Essential Biochemistry - S PDF
Essential Biochemistry - S PDF
ESSENTIAL
BIOCHEMISTRY
FOR MEDICAL STUDENTS
WITH PROBLEM-SOLVING EXERCISES
-GEOTAR-Media-
PUBLISHIHQ GROUP
tWfAMMe iiriwMM Mpuduicjimoe
ptpj'ijfiiaK: iMciicro tififtjouAEt rifpiMi Moe14veto
2> fkjwcmmtri whiumcvm fwiwiur mm <M Grama
MKTEpCTU lOJLMM-LHMfWI PKUKUI ft#;MUM
SKUf-.V »VlM\hKMlY fframeut WmpHivJ
TEXTBOOK
ESSENTIAL
BIOCHEMISTRY
FOR MEDICAL STUDENTS
WITH PROBLEM-SOLVING EXERCISES
Moscow
►GHOTAR-Mcdla-
PUbushing Group
2020
VUK 577.1(075.8) OI-VME-3473
EEK 28.707.2h73-1
E80
Editors:
4/exander JvanavicA G7ukhav — Doctor of Biological Sciences. Professor, Head of the
Department of the Biological Chemistry, I.M. Scchenov First Moscow State Medical
University (Scchenov University);
4/erandro E'vgcn/cvvja Gnfeareva — Candidate of Medical Sciences, Docent in the
Department of the Biological Chemistry, I.M. Scchenov First Moscow State Medical
University (Scchenov University).
YAK 577.1(075.8)
BBK28.70Z2fl7.3-L
4// RjgAfs Reserved. A'o part q/rAis book, may fee reproduced, disfrifeiafed or copied in any/orm or
fey any means w/Aowf /Ac written permixs/on a//Ae original capvrigAf Ao/dcr. GE0T4R-.Media
Pirfe/isAfng Growp.
I JIT — diiodoth.yroni.ncs
DNA pol — DNA polymerase
DNP — 2,4-di nitrophenol
DOPA — di hydroxyphenylalanine
DSB — DNA double-stranded break
DT — diphteria toxin
cEF — elongation factor
ETC — electron transport chain
FAD — flavin adenine di nucleotide
FH4 — tetra hydrofolatc
FMN — flavin mononucleotide
5-FU — 5-fluorouracil
fcjalNAc — N-acetylgalactosamine
GDH - L-glutamate dehydrogenase
GF — growth factor
GH — growth hormone
Gi — inhibitory GTP/GDP-binding protein
GLUT — glucose transporter
GOT - glutamate oxaloacclaLc transaminase
GPT — glutamate-pyruvate transaminase
Grt>2-protein — specific adaptor protein
Gs — activating GTP/G DP-binding protein
GSH — glutathione
GSSG — oxidized glutathione
H4BP(BH4) — tetrahydrob iopic rin
HAT — histone acetyl transferase
Hb — hemoglobin
HI JAC — histone deacetylase
HOL — high density lipoproteins
HIV - human immunodeficiency virus
HLGL — hepatic triacylglycerol lipase
HMG Co A — hydroxy-mcthylglutaryl Co A
H PETE - hydropcroxycicosatctracnioc acid
HSL - hormone sensitive lipase
HSV — herpes simplex virus
II) DM — insulin-dependent diabetes mcllitus
IDE — intermediate density lipoproteins
IMP — inosine monophosphate
IP3 — inositol 1,4,5-trisphosphate
IRS — insulin receptor substrate
a-KG — o-Kctogl uta rate
kB — kilobase
kIJA — kilodaltons
Km — Michaelis constant
LCA1 — lecithin cholesterol acyl transferase
LOH — lactate dehydrogenase
LDL — tow density lipoproteins
List of abbreviations 7
Protein functions
► Structural hi net ion. Proteins arc directly involved in the construction of the
cell membranes and cytoskeleton (integral, sc mi-integral and surface proteins).
The substance of connective tissue and the extracellular matrix form proteins
collagen, elastin, keratin, proteoglycans.
► Enzymatic function All enzymes arc proteins. Enzymes catalyze
the transformations of various molecules in. the cells of the body. Enzymes
constitute more than 50% of all proteins.
► Receptor function. This function isthc selective bindingofhorniones, biologically
active substances and mediators on the surface of membranes or inside the cells.
► Transport function. Only proteins transport some substances in the blood. For
example, lipoproteins (lipid transfer), hemoglobin (oxygen transport), transferrin
(iron transport). Proteins transport cations of calcium, magnesium, sodium and
other ions into the blood.
10 Cnaptef 1. Tne structure, properties and functions of proteins
Side Chain
In aqueous solutions at neutral pH, amino acids exist as bipolar ions. All amino
acids (with the exception of glycine) contain an asymmetric carbon atom, therefore
they can exist as L- and II-stereoisomers (Fig. I.2). For the synthesis of human
proteins only L-a mi no acids arc used. In some protcinswithalong lifetime, L-isomers
can be converted to D-isomers.
All 20 amino acids in the human body differ in structure, size and physicochemical
properties of radicals (side chains). Amino acid radicals arc variable parts of a
polypeptide backbone and may contain various functional groups.
1.2. Structures, Properties and Classification of Amino Acids 11
L-alanine D-alanine
► Polar (hydrophilic):
* Hydroxyl group-OH:
* Carboxyl group—COOH;
* Amino group -NH,_
* Imino group =NH2;
* Amide group -CO-NH,;
* Thiol group —SH.
► Non-polar (hydrophobic):
* Methyl group -CHa;
* Aromatic group.
To designate amino acids in proteins, their trivial names arc usually used. In
addition, for the convenience of recording the amino acid sequence of peptides and
proteins, their three-letter and one-letter designations arc used (Table LI).
Amino acids are classified according to the physicochemical properties of their
radicals. All amino acids can be divided into 4 groups
Amino acids can be divided into groups according to their ability to dissolve
in water. The solubility of amino acid radicals is determined by the polarity of the
functional groups. Polar groups attract water, non-polar repel it.
Amphotcricily is the main physicochemical property of amino acids. Amphoteric
means that the substance combines the properties of both acids and bases. In an
aqueous solution, amino acids simultaneously behave like acids — proton donors and
as bases — proton acceptors. Amino acids with polar negatively charged radicals have
an additional carboxyl group in the radical. Ai a physiological pH of 7.0, it dissociates
to form COO and H .The radicals of such proteins arc an ions. Ami no acids with polar
positively charged radicals have a second amino group in the radical. At physiological
pH, these groups arc positively charged. Radicals of such proteins arc cations.
Chapter 1. Tnb structure, properties and functions of proteins
Table 1.1. C lassification of ami no acids on the c hemic al structure of their radicals
NONPOLAR +CHARGE
H „
’i’ 0 H,N‘-C-C*
H.M4-C-C*
RjN'-f-C*0 RsH4-i-C*° H.M'-C-C* ru °
h/
A
hi
ch^cHj SHj uC , 0 h
CH,
1
CH,
Glycine Alanine Valine Cy&Teine Praline I
NH
(flaw (VaLV| (CyS/C) [PrctfP)
C-NWj'
MH,' NH,
Lysine Arginine
t
HshC-OC*
L HjN'-C-C*
H3N+-C-C* h^w-c-c; CH, °
H,N+-C -C’
0 H^-CH ** MjN'-C-C*0
I ’
CH
*V
CH,
1
s
"S
dH, 0
6 1
CHjCHj
Leucine
SHj
Iso leucine
£H,
POLAR -CHARGE
H^JT-C-C*0
1 H.N*-C-C’
H.N4-C-C* HJN--C-C*0
HjN^-C-C* i H, 0
H^’-C-C*0 I J
^Hj,
1
CH,
1
CH,
I I
CH,
OH CH CHj
V
OH
NH^b A
HH,0 O
A
0
A
o o
Serine nweonine TyraEine Asparagine GjhjEamine Aspartic Acid Glutamic Acid
fSerffi) 0^ lAsfL-Ni (GWQ| (Asp®) (GUE)
R R R
O II □ I
II I II
4
f
f
II
Hi H
\l
]HH
I ’eptide bonds
Pqptide
backbone
N — terminal C — terminal
a mino acid amino acid
The peptide bond formed by the imino group of the proline di Ilers from other
peptide bonds, since the nitrogen atom of the peptide group is associated not with
hydrogen, but with a radical (Fig. I.5).
CH
I / \
H HPC CHj
h3c^ "'oh X /
ch2
The pc pride bond is a strong covalent bond, It has a partial double bond character.
The peptide bond's length is [css than a single bond, it is a rigid (planar) struct Lire,
and rotation around it is difficult. But since, in addition to the peptide, there arc other
bonds in the protein, the chain of amino acids is able to rotate around the main axis,
which gives proteins a different conformation (the spatial arrangement of atoms). All
atoms in the peptide group arc in the same plane, while the atoms H and 0 arc located
on opposite sides of the peptide bond (Fig. 1.6 A). The oxygen and hydrogen atoms
in the peptide group have the ability to form hydrogen bonds with the oxygen and
hydrogen atoms of other peptide groups (Fig. 1.6 U). Amino acid radicals in relation
to the axis of the peptide C-N bonds arc on opposite sides, in the trans-position
(Fig. 1.6 C).
RO RO
B. I II ll
HO O
I I 1
— N — CH — C—N—CH — C-
I I I
R HR
These properties of the peptide bond determine the ability of amino acids to
interact with each other within one protein, as welt as with, other proteins. Peptide
bonds arc very strong and under physiological conditions they do not spontaneously
break. In laboratory conditions, the peptide bonds arc hydrolyzed in the presence of
concentrated hydrochloric acid at 105° C within a day. In living organisms, peptide
bonds in proteins are destroyed with the help of special proteolytic enzymes —
proteases. To delect proteins and peptides in a solution, the color biuret reaction is
used.
NH2—CH—CH2—CH2—C—NH—CH—C—NH—CH2—COOH
Some ami no acids a nd severs I peptides arc important human hormones. Hormones,
in general, arc biological molecules used in multicellular organisms io direct and
regulate biological processes, such as growth, reproduction and metabolism. A peptide
hormones are chains of amino acids, which serve as a biological communication
molecules. Peptide hormones have a short half-life, meaning they break apart quickly.
This allows organisms to use peptide hormones to direct processes quietly and
efficiently, without the signal lingering fora long time.
The antino acid-derived hormones arc relatively small molecules derived from
the amino acids tyrosine and tryptophan. If a hormone is antino acid-derived, its
chemical name will end in «-inc*. Examples of amino acid-derived hormones include
epinephrine and norepinephrine, which arc synthesized in the medulla of the adrenal
glands, and thyroxine, which is produced by the thyroid gland. The pineal gland in the
brain makes and secretes melatonin, which regulates sleep cycles. The formulas antino
acid-derived hormones arc below:
16 Cnaptef 1. Tn e structure, properties and functions of proteins
Tryptophan Melatonin
Heme
n pctypeptide
ft polypeptide
Primary structure
/Imino acid residues in the peptide chain arc not randomly located, but arranged
in a specific order. The linear sequence of amino acid residues in a protein is called the
primary structure of (he protein (Fig. 1.8).
The primary structure of proteins, i.c. the ami no acid sequence in it is programmed
by the nucleotide sequence in the DMA. The deletion, insertion, replacement of a
nucleotide in DNA leads to a change in the amino acid composition and. therefore,
the structure of the synthesized protein. If a change in the amino acid sequence is
not lethal, but adaptive oral least neutral, then the new protein can be. inherited and
remain in the population. Asa result, new proteins appear with similar functions. This
phenomenon is called, protein polymorphism.
For example, there are about JOO different types of hemoglobin, some of them arc
necessary al different stages of ontogenesis: for example. HbP — embryonic, formed
in the first month of development, HbF — fetal, necessary in the later stages of fetal
development, HbA and HbA2 — adult hemoglobin. The diversity is provided by the
polymorphism of globin chains: there are 2g and 2r chains in hemoglobin P. 2a and
2y chains in HbF. 2a and 20 chains in HbA. and 2a and 2d chains in HbA2. Proteins
ofthe main histocompatibility complex provide tissue transplantation incompatibility.
They have extremely high polymorphism, in general, there arc several million alleles
of those proteins. Due to this diversity, each person has an almost unique set of alleles.
In addition, many genetic diseases result from the amino acid sequence violation,
of proteins. Information about the primary structure of a normal and mutant protein
is needed to diagnose and predict the development of a disease.
Secondary structure
The secondary structure of a protein is a spatial structure resulting from in teracl Ions
between the functional groups that make up the peptide backbone. The secondary structure
1.3. The Levels of Protein Structures: Primary, Secondary, Tertiary 19
is formed only with the participation of hydrogen bonds between peptide groups: the
oxygen atom of one group reacts with, the hydrogen atom of the second, while the
oxygen of the second peptide group is bound to the third hydrogen, etc. (Pig. I.9).
