body fluids, buffers bind with the hydrogen
ion, minimizing the change in pH. When body fluids become too alkaline, buffers
can release hydrogen ion, again minimizing the change in pH. The action of a
buffer is immediate, but limited in its capacity to maintain or restore normal acid–
base balance. The major buffer system in extracellular fluids is the bicarbonate
(HCO3-) and carbonic acid (H2CO3) system. When a strong acid such as
HydroChloric Acid (HCl) is added, it combines with bicarbonate and the pH
drops only slightly. A strong base such as sodium hydroxide combines with carbonic
acid, which is the weak acid of the buffer pair, and the pH remains within the
narrow range of normal.
The amounts of bicarbonate and carbonic acid in the body vary; however,
as long as a ratio of 20 parts of bicarbonate to 1 part of carbonic acid is maintained,
the pH remains within its normal range of 7.35–7.45. Adding a strong acid to
ECF (ExtraCellular Fluid)can change this ratio as bicarbonate is depleted in
neutralizing the acid. When this happens, the pH drops, and the individual suffers
from a condition called acidosis. The ratio can also be upset by adding a strong
base to ECF, depleting carbonic acid as it combines with the base. In this case the
pH rises and the individual has alkalosis. In addition to the bicarbonate carbonic
acid buffer system, plasma proteins, haemoglobin and phosphates also function as
buffers in body fluids.
13.3.2 Respiratory and Renal Regulation
In respiratory regulation, the lungs can also help you to regulate the acid–base
balance by eliminating or retaining carbon dioxide (CO2), a potential acid.
Combined with water, carbon dioxide forms carbonic acid (CO2 + H2O o
H2CO3). This chemical reaction is reversible; carbonic acid breaks down into
carbon dioxide and water. Working together with the bicarbonate–carbonic acid
buffer system, the lungs regulate acid–base balance and pH by altering the rate
and depth of respirations. The response of the respiratory system to changes in
pH is rapid, occurring within minutes. Carbon dioxide is a powerful stimulator of
the respiratory centre. When blood levels of carbonic acid and carbon dioxide
rise, the respiratory centre is stimulated and the rate and depth of respirations
Self-Instructional
330 Material
increase. Carbon dioxide is exhaled, and carbonic acid levels fall. By contrast, Acid-Base Balance

when bicarbonate levels are excessive, the rate and depth of respirations are
reduced. This causes carbon dioxide to be retained, carbonic acid levels to rise
and the excess bicarbonate to be neutralized.
NOTES
Carbon dioxide levels in the blood are measured in terms of PCO2, or
partial pressure of the dissolved gas in the blood. The pressure of carbon dioxide
present in the venous blood is termed as PvCO2 (range 40–50 mmHg), whereas
in the arterial blood it is termed as PaCO2 (35–45 mmHg).
Renal Regulation
Although buffers and the respiratory system can compensate for changes in pH, the
kidneys are the ultimate long-term regulator of acid–base balance. They are slower
to respond to changes, requiring hours to days for correcting imbalances, but
according to Yucha (2004), their response is more permanent and selective than that
of the other systems. Selective excretion or conservation of bicarbonate and hydrogen
ions aid the kidneys in maintaining the acid–base balance. When excess of hydrogen
ions are present and the pH falls (acidosis), the kidneys reabsorb and regenerate
bicarbonate and excrete hydrogen ions. In the case of alkalosis and a high pH,
excess bicarbonate is excreted and hydrogen ion is retained. The normal serum
bicarbonate level is within the range 22–26 meq/L. The lungs and kidneys are the
two major systems that are working on a continuous basis to help regulate the acid–
base balance in the body (Refer Figure 13.5). In the biochemical reactions above,
the processes are all reversible and move back and forth as the body’s needs
change. The lungs can work very quickly and do their part by either retaining or
getting rid of carbon dioxide by changing the rate and depth of respirations. The
kidneys work much more slowly; they may take hours to days to regulate the
balance by either excreting or conserving hydrogen and bicarbonate ions. Under
normal conditions, the two systems work together to maintain homeostasis.
Lungs Kidneys
CO 2  H2O l H2CO 3 l H  HCO 2
Carbon dioxide Hydrogen
+ Cabonic acid +
water bicarbonate

Figure 13.5 Acid–Base Balance Maintained by the Lungs and the Kidneys

13.4 ACID–BASE DISORDERS

When your body fails to maintain its pH or the acid–base balance, it results in the
development of various acid–base disorders as will be discussed in this section.
Acid–base disorders generally are classified as respiratory or metabolic by
the general or underlying cause of the disorder. Carbonic acid levels are normally
regulated by the lungs through the retention or excretion of carbon dioxide, and
Self-Instructional
Material 331
Acid-Base Balance problems of regulation lead to respiratory acidosis or alkalosis. Bicarbonate and
hydrogen ion levels are regulated by the kidneys, and problems of regulation lead
to metabolic acidosis or alkalosis. Healthy regulatory systems will attempt to correct
acid–base imbalances, a process called compensation.
NOTES
Respiratory Acidosis: Hypoventilation and carbon dioxide retention cause
carbonic acid levels to increase and the pH to fall below 7.35, a condition known
as respiratory acidosis. Serious lung diseases such as asthma and COPD (Chronic
obstructive pulmonary disease) are common causes of respiratory acidosis. Central
nervous system depression due to anaesthesia or a narcotic overdose can
sufficiently slow the respiratory rate so that carbon dioxide is retained. When
respiratory acidosis occurs, the kidneys retain bicarbonate to restore the normal
carbonic acid to bicarbonate ratio. Recall, however, that the kidneys are relatively
slow to respond to changes in acid–base balance, so this compensatory response
may require hours to days to restore the normal pH.
Respiratory Alkalosis: When a person hyperventilates, more carbon dioxide
than normal is exhaled, carbonic acid levels fall, and the pH rises to above 7.45.
This condition is termed respiratory alkalosis. Psychogenic or anxiety-related
hyperventilation is a common cause of respiratory alkalosis. Other causes include
fever and respiratory infections. In respiratory alkalosis, the kidneys will excrete
bicarbonate to return the pH to within the normal range. Often, however, the
cause of the hyperventilation is eliminated and the pH returns to normal before
renal compensation occurs.
Metabolic Acidosis: When bicarbonate levels are low in relation to the amount
of carbonic acid in the body, the pH falls and metabolic acidosis develops. This
may develop because of renal failure and the inability of the kidneys to excrete
hydrogen ion and produce bicarbonate. It also may occur when too much acid is
produced in the body, for example, in diabetic ketoacidosis or starvation when fat
tissue is broken down for energy. Metabolic acidosis stimulates the respiratory
centre, and the rate and depth of respirations increase. Carbon dioxide is eliminated
and carbonic acid levels fall, minimizing the change in pH. This respiratory
compensation occurs within minutes of the pH imbalance.
Metabolic Alkalosis: In metabolic alkalosis, the amount of bicarbonate in the
body exceeds the normal 20-to-1 ratio. Ingestion of bicarbonate of soda as an
antacid is one cause of metabolic alkalosis. Another cause is prolonged vomiting
with loss of hydrochloric acid from the stomach. The respiratory centre is depressed
in metabolic alkalosis, and respiration become slow and more shallow. Carbon
dioxide is retained and carbonic acid levels increase, helping to balance the excess
bicarbonate.

13.5 ANION GAP

The concentration of all the unmeasured anions present in the plasma is denoted
by the term Anion Gap (AG). When the conditions are normal, proteins which
Self-Instructional
332 Material
have negative charge constitute about 10% of the plasma anions and this represents Acid-Base Balance

the major portion of the unmeasured anion or the AG. As per the usual laboratory
biochemical profile, the acid anions (for example lactate, acetoacetate, sulphate)
which are produced as a result of metabolic acidosis are not measured. Reaction
takes place between the hydrogen ions produced and the bicarbonate anions NOTES
(buffering); this results in the production of CO2 which is excreted through the
lungs (also known as respiratory compensation). The complete procedure leads
to a decrease in the measured anion concentration (i.e., HCO3) and an increase in
the concentration of the unmeasured anions (the acid anions), as a result of which
the AG increases.
The calculation of AG is done through the following formula:
AG = [Na+] - [Cl-] - [HCO3-]
The reference range is 8–16 mmol/L. Nephrologists at times use an
alternative formula which includes K+. In Renal Units, K+ can vary over a wider
range and have more effect on the measured AG. The alternative formula is given
by:
AG = [Na+] + [K+] - [Cl-] - [HCO3-]
The reference range is slightly higher with this alternative formula.
Principal Clinical Uses of the AG
x To indicate the presence of metabolic acidosis and establish other findings.
x To aid in distinguishing among various causes of metabolic acidosis: high
versus normal AG metabolic acidosis. In inorganic metabolic acidosis (for
example, due to HCl infusion), the infused Cl- replaces HCO3 and the AG
remains normal. On the contrary, in an organic acidosis, the lost bicarbonate
is replaced by the acid anion which is not normally measured. This means
that the AG is increased.
x To assist in assessing the biochemical severity of the acidosis and follow the
response to treatment.
The AG can be Misleading
In AG determination the calculation includes additional three measured ions, as a
result of which the error in case of an AG is much higher than that of a single
electrolyte determination. The most common reason for a low AG is laboratory
error in the electrolyte determinations. The 95% error range for AG is approximately
±5 mmol/L (i.e., 10 mmol/L range)
x If AG > 30 mmol/L, then it confirms the presence of metabolic acidosis.
x If the AG range is 20–29 mmol/L, then metabolic acidosis will not be present
in about one-third of the patients.
The AG is used especially if very elevated or used to establish other findings:
x Lactate
x Creatinine Self-Instructional
Material 333
Acid-Base Balance x Plasma Glucose
x Urine Ketone Test
The principal unmeasured anion is albumin and contributes almost the entire
NOTES value of the AG. With each 1 g decrease in albumin the AG will also decrease by
2.5–3 mmol. A normally high AG acidosis in a patient with hypoalbuminaemia
may appear as a normal AG acidosis. This is particularly applicable in case of
Intensive Care patients for whom reduced levels of albumin are common. Lactic
acidosis in a patient suffering from hypoalbuminaemia in the ICU will generally be
associated with a normal AG.

13.6 ASSESSMENT OF ACID–BASE HOMEOSTASIS

Acid–base homeostasis is a part of human homeostasis concerning the proper


balance between acids and bases, also known as body pH. The assessment of
acid-base homeostasis using bicarbonate buffering system and the Henderson–
Hasselbalch equation is discussed below.
The Bicarbonate Buffering System and the Henderson–Hasselbalch
Equation
In assessing acid–base homeostasis, components of the bicarbonate buffering
system are measured and calculated. From the data, inferences can be made
pertaining to the other buffers and the systems that regulate the production, retention
and excretion of acids and bases. For the bicarbonate buffering system, the dissolved
CO2 (dCO2) is in equilibrium with CO2 gas, which can be expelled by way of the
lungs. Therefore, the bicarbonate buffering system is referred to as an open system,
and the dCO2, which is controlled by the lungs, is the respiratory component. The
lungs participate rapidly in the regulation of blood pH through hypoventilation or
hyperventilation. Mainly, the kidneys, the non-respiratory or formerly known as
the metabolic component, control the bicarbonate concentration.
The Henderson–Hasselbalch equation expresses acid–base relationships
in a mathematical formula:
cA 
pH  pK c  log
cHA
where A- is a proton acceptor or base (for example, HCO3-), HA is a proton
donor or weak acid (for example, H2CO3) and pK’ is the pH at which there is an
equal concentration of protonated and unprotonated species. Knowing any of the
three variables allows the calculation of the fourth.
In plasma and at body temperature (37°C), the pK¢ of the bicarbonate
buffering system is 6.1. The equilibrium between H2CO3 and CO2 in plasma is
approximately 1:800. The concentration of H2CO3 is proportional to the partial
pressure exerted by the dissolved CO2. In plasma at 37°C, the value for the
combination of the solubility constant for pCO2 and the factor to convert mmHg
Self-Instructional
334 Material
to millimoles per litre is 0.0307 mmol/L/mmHg. Temperature and the solvent affect Acid-Base Balance

the constant. If either of these changes, the solubility constant also will change.
Both pH and pCO2 are measured in blood gas analysis and the pK¢ is a constant;
therefore, HCO3 can be calculated as:
NOTES
cHCO 3
pH  pK c  log
0.0307 u PCO 2

In healthy condition, when the kidneys and lungs are functioning properly, a
20:1 ratio of HCO3- to H2CO3 will be maintained (resulting in a pH of 7.40).

Check Your Progress


1. What is the use of Henderson Hassebalch equation?
2. What is calculated with the use of Henderson-Hasselbach equation?
3. What is a buffer?
4. How many buffer systems does human body consists of?
5. How is calculation of Anion Gap (AG) is done?

13.7 ANSWERS TO CHECK YOUR PROGRESS


QUESTIONS

1. The Henderson–Hasselbalch equation aids in studying the nature of the


titration curve of a weak acid, which is crucial for understanding the buffer
action and the acid–base equilibrium in the blood and tissues of vertebrates.
2. Through the Henderson–Hasselbalch equation you can calculate the
following:
x pKa, when pH and the molar ratio of proton donor and acceptor are
given.
x pH, when pKa and the molar ratio of proton donor and acceptor are
given.
x Molar ratio of proton donor and acceptor, when pH and pKa are given.
3. A buffer, which is the combination of a weak acid or weak base and its salt,
is a system that resists changes in pH. The effectiveness of a buffer depends
on the pK of the buffering system and the pH of the environment in which it
is placed. In plasma, the bicarbonate–carbonic acid system, having a pK of
6.1, is one of the principal buffers.
4. The human body consists of three important buffer systems which are as
follows:
x Bicarbonate Buffer System
x Phosphate Buffer System
x Protein Buffer System
Self-Instructional
Material 335
Acid-Base Balance 5. The calculation of AG is done through the following formula:
AG = [Na+] - [Cl-] - [HCO3-]