O RO
II ] II
-N — CH — C — N—CH — C — N—
I I
HR H H
R O 0
|
II
-N — CH —C — N— CH — C — N—
| I I
H H R H
■
RO O
I II II
«N —CH—C —N —CH —C —N—
I
H
III
H R ft
Fig. 1.9. Hydrogen bonds between peptide backbone groups form a secondary protein structure
When the secondary structure is formed, the peptide accepts the conformation
with the largest number of bonds between the peptide groups. The type of secondary
structure depends on the stability oft he peptide bond, the mobility of the bond between
the central carbon, atom and the carbon of the peptide group, the size of the amino
acid radical. All of this, together with the amino acid sequence, will subsequently lead
to a strictly defined configuration of the protein. I n this case, peptide chains can form
regular structures of two types: a-helix and the P-shcct.
I none protein, asa rule, both structures a re simultaneously present, but indifferent
proportions. In globular proteins, the a-helix predominates, in fibrillar proteins, the
p-structure prevails.
aHeiix
The most common form of the secondary structure is the a-hclix (the polypeptide
chain seems to be twisted clockwise on an imaginary cylinder, which is due to the
L-amino acid composition of natural proteins). Al each turn (step) of the helix there
arc 3.6 amino acid residues, the helix pilch is 0.54 nni per turn, and one amino acid
residue isO.IS nm(Fig. 1.10).
Not all globular proteins arc helical across the entire length of the polypeptide
chain. In the protein molecule, the a-helical regions alternate with the linear ones.
Practically all atoms of the oxygen and hydrogen peptide groups arc involved in the
formation of hydrogen bonds. Since all the hydrophilic groups of the peptide core arc
occupied, the hydrophilicity a-helix (the ability lo form hydrogen bonds with water)
decreases, and the hydrophobicity increases.
20 Cnaptef 1. Tn e structure, properties and functions of proteins
The a-hclix is a ver)' stable conformation of the peptide backbone. The amino
acid radicals arc on the outside of the a-helix and arc directed away from the peptide
backbone. They do not participate in the formation of hydrogen bonds characteristic
of the a-helix, bin some may disrupt its formation, proline and hydroxyprolinc cause
chain bending, for example, in collagen.
fkSbeet
A p-shect is formed by the formation of hydrogen bonds between the atoms of
the peptide groups of the linear regions of one polypeptide chain. making bends or
between many different polypeptide chains (Fig. I JI). It looks like a folded sheet. In
this way of laying the protein the molecule makes a snake-shaped form, the remote
segments of the chain arc going close to each other. As a result, peptide groups
that previously were remote amino acids of the protein chain arc able to interact by
hydrogen bonds.
The orientation of the reactive sites may be parallel (i.c. the direction of N-terminal
to C-terminal ends is the same) or anti parallel where chains go in the opposite
direction (Fig. 1.12). Such sites of one protein interacting with each other can be from
two to five.
1.3. Trie Levels of Protein Structures: Primary, Secondary, Tertiary 21
The p-shests
Tertiary structure
The tertiary structure of proteins is a three-dimensional spatial structure formed
due to interactions between amino acid radicals, which can be located at a considerable
distance from each, other in the polypeptide chain. Secondary structures of proteins
often constitute distinct domains. A domain is the basic unit ofst rue lure and function.
Tertiary structure describes the relationship ofdilfercni domains to one another within
a protein. Pour types of chemical bonds arc involved in the formation of the tertiary
structure: hydrophobic, ionicT hydrogen and disulfide (Fig. 1.13).
Hydrophobic inreractiotis
{clustering of hydrophobic
groups away from water)
and van der Waals
interactions
Ch2 —------- - Polypeptide
zCH3 backbone
Hydrogen CH
bond
— CHj —S—S“CH2 —
Disulfide bridge
Fig. 1.11 Types at chemical bonds are involved in the formation of the tertiary structure
► Ionic bonds: Ionic bonding can occur between negatively charged (anionic)
carboxyl groups of aspartic and glutamic acid radicals and positively charged
(cationic) groups of lysine, arginine and histidine radicals.
peptide
chain CH3
l z Glu-LyS
CHa -R-C^o- Hahr— R—
CHg nh2
1
CH2 LyS 1 Glu-Arg
ionic pond ■ rn ‘“-Hghr—C — NH—R —
: 1 :
I 1
i 1 n n Asp-Hs
o-
l HNt NH
V
4=O.J
CH2 Asp
— R—C=O*ii™h — O— R—;
s— R ■—■ N — H ■■■■■1111*“ O-— R —1 H — R— GyS-Glu
I I
H H
hydrogen Ponds Tyr-Lys
24 Cnaptef 1. Tn e structure, properties and functions of proteins
► Disulfide bonds: The covalent disulfide bond is formed between two -SH groups
of cysteine radicals located in different places of the polypeptide chain. Disulfide
bonds can. stabilize the spatial structure of a single polypeptide chain or link lwo
chains together, such as in an insulin molecule.
ww NH — CH — CO 'xyx/w
peptide 1
chain CHg
: s ■
disulfide
.oJi
bond
S ■
CHj
I
'wvv NH — CH — CO "ww
All proteins with the same primary structure and under the same conditions
acquire the same conformation, which determines their function. The functionally
active protein conformation is called the native structure. Hydrophobic, ionic and
hydrogen bonds arc weak bonds, therefore their breaking is possible even under
physiological conditions. This fact ensures the conformational lability of proteins,
i.c. they arc capable of small changes due to the breaking of some weak bonds and the
formation of others. Protein conformation can change with changing the chemical
and physical properties of the medium, as well as when interacting with other
molecules. Conformational changes play a huge role in the functioning of proteins
in a living cell
The breaking of a large number of weak bonds leads to the destruction of the native
conformation of the protein. The loss of the native conformation is accompanied by the
loss of the specific function of the protein. This process is called protein denaturation.
When denaturation occurs, an occasional break of weak bond's happen and protein
molecules acquire a random conformation.
Initially the weakest bonds are lorn and when conditions arc tightened, stronger
ones arc also broken. Therefore, al first, the quaternary, then the tertiary and secondary
structures arc lost. Denaturation docs not break the peptide bonds, i.c. the primary
structure of the protein is not disturbed.
Denaturation may be reversible., if restoration of the protein-characteristic structure
is possible (for example, membrane receptors): or irreversible, if the restoration of
the spatial configuration of the protein is impossible. Irrcvcsible denaturation usually
occurs when a large number of bonds arc broken, for example, when eggs arc boiled.
If the protein has undergone reversible denaturation, then when normal conditions
of the environment arc restored, it is able to completely restore its structure and,
accordingly, its properties and functions. The process of restoring the protein structure
after denaturation is called maturation (Fig. 1.14).
1.3. Trie Levels of Protein Structures: Primary, Secondary, Tertiary 25
a «iA|l p
Fig. 115. Examples of protein domains classified by CATH (class, architecture, topology. homology)
26 Cnaptef 1. Tn e structure, properties and functions of proteins
Quite often, domains arc assigned separate names, since their presence directly
affects the biological functions performed by the protein, for example, the Ca2‘
binding domain of calmodulin, a homcodomain responsible for binding to ONA in
various transcription factors. Different domains in the protein can move relative to
each other when interacting with the ligand, which facilitates (he further functioning
of the protein.
The formation of the three-dimensional structure of the protein in the cell
is the most important process, since its biological function depends on the spatial
structure of the protein. The process of packing the polypeptide chain into the correct
spatial structure is called protein folding. The concentration of proteins in a cell is
very high, so an abnormal protein conformation may occur. For many proteins with
high molecular weight and complex spatial structure, folding occurs with the help of
special chaperone the participation proteins. Chaperones isolate the protein from the
environment and allow it to accept the native conformation.
Protein functioning
Each protein with a unique primary structure and conformation has a unique
function. Proteins perform many different functions in a cell. A prerequisite for the
functioning of a protein is the binding of a not her sub stance called a ligand. Ligands can.
be both low molecular weight substances, such as metal ions, small organic molecules
and macromolecular substances, such as other protein molecules. The interaction of
the protein with the ligand is highly specific, which is determined by the structure of
the protein site, called the active she (active center) of the protein.
The active site (active center) of proteins is a specific part of a protein molecule,
usually located in its recess pocket*), formed by the ami.no acid radicals collected
in a certain spatial region during the formation of the tertiary structure and capable of
complementary binding to the ligand (Fig. 1.16).
Fig. 1.16. The active site of me protein and its imeradion with me ligand
In the linear sequence of the polypeptide chain, the radicals forming the active
center can be located al a considerable distance from each other. The high specificity
of protein binding to the ligand is ensured by the complementary structure of the
active center of the protein to the ligand structure.
1.3. Trie Levels of Protein Structures: Primary, Secondary, Tertiary 37
Fig. 117. The interaction of protein with a ligand in me active site. A and B are incomplete interaction.
C is complementary interaction. L-ligaiM
The unique properties of the active center depend not only on the chemical
properties of the amino acids forming it, but also on their exact mutual orientation
in space. Therefore, even minor violations of the general conformation of the protein
as a result of point changes in its primary structure or environmental conditions can
lead to a change in the chemical and functional properties of the radicals that form the
active center, disrupt the binding of the protein to the ligand and its function. During
denaturation, the active center of proteins is destroyed, and their biological activity
is lost. Often the active center is formed in such a way that the access of waler to the
functional groups of its radicals is limited, i.e. conditions arc created for the binding
of the ligand to amino acid radicals. In some eases, the ligand is attached to only one
28 Cnaptef 1. Tn e structure, properties and functions of proteins
of the atoms with a certain reactivity, for example, the addition ofO2 to the iron ion of
myoglobin, or hemoglobin.
The main property of proteins that underlies their functions is the selectivity of
specific ligands to attach to certain sections of the protein molecule.
There arc ligands that can change their chemical structure upon attachment to the
active center of the protein, for example, changes in the substrate in the active center
of the enzyme. There arc ligands that can attach, to the protein only at the moment of
functioning, for example, O., transported by hemoglobin. Ligands can be constantly
associated with the protein, which plays an auxiliary role in the functioning of proteins,
for example, iron which is part of hemoglobin. The connection of protomers in an
oligomeric protein is an example of the interaction of high molecular weight ligands.
Each protomer connected lo other protomers serves as a ligand for them, just as they
do for it. Sometimes the attachment of a ligand changes the conformation of the
protein, resulting in the formation ofa binding center with other ligands. Tor example,
the protein calmodulin, after binding to four Ca2 ions in specific areas, acquires the
ability to interact with some enzymes, changing their activity.
Protein classification
Protein classification is based on chemical composition, structure, functions,
and solubility in different solvents. Based on their chemical composition, proteins
may be divided into two classes: simple and complex. Simple proteins also known
as homop rote ins, they arc made up of only amino acids. For examples, arc plasma
albumin, collagen, and keratin. Complexprotc ins sometimes also called hctcroprotcins,
except amino acids, have non-protcin components. The non-protcin part is called the
prosthetic group, and the protein part — apoprotein. Complex protein — holoprotein —
can dissociate into components:
The direction of the reaction depends on the bond strength of the holoprotcin
components. The prosthetic group can. be organic substances, metal ions, nucleic
acids, carbohydrates, lipids and other substances (Table 1.2).
Table 12. Complex protein examples
Flavoproteins Flavinnucleotides
Proteoglycans Glycosaminoglycans
Nucleoproteins:
ribo nucleoproteins (ribosomes, etc.); RNA
deoxyribonuefeoproteins ; chromatin i DMA
1.3. Trie Levels of Protein Structures: Primary, Secondary, Tertiary 29
Glycoproteins arc 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. Examples of glycoproteins arc: glvcophorin, the best known
among erythrocyte membrane glycoproteins; fibronectin, 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. Phosphoprotcins are proteins
that bind phosphoric acid to serine and threonine residues. Generally, they have a
structural function, such as tooth den Lin, or reserve function, such as milk caseins
( alpha, beta, gamma and delta), and egg yolk phosvitin.
Based on their shape, proteins may be divided into two classes: fibrous and globular.
Fibrous proteins have primarily mechanical and structural functions, providing
support to the cells as well as the whole organism. These proteins arc insoluble in water
as they contain, internally as well as on their surface, many hydrophobic amino acids.
The presence on their surface of hydrophobic amino acids facilitates their packaging
into very complex supra molecular structures. In this regard, it should be noted that
their polypeptide chains form long filaments or sheets, where in most cases only one
type of secondary structure, that repeats itself, is found.
Here arc some examples, collagen which constitutes the main protein component
of connective tissue, and more generally, the extracellular scaffolding of multicellular
organisms, a-Ke rati ns 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. Most of the
proteins belong to the class of globular proteins.