NOTES 13.8 SUMMARY


x The chemical balance or homeostasis of body fluids is maintained by regulating
their pH (which gives a measure of how acidic or basic a substance is).
x Substances releasing hydrogen ions (H+) in solution are referred to as acids.
Acids which release most of their ions are called strong acids (for example
hydrochloric acid) and those which release only some of the hydrogen ions
are called weak acids (for example, carbonic acid).
x Bases or alkalis generally have a low hydrogen ion concentration and can
accept hydrogen ions in solution.
x The pH reflects the hydrogen ion concentration of the solution: the higher
the hydrogen ion concentration (and the more acidic the solution), the lower
will be the pH.
x The Henderson–Hasselbalch equation aids in studying the nature of the
titration curve of a weak acid, which is crucial for understanding the buffer
action and the acid–base equilibrium in the blood and tissues of vertebrates.
x An acid is a substance that can yield a hydrogen ion (H+) or hydronium ion
when dissolved in water. On the other hand, a base is a substance that can
yield hydroxyl ions (OH-).
x The relative strengths of acids and bases, and their ability to dissociate in
water are described by their dissociation constant (also known as ionization
constant or K value).
x The pK value, which is defined as the negative log of the ionization constant,
is also the pH in which the protonated and unprotonated forms are present
in equal concentrations. Strong acids have pK values of less than 3.0,
whereas strong bases have pK values greater than 9.0.
x A buffer, which is the combination of a weak acid or weak base and its salt,
is a system that resists changes in pH.
x The effectiveness of a buffer depends on the pK of the buffering system
and the pH of the environment in which it is placed.
x Human body consists of three important buffer systems which are as follows:
R Bicarbonate Buffer System
R Phosphate Buffer System
R Protein Buffer System
x Acid–base disorders generally are classified as respiratory or metabolic by
the general or underlying cause of the disorder.
x Hypoventilation and carbon dioxide retention cause carbonic acid levels to
increase and the pH to fall below 7.35, a condition known as respiratory
acidosis.
Self-Instructional
336 Material
x When a person hyperventilates, more carbon dioxide than normal is exhaled, Acid-Base Balance

carbonic acid levels fall, and the pH rises to above 7.45. This condition is
termed respiratory alkalosis.
x In metabolic alkalosis, the amount of bicarbonate in the body exceeds the
NOTES
normal 20-to-1 ratio. Ingestion of bicarbonate of soda as an antacid is one
cause of metabolic alkalosis. Another cause is prolonged vomiting with loss
of hydrochloric acid from the stomach.
x The concentration of all the unmeasured anions present in the plasma is
denoted by the term Anion Gap (AG).

13.9 KEY WORDS

x Acid: Substance capable of releasing hydrogen ions are term as acids.


x Base: Substance capable of releasing hydroxyl ions are called as bases.
x Buffer: A combination of a weak acid or weak base and its salt which
helps to maintain the pH is called as buffer.
x Coma: A state of deep unconsciousness that lasts for a prolonged or
indefinite period, especially caused by severe injury or illness.
x Homeostasis: The tendency toward a relatively stable equilibrium between
interdependent elements, especially as maintained by physiological
processes.
x Metabolism: The chemical processes that occur within a living organism
in order to maintain life.
x Narcotic: A drug or other substance affecting mood or behaviour and sold
for non-medical purposes.
x Protonated: Transfer a proton to a molecule, group or atom so that a
coordinate bond to the proton is formed.
x Titration: A measured amount of a solution of unknown concentration added
to a known volume of a second solution until the reaction occurs between
them.
x Tetany: A condition marked by intermittent muscular spasms, caused by
malfunction of the parathyroid glands and a consequent deficiency of calcium.
x Respiratory acidosis: Condition in which hypoventilation and carbon
dioxide retention cause carbonic acid levels to increase and the pH to fall
below 7.35.
x Respiratory alkalosis: Condition in which during hyperventilation, more
carbon dioxide than normal is exhaled by an individual, carbonic acid levels
fall and the pH rises to above 7.45.
Self-Instructional
Material 337
Acid-Base Balance
13.10 SELF ASSESSMENT QUESTIONS AND
EXERCISES

NOTES Short Answer Questions


1. What is blood buffer system?
2. What is respiratory regulation?
3. Write a short note on renal regulation.
4. Explain about acid-base disorders.
Long Answer Questions
1. Explain and derive the Henderson–Hasselbalch equation.
2. Explain the acid–base balance in the human body.
3. What do you understand by the blood buffer system? Describe the different
types of buffer systems present inside the human body.
4. Briefly describe the mechanisms of respiratory and renal regulation.
5. What are acid–base disorders? Briefly describe the various types of acid–
base disorders.

13.11 FURTHER READING


Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.
Self-Instructional
338 Material
Fluid and Electrolyte

UNIT 14 FLUID AND ELECTROLYTE Balance

BALANCE
NOTES
Structure
14.0 Introduction
14.1 Objectives
14.2 Electrolytes
14.2.1 Sodium
14.2.2 Potassium
14.2.3 Calcium
14.2.4 Magnesium
14.2.5 Chloride
14.2.6 Bicarbonate
14.3 Electrolyte and Fluid Balance
14.4 Answers to Check Your Progress Questions
14.5 Summary
14.6 Key Words
14.7 Self Assessment Questions and Exercises
14.8 Further Reading

14.0 INTRODUCTION

Did you know that when you exercise vigorously and sweat, your body is actually
losing electrolytes like sodium and potassium? This loss of electrolytes through
sweat needs to be replenished. You may not be aware of the severe consequences
that can result from an imbalance of electrolytes. In this unit, you will find out what
electrolytes are, their importance and how to replenish them.
Electrolytes are salts that become charged molecules, called ions, when
they are dissolved in a liquid. Their electrical charges and the ability to conduct
electricity help the body to send electrical signals from one cell to another. The
different types of electrolytes include sodium, potassium, chloride, bicarbonate,
calcium, sulphate, magnesium and phosphate and we will discuss these in detail in
this unit.
Electrolytes are ions capable of carrying an electric charge. They are classified
as anions or cations based on the type of charge they carry. These names were
determined years ago based on how the ion migrates in an electric field. Anions
have a negative charge and move toward the anode, whereas cations migrate in
the direction of the cathode because of their positive charge.
Electrolytes are an essential component in numerous processes, including
volume and osmotic regulation [Sodium (Na+), Chloride (Cl-), Potassium (K+)];
myocardial rhythm and contractility [(K+), Magnesium (Mg2+), Calcium (Ca2+)];
cofactors in enzyme activation (for example, Mg2+, Ca2+, Zn2+); regulation of
Self-Instructional
Material 339
Fluid and Electrolyte Adenosine Triphosphatase (ATPase) Ion pumps (Mg2+); acid–base balance
Balance
(Bicarbonate [HCO3-], K+, Cl-); blood coagulation (Ca2+, Mg2+); neuromuscular
excitability (K+, Ca2+, Mg2+); and the production and use of ATP from Glucose
(for example, Mg2+, Phosphate [PO4-]). Because many of these functions require
NOTES electrolyte concentrations to be held within narrow ranges, the body has complex
systems for monitoring and maintaining electrolyte concentrations.
In this unit, you will study about the metabolic physiology and regulation of
each electrolyte in the body, and relate these factors to the clinical significance of
electrolyte measurements. In addition, methodologies used in determining
concentrations of the individual analytes will also be discussed.