They have a compact and more or less spherical structure, more complex than
fibrous proteins. In this regard, motifs, domains, tertiary and quaternary structures
arc found, in addition to the secondary structures. They arc 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:
* enzymes:
* hormones;
► membrane transporters and receptors:
» transporters of triglycerides, fatly acids and oxygen in the blood:
► immunoglobulins or antibodies;
ligand. It also interacts with the active center of a protein. An analog that replaces
a natural ligand in an active center protein and reduces its function is called a
competitive protein inhibitor
Analogs of natural protein ligands arc used as medicines. Such drugs a re widely used
in the regulation of transmission of excitation through synapses. The neurotransmitter
secreted during the passage of the pulse by the nerve endings should interact highly
with receptor proteins on the postsynaptic membrane. However, by modifying the
chemical structure of the neurotransmitter, it is possible to obtain substances that
would also bind to the receptor, but the physiological effect changed, decreased or
intensified. In pharmacology; such substances arc called *antagonisls*and ^agonists*,
respectively.
Dilhilin (a pharmacological substance) is an analog of acetylcholine, which
ensures the transmission of a nerve impulse to a muscle, being a synapse mediator.
When dilhilin is introduced into the body; it binds to the N-cholinergic receptors
of cholinergic synapses, being an antagonist of acetylcholine, a neurotransmitter
that causes muscle contraction. As a result, the transmission is blocked and muscle
relaxation (paralysis) occurs. Therefore, dilhilin is used as a muscle relaxant in.
operations and endoscopic examinations. Another drug that acts as a protein inhibitor
is atropine, an alkaloid of plant origin, which is also a natural analog of acetylcholine,
but interacts with M-cholinergic receptors of the membrane of effector cells. Being an
acetylcholine antagonist, it eliminates the irritation of parasympathetic nerves, and
instead of smooth muscle contraction, which, is stimulated by acetylcholine, relieves
their spasm.
Some poisons, getting into the human body, firmly bind to certain proteins,
inhibit them and thereby cause disturbances in biological Junctions. For example,
cobra neurotoxins specifically interact with, cholinergic receptors of postsynaptic
membranes, blocking their work. The affinity of neurotoxins for cholinergic receptors
causes to the formation of many bonds between the toxin and the receptor, which
leads to their practically irreversible connection. It must be remembered that there is
often a transparent border between drugs and poisons, and the effect of their action
depends on the dose of the substance administered. Thus, drugs prescribed in doses
greater than therapeutic can act as poisons, i.e. cause serious metabolic disorders
and disruptions of body functions, and poisons in microdoscs arc often used as
medications. For example, atropine, which is widely used to relieve spasms of smooth,
muscles, in high doses causes CNS excitement, and in even higher doses, it causes
a sleep that goes into a coma. Known hypotensive agent clonidinc in the case of an
overdose causes collapse.
1.3. The Levels of Protein Structures: Primary, Secondary, Tertiary
Review tests
I. Choose one correct answer. The peptide contains:
H o H C
l I !
COOH
A. Renailiration.
B. Disulfide bonds.
C. Native protein.
D. Denaturation.
E. Cysteine radicals.
6. Match the figure with the Idler Secondary struct urc of protein:
A. N-terminal end.
B. Parallel p-sheet.
C. C-terminal end.
D. Antiparalfcl p-shecl.
E. Hydrogen bonds between the atoms of the peptide backbone.
7. Match the figure with the letter. Levels of structural organization of proteins:
1. The primary structure.
2. The secondary structure.
3. Tertiary structure:
a) spatial folding of the polypeptide chain:
b) the order of alternation of amino acids:
c) a structure formed by imcrradical interactions:
d) spatial stacking of the peptide backbone:
e) the specific arrangement of secondary structures.
8. Match the figure with the letter. Protein-ligand interaction:
A. ProLein.
B. A ligand.
C. Peptide bonds.
D. Active center.
E. Weak connections.
1.4. Quaternary Structure of Proteins. Hemoglobin 33
Situational Problems
1. The use of natural proteins as drugs requires compliance with certain storage
and use conditions, which arc clearly prescribed in. the instructions. So, most
protein preparations must be stored in a refrigerator al a temperature not excee
ding 10 ’C3 and dry prepara Lions should be dissolved in boiled water cooled to
room temperalLire. Some protein preparations are stored in hermetically scaled
ampoules, from which air is removed. Why do protein drugs require strict
adherence to all the conditions prescribed in the instructions, and what can
happen if these conditions arc violated?
2. Atropine is an alkaloid found in some plants: belladonna, dope, and bleach. Il is
pan. of the drugs belonging to the group of antispasmodics that relieve smooth
muscle spasms. Such, drugs are used for spastic pain and stomach, intestines, to
relax the smooth, muscles of the bile ducts. What is the mechanism of action of
this substance?
Myoglobin
Myoglobin is classified as he me-containing proteins, i.e. it contains a prosthetic
group - heme, quite firmly connected with the protein part. Myoglobin is classified
34 Cnaptef 1. Tn e structure, properties and functions of proteins
as a globular protein; it has only one polypeptide chain. Myoglobin is found in red
muscles and is involved in oxygen storage. In conditions of intense muscular work,
when the partial pressure of oxygen in the tissue drops, O2 is released from the complex
with myoglobin and is used in the mitochondria of cells to obtain the energy necessary
for muscle work.
Heme is a molecule with the structure of cyclic tctrapyrrolc, where 4 pyrrole
rings are connected by methylene bridges. This organic part of the heme is called
protoporphyrin.
In heme. 4 nitrogen atoms of the pyrrole rings of protoporphyrin IX arc linked by
four coordination bonds with Fe2‘ located in the center of the molecule {Fig. 1.18).
Fig. 11tL Myoglobin structure. Heme is a complex of porphyrin and ferrous iron (IV-
His E7, although not associated with he me, is necessary for the proper orientation
and attachment of another ligand O2 to myoglobin. The amino acid environment of
the heme creates the conditions fora rather strong, but reversible binding of O, to Fc2'
myoglobin. The hydrophobic amino acid residues surrounding heme prevent water
from entering the myoglobin, binding site and the oxidation of Fc3, to Fc3\. Trivalent
iron in the heme is not able to attach oxygen.
Fia 119- The location of the heme in me active center ol apomioglehin and he mo globin protomers
Hemoglobin
Hemoglobins are related proteins found in. human and vertebrate erythrocytes.
They are also classified as heme-containing proteins. These proteins perform
2 important functions:
► transfer of oxygen from the lungs to the peripheral tissues;
► participation in the transfer of CO., and protons from peripheral tissues to the
lungs for subsequent excretion from the body.
The most important characteristic of hemoglobin is its ability to regulate the
affinity for O3depending on tissue conditions. Hemoglobins arc similar in structure
to myoglobin, but they have a quaternary structure (they consist of 4 polypeptide
chains), which makes it possible to regulate their functions. In adult red blood cells,
hemoglobin accounts for 90% of all proteins in a given cell.
Adult Hemoglobins:
► hemoglobin A (HbA) the main adult hemoglobin, accounts for about 98% of the
total hemoglobin, tetramer, consists of 2 polypeptide chains a and 2p (2a2p):
* hemoglobin A2 is in the adult body in a lower concentration, it accounts for about
2% of total hemoglobin. It consists of 2a and 20 chains (2a2O);
* hemoglobin Ale is hemoglobin A modified by the covalent addition of glucose to
it (the so-called glycosylated hemoglobin).
Hemoglobins synthesized during fetal development:
► embryonic hemoglobin (HbE) is synthesized in the fetal yolk sac a few weeks
after fertilization. h is a tetramer 2g2e. After 2 weeks of formation of the fetal
36 Chapter 1. Tnb structure, properties and functions of proteins
or alkaline side. the bonds between the dimers arc first destroyed. In addition, dimers
arc able to easily move relative to each other. Since the surface of the protomers is
uneven, the polypeptide chains in the central region cannot adhere closely to each
other; as a result, a ^central cavity* is formed in the center, which passes through
the entire hemoglobin molecule. The weaker interactions between dimers result in
the two dimers occupying different relative positions in deoxyhemoglobin (T form) as
compared with oxyhemoglobin (R form) (Fig. 1.21).
Heme
Heme
fl - polypeptide ci - polypeptide
(globin) chain (globin) chain
r
4O2
protein pan, protrudes from the heme plane in the direction of His F8. The addition
of O2 to the Fc2- ion of one protomer causes it to move to the heme plane; the His
F8 residue and the polypeptide chain of which it enters also move behind it. Since
proteins have conformational lability, the conformation of the whole protein changes.
Conformational changes that occurred in other protomers facilitate the attachment of
the next O2 molecule, which, causes new conformational changes in the protein and
accelerates the binding of the next ()., molecule. The fourth ()., molecule attaches to
hemoglobin 300 times easier than the first molecule (Fig. 1.22).
\ /H \
. . . C— N . . . C—N
Histidine a Histidine
tZ \A
Fe
Heme is domed
(nonplanar)
Deoxygenated Oxygenated
Increasing
affinity
Fig. 122. Cooperative changes in the conformation of hemogl oh in protomers upon addition of 0,
1.4. Quaternary Structure of Proteins. Hemoglobin 39
pO2 in pO£ in
Fig. 1-23.Oxygen tfissociatiou curves for myoglobin and hemoglobin depending on the partial pressure
of oxygen
Myoglobin binds oxygen, which releases hemoglobin in the tissue capillaries, and
myoglobin itself can release OJn response to increased demand for muscle tissue and
intensive use of 07 during exorcise. Myoglobin has a very high affinity forO.,. Even al
a partial pressure ofO,, equal to 1.-2 mm Hg myoglobin remains bound to O by 50%.
Hemoglobin has a significantly lower affinity for O:,; half-saturation of hemoglobin
occurs at a higher pressure of O3 (about 26 mm Hg). The hemoglobin dissociation
curve has a sigmoid shape (S-shaped). This indicates that the hemoglobin protomers
work cooperatively: the more is released by the protomers, the easier is the cleavage
of subsequent O2 molecules.
40 Cnaptef 1. Tn e structure, properties and functions of proteins
Due to the unique structure, each of the considered proteins is adapted to perform
its function: myoglobin — attach O2 released by hemoglobin, accumulate it in. the coll
and give it up, if necessary. Hemoglobin is an additive in the lungs to O2, where its
saturation reaches 100%, and release O, in die capillaries of tissues, depending on. the
change in O2 pressure, in them.
20-■
0 20 40 60 SO 100
Po2(mmHg)
In the lung capillaries. the high partial pressure of O] leads to oxygenation of the
hemoglobin and removal of 6 protons. The reaction CO2+ + HCO2
is shifted to the left and the formed CO2 is released into the alveolar space and is
removed with exhaled air (Pig. 1.25). During evolution, the hemoglobin molecule
acquired the ability to perceive and respond to environmental changes. An increase in
the concentration of protons in the medium decreases the affinity ofO3 for hemoglobin
and enhances its transport into tissues.
Most CO2 is transported by blood in the form of HCO, bicarbonate. A small
amount of CO (about 15-20%) can be transferred to the lungs, reversibly attaching
to non-ionized terminal ot-amino groups. As a result, carboxyhemoglobin is formed.
The addition of CO2 to hemoglobin also reduces its affinity for Or
>.—J V '* ‘J
Hz'dz |‘ ci ■
Oxyhemoglobin | Deojcyhemoqlobin I
«.,4
$A
sutjunrt
Vj
oo
p2 subunh *
Lys92
His 2
His 143
In the central cavity; there arc positively charged radicals of amino acid residues
and positively charged a-amino groups of the N-terminal partofp-chains. 2,3-BPG,
which has a strong negative charge, is attached to the expanded deoxyhemoglobin
1.4. Quaternary Structure of Proteins. Hemoglobin 43
cavity by means of ionic bonds formed with positively charged functional groups. The
addition of 2,3-B PG even more stabilizes the taut form structure of deoxyhe moglobin
and reduces its affinity for O2. The addition of2,3-BPG to dcoxyhcmoglobin occurs in
a different site, compared with the active site where O2 binding occurs. Such a ligand
is called allosteric and the center where the allosteric ligand is bonded is the allosteric
center («allos* — is another, different, micros* — is spatial). In the lungs, a high partial
pressure of O, leads to hemoglobin oxygenation. The rupture of ionic bonds between
dimers al pl and a2p2 leads to relaxation of the protein molecule, a decrease in the
central cavity and the displacement of 2,3-BPG.
The concentration of 2,3-BPG in the erythrocytes of people living in certain
climatic conditions is a constant value. However, in the period of adaptation to high
mountains, when a person rises to an altitude of more than 4000 m above sea level,
the concentration of 2,3-BPG in almost 2 days increases almost twice (from 4.5 to
7.0 mM). This reduces the affinity of hemoglobin for O3 and increases the amount of
O., transported to the tissue (Fig. 1.27) Changing the concentration of 2,3-BPG works
as a mechanism for adapting the body to hypoxia. The same adaptation is observed in
patients with lung diseases, in which general tissue hypoxia develops.
Y-chains. Some of the positively charged amino acid radicals arc absent in the primary
struct tire of the y chains. In a medium lacking 2,3-11 PG, HbA and HbF exhibit the
same high affinity forO3.
Hereditary hemoglobinopathy
The importance of the primary structure of proteins for the formation of their
conformation and function can be Lraccd to the examples of hereditary diseases
associated with a change in the primary structure of hemoglobin. At present, about
.300 HbA variants arc known that have only small changes in the primary structure of
the a or p chai ns. Some of them ba rely affect protein function and human health, others
reduce protein function, and especially in extreme situations, reduce the possibility
of human adaptation, others cause significant impairment of HbA. functions and the
development of anemia, which leads to serious clinical consequences.
In 1904, a Chicago doctor, J. Herrick, described severe anemia in a student with
the discovery of many elongated, sickle-1 ike red blood cells in his blood. The disease
was called sickle cell anemia, and only in 1949 did L. Pauling and his colleagues prove
that it was caused by a change in the primary structure of HbA. in the hemoglobin
S (HbS) molecule (the so-called abnormal hemoglobin), 2 P-chains arc mutant, in
which glutamate, the highly polar negatively charged amino acid at 6,k position, was
replaced by a valine containing a hydrophobic radical (Fig. 1.28).
delivery io tissues causes pain and even cell necrosis in. this area. Sickle cell anemia is a
homozygous recessive disease; it only manifests itself in the case when 2 mutant genes
p-chains of globin arc followed from both parents (Fig. 1.29).
it ii it it Sickle Cell
il #1 ♦t li
Unaffected [Sickle Cell Trait] Sickle Cell
Fig. 129 inheritance of sickle cell anemia and manifestation of the disease
After the birth of a baby, the disease docs not appear until significant amounts
of HbF arc replaced: by HbS. Patients reveal clinical symptoms characteristic for
anemia: dizziness and headaches, shortness of breath, palpitations, pain in the limbs.
46 Chapter 1. Tnb structure, properties and functions of proteins
4.S-5.4. which indicates the predominance of glutamine and aspartic amino acids
in their composition. If the protein is dominated by basic amino acids (lysine and
arginine), then al neutral pH the protein charge is positive and it is due io these
positively charged amino acids.
Amphocericity is important for proteins to perform certain functions. For example,
the buffer properties of proteins? i.e the ability to maintain unchanged blood pH .
based on the ability io attach H ions during acidification of the environment or give
them when alkalizing. On the practical side, the presence of amphotericity makes it
possible to separate proteins by charge (electrophoresis) or use a change in the pH
of the solution to precipitate any known protein. The presence of both positive and
negative charges in a protein determines their ability to salting out, which is convenient
for isolating proteins in a native (living) conformation.
As the pH in the solution changes, the concentration of H ions changes loo.
When the medium is acidified (with a decrease in pH) below the isoelectric point,
H ions attach to the negatively charged groups of glutamic and aspartic acids and
neutralize them. The protein charge in this case becomes positive. With increasing pH
in the solution above the isoelectric point, the concentration of H ions decreases and
positively charged protein groups (N H, groups of lysine and arginine ► lose protons,
their charge disappears. The total charge of the protein becomes negative 4 Fig. 1.30).
+H* +c+r
NH.— Ch— COOH * - nhZ— Ch — COO" * ■ Ch — COO"
nonpolar and I
polar radicals H H Fl
| net charge = +l 1 | ne* charge = 0 | net charge =-■!
+H* 1-CH"
NHg‘— CH — COOH - - nhZ—Ch — COO" * NFk—CH — COO-
1 |i |
- charge CH.
radicals I
COOH COCr COO"
[ net charge = +l netchajge=-l i | net charge =-’)
t+T tOH"
NH3 Ch COOh * NH3 CH COO" * nh2— Ch—■COG-
The pH value al which the protein acquires a total zero charge is called the
isoelectric point and is denoted as pl. At the isoelectric point, the number of positively
and negative ly charged protein groups is the sameT i.e. the protein is in an isoelectric
state. Since most of the proteins in the cell contain more anionic groups (-COO-),
48 Cnaptef 1. Tn e structure, properties and functions of proteins
the isoelectric point of these proteins lies in a slightly acidic medium. The isoelectric
point of the proteins, in which cat io nogen ic groups predominate, is in an alkaline
environment. The most striking example of such intracellular proteins containing a lot
of arginine and lysine arc histones, which arc part of chromatin.
Proteins having a total positive or negative charge arc better soluble than proteins
located al an isoelectric point. The total charge increases the number of water dipoles
that can bind to a protein molecule, and prevents the contact of the same charged
molecules, as a result, the solubility of proteins increases. Charged proteins can move
in an electric field: anionic proteins having a negative charge will move to a positively
charged anode (+), and cationic proteins to a negati vely charged cathode (-). Proteins
in an isoelectric state do not move in anelectric field. At the isoelectric point, proteins
are the least stable in a solution and easily precipitate. The isoelectric point of the
protein is highly dependent on the presence of salt ions in Lhc solution; al the same
time, its value is not affected by protein concentration.
With a shift in the acid-base balance of the body towards an increase in acidity
(decrease in pH), acidosis occurs. For example, with diabetes, there is an increase in
the production of ketone bodies (ketoacidosis). This is a dangerous condition for the
body, proteins lose their charge and their solubility decreases, while they can aggregate.
Protein solubility
Most proteins carry many charged groups on the surface; therefore, they arc
soluble in waLcr. Solubility is due to the presence of a charge, as well as the repulsion of
charged protein molecules. Besides,, the presence ofa hydration shell, i.c. environment
of a protein molecule with water dipoles and their interaction with polar and charged
groups on the surface ofa protein globule.
exchanger is determined by the charge of the protein released. So, to isolate a negatively
charged protein, an anion exchanger is used. Wien a protein solution is passed through
a column, the binding strength of the protein to the anion exchanger depends on the
number of negatively charged carboxyl groups in the molecule. Proteins adsorbed on
the anion exchanger can be washed oil (eluted} with buffer solutions with different salt
concentrations, most often NaCI, and different pH values. Chlorine ions bind lo the
positively charged functional groups of the anion exchanger and displace the carboxyl
groups of the proteins. At low salt concentrations, proteins weakly bound to the anion
exchanger elute. A gradual increase in salt concentration ora change in pH, which
changes the charge of a protein molecule, leads to the release of protein fractions, one
of which contains the desired protein (Fig. 1.31).
Electrophoresis
The method is based on the property that, al a certain pH and ionic strength of a
solution, proteins move in an electric field at speed proportional to their total charge.
Proteins with a total negative charge move to the anode (+), and positively charged
proteins move to the cathode (-). Electrophoresis is carried out on various media:
paper, starch gel, polyacrylamide gel, etc. Unlike paper electrophoresis, where the
speed of proteins is proportional only to their total charge, in polyacrylamide gel the
r
speed of movement of proteins is proportional Lo their molecular weights (Fig, 1.32).
c
J2
n Well ■
E Langer
■C
u fragments
e
Cl
s-
£
u
fragments
1■
1
Fig. 132. The principle of separation of proteins using electrophoresis gel. Gel stained by Coomassie
brilliant blue reagent
1.5. Physical Chemical Properties of Proteins 51
Affinity chromatography
This is the most specific method for isolating individual proteins, based on the
selective interaction of proteins with ligands attached (immobilized) to a solid carrier.
As a ligand, a substrate or coenzyme can be used if any enzyme, antigens for the
isolation of antibodies, etc. arc isolated. A solution coma ini ng a protein mixture
is passed through a column filled with an immobilized ligand. Only a protein that
specifically interacts with it is attached to the ligand; all other proteins come out with
the eluate. The protein adsorbed on the column can be removed by washing it with
52 Cnaptef 1. Tn e structure, properties and functions of proteins
A
elution
A A A
» A .La
pH 2,0
A-L
Secondary proteinopathy
Any disease is accompanied by a change in the protein composition of the body; i.c.
secondary proteinopathy develops. At the same Lime, the primary structure of proteins
is not disturbed, and usually there is a quantitative change in proteins, especially in
those organs and tissues in which the pathological process develops. For example,
pancreatitis reduces the production of enzymes necessary for the digestion of nutrients
in the gastrointestinal tract. I n some cases, acquired proteinopathy develops as a result
of changes in the conditions in which proteins function. So. when the pH of the
medium changes to the alkaline side < alkalosis of different natures, the conformation
of hemoglobin changes, its affinity for O2 increases, and the delivery of O? to tissues
(tissue hypoxia) decreases.
Sometimes as a result of the disease, the level of metabolites in the cells and blood
serum increases, which leads to the modification of certain proteins and the disruption
of their function. Thus, elevated blood glucose concentrations in diabetes mcllitus
lead to non-enzymatic attachment of glucose to proteins (glycosylation of proteins!.
An example is an increase in the level of glycated hemoglobin in red blood colls, which
increases its affinity for O, and reduces the transport of O, into tissues. Glycosylation
of proteins of the lens of the eye leads to its clouding and the development of cataracts.
In some cases, biochemical data on changes in the protein composition of the
blood or urine can be leading in the diagnosis. For example, in myeloma (malignant
degeneration of plasma cells that synthesize immunoglobulins}, Bcrts-Jones proteins
appear in the blood and urine, they’ arc present in low- concentrations in the blood of
healthy people. These proteins arc the light chains of immunoglobulin G. the synthesis
of which is enhanced in malignantly reborn ceils.
Conformational disorders
Some water-soluble proteins, when the conditions change, can acquire the
conformation of poorly soluble molecules capable of aggregation, forming fibrillar
deposits in cells called amyloid (from lai. amylum — starch). Like starch, amyloid
deposits arc delected by iodine staining of the tissue. This may occur
► wit h t h e overproduct ion of ccrtai n proici ns. as a resul t ofwrhich their concent rat ion
in ihc cell increases:
► when proteins enter the cells or form proteins in them that ean affect the
conformation of other protein molecules:
► upon activation of proteolysis of normal body proteins, with the formation of
insoluble fragments prone to aggregation:
► as a result of point mutations in the protein structure.
54 Cnaptef 1. Tn e structure, properties and functions of proteins
As a result of the deposition of amyloid in the organs and tissues, the structure
and function of cells arc disrupted, their degenerative changesand the proliferation of
connective tissue- cells arc observed. A disease called amyloidosis develops. Each type
of amyloidosis is characterized by a specific type of amyloid. Currently, more than
15 such diseases are described.
Alzheimer's disease is the most frequently observed p-amyloidosis of the nervous
system, usually affecting elderly people and characterized by progressive memory
disorder and complete personality degradation. In the brain tissue, 0-amyioid is
deposited — a protein that forms insoluble fibrils that disrupt the structure and
function of nerve cells. Protein p-amyloid is a product of a change in the conformation
of a normal protein of the human body, h is formed from a larger precursor by partial
proteolysis and is synthesized in many tissues. Protein p-amyloid, unlike its normal
precursor, which contains many a-heli cal regions, has a secondary P- fold structure
that can aggregate with the formation of insoluble fibrils, and is resistant to proteolytic
enzymes.
Parkinson’s disease was first described in 1817 by the English physician
James Parkinson (he called it «trembling paralysis*) and is one of the most
common neuro degenerative disorders. Most often, parkinsonism and other
ncu rod cgenc rad vc disorders (such as Alzheimer s disease) arc found in the elderly
and along with oncological diseases occupy a leading position among the causes of
death. Bui parkinsonism is not only a disease of the elderly: with the improvement
of diagnostic methods, there is more and more evidence that the disease affects
people under 40 years of age. As follows from the original name of the disease, its
characteristic symptoms arc motor disorders: trembling (tremors) of fingers, lower
jaw and tongue, head and eyelids, slowness and impoverishment of the pattern of
movements, stillness of the body, difficulty in starting and stopping movement,
impaired coordination, etc. Such disorders are caused by the flexibility of nerve
cells, primarily the loss of pigment-containing neuro n s of the which
produce the dopamine neuro transmit ter. Dopamine is the biochemical precursor
of norepinephrine and adrenaline. In many patients who died from Parkinson’s
disease, protein clusters are found during an autopsy in the ju&j/an/Kr (they
arc called Levy bodies by the name of the German pathologist who discovered them
in 1912). Parkinson’s disease develops as a result of disturbances in the functioning
of the chaperone and ubiquitin-protcasomc systems. Apparently, the situation is
as follows. Some damage in the neurons of the suistoitaj w/gra triggers a cascade
of reactions leading to the appearance of a large nu mber of improperly packaged
proteins.