14.1 OBJECTIVES

After going through this unit, you will be able to:


x Explain different types of electrolytes and electrolytic balance
x Discuss the determination and regulation of sodium, potassium, calcium,
magnesium, chloride and bicarbonate
x Explain the fluid balance mechanism inside the human body
x Discuss about the distribution, daily input and output, and regulation of
fluids in the human body
x Explain the concepts of hypovolemia and hypervolemia

14.2 ELECTROLYTES

Electrolytes maintain the electric voltage throughout your cells so that signals can
pass easily. Several bodily functions are dependent on this electrical communication
that electrolytes help to carry. They include regulating nerve and muscle function,
acidity levels and fluid levels. An imbalance of electrolytes has very serious
consequences. For example, bicarbonate is an electrolyte that is responsible for
regulating muscles like the heart. Insufficient levels of bicarbonate would result in
irregular heartbeats, which may be fatal.
The average water content of the human body varies from 40 to 75% of
total body weight, with values declining with age and especially with obesity. Women
have lower average water content than men as a result of a higher fat content.
Water is the solvent for all processes in the human body. It transports nutrients to
cells, determines cell volume by its transport into and out of cells, removes waste
products by way of urine and acts as the body’s coolant by way of sweating.
Water is located in intracellular and extracellular compartments. IntraCellular Fluid
(ICF) is the fluid inside the cells and accounts for about two-thirds of the total
body water. ExtraCellular Fluid (ECF) accounts for the other one-third of the
total body water and can be subdivided into the intravascular ECF (plasma) and
Self-Instructional
340 Material
the interstitial cell fluid that surrounds the cells in the tissue. Normal plasma is Fluid and Electrolyte
Balance
about 93% water, with the remaining volume occupied by lipids and proteins. The
electrolytes present in ExtraCellular Fluid (ECF) and IntraCellular Fluid (ICF) are
shown in Figure 14.1.
NOTES

Fig. 14.1 Electrolytes in ECF and ICF

The concentrations of ions within cells and in plasma are maintained both by
energy-consuming active transport processes and by diffusion or passive transport
processes. Active transport is a mechanism that requires energy to move ions
across cellular membranes. For example, maintaining a high intracellular
concentration of K+ and a high extracellular (plasma) concentration of Na+ requires
use of energy from ATP in ATPase-dependent ion pumps. Diffusion is the passive
movement of ions across a membrane. It depends on the size and charge of the
ion being transported and on the nature of the membrane through which it is passing.
The rate of diffusion of various ions also may be altered by physiologic and hormonal
processes.
By maintaining the concentration of proteins and electrolytes in a controlled
yet somewhat flexible environment, the distribution of water in these compartments
also can be controlled. Most of the biologic membranes are freely permeable to
water but not to ions or proteins. The concentration of ions and proteins on one
side of the membrane or the other, influence the flow of water across a membrane
(an osmoregulator). In addition to the osmotic effects of Na+, other ions, proteins
and blood pressure influence the flow of water across a membrane.
14.2.1 Sodium
Sodium ion (Na+) is the most abundant cation in the ECF, representing 90% of all
extracellular cations, and largely determines the osmolality of the plasma. Osmolality
of normal plasma is approximately 295 mmol/L, with 270 mmol/L being the result
of Na+ and associated anions. Na+ concentration in the ECF is much larger than
inside the cells. Because a small amount of Na+ can diffuse through the cell
membrane, the two sides would eventually reach equilibrium. To prevent equilibrium
from occurring, active transport systems, such as ATPase ion pumps are present
in all cells. K+ is the major intracellular cation. Like Na+, K+ would eventually
diffuse across the cell membrane until equilibrium is reached. The Na+–K+ ATPase
ion pump moves three Na+ ions out of the cell in exchange for two K+ ions moving
Self-Instructional
Material 341
Fluid and Electrolyte into the cell as ATP is converted to ADP. Because water follows electrolytes across
Balance
cell membranes, the continual removal of Na+ from the cell prevents osmotic rupture
of the cell by also drawing water from the cell.

NOTES Determination of Sodium: Specimen


Serum, plasma and urine are all acceptable for Na+ measurements. When plasma
is used, lithium heparin, ammonium heparin and lithium oxalate are suitable
anticoagulants. Haemolysis does not cause a significant change in serum or plasma
values as a result of decreased levels of intracellular Na+. However, with marked
haemolysis, levels may be decreased as a result of a dilution effect.
Whole blood samples may be used with some analysers. Consult the
instrument operation manual for acceptability. The specimen of choice in urine
Na+ analyses is a 24-hour collection. Sweat is also suitable for analysis.
Methods
Na+ can be measured in various ways, including chemical methods, Flame Emission
Spectrophotometry (FES), Atomic Absorption Spectrophotometry (AAS) and
Ion-Selective Electrodes (ISEs). Chemical methods are outdated because of large
sample volume requirements and lack of precision. ISEs are the most routinely
used methods in clinical laboratories.
ISE method uses a semipermeable membrane to develop a potential
produced by having different ion concentrations on either side of the membrane.
Here, two electrodes are used. One electrode has a constant potential, making it
the reference electrode (Refer Figure 14.2). The difference in potential between
the reference and measuring electrodes can be used to calculate the ‘concentration’
of the ion in solution. However, it is the activity of the ion, not the concentration
that is being measured.

Fig.14.2 Ion-Selective Method


Self-Instructional
342 Material
Most analysers use a glass ion-exchange membrane in its ISE system for Fluid and Electrolyte
Balance
+
Na measurement. There are two types of ISE measurement, based on sample
preparation: direct and indirect. Direct measurement provides an undiluted sample
to interact with the ISE membrane. With the indirect method, a diluted sample is
used for measurement. NOTES
One source of error with ISEs is protein build-up on the membrane through
continuous use. The protein coated membranes cause poor selectivity, which results
in poor reproducibility of results.
14.2.2 Potassium
Potassium ion (K+) is the major intracellular cation in the body, with a concentration
20 times greater inside the cells than outside. Many cellular functions require that
the body maintain a low ECF concentration of K+ ions. As a result, only 2% of the
body’s total K+ circulates in the plasma. Functions of K+ in the body include
regulation of neuromuscular excitability, contraction of the heart, ICF volume and
H+ concentration. The K+ concentration has a major effect on the contraction of
skeletal and cardiac muscles. Elevated plasma K+ decreases the Resting Membrane
Potential (RMP) (Refer Figure 14.3) of the cell (the RMP is closer to zero), which
decreases the net difference between the cell’s resting potential and threshold
(action) potential. A lower-than-normal difference increases cell excitability, leading
to muscle weakness. Severe hyperkalemia can ultimately cause a lack of muscle
excitability (as a result of a higher RMP than action potential), which may lead to
paralysis or a fatal cardiac arrhythmia. Hypokalemia decreases cell excitability by
increasing the RMP, often resulting in an arrhythmia or paralysis.

Fig. 14.3 Resting Membrane Potential

Determination of Potassium: Specimen


Serum, plasma and urine may be acceptable for analysis. Haemolysis must be
avoided because of the high K+ content of erythrocytes. Heparin is the anticoagulant
Self-Instructional
Material 343
Fluid and Electrolyte of choice. Whereas serum and plasma generally give similar K+ levels, serum
Balance
reference intervals tend to be slightly higher. Significantly elevated platelet counts
may result in the release of K+ during clotting from rupture of these cells, causing
a spurious hyperkalemia. In this case, plasma is preferred. Whole blood samples
NOTES may be used with some analysers. Consult the instrument’s operations manual for
acceptability. Urine specimens should be collected over a 24-hour period to
eliminate the influence of diurnal variation.
Methods
As with Na+, the current method of choice is ISE. For ISE measurements, a
valinomycin membrane is used to selectively bind K+, causing an impedance change
that can be correlated to K+ concentration. KCl- is the inner electrolyte solution.
14.2.3 Calcium
In 1883, Ringer showed that Ca2+ was essential for myocardial contraction.
Because decreased ionized Ca2+ impairs myocardial function, it is important to
maintain ionized Ca2+ at a near-normal concentration during surgery and in critically
ill patients. Decreased ionized Ca2+ concentrations in blood can cause neuromuscular
irritability, which may become clinically apparent as irregular muscle spasms, called
tetany.
Determination of Calcium: Specimen

The preferred specimen for total Ca2+ determinations is either serum or lithium
heparin plasma collected without venous stasis. Because anticoagulants such as
EthyleneDiamineTetraacetic Acid (EDTA) or oxalate bind Ca2+ tightly and interfere
with measurement, they are unacceptable for use.
The proper collection of samples for ionized Ca2+ measurements requires
greater care. Because loss of CO2 will increase pH, samples must be collected
anaerobically. Although heparinized whole blood is the preferred sample, serum
from sealed evacuated blood collection tubes may be used if clotting and
centrifugation are done quickly (30 minutes) and at room temperature. No liquid
heparin products should be used. Most heparin anticoagulants (Na+, lithium)
partially bind to Ca2+ and lower ionized Ca2+ concentrations. A heparin
concentration of 25 IU/mL, for example, decreases ionized Ca2+ by about 3%.
Dry heparin products are available titrated with small amounts of Ca2+ or Zn2+ or
with small amounts of heparin dispersed in an inert ‘puff’ that essentially eliminates
the interference by heparin.
For analysis of Ca2+ in urine, an accurately timed urine collection is preferred.
The urine should be acidified with 6 mol/L HCl, with approximately 1 mL of the
acid added for each 100 mL of urine.
Self-Instructional
344 Material
Methods Fluid and Electrolyte
Balance
The two commonly used methods for total Ca2+ analysis use either ortho-
CresolPhthalein Complexone (CPC) or Arsenazo-III dye to form a complex with
Ca2+. Prior to the dye-binding reaction, Ca2+ is released from its protein carrier NOTES
and complexes by acidification of the sample. The CPC method uses 8-
hydroxyquinoline to prevent Mg2+ interference.
14.2.4 Magnesium
Magnesium (Mg2+) is the fourth most abundant cation in the body and second
most abundant intracellular ion. The average human body (70 kg) contains 1 mol
(24 g) of Mg2+. Approximately 53% of Mg2+ in the body is found in bone, 46% in
muscle and other organs and soft tissue, and less than 1% is present in serum and
red blood cells. Of the Mg2+ present in serum, about one-third is bound to protein,
primarily albumin. Of the remaining two-thirds, 61% exists in the free or ionized
state and about 5% is combined with other ions.
The role of Mg2+ in the body is widespread. It is an essential cofactor of
more than 300 enzymes, including those important in glycolysis, transcellular ion
transport, neuromuscular transmission, synthesis of carbohydrates, proteins, lipids,
and nucleic acids, and release of and response to certain hormones.
Determination of Magnesium: Specimen
Non-haemolysed serum or lithium heparin plasma may be analysed. Because the
Mg2+ concentration inside erythrocytes is 10 times greater than that in the ECF,
haemolysis should be avoided and the serum should be separated from the cells as
soon as possible. Oxalate, citrate and EDTA anticoagulants are unacceptable
because they will bind with Mg2+. A 24-hour urine sample is preferred for analysis
because of a diurnal variation in excretion. The urine must be acidified with HCl-
to avoid precipitation.
Methods
The three most common methods for measuring total serum Mg2+ are: colorimetric–
–calmagite, formazen dye and methylthymol blue. In the calmagite method, Mg2+
binds with calmagite to form a reddish-violet complex that may be read at 532
nm. In the formazen dye method, Mg2+ binds with the dye to form a coloured
complex that may be read at 660 nm. In the methylthymol blue method, Mg2+
binds with the chromogen to form a coloured complex. Most methods use a Ca2+
shelter to prohibit interference from this divalent cation.
The reference method for measuring Mg2+ is AAS. Although the measurement
of total Mg2+ concentrations in serum remains the usual diagnostic test for detection
of Mg2+ abnormalities, it has limitations. First, because approximately 25% of
Mg2+ is protein-bound, total Mg2+ may not reflect the physiologically active free
ionized Mg2+. Second, because Mg2+ is primarily an intracellular ion, serum
Self-Instructional
Material 345
Fluid and Electrolyte concentrations will not necessarily reflect the status of intracellular Mg2+. Even
Balance
when tissue and cellular Mg2+ is depleted by as much as 20%, serum Mg2+
concentrations may remain normal.

NOTES 14.2.5 Chloride


The exact function of chloride ion (Cl-), which is a major extracellular anion, in the
body is not well understood. But, it is clear that it is involved in maintaining osmolality,
blood volume and electric neutrality. In most processes, Cl shifts secondarily to a
movement of Na+ or HCO3-.
Cl- ingested in the diet is almost completely absorbed by the intestinal tract.
Cl- is then filtered out by the glomerulus and passively reabsorbed, in conjunction
with Na+, by the proximal tubules. Excess Cl- is excreted in the urine and sweat.
Excessive sweating stimulates aldosterone secretion, which acts on the sweat glands
to conserve Na+ and Cl-. Cl- maintains electrical neutrality in two ways. First, Na+
is reabsorbed along with Cl- in the proximal tubules. In effect, Cl- acts as the rate-
limiting component, in that Na+ reabsorption is limited by the amount of Cl- available.
Electroneutrality is also maintained by Cl- through the chloride shift. In this process,
carbon dioxide (CO2) generated by cellular metabolism within the tissue diffuses
out into both the plasma and the red cell.
Determination of Chloride: Specimen
Serum or plasma may be used, with lithium heparin being the anticoagulant of
choice. Haemolysis does not cause a significant change in serum or plasma values
as a result of decreased levels of intracellular Cl-. However, with marked haemolysis,
levels may be decreased as a result of a dilution effect.
Whole blood samples may be used with some analysers. Consult the
instrument’s operation manual for acceptability. The specimen of choice in urine
Cl- analyses is 24-hour collection because of the large diurnal variation. Sweat is
also suitable for the analysis.
Methods
There are several methodologies available for measuring Cl-, including ISEs,
amperometric coulometric titration, mercurimetric titration and colourimetry. The
most commonly used is ISE. For ISE measurement, an ion-exchange membrane
is used to selectively bind Cl- ions.
Amperometric–coulometric titration is a method using coulometric generation
of silver ions (Ag+), which combine with Cl- to quantitate the Cl- concentration.
Ag2  2CI o AgCI2
When all Cl- in a patient is bound to Ag2+, excess or free Ag2+ is used to
indicate the endpoint. As Ag2+ accumulates, the coulometric generator and timer
Self-Instructional
346 Material
are turned off. The elapsed time is used to calculate the concentration of Cl- in the Fluid and Electrolyte
Balance
sample. The digital (Cotlove) chloridometer (Labconco Corporation) uses this
principle in Cl- analysis.
14.2.6 Bicarbonate NOTES
Bicarbonate is the second most abundant anion in the ECF. Total CO2 comprises
the bicarbonate ion (HCO3-), carbonic acid (H2CO3) and dissolved CO2, with
HCO3- accounting for more than 90% of the total CO2 at physiologic pH. Because
HCO3- composes the largest fraction of total CO2, total CO2 measurement is
indicative of HCO3- measurement.
HCO3- is the major component of the buffering system in the blood. Carbonic
anhydrase in RBCs converts CO2 and H2O to carbonic acid, which dissociates
into H+ and HCO3-.
CO2 + H2O <-CA-> H2CO3 <-> H + HCO3
HCO3- diffuses out of the cell in exchange for Cl- to maintain ionic charge
neutrality within the cell. This process converts potentially toxic CO2 in the plasma
to an effective buffer: HCO3-.
HCO3- buffers excess H+ by combining with acid, then eventually dissociating
into H2O and CO2 in the lungs where the acidic gas CO2 is eliminated.
Determination of Carbon Dioxide: Specimen
For discussion of arterial and whole blood pCO2 measurements, serum or lithium
heparin plasma is suitable for analysis. Although specimens should be anaerobic
for the highest accuracy, many current analysers (excluding blood gas analysers)
do not permit anaerobic sample handling. In most instances, the sample is capped
until the serum or plasma is separated and the sample is analysed immediately. If
the sample is left uncapped before analysis, CO2 escapes. Two common methods
of CO2 measurement are ISE and an enzymatic method. One type of ISE for
measuring total CO2 uses an acid reagent to convert all the forms of CO2 to CO2
gas and is measured by a pCO2 electrode.
The enzyme method used for CO2 determination alkalinizes the sample to
convert all forms of CO 2 to HCO 3-. HCO 3- is used to carboxylate
PhosphoEnolPyruvate (PEP) in the presence of PhosphoEnolPyruvate (PEP)
carboxylase, which catalyses the formation of oxaloacetate.