1.6. Diseases Associated with Structure and Function Proteins Disorders S5
Review tests
1. Choose one correct answer. I IBS in contrast to I IbA:
A. Has a 6-posilion a-chain Vai.
B. Contains 2a and 2.y chains.
C. Deoxyform is poorly soluble in water.
D. Has a high affinity for (\.
E. In the active center there is an amino acid substitution.
2. Choose one correct answer. Isoelectric point of proteins:
A. The amount o! protein charge.
B. The ratio of polar and nonpolar amino acid radicals.
C. The pH value at which the protein has a charge equal to zero.
D. The pH value at which the protein is best soluble in water.
E. The charge of the protein at which it is most actively moving in the electric field.
3. Match the figure with the letter. Hemoglobin molecule:
A. a chain Hb.
B. p chain Hb.
C. .Active center.
IX Central cavity.
E. Domain.
■4. Match the figure with the letter. Protein
separation methods:
1. Ci cl filtration.
2. Electrophoresis.
3. .Affinity chromatography.
The principles of separation by difference:
A. Charges.
B. Solubility in water.
C. Molecular weight.
D. Sedimentation rate in solution.
E. .Affinity for a specific ligand.
5. Choose all correct answers. The affinity of I I B to oxygen decreases with:
A. An increase in rhe concentration of protons.
B. A decrease in the concentration of protons.
C. .A decrease in the concentration of 2,3-BPH.
D. An increase in the concentration of 2.3-B PH.
E. Sequential cleavage of oxygen molecules.
6. Match the figure with the letter. Functional parts of the oligomeric protein:
Active site
1
56 Cnaptef 1. Tn e structure, properties and functions of proteins
A. Subunit.
B. An allosteric center.
C. Active center.
D. An oligomer.
E. An effector.
Situational problems
1. In high altitude conditions, climbers usually led the clinical signs of hypoxia:
headache, shortness of breath, nausea, increased heart rate. However, after
2 days of rest in the base camp, the symptoms disappear. How docs the body
adapt to high altitude conditions and increase oxygen delivery to tissues?
2. The fetal HbF has a higher affinity for oxygen than the mother HbA. What
structural features of these proteins determine their difference in the ability to
bind oxygen and what role docs 2,3-bisphosphoglyccrate play in this? What is the
physiological meaning of t he different affinities of these Hb forms for oxygen?
3. The patient went to the clinic with complaints of dizziness, shortness of breath,
palpitations and pain in the limbs, which sharply worsened after a short rest in
the mountains. A reduced number of red blood cells were found in the patient's
blood, as well as immature crescent-shaped cells and red blood cells. What arc
the causes of this pathology? Why did the disease worsen in conditions of low-
partial pressure of oxygen?
Chapter 2
ENZYMES
Figu 2.1 Graphs ol me reaction energy in me absence anti presence of the enzyme
Similarity
1. Catalyze only energetically possible reactions;
2. Do not change the direction of the reaction:
3. Accelerate the onset of reaction equilibrium, but do not shift it;
4. Not consumed during the reaction.
Differences
1. The velocity of the enzymatic reaction is much higher:
2. High, specificity;
3. Mild working conditions (intracellular);
4. The ability to control the reaction rate;
5. The velocity of the enzymatic reaction is proportional to the amount of enzyme.
The catalytic power of enzymes (the ratio of the reaction rate in the presence of
a catalyst to the reaction rate without a catalyst) is in the range from 106 to 1014, so
they can provide rapid processes, such as heartbeats or conduction of nerve impulses.
Enzymatic catalysis
Enzymes reduce the activation energy through a process called catalysis. A
biochemical reaction when an enzyme is present is called a catalyzed reaction.
Catalysis can happen in di fie rent ways. Enzymes can use the transfer of protons or
electrons to the reactants to modify the state of the reactants. Enzymes also use electric
charges to stabilize the state of the reactants. Enzymes, however, do not modify the
final products of the reaction.
2.1. Enzymes as Catalysts. Active Site cf the Enzyme 59
The following steps arc distinguished in the enzymatic reaction (Fig. 2.2-23):
12 3 4
E + S -<------ ► E-S <------ ► E-X ■*------ ► E-P * * E +P
The cofactor, in turn, can be called a coenzyme (NAD-F, NADP+, FMN, FAD,
biotin) ora prosthetic group (heme, oligosaccharides, ions of metals Fc2\ Mg1*, Ca2',
Zn2'). If Lire connection between the cofactor and the protein is strong, then, in this
case, it is said that there is a prosthetic group, but if a vitamin derivative is used as a
cofactor, it is called a coenzyme, regardless of bond strength. For the implementation
of catalysis, a full-fledged complex of the apoprotein and a cofactor is necessary;
separately, they cannot carry out catalysis. A cofactor is pari of the active center, is
involved in the binding of the substrate or in its transformation. Examples of complex
enzymes are succinate dehydrogenase (contains FAD), aminotransferases (contain
pyridoxal phosphate), peroxidase (contains heme), lactate dehydrogenase (contains
Zn2*), amylase (contains Ca2). Like many proteins, enzymes can be monomers, i.c..
consist of one subunit and polymers consisting of several subunits.
As part of the cnzvmc active site, isolated areas that perform a different function.
(Fig. 2.6).
Active site (active center) — a combination of amino acid residues (usually 12—16)
providing direct binding lo the substrate molecule and performing catalysis. Amino
acid radicals in the active center can be in any combination, with the amino acids
located far from each other in a linear chain located nearby. In the active center, there
arc two sites — binding site and catalytic site. Binding site (contact, anchor) — is
responsible for binding and orientation of the substrate in the active center. Catalytic
site — is directly responsible for the implementation of the reaction.
Enzymes containing several monomers may have several active centers in the
number of subunits. Also, two or more subunitscan form one active center. I n complex
enzymes, functional groups of the cofactor are necessarily located in the active center
(Fig. 2.7).
2.1. Enzymes as Catalysts. Active Site of the Enzyme 61
Protein structure
Stafford to support and
position active site
Active site
Binding sites Catalytic site
Fig. 2 J. The structure of the active site of We complex enzyme. A coenzyme or cotactor is involved
in its formation
Allosteric center (alios —alien) is the center of the activity of the enzyme regulation,
which is spatially separated from the active center and is not available for all enzymes.
Ilinding to the allosteric center of any molecule (called an activator or inhibitor, as
well as an. effector, modulator, regulator) causes a change in the protein-enzyme
configuration and, as a consequence, the rate of the enzymatic reaction {Fig. 2.8).
62 Chapter 2. Enzymes
Rg. 2.8. Allosteric site of the enzyme allows to regulate the activity of the enzyme
Allosteric enzymes arc oligomeric proteins, active and regulatory centers are
located in diflerent subunits. Such a regulator may be the product of this or one of the
subsequent reactions, the substrate of the reaction, orothersubstan.ee.
Enzyme specificity
One of the important characteristics of enzymes is their high specificity, it lies
in the fact that each, enzyme catalyzes the transformation of a particular substrate
or a group of substrates that arc similar in structure. The specificity of the action of
enzymes determines the directional metabolism in the body.
Specificity, i.c., the high selectivity of the action of enzymes is based on the
complementarity, it’s mean structural and chemical correspondence of the substrate
structure and the active center of the enzyme (Fig. 2.9).
Absolute specificity — the enzyme produces catalysis of only one substrate. For
example, urease cleaves only urea and docs not act on other compounds. Il acts on
urea, but not at thiourea, mcthylurca. biuret.
Group or relative specificity - catalysis of substrates with common structural
features, i.c., in the presence of a specific bond or a chemical group. For example,
pancreatic lipase hydrolyzing triacylglyccrols (TAG) with different fatty acid
composition to 2-monoacylglycerols (2-MAG) and fatty acids breaks down ester
bonds in various food fats.
O
Urease
h2n c — nh2 + h2o CO2 + 2NH3
urea
o S O O
II
II I 11
HSN- C- NHCHj h2n -c- ■ nh2 h2n — C —NH— C — NFfe
Methylurea Thiourea Biuret
h2o --O
HC—O— R‘
Lipase upase
O
|l
H2C—-O—C—R" H2C — O— H2C—OH
Triacygiyceroi Diacygfyceroi Monoacylglycerol
Pepsin
R and R1 = Leu. Phe, Trp, and Tyr (preferred); also hydrolyzes esters
64 Chapter 2. Enzymes
H NH./ H
I Aspartase
C— C---- CO< C + NH/
I I
H H CO2“
L Aspartate Fumarate
Catalytic specificity or specificity of transformations — the specificity of the
transformation paths lies in the fact that a single substrate under the action of
di fie rent enzymes can turn into products that differ in the structure and their role
in metabolism. For example, the transformation of malate by two different enzymes:
malate dehydrogenase and malic enzyme.
Two models of the enzymes specificity. There arc two main models explaining the
specificity of enzymes. In 1890 the chemist Emil Fischer proposed that the substrate
of an enzyme fits into the enzyme’s active site, the physical location on an enzyme
where the reaction takes place, to form an enzyme-substrate complex. The analogy he
used was of a lock and key. The key (substrate) has a specific shape (arrangement of
functional groups and other atoms) that allows it and no other key to fit into the lock
(the enzyme). A lock-and-kcy model explains absolute specificity (Fig. 2.10).
Enzyme kinetics
Under optimal conditions, the enzyme activity depends on:
* the amount of enzyme (E);
> substrate amounts (S):
> the amount of product (P);
* cofactor concentrations:
> presence of activators or inhibitors.
Temperature and pH also affect the rate of enzymatic reactions.
Fig. 2.12. The dependence of the enzymatic reaction rate on the concentration of me enzyme
66 Chapter 2. Enzymes
o
Substrate concentration's
Fig. 2J3. The dependence of the enzymatic reaction rate on die concentration of me substrate
The general theory of enzymatic kinetics and the dependence of enzyme activity
on the substrate described by L. Michaelis and M.L. Menton, gives it in their equation.
The Michaelis-Men ten equation shows the relationship between the maximum possible
speed, the real reaction rate, the Michaelis constant, and the substrate concentration.
Km (the Michaelis constant) is visible in. the graph as the concentration of a substrate
al which the initial velocity ishalfoft.be maximum velocity (Fig. 2.14).
Fig: 2.14 Delernri nation of Km and Vjiai according to the graph of dependence of tne enzymatic
reaction rate on the concentration of the substrate
2.1. Enzymes as Catalysts. Active Site cf the Enzyme &7
where: Vfl = the velocity al any time: [S]l = the substrate concent ration at this
limejV^ = the highest under this set of experimental conditions (pH, temperature,
etc.); Km = the Michaelis constant for the particular enzyme being investigated.
Maximal Velocity (V^j: Increasing the substrate concentration indefinitely docs
not increase the rate of an enzymc-cataiyzed reaction beyond a certain point. This
point is reached when there arc enough substrate molecules to completely fill (saturate)
the enzyme’s active sites. The maximal velocity, or V^, is the rate of the reaction
under these: conditions. Vnia* reflects how fast the enzyme can catalyze the reaction.
Michaelis Constant (Km): Enzymes have varying abilities to bind their substrates
(affinities). An enzyme’s Km describes the substrate concentration al which half the
enzyme’s active sites arc occupied by substrate. A high Km means that a lot orsubsiratc
must be present to saturate the enzyme, meaning the enzyme has a low affinity for the
substrate. On the other hand, a low Km means that only a small amount of substrate is
needed to saturate the enzyme, indicating a high affinity for substrate.
These constants arc important to understand enzyme activity in general, as well as
to understand the effects of different types of enzyme inhibitors.
This feature of enzymes is essential for the organism in its adaptation to changing
external and internal conditions. Shifts in pH outside and inside the cell plays a role in
the pathogenesis of diseases, changing the activity of enzymes of different metabolic
pathways. For each enzyme, there is a certain narrow range of pH. which is optimal for
the manifestation of its highcractivity. For example, the optimum pH values for pepsin
are 1.5-2.5, trypsin 8.0-8.5, amylase of saliva 7.2, arginase 9.7, acid phosphatase 4.5-
5.0, succinate dehydrogenase 9.0 (Fig. 2.I7, Table 2.1).
Table 2.1. Optimum pH values tor various enzymes with different location and substrates
Enzyme activity
The Commission on Enzymes of the International Biochemical Union gives an
idea of the standard unit of activity. The unit of activity (U) is the amount of enzyme
that catalyzes the conversion of one micromole of substrate per minute under standard
conditions (at optimum pH, with an excess of substrate, at a temperature of 37 or
20 °C).
Enzyme activity = moles of substrate convened per unit time = rate x 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 kata!,
I katal = I mol s but this is an excessively large unit. A more practical and commonly
used value is enzyme unit (U) = 1 pmol min ]. I U corresponds to 16.67 nanokatals.
An increased amount of substrate will increase the rale 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.