14.3 ELECTROLYTE AND FLUID BALANCE

In electrolyte balance, inside the body electrolytes have a very major role to play
in maintaining homeostasis. They aid in regulating myocardial and neurological
function, fluid balance, oxygen delivery, acid–base balance and so on. Electrolyte
Self-Instructional
Material 347
Fluid and Electrolyte imbalances are produced because of the following factors: excessive ingestion,
Balance
diminished elimination of an electrolyte, diminished ingestion or excessive elimination
of an electrolyte. Kidney failure is the most common reason behind electrolyte
disturbances.
NOTES
The most serious electrolyte disturbance develops when the levels of sodium,
potassium and calcium become abnormal. Other types of electrolyte imbalances
are not so prevalent. Chronic laxative abuse or severe diarrhoea or vomiting
(gastroenteritis) can lead to electrolyte disturbances along with dehydration. People
suffering from bulimia or anorexia nervosa are at especially high risk for an
electrolyte imbalance.
Electrolytes are important because they are what cells (especially nerve,
heart, muscle) use to maintain voltages across their cell membranes and to carry
electrical impulses (nerve impulses, muscle contractions) across themselves and
to other cells. Kidneys work to keep the electrolyte concentrations in blood constant
despite changes in your body. For example, during heavy exercise, electrolytes
are lost in sweat, particularly sodium and potassium. These electrolytes must be
replaced to keep the electrolyte concentrations of the body fluids constant.
Fluid Balance

There are different compartments in the body through which water moves freely.
Two forces, viz. hydrostatic pressure and osmotic pressure, are responsible for
this motion. The motion of fluid across the capillary membrane is contributed by
these pressures, but osmotic pressure plays a major role in moving the fluid across
the plasma membrane of cells.
Total Body Fluid or Total Body Water (TBW) = 60% x Body Weight
x Extracellular Fluid: Extracellular fluid is formed by equal concentrations
of cations, i.e. positively charged ions (Ca2+, Mg2+, Na+), anions, i.e.,
negatively charged ions (Cl-, HCO3-), and proteins and is found outside the
cells.
x Intracellular Fluid: The intracellular fluid is formed by equal concentrations
of cations, i.e., positively charged ions (Ca2+, Mg2+, Na+), anions, i.e.,
negatively charged ions (Cl-, HCO3-) and proteins and is found inside the
cells.
x Fluid Movement between Compartments: The osmolality of the ECF
changes with intake of water and salt. Subsequently, water moves from
lower solute concentration to higher solute concentration between ECF
and ICF. This movement of water towards the compartment having higher
solute concentration takes place till the equilibrium is reached between these
two compartments.
Self-Instructional
348 Material
x Role of Kidney in Fluid Regulation: Nephron, a functional unit of kidney Fluid and Electrolyte
Balance
is found inside the renal medulla. Nephrons drive the fluid collectively into
the collecting duct system to which they are connected. Finally the fluid is
excreted from the collecting duct system as urine.
NOTES
Fluid Distribution
The body’s fluid is divided into two major compartments: intracellular and
extracellular. IntraCellular Fluid (ICF) is found within the cells of the body. It
constitutes approximately two-thirds of the total body fluid in adults. ExtraCellular
Fluid (ECF) is found outside the cells and accounts for about one-third of total
body fluid. It is subdivided into compartments. The two main compartments of
ECF are intravascular and interstitial. Intravascular fluid, or plasma, accounts for
approximately 20% of the ECF and is found within the vascular system. Interstitial
fluid, accounting for approximately 75% of the ECF, surrounds the cells. The
other compartments of ECF are the lymph and transcellular fluids. Examples of
transcellular fluid include cerebrospinal, pericardial, pancreatic, pleural, intraocular,
biliary, peritoneal, and synovial fluids. Fluid distribution within a human body is
represented in Figure 14.4.

Fig. 14.4 Distribution of Fluids in the Body

Intracellular fluid is vital to normal cell functioning. It contains solutes such


as oxygen, electrolytes, and glucose, and it provides a medium in which metabolic
processes of the cell take place. Although extracellular fluid is in the smaller of the
two compartments, it is the transport system that carries nutrients to and waste
products from the cells. For example, plasma carries oxygen from the lungs and
glucose from the gastrointestinal tract to the capillaries of the vascular system.
From there, the oxygen and glucose move across the capillary membranes into the
interstitial spaces and then across the cellular membranes into the cells. The opposite
route is taken for waste products, such as carbon dioxide going from the cells to
Self-Instructional
Material 349
Fluid and Electrolyte the lungs and metabolic acid wastes going eventually to the kidneys. Interstitial
Balance
fluid transports wastes from the cells by way of the lymph system as well as directly
into the blood plasma through capillaries.

NOTES Regulating Body Fluids


In a healthy person, the volumes and chemical composition of the fluid
compartments stay within narrow safe limits. Normally fluid intake and fluid loss
are balanced. Illness can upset this balance so that the body has too little or too
much fluid.
Fluid Input
During periods of moderate activity at moderate temperature, the average adult
drinks about 1,500 mL per day but needs 2,500 mL per day, an additional 1,000
mL. This added volume is acquired from foods and from the oxidation of these
foods during metabolic processes. Interestingly, the water content of food is
relatively large, contributing about 750 mL per day. The water content of fresh
vegetables is approximately 90%, of fresh fruits about 85%, and of lean meats
around 60%. Water as a by-product of food metabolism accounts for most of the
remaining fluid volume required. This quantity is approximately 200 mL per day
for the average adult.
The thirst mechanism is the primary regulator of fluid intake. The thirst centre
is located in the hypothalamus of the brain. A number of stimuli trigger this centre,
including the osmotic pressure of body fluids, vascular volume, and angiotensin (a
hormone released in response to decreased blood flow to the kidneys). For
example, a long-distance runner loses significant amounts of water through
perspiration and rapid breathing during a race, increasing the concentration of
solutes and the osmotic pressure of body fluids. This increased osmotic pressure
stimulates the thirst centre, causing the runner to experience the sensation of thirst
and the desire to drink to replace lost fluids.
Thirst is normally relieved immediately after drinking a small amount of fluid,
even before it is absorbed from the gastrointestinal tract. However, this relief is
only temporary, and the thirst returns in about 15 minutes. The thirst is again
temporarily relieved after the ingested fluid distends the upper gastrointestinal tract.
These mechanisms protect the individual from drinking too much, because it takes
from 30 minutes to 1 hour for the fluid to be absorbed and distributed throughout
the body.
Fluid Output
Fluid losses from the body counterbalance the adult’s 2,500-mL average daily
intake of fluid, as there are four routes of fluid output:
x Urine.
x Insensible loss through the skin as perspiration and through the lungs as
water vapour in the expired air.
Self-Instructional
350 Material
x Noticeable loss through the skin. Fluid and Electrolyte
Balance
x Loss through the intestines in faeces.
x Urine: Urine formed by the kidneys and excreted from the urinary
bladder is the major avenue of fluid output. Normal urine output for an NOTES
adult is 1,400 to 1,500 mL per 24 hours, or at least 0.5 mL/kg/hour. In
healthy people, urine output may vary noticeably from day to day. Urine
volume automatically increases as fluid intake increases. If fluid loss
through perspiration is large, however, urine volume decreases to maintain
fluid balance in the body.
x Insensible Losses: Insensible fluid loss occurs through the skin and
lungs. It is called insensible because it is usually not noticeable and cannot
be measured. Insensible fluid loss through the skin occurs in two ways.
Water is lost through diffusion and through perspiration (which is
noticeable but not measurable). Water losses through diffusion are not
noticeable but normally account for 300 to 400 mL per day.
x Skin: This loss can be significantly increased if the protective layer of
the skin is lost as with burns or large abrasions. Perspiration varies
depending on factors such as environmental temperature and metabolic
activity. Fever and exercise increase metabolic activity and heat
production, thereby increasing fluid losses through the skin. Another
type of insensible loss is the water in exhaled air. In an adult, this is
normally 300 to 400 mL per day. When respiratory rate accelerates, for
example, due to exercise or an elevated body temperature, this loss can
increase.
x Faeces: The chyme that passes from the small intestine into the large
intestine contains water and electrolytes. The volume of chyme entering
the large intestine in an adult is normally about 1,500 mL per day. Of this
amount, all but about 100 mL is reabsorbed in the proximal half of the
large intestine. Certain fluid losses are required to maintain normal body
function. These are known as obligatory losses. Approximately 500 mL
of fluid must be excreted through the kidneys of an adult each day to
eliminate metabolic waste products from the body. Water lost through
respirations, through the skin, and in faeces also are obligatory losses,
necessary for temperature regulation and elimination of waste products.
The total of all these losses is approximately 1,300 mL per day.
Hypervolemia
Hypervolemia is an abnormal increase in the body’s blood volume, particularly in
the sense of blood plasma. To put in other words, hypervolemia occurs when the
volume of fluid in the blood is too high.
Self-Instructional
Material 351
Fluid and Electrolyte Symptoms
Balance
Signs and symptoms of hypervolemia may be different in each case. Some possible
symptoms are as follows:
NOTES x Acites (fluid build-up in the abdomen).
x Crackles on auscultation.
x Oedema (swelling) - particularly hands, feet, and ankles.
x Difficulty in breathing while lying down.
x High blood pressure.
x Irritated cough.
x Jugular vein distension.
x Shortness of breath (Dyspnoea).
x Strong, rapid pulse.
Causes
There are different causes of hypervolemia. Some of the causes are as follows:
x Chronic liver disease.
x Blood transfusion reaction.
x Congestive heart failure.
x Cushing’s syndrome.
x Glomerulonephritis.
x Acute.
x Focal or embolic.
x Membranoproliferati.
x Post-infectious.
x Post-streptococcal.
x Heart problems.
x Hyperaldosteronism–Kidney Failure.
x Liver failure.
x Lung problems.
x Nephritis (Kidney Inflammation).
x Familial interstitial.
x Hereditary.
x Lupus.
x Secondary.
x Nephropathy.
Self-Instructional
352 Material
x Nephrotic syndrome. Fluid and Electrolyte
Balance
x Preeclampsia.
x Pregnancy.
x Surgery/operation complications. NOTES
Diagnosis and Treatment: There are different ways to diagnose hypervolemia.
Diagnosis may be done by observing medical signs and symptoms, or medical
history of the patient or by conducting some tests and exams such as abdominal
ultrasound, albumin, blood chemistry, cholesterol, ElectroCardioGram (ECG or
EKG), Glomerular Filtration Rate (GFR), liver enzymes, and urine analysis.
The method for treating hypervolemia may be based on the primary cause.
It may be essential to treat the cause also. Some of these treatments include sodium
restriction, fluid restriction, and taking diuretics.
Hypovolemia
In hypovolemia, there is a considerable decrease in the blood volume as a
consequence of loss of blood or body fluid. In other words, the volume of fluid in
the blood significantly decreases in case of hypovolemia.
Symptoms
The onset of hypovolemia is gradual or it can occur all of a sudden. The general
initial symptoms of hypovolemia are as follows:
x Dry mucous membranes such as the mouth and nose.
x Loss of skin elasticity.
x Reduced urine excretion.
To compensate for this loss, the body increases the heart rate and the
strength of heart contractions, and constricts blood vessels present in the periphery
while ensuring the blood flow to the brain, heart and kidneys. If the loss continues,
the body is unable to compensate the loss, resulting in drop in blood pressure. At
this stage, the heart cannot supply sufficient amount of blood to the vital organs,
which may result in tissue damage.
When a fifth of the blood volume is lost, hypovolemic shock occurs. It is a
medical emergency that needs immediate attention. Its common symptoms are
cold, sweating, nervousness, increased breathing and heart rate, weakness, reduced
or no urine excretion, confusion and unconsciousness.
In the later stages of hypovolemia, more serious symptoms are observed.
In some circumstances, hypovolemia can be very serious, that requires immediate
medical help. These symptoms are as follows:
x Bleeding during pregnancy.
x Developing bluish colour on lips and fingernails.
x Change in level of consciousness or alertness, such as passing out or
unresponsiveness.
Self-Instructional
Material 353
Fluid and Electrolyte x Chest pain, chest tightness, chest pressure, palpitations.
Balance
x Large burns with blisters.
x Not producing any urine, or an infant who does not produce the usual
NOTES amount of wet diapers.
x Rapid breathing (tachypnoea).
x Rapid heart rate (tachycardia).
x Trauma such as bone deformity, burns, eye injuries and other injuries.
x Uncontrolled or heavy bleeding, haemorrhage.
x Uncontrolled vomiting.
x Vomiting blood, major rectal bleeding or bloody stool.
x Weak pulse.
Causes
In hypovolemia, the loss of blood may be due to external injuries, internal bleeding
or certain obstetric emergencies. Although the common reasons for loss of body
fluid are continuous or severe diarrhoea and vomiting, large burns, and excessive
perspiration also may lead to depletion of fluid. Use of diuretics depletes body
fluid by increasing urine excretion. Insufficient intake of fluid may also be one of
the causes of hypovolemia.
Diagnosis and Treatment
Severity of hypovolemia defines the line of treatment. In severe conditions,
intravenous fluids and possibly blood transfusions may need to be carried out to
raise the blood volume rapidly. Treatments for normalizing blood pressure, heart
rate and strength of heart contraction are also necessary. If there is any underlying
cause of hypovolemia, then it must also be treated to stop the ongoing fluid loss.
In some cases, the hypovolemia, if it is due to vomiting and diarrhoea, can
be prevented by increasing fluid intake to compensate the loss of body fluid.
However, in other cases, the loss of body fluid is so severe that it requires urgent
medical attention.