Specific activity
The specific activity ol’an enzyme is another common unit. This is the activity of an
enzyme per milligram of total protein (expressed in pmoi min 'mg '). Specific activity
gives a measurement of enzyme purity in the mixture. It is the micromoles of 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 pmol mg 1 min '. Specific activity is a measure of
enzyme process! vity, at a specific (usually saturating) substrate concentration, and is
usually constant for a pure enzyme. The specific activity should then be expressed as
70 Chapter 2. Enzymes
limol min 1 mg 1 of active enzyme. If the molecular weight of the enzyme is known,
the turnover number, or pmol product per second per pmol 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.
Classical units:
Unit of enzyme activity:
pmol substrate iransformed/'min = unit
Specific activity:
pmol subst rate/min-mg E = unil/mg E
Review tests
1. Match the figure with the letter;
On the image, which letter represents the enzyme a), the
substrate b), the product of the reaction c)?
A.
B
C
2. The part of the enzyme where the substrate binds is called
the;
A. Active site.
B. Catalyst.
C. Inhibitor.
D. Large subunit.
3. Enzymes have a distinct advantage over non-biological catalysts because they:
A. Arc made of protein.
B. Arc very efficient, specific, and sensitive to control
C. Arc all allosteric.
D. Can't be denatured by heating.
4. Choose all the correct answers. In the process of enzymal k catalysis occurs: E + S >
ES > EX > EP > E + P:
A. Establishing rhe induced correspondence between the substrate and the active
center of the enzyme.
B. The formation of covalent bonds between the substrate and the active center.
C. A change in the conformation of the enzyme.
D. The formation of an enzyme-subst rate complex.
E. Destabilization of bonds in the substrate molecule.
5. Find a match. Specificity:
1. Absolute.
2. Relative.
3. Catalytic.
Enzyme:
A. Catalyzes the transformation of the substrate in one of the ways of transformation.
B. Interacts with only one substrate.
C. Catalyzes several different transformations of the same substrate.
2.1. Enzymes as Catalysts. Active Site of tne Enzyme 71
Reaction
Situational Problems
1. The optimal conditions for salivary lysozyme (hydrolyzing glycoproteins of bacterial
wail) are 37 C — temperature and pH is 5.2. Explain the decrease in this enzyme
activity if the temperature will rise up to 60 T and pH will be changed to 8.0. To
answer the question:
a) draw the graph of the velocity dependency on temperature and pH:
b) calculate the relative enzyme activity if 10 mg of lysozyme catalyzes the
formation of 5 pM of the product per 2 minutes.
2. Consider lhe enzy matic reaction scheme: Asparagine + H20 > Aspartate + NH3:
a) calculate the specific activity of the enzyme, if in 30 seconds as a result of
a reaction involving 3 mg of the enzyme under optimal conditions (pH 8.0,
37 *C) 75 pmol aspartate is obtained;
b) describe the masons lor the decrease: in enzyme activity after incubation for
10 minutes at 70 °C (provide an appropriate graph).
72 Chapter 2. Enzymes
3. The figure below shows the reaction velocity catalyzed by an enzyme as a function
of the enzyme substrate concentration. Such a relationship is typically referred
to as a dose-response relationship. Based on the data shown, what is the Km
(Michaelis constant) of this enzyme for its substrate? Note that the reaction
velocity is normalized to the maximum velocity (VmHj. Therefore, in the plot
shown, VmaK = 100%:
a) 2.5 nM;
b) 5.0 nM:
c) 10.0 nM:
d) 20.0 nM.
Vitamins classification
Vitamins were originally classified according io their solubility in water or fats, and
later as more were discovered, they wore also classified alphabetically. The fat-soluble
vitamins arc A, D, E,and K: the B complex and C vitamins arc water soluble. A group
of substances that decrease blood capillary fragility, called the vitamin P group, are no
longer considered to be vitamins.
1. Pat-soluble vitamins: D (calciferol), E (tocopherol), F (polyunsaturated fatty
acids), K (naphthoquinone), A (retinol). The function of fat-soluble vitamins
can be coenzyme (vitamin K), antioxidant ( vitamins A and E), or hormonal
(vitamins A and D).
2. Water soluble vitamins: B, (thiamine), B3 (riboBavin), B3 (nicotinamide),
B. (pantothenic acid), B (pyridoxine), B, (BC, folic acid), B|2 (cobalamins),
H (B7, biotin), C (vitamin C).
3. Vitamin-like substances:
* fat soluble — Q (ubiquinone):
* water soluble — B4 (choline), P (bioflavonoids), Bg. (inositol), B|o (para-
anti no benzoic acid), Bn (BT, carnitine).
Vitamin Bi (Thiamine)
Vitamin B, or thiamine, enables the body to use carbohydrates as energy. It is pan
of thiamine di phosphate (TOP), which is the coenzyme of transketolase, the enzyme
of [he pentose phosphate pathway. Thiamin serves as a cofactor fora series of enzymes
in different metabolic pathways and is required for the production of ATP (Ch. 5),
ribose, NAD, and DNA. Il is essential for glucose metabolism, and it plays a key role
in nerve, muscle, and heart function. Vitamin B, is a water-soluble vitamin, as arc all
vitamins of the B complex.
2.2. Vitamins. Cofactors ano Coenzymes 75
nh2
Vitamin (Riboflavin)
Vitamin Bp or riboflavin, is one of eight I? vitamins that are essential for human
health. It can be found in grains, plants, and dairy products. Riboflavin (Vitamin Bj
is the precursor of two coenzymes known as flavin mononucleotide (FMN) and
flavin adenine di nucleotide (FAD). Both arc essential for tissue respiration and the
generation of energy front the metabolism of carbohydrates, amino acids and fats.
Riboflavin is vital for normal reproduction., growth, repair and development of
body tissues, including the skin, hair, nails, connective tissue and immune system.
Riboflavin is mainly convened into FMN and FAD in the small intestine, liver, heart,
and kidneys.
FAD + 2H*>2e"-FADH2
Vitamin B3 (Niacin)
Vitamin Br also known as niacin, plays a key role in skin, digestive, and mental
health, and supports the functions of more than 200 enzymes in the body. Vitamin B3
is a combination of two chemicals: nicotinic acid and nicotinamide. The body breaks
these chemicals down to produce two additional chemicals: NAD and NADP.
NAD and NADP arc coenzymes of most dehydrogenases, participate in the
reactions:
► synthesis and oxidation of carboxylic acids:
► cholesterol synthesis:
► exchange of glutamic acid and other amino acids;
► carbohydrate metabolism: pentose phosphate pathway, glycolysis;
► oxidative decarboxylation of pyruvic acid;
► Krebs cycle, etc.
NAD and NADP play a role in a variety of chemical reactions inside the body
and also support cell metabolism. So, people who don’t get enough vitamin B3 can
experience a range of health problems and symptoms, ranging from minor to I ire-
threatening. Vitamin B3 deficiency can disrupt dozens of processes in the body and can
lead to a disease called pellagra.
2.2. Vitamins. Cofactors ano Coenzymes 77
■
■
i
■
cooh!
ch3 h
■ i
I
Coenzyme A
78 Chapter 2. Enzymes
Vitamin H or B? (Biotin)
Biotin is a water-soluble vitamin, also called vitamin B? and formerly known as
vitamin II or coenzyme R. It is involved in a wide range of metabolic processes,
both in humans and in other organisms, primarily related to the utilization of fats,
carbohydrates, and amino acids. Biotin is important in fatty acid synthesis, branched-
chain amino acid catabolism, and gluconeogenesis. Biotin is involved in the transfer
of OO2 cither from HCO3 (carboxylation reaction) or from R-COOH (trans
carboxylation reaction). Such a reaction is necessary for the synthesis ofoxaloacctale —
biotin is a part of pyruvate carboxylase complex, which ensures the maintenance of the
activity of the tricarboxylic acid (TCA) cycle and gluconeogenesis. In the synthesis of
fatty acids — biotin is in the composition of acetyl-CoA — carboxylase a key enzyme
of th is process.
(CH2)4—COOH
Class 1 — Oxidoreductases
The enzymes of this class catalyze the redox reactions underlying biological
oxidation. The class has 22 subclasses. Coenzymes of this class arc NAD, NAD PH,
PAD, FMN, ubiquinone, glutathione, lipoicacid. Exam pics of subclasses arc enzymes
that act on the CH-OH group of donors, on the CH-CH group of donors, on the
CH-NH3 group of donors, on heme-containing donors.
The most common working names for oxidoreductases arc:
► dehydrogenases — oxidoreductases catalyzing the dehydrogenation of the
substrate using any molecules other than oxygen as the acceptor of hydrogen.
For example, the enzyme lactate dehydrogenase, which catalyzes the conversion
of lactate into pyruvate.
| L-Lactate ] | Pyruvate J
COO" COO-
Succinate 1
CH-. dehydrogenase
CH
| 2 + FADH + fadh,
CHj CH
COO- COO-
! Succinate j [ Fumarate ]
Xanthine oxidase
O
Z. IE
Carbon
monoxide
82 Chapter 2. Enzymes
H2O2 + 2 H2O
Class 2 — Transferases
Transferases catalyze the transfer of various groups Prom one substrate (donor)
to another (acceptor), participate in the intcrconvcrsion reactions of various
substances, neutralization of natural and foreign compounds. Coenzymes arc
pyridoxal phosphate, coenzyme A. tetra hydro folic acid, methylcobalamin. The
class is divided into 9 subclasses depending on the structure of the transferred
groups. Examples of subclasses arc enzymes that carry one-carbon fragments,
aldehyde or keto-residues, acyl residues, nitrogen-containing groups, phosphorus-
containing groups.
An example of the transferases action is the reaction of transamination in the
synthesis of amino acids.
COO" COO"
Alanine
:oo" COO"
ansaminase
H3N+— C----- H c=o :=o + HJM*------C----- H
1 I
Often the working name of transferase — kinase is used. These arc transferases
catalyzing the transfer of phosphate Prom ATP to a substrate (monosaccharides,
proteins, etc.), i.c.T phosphotransferase. For example, the fust step of glucose
activation in glycolysis is the conversion of D-glucose into glucose-6-phosphate.
2.3. Enzymes Classification and Nomenclature
Kinases arc the most important regulatory enzymes of the metabolic pathways.
Class 3 — Hydrolases
Hydrolases — enzymes that break intramolecular bonds in a substrate by attaching
elements of H,O, arc divided into 13 subclasses. Due to the complexity of many
substrates, a. number of enzymes retain trivial names, for example, pepsin, trypsin.
Coenzymes arc absent. Hydrolases arc widely represented by the gastrointestinal
tract enzymes (pepsin, trypsin, lipase, amylase, and others) and lysosomal enzymes.
They carry out the decay of macromolecules, forming easily adsorbed monomers.
Examples of subclasses arc groups of enzymes acting on esters, on ethers, on peptides,
on carbon-carbon bonds. Historically, the names of hydrolases evolved from the name
of the substrate with the end of the <-asc* — collagenase, amylase, lipase. DNA-asc.
The following working hydrolase names are most common:
► esterase - hydrolyzis of ester bonds. Cholesterol esterase catalyzes the hydro I yzis
of sterol esters into their component sterols and fatty acids;
84 Chapter 2. Enzymes
rafty acid
O^Y^O
rRj
AR,°y“
r2
[Phosphatidate] 'Diacylglyccrol (DAG).
Diacylglycerol kinase
OH
ho—P=O
O H(OH)
HO!— P=O
oI
o
86 Chapter 2. Enzymes
Class 4 — Lyases
Lyases arc enzymes catalyzing the cleavage of C-O, C-C, C-N, and other bonds,
as well as reversible cleavage reactions of various groups in a non-hydrolytic manner.
There are 7 subclasses. These reactions arc accompanied by the formation of a double
bond or the addition of groups to the double bond site. Lyases are complex enzymes.
Coenzymes arc pyridoxal phosphate, thiamine diphosphate, magnesium and cobalt
arc involved.
Enzymes arc divided into subclasses depending on the nature of the attacked
connection. Examples include enzymes acting on carbon-carbon bonds, carbon
oxygen bonds, carbon-nitrogen bonds. For example, glutamate decarboxylase
converts glutamate to gamma-aminobutyric acid, the most important inhibitory
neurotransmitter of the central nervous system.
COO" NH3+
H3N+----- CH CH2
H+ CO2
ch2 A A. ch2
I
ch2
Glutamate
decarboxylase ch2
COO” COO”
[GABA]
Class 5 — Isomerases
Isomerases are enzymes that catalyze isomeric transformations within a single
molecule. Isomerases arc complex enzymes. Their coenzymes include pyridoxal
phosphate. deoxyad cnosyl cobala mi nT g I utat h ionc. monosaccharide phosphates
(glucose-1, 6-diphosphatc), etc. Isomerase subclasses arc distinguished depending
on the type of reaction. For example, racemases (reversible transformation of L-
and D-stcrcoisomers, epimerase (transformation of isomers with more than one
asymmetry center, for example, a-D-glucose into p-D-glucose), mutases (transfer
of chemical groups inside molecules). For example, phosphoglucomutasc converts
glucose-1.-phosphate to glucose-6-phosphate:
COO” COO”
Alanine
racemase
H3N+------ C------ H H------ C------ NH3+
I
ch3
I
ch3
[ L-Alanine | [p-Alanine]
2.3. Enzymes Classification and Nomenclature 37
Class 6 — Ligases
Ligases (synthetases) arc enzymes that catalyze the attachment of two molecules
to each other using the energy of high-energy bonds of ATP (or other macroergs}.