Check Your Progress


1. What is the function of electrolytes?
2. What is the average water content of human body?
3. How is the concentration of ions maintained in the cell and plasma?
4. Define the term active transport.
5. What is bicarbonate?
6. How is intracellular fluid formed?
Self-Instructional
354 Material
Fluid and Electrolyte
14.4 ANSWERS TO CHECK YOUR PROGRESS Balance

QUESTIONS

1. Electrolytes maintain the electric voltage throughout your cells so that signals NOTES
can pass easily.
2. The average water content of the human body varies from 40 to 75% of
total body weight, with values declining with age and especially with obesity.
Women have lower average water content than men as a result of a higher
fat content.
3. The concentrations of ions within cells and in plasma are maintained both
by energy-consuming active transport processes and by diffusion or passive
transport processes.
4. Active transport is a mechanism that requires energy to move ions across
cellular membranes. For example, maintaining a high intracellular
concentration of K+ and a high extracellular (plasma) concentration of Na+
requires use of energy from ATP in ATPase-dependent ion pumps.
5. Bicarbonate is the second most abundant anion in the ECF. Total CO2
comprises the bicarbonate ion (HCO3-), carbonic acid (H2CO3) and
dissolved CO2, with HCO3- accounting for more than 90% of the total CO2
at physiologic pH. Because HCO3- composes the largest fraction of total
CO2, total CO2 measurement is indicative of HCO3- measurement.
6. The intracellular fluid is formed by equal concentrations of cations, i.e.,
positively charged ions (Ca2+, Mg2+, Na+), anions, i.e., negatively charged
ions (Cl-, HCO3-) and proteins and is found inside the cells.

14.5 SUMMARY

x A balance of fluids, electrolytes, acids, and bases in the body is necessary


for health and life.
x The body fluid is divided into two major compartments. The IntraCellular
Fluid (ICF) inside the cells and ExtraCellular Fluid (ECF) outside the cells.
x Extracellular fluid is further subdivided into two compartments - intravascular
(plasma) and interstitial.
x ECF constitutes about one fourth to one-third of total body fluid. ECF is in
constant motion throughout the body. It is the transport system that carries
nutrients to and waste products from the cells.
x The percentage of total body fluids varies according to the individual’s age,
body fat, and sex. The younger the person, the higher the proportion of
water in the body. The less body fat present, the greater the proportion of
Self-Instructional
Material 355
Fluid and Electrolyte body fluid. Post-adolescent females have a smaller percentage of fluid in
Balance
relation to total body weight than do men.
x There are two types of body electrolytes (ions): positively charged ions
(cations) and negatively charged ions (anions).
NOTES
x The principal ions of ECF are sodium and chloride, and the principal ions
of ICF are potassium and phosphate.
x Fluids and electrolytes move among the body compartments by osmosis,
diffusion, filtration, and active transport.
x The major fluid pressures exerted as part of the movement of fluid and
electrolytes from one compartment to another are osmotic pressure and
hydrostatic pressure.
x The three sources of body fluid are fluids taken orally, food ingested, and
the oxidation of food.
x Fluid output occurs chiefly through excretion of urine, although body fluid is
also lost through sweat, faeces, and insensible vapour loss.
x In healthy adults, measurable fluid intake and output should balance (about
1,500 mL per day). The output of urine normally approximates the oral
intake of fluids. Water from food and oxidation is balanced by fluid loss
through the skin, respiratory process, and faeces.
x A number of body systems and organs are involved in regulating the volume
and composition of body fluids including the kidneys, the endocrine system,
the cardiovascular system, the lungs, and the gastrointestinal system.
x The kidneys are the primary regulator of fluid and electrolyte balance.
Substances such as the antidiuretic hormone, the rennin-angiotensin-
aldosterone system, and the atrial natriuretic factor are also involved in
maintaining fluid balance.
x The most common electrolyte imbalances are deficits or excesses in sodium,
potassium, and calcium.

14.6 KEY WORDS

x Equilibrium: A state in which opposing forces or influences are balanced.


x Extracellular: Situated or taking place outside a cell.
x Intracellular: Situated or taking place inside a cell.
x Hydrostatic: Relating to or denoting the equilibrium of liquids and the
pressure exerted by liquid at rest.
x Electrolyte: Ion capable of carrying an electric charge.
x Diffusion: Passive movement of ions across a membrane.
x Nephron: Functional unit of kidney found inside the renal medulla.
Self-Instructional
356 Material
x Angiotensin: Hormone released in response to decreased blood flow to Fluid and Electrolyte
Balance
the kidneys.
x Hypokalemia: Deficiency of potassium in the bloodstream.
x Myocardial: Pertaining to the muscular tissue of the heart. NOTES
x Obligatory losses: Fluid losses which are essential for maintaining normal
body function.
x Hypovolemia: Considerable decrease in the body’s blood volume due to
loss of blood or body fluid.
x Hypervolemia: Abnormal increase in the body’s blood volume.
x Auscultation: Auscultation is the term for listening to the internal sounds of
the body, usually using a stethoscope.

14.7 SELF ASSESSMENT QUESTIONS AND


EXERCISES

Short Answer Questions


1. What are electrolytes?
2. Write a short note on electrolyte balance.
3. What is fluid balance?
4. Distinguish between hypervolemia and hypovolemia.
Long Answer Questions
1. Describe the method of sodium determination.
2. How is the level of potassium and calcium determined inside the human
body? Explain.
3. Explain the mechanism of fluid balance inside the body.
4. Elaborate a note on regulation of body fluids.
5. Describe the phenomenon of hypervolemia and hypovolemia.

14.8 FURTHER READING

Goyal, Shashi and Pooja Gupta. 2012. Food, Nutrition and Health. New Delhi:
S. Chand And Company Limited.
Garbutt, John. 1997. Essentials of Food Microbiology. London: Arnold –
International Students Edition.
Jay, J. M. 2000. Modern Food Microbiology, 6th Edition. New York: Chapman
& Hall.
Self-Instructional
Material 357
Fluid and Electrolyte Prescott, L. M., J. P. Harley and D. A. Klein. 2014. Microbiology, 9th Edition.
Balance
New York: McGraw Hill.
Ray, Bibek and Arun Bhunia. 2013. Fundamental Food Microbiology, 5th
NOTES Edition. . New York: CRC Press.
Blackstock, James C. 2014. Guide to Biochemistry. Oxford: Butterworth-
Heinemann.
Fromm, Herbert J. and Mark Hargrove. 2012. Essentials of Biochemistry. Berlin:
Springer.
Fearon, William Robert. 2014. An Introduction to Biochemistry. Amsterdam:
Elsevier.
Jain, J. L. 2008. Fundamentals of Biochemistry, 5th Edition. New Delhi: S.
Chand & Company Ltd.
Park, K. H. 2008. Carbohydrate-Active Enzymes: Structure, Function and
Applications. Amsterdam: Elsevier.

Self-Instructional
358 Material

You might also like