Ligases arc complex enzymes. They contain nucleotide (UTP), biotin {vitamin HL
folic coenzymes. There arc 6 subclasses.
An example of subclasses is the group of enzymes by the type of the formed bond :
carbon-oxygen (C-O), carbon-sulfur (C-S), carbon-nitrogen (C-N), carbon
carbon (C-C):
CQO
I
CH,
Z\ Z\
I
o o o nh5
[L-Glulamate ] [ L-Glutamine ]
For example, DNA ligase is a specific type of enzyme that facilitates the joining of
DNA strands together by catalyzing the formation of a phosphodicstcrbond. h plays a
role in repairing single-strand breaks in duplex DNA in living organisms:
3'
DMA strand
UNA strand
I DMA strand
S3 Chapter 2. Enzymes
Review tests
1. Choose alt I he correct answers. Enzyme catalyzing Lhis reaction:
COO COO
HO—C—'H C=O
+ NAD +--------- ► I + NADH + H*
CH2 ch2
I
coo coo
f L-Malatej (jOxalgaceLate J
2 2
Coenzyme NAD" composition:
A. Adenyl nucleotide.
B. Nicotinamide coenzyme.
C. Adenine.
D. Ribose.
E. Nicotinamide.
Coenzyme:
1. NAD \
2. FAD
3. Coenzyme A.
Vitamin:
A. Pantothenic acid.
B. B,
C. Niacin.
D. Biotin.
E. B,.
7. During replication, a number of DNA fragments are produced. Energy is required for an
enzyme to join these fragments together to form an intact strand of DNA. The enzyme
is classified as an/a:
A. Transferase.
B. Hydrolase.
C. Lyase.
D. Isomerase.
E. Ligase.
90 Chapter 2. Enzymes
8. What is the class of the enzyme catalyzing the reaction of glucose activation?
A. Oxidorcductascs.
B. Transferases.
C. Isomerases.
EX Hydrolases.
E. Lyases.
Situational Problems
1. With the prevalence in food of peeled cereals or bread made from high-grade flour,
hypovitaminosis B l may occur. Explain the rote that vitamin Bs plays in the body.
For this:
a) name the coenzyme which contains vitamin BL and enzymes, which require
this coenzyme to function:
b) write the process in which these enzymes arc involved and explain how the
process speed will change with a lack of B :
d) what disease develops in the absence of vitamin B,.
2. The patient has signs of pellagra, symmetrical dermatitis on the rear surface of the
hand, neck, face, stomatitis. The patient complains of nausea, abdominal pain,
diarrhea, lack of appetite, headaches, dizziness, depression.
a) what vitamin deficiencies cause these symptoms?
b) what coenzyme synthesis is reduced in this situation?
stops or starts (he reaction. An important parameter for the occurrence of enzymatic
reactions is also the presence ofcoenzymes necessary during the reaction. For example,
for the TCA cycle, this substrate isoxaloacctatc. The presence of oxaloacclate triggers
cycle reactions, which makes it possible to involve acctyl-CoA molecules in the
oxidation.
Genetic regulation
Genetic regulation (change in the amount of enzyme) can occur as a result of an
increase or decrease in its synthesis. From this point of view, enzymes can be divided
into several groups:
Constitutive — such enzymes that arc constantly formed in. the cell, regardless of
the presence of the substrate. These arc enzymes necessary to maintain cell activity,
for example, glycolysis enzymes, p-oxidation of fatly acids, DNA synthesis and repair.
Induced (adaptive) — the synthesis of these enzymes increases with the presence of
appropriate stimuli or inductors. For example, during pregnancy and after childbirth,
synthesis of the enzyme lactose synthase is induced under the influence of lactotropic
hormone, glucocorticoid hormones stimulate the synthesis of gluconeogenesis
enzymes, which ensures the stability of blood glucose concentration during prolonged
fasting and the central nervous system resistance to stress.
Repressed — the formation of such enzymes in. the cell, if necessary, is suppressed.
For example, in the liver, repression of the enzyme HMG-CoA reductase under the
influence of cholesterol. It affects the repression of gluconeogenesis enzyme synthesis
by insulin.
The change in the rate of enzyme synthesis (induction or repression) usually
depends on the amount of certain hormones or metabolites of the process.
92 Chapter 2. Enzymes
Com partmentalization
The compartmentalization is the accumulation of enzymes and their substrates
in one cell compartment (one organelle — endoplasmic reticulum, mitochondria,
nucleus, etc.). For example, the enzymes of the TCA cycle and [J-oxidation of fatly
acids arc located in the mitochondria, enzymes of protein synthesis on the ribosomes.
The regulation of enzyme activity is also carried out in different ways.
□
HO—1—•□*■
Target — Target
Vu Phosph Ofylated
UiiptiosphoryHtecl
Phosphatase
o
I1Q— h£o
L
Transferred | water ]
phosphate group
For example, in this way, glycogen phosphorylase and glycogen synthase enzymes
arc regulated in muscles. Du ringcxcrcisc, whileglycog.cn phosphoiwlascbecomcsactivcand
starts breaking down glycogen and burning glucose, during this lime glycogen synthase is
not active. When glycogen synthase active, glycogen phosphorylase — inactive (Fig. 2.20).
AcLivabon or glycogen phosphorylase and inaclivaLion
ot glycogen synthase by covalent modification
Protein kinase A
ATI*
Glycogen phosphorylase
(inactive)
A Glycogen synthase
(active)
Fig. 2.20. Regulation of glycogen synthase and glycogen phosphorylase Dy covalent modification
A-Jlocatatybc adivabor-
Activation
at low pH
Pepsin [
Fig. 221. The scheme of activation of pepsinogen and the formation of me active form of pepsin
94 Chapter 2. Enzymes
Protein-protein interactions
The term protein-protein interaction refers to a situation, where specific
metabolites, rather than metabolites of biochemical processes, act as a regulator. After
the influence of any factors on specific proteins, the activities of the sc proteins change,
and they, in turn, affect the given enzyme. Also, a change in catalytic activity can be
caused by the association or dissociation of the enzyme subunits. This is a reversible
form of regulation. An example of protein-protein interaction can be the regulation of
the activity of protein kinase A through the mechanism of association-dissociation.
Protein kinase A is a tetrameric enzyme consisting of 2 catalytic (C) and 2 regulatory
(R) subunits. Activator for protein kinase A is cAMP (cyclic AMP). The addition of
c.AM P to the regulatory subunits of the enzyme causes their release from the catalytic
subunits. Catalytic subunits are activated (Fig. 2.22).
Cid ic AMP
inactive
protein kinase A
Regulatory
subunits
Catalytic
subunits
Inactive catalytic
subunits
Fiql 222. Scheme of regulation of me activity of prole in kinase A using protein-protein interactions
Allosteric regulation
Allosteric enzymes arc built from two or more subunits: some subunits contain a
catalytic center, others have an allosteric center. Attaching the effector molecules to an
allosteric (regulatory) subunit changes the protein conformation and. accordingly, the
activity of the catalytic subunit. These effector molecules can function as activators or
inhibitors, meaning that the change to the protein will increase the rate of the reaction,
or prevent further binding of substrates at the active site, respectively. In short, an
activator will increase the reaction rate, while an inhibitor will decrease the rate or stop
it entirely (Fig. 2.23). A regulation of allosteric enzymes is reversible.
2.4. Regulation of Enzymes and MetaDolic Pathways, inhibition of Enzymes... 95
tnnibitor site
activator Allosteric
inhibitor
Activator site
T njctosc-6-phosphate
Hatp
I L<ATP
{
H Citrate
HAMP y Fructose I I Phosphofructokinase
{-) Fructosc-2,6-bisP bisphosphatase
I *^ADP
T riKtosc-1,6-biphosphatc
(—) Phosphoeno Ip yru
(+} AMP
Fa 225. Scheme of glucose metabolism regulation
(+} Fructose-?,6-bisP
Inhibition of enzymes
Enzymes need to be tightly regulated to ensure that levels of the product do not rise
to undesired levels. This is accomplished by enzyme inhibition.
There arc two main directions of i nhibition. (Fig. 2.26):
► in terms of the strength of binding the enzyme to the inhibitor, inhibition is
reversible and irreversible;
► in relation to the inhibitor to the active center of the enzyme, the inhibition is
divided into competitive and noncompetitive.
Reversible inhibition
Reversible inhibitors bind to enzymes with non-covalent interactions such as
hydrogen bonds, hydrophobic interactions and ionic bonds. Multiple weak bonds
between the inhibitor and the active site combine to produce strong and specific
2.4. Regulation of Enzymes and Metaooiic Painways, inhibition of Enzymes... 97
Normal
Diisopropyl phospnofkioridate
H
I
H3q..-C — GHa
0
1
C—CH3
H
Diisopropyl phosphoryl
Enzyme
(inactivated)
Ry. 229. Reaction of me irreversible iihibilor diisopropyl pnospnofluondate |DFP) with a serine protease
inhibitors as drugs to treat disease. Many of these inhibitors target a human enzyme
and aim to correct a pathological condition. However, not all drugs arc enzyme
inhibitors. Sonic, such as anti-epileptic drugs, alter enzyme activity by causing more
or less of the enzyme to be produced. These effects arc called enzyme induction and
inhibition and act through alterations of gene expression, which is unrelated to the
type of enzyme inhibition discussed here. Other drugs interact with cellular targets
that arc not enzymes, such as ion channels or membrane receptors.
J
100 Chapter 2. Enzymes
Aminotransferases
Aspartate aminotransferase (AST, or SGOTl Myocardial infarction
Alanine aminotransferase (ALT, or SGOT) Viral hepatitis
One of the examples is lactate dehydrogenase (LDH) which catalyzes the reaction
of convening pyruvate into lactate:
0 OH
NADH •+ H
II OH
t NAD+ II n
HaC- -C| -v UH
II Lactate H II
0 Dehydrogenase o
Pyruvate Lactic Acid
This enzyme is composed of four subunits of two types: type M (specific for skeletal
muscle ) and type H (specific for heart). Different tissues produce different amounts
of the two subunits, which then combine to form five different tetramers (Fig. 230).
102 Chapter 2. Enzymes
DNA -------------------
1
Fi^ 2JO. Lactate dehydrogenase isofarms composition: LDffl (4H) - in the heart and in REC (red
blood cellsas well as the Dram LDH2 (3H1M} - in the reticuloendothelial system; LDH3 (2H2M) - in
lhe lungs; LDH4 (1H3M) - in the kidneys, placenta, and pancreas: LDH5 (4M) - specific for skeletal
muscle and liver
The activity of the enzyme in scrum blood of healthy people is low, because
the proteins can’t, penetrate through cell membrane to blood. If the membrane is
damaged, the enzymes leak to blood. The determination of tissue-specific enzymes
(or isoenzymes? activity in scrum blood allows us to diagnose different diseases, such
as myocardial infarction, hepatitis, and many others.
A B C
normal Acuw Aojie
serum Tyx&'Ua.- hepaaicis
irTaoion
e
LDH| -___ _
LDHs ------
LDHg
Fig. 2.31. Detection of different isoforrns of Isolate dehydrogenase in me serum of a healthy patient
a patient with myocardial infarction and hepatitis
On the other hand, diseases of various organs arc always accompanied by a specific
^enzymatic profile*. For example, myocardial infarction is accompanied by increased
activity in the blood of the isoenzymes: creatine phosphokinase (CK-M B). isoenzymes
lactate dehydrogenase I and 2 (LDH-1 and LDH-2), aspartate aminotransferase
(AST); troponin Tand I.
2.5. Clinical Applications of Enzymes. Plasma Proteins and Enzymes in Diagnosis of Diseases 103
Enzymopathies
If the enzyme cannot perform its function, it is called cnzymopathology or
enzymopathy. Enzymopathies arc conditions associated with an abnormal increase
or decrease in the activity of enzymes. The most common decrease in their activity
is a violation of the relevant metabolic processes. With cnzymopathology, clinical
significance may be:
► accumulation of subsume reaction. For example, phenylalanine in
phenylketonuria, free bilirubin in physiological jaundice of the newborn, some
fats in diseases of lysosomal accumulation (lipidosis):
► product deficiency. For example, melanin in albinism, catecholamines in
parkinsonism;
► both features simultaneously, as in glycogenosis, accompanied bv hypoglycemia
with an excess of glycogen in the liver:
By the nature of the violation, primary and secondary enzymopathies arc
distinguished. Lack of vitamins and their coenzyme forms is also the cause of acquired
enzymopathies.
Review tests
1. In a patient's blood, the activities of lactate dehydrogenase (LDII4. LDII 5), alanine
aminotransferase. carbamoyl ornithine transferase are increased. What organ is affected
by the pathological process?
A. In skeletal muscles.
B. In the myocardium (myocardial infarction is possible).
C. In the liver (hepatitis is possible).
D. In kidneys.
E. In connective tissue.
2.5. Clinical Applications of Enzymes. Plasma Proteins and Enzymes in Diagnosis of Diseases 105
Chart Analysis:
A. Curve with inhibitor.
B. Km with an inhibitor.
C. Initial speed.
D. Km. without inhibitor.
E. Curve without inhibitor.
6. A 42-year man suffering from gout has increased level of uric acid in the blood.
Allopurinol was prescribed to decrease lhe level of uric acid. Competitive inhibitor of
what enzyme is allopurinol?
A. Xanthine oxidase.
B. Adenosine deaminase.
C. Adenine phosphoribosyltransrerasc.
D. Hypoxantin phosphoribosyl transferase.
E. Guanine deaminase.
7. Match the figure and the letter.
The mechanism of regulatio n of enzyme activity:
1. Allosteric regulation.
2. Ph osph.oryialion - dephosp horyliatio n.
3. Protein-protein interactions.
Features of regulation:
A. There is a change in the primary structure of the enzyme.
B. Inhibition is irreversible.
C. Is carried out by activating or inactivating protein kinases and protein phosphatases.
I>. Is accompanied by dissociation or association of protomers or protein complexes.
8. A 47-year-old patient was brought to an emergency department with the diagnosis of
myocardial infarction. What lactate dehydrogenase (LDI1) fraction's activity would
prevail in the patient s blood serum during lhe first two days after hospitalization?
A LDH4.
B. LDH6.
C. LDH3.
D. LDH1.
E. LI)H5.
Situational Problems
1. Pepsinogen (an inactive form of the enzyme) that forms in the main cells of the
stomach has a molecular weight of 42.000 D. I n the gastric juice, the pepsinogen
turns into lhe active enzyme pepsin, while its molecular weight decreases to
35.000 D. Explain the mechanism of regulation of enzyme activity. To do this:
a) draw a scheme for regulating the activation of pepsin:
b) name the class of the enzyme that converts pepsinogen to pepsin:
c) indicate which levels of protein structural organization change when pepsin
is activated.
2. The interaction of the neurotransmitter acetylcholine with M-cholinergic receptors
causes a smooth muscles of lhe internal organs to contract — the intestines,
stomach, gall and bladder, bronchi, as well as constriction of (he pupils. In addition,
M-cholinergic receptors are found in the central nervous system. Atropine is a
2.5. Clinical Applications of Enzymes. Plasma Proteins and Enzymes in Diagnosis of Diseases 107
drug used in clinical practice Io relieve smooth muscle spasms and belongs to the
group ofanlispasmodics. What is the mechanism of action of this drug?
a) comparison with neurotransmitters and drugs, detection of similar functional
groups;
b) suggest the mechanism of action of atropine as a drug that relieves smooth
muscle spasms:
c) explain why an overdose of atropine can cause motor and mental agitation.
Chapter 3
GENE EXPRESSION AND PROTEIN
SYNTHESIS
Nucleic acids are an important class of macromolecules found in all cells and
viruses. Nucleic acids arc responsible for the storage and expression of genetic
information. The three main processes used by ail cells to maintain and propagate
their genetic information are replication, transcription, and translation.
Nitrogenous base
Fig. 3.1. A simple diagram of a nucleotide. Eacn nucleotide is composed of three parts: a 5-carbon
sugar, a phosphate group, and a nitrogenous base. Bom the phosphate group and nitrogenous base
are attached to the central pentose sugar: me nitrogenous base is attached to me T carbon atom via a
glycosidie bond ano the phosphate groups are covalently linked to me 5' carbon atom
Fig. 3.2. Nitrogenous bases molecular structures. The base is attached to a ribose or deoxyribose
molecule through a bond to me nitrogen atom shewn in color
[ [W-2-Deoxynbcise]
Table 3.1. Name of me bases and their corresponding nucleosides and nucleotides
RNA
DNA
Fig. 3.4. A structure of a polynucleotide chain Sugars are joined together Dy pnosphate groups
via pnosphodiester bonds Del ween me third and fifth carbon atoms of adjacent sugar rings. The
phospnodiester bond is indicated by red arrows. The prime or {*) designation is used when me sugar
is attached to me base to distinguish me base atoms from the sugar atoms
Guanosine triphosphate degxyribo nucleotide (dQTP)
t------------------------------------------------------- X------------------------------------------------------- V
Diphosphate released,
energy used for synthesis
Fig. 3.5. Nucleic acid synthesis. (A) NTPJS are the precursors used In DNA synthesis. A nuclei tide formed from phosphoric
acldr deoxyribose, and an erganIc base, guanine. <B) NMP moiety of NfTP is added to the3!-0H end of a polynucleotide chain
3.1. Nucleic Acid Structure 113
Sugar-phosphaSe
bacWxcte
Phosphorus
Carbon in
,34 nm “backbone7
3.4 nm
Bases
2 nm
Fig. 36 The 3D double helix structure ol DMA. Complemenlary base pairings are responsible for the
double-helix structure of DNA. f A) The structure of a douflle-stranded ONA. The two strands of ONA
run in opposite directions to each otner and are thus anti parade I. (B) Hydrogen bonding between
complementary base pairs. Adenine and thymine form two hydrogen bonds, and cytosine and
guanine form three hydrogen bonds
RNA molecule is quite similar to DNA. However, whereas DNA molecules arc
usually long and double-stranded, RM A molecules arc much shorter and single
stranded. RNA molecules play many critical roles in the cell but arc mainly involved
in protein synthesis (translation) and its regulation.
One of the key features of any RNA is that it can fold upon, itself via base pairs
between complementary nucleotides to form a three-dimensional structure (Fig. .3.7).
The secondary structure of RNA can be predicted by experimental data on the
secondary structure elements.
114 Chapter 3. Sene expression and protein syiitnesis
I
o—f
A Bulge loop
20 GUCC T
i
GCGCGGUG CACCUGA U-4O
I I. - I I I * I I I I I I I t
G UGG A C U
Fig. 3.7. Different types of RNA secondary structures. (A) The first structure (upper row, from [eft to
right) represents a stem with six paired Dases. Tne second structure represents a nairpin loop having
three paired and seven unpaired Dases. Tne tnird structure is a pseudoknot which is one of the most
common type in whicn tne unpaired base of two hairpins structure get paired with tne Diner one. Tne
forth structure (bottom row, from left to right) represents a bulge loop. The fiftn structure represents
an internal loop. Tne sixth structure represents a brancn loop, a multi-loop tn at contains al least three
branches. (B) RNA secondary structure of Estf?eriitt?ra 5S rRNA
3.1. Nucleic Acid Structure 115
The Ihree major types of RNA include: messenger RNA (mRNA), ribosomal
RIMA (rRNAh transfer RNA ([RNA). These RNAs participate in protein synthesis
(Table 3.2).
Messenger RNA (m RNA) carries the message from the DNA, which controls most
cellular activities in a cell. mRN.A accounts for just 5% of the lota! RNA in a cell.
mRN.A transcripts contain the sequence information that ribosomes need to translate
io prole in. mRN.A is relatively short-lived in a cell.
Eukaryotes have devised modifications at both termini to prevent mRNAs from
exonuclease degradation from both 5’- and 3’-cnds and, thus, stabilize the mRNA
(discussed below).
Transfer RNA (tRNA) is the smallest of the three types of RNA. It acts as an
adapter in. the translation of mRNA into proteins. The main function is the transfer of
amino acids during protein synthesis. Each of the 20 amino acids has a specific tRNA
that binds with it and transfers it to the growing polypeptide chain. All tRNAs have
certain common features, including the secondary and tertiary structures (Fig. 3.8).
Fig. 3.8. The tRNA structure. The canonical tRNA (left) secondary structure has a clover-leaf shape.
In me celL it folds into a compact L shape (right). tRNA mole cutes fold into a cloverleaf structure with
four key regions. The acceptor stem (3'-CCA) carries an amino acid; the anticodon associates wim the
mRNA codon (via complementary base pairing): the T arm associates with the ribosome (via the E, P
and A binding sites}; me D arm associates with me tRNA activating enzyme (responsible for adding the
amino acid to me acceptor stem) •; discussed in Ch. 3.5r 3.6)
with each other, stabilizing the overall structure of the molecule. Three adjacent
nucleotides constitute a unit known as the codon, which codes for an amino acid,
whereas an anticodon is a sequence of nucleotides that is complimentary to codon
(discussed in Ch. 3.5). Importantly, tRNAs have two properties:
> it represents a single amino acid, to which it covalently attached;
► each tRNA contains a set of three nucleotides called an anticodon. The
anticodon of a given tRNA can bind lo one specific mRNA codons (Fig. 3.9).
The anticodon enables the tRNA to recognize the codon via complementary
pairing.
3*
Amino acid
attachment sits
tRNA
AniicoOon kx»p
Anticodon
cue
LLL
mRNA
Fig. 3.9. Pairing of me tRNA anticodon with me mRNA codon proceeds from me S' end of the codon
The ribosome (Fig. 3.10) is an organelle that is present in large numbers in all
living cells and serves as the site of protein synthesis. Ribosomal RNA (rRNA) is
the major structural component of the ribosome and accounts for 80% of the total
RNA present in the cell. rRNAs perform critical functions in the ribosome that a How-
protein synthesis to occur: they arc involved in binding to mRNA, recruiting tRNA,
catalyzing the formation of a peptide bond between two amino acids.
Cytoplasmic ribosomes in eukaryotes contain four types of rRNA molecules,
whereas prokaryotic ribosomes contain only three types of rRNA molecules (Fig. 3.10).
3.2. DNA Replication 117
Mammalian mitochondrial ribosomes have small 28S and large 39S subunits,
together forming a 55S ribosome. Mitochondrial ribosomes have only two
mitochondrial, IlSand I6S, rRNA molecules.
70S SOS
SOS
subunit 60S
subunit
90S 40S
subunit subunit
Fig. 3.1 G. Tne ribosome is a comp tex subcellular structure or particle made of rRNA molecutes and
proteins trial form a factory for protein synthesis in cells. Prokaryotes nave 70S ribosomes respectively
suDanils comprising the small subunit of 30S and tne large subunit of 50S. Eukaryotes nave SOS
ribosomes respectively comprising of tne small AOS ano tne large 60S subunits. Su Du nils are named
according to their rate of sedimentation, measured in SvedDerg units [ST SuDunits are composed
of numerous rRNA and protein molecules: SOS: 5S RNA, 23S RNA. 34 proleins; 30 S: 16s RNA,
21 proteins; 60S: 5S RNA, 28S RNA, 5.8 RNA, about 49 proteins, 40S: 18S RNA, about 33 proteins
Table 32. Three main types of RNA molecules: structure and function
Function Serves as an intermediary Ensures the proper alignment Transports the correct
between DNA and protein: of mRNA tRNA. and ribosome amino acid to the site of
used by ribosome to direct during protein synthesis; protein synthesis in the
synthesis of protein it catalyzes peptide bond ribosome
encodes formation between amino acids
A T A T A I
G C G C G C
C G C G C G
T A T A I A
T A
T A T
T A
+ A T
T A
A T A T A T
G C G C G C
G C G C G C
C G C G C G
Orqin 3
Fig. 3.11. DNfl replication.. (A) semi-conservative replication: earn strand in the double helix acts as
a template for synthesis of a new, complementary strand. The result is two DNA molecules with one
original strand and one new strand. (B) part of a eukaryote chromosome showing multiple Origins
(1.2, 3) of replication, each defining a repficon (1, 2,3}. Replication may start al different times in
S-prtase. Here 1 and 2 begin first, men 3. as me replication forks proceed bidirectionally (indicated
by a Diack arrows}, mey create replication bubbles (indicated by a red arrow] mat meet and form larger
bubbles. Tbe end result is two semi-conservatively replicated duplex DNA strands
3.2. DNA Replication 119
3 The leading DMA strand is synthesized continuously in the 5' to 3" direction by DMA pot
5 RNA primers are removed, gaps are filled by DNA pol. then DNA ligase joins the Okazaki fragments
Each primer, bound to its complementary DNA strand, is elongated by DNA pol,
thereby synthesizing a now daughter strand. Importantly, DNA pol adds nucleotides
to 3’-end of the primer; therefore, DNA synthesis always proceeds in a 5’ to 31
direction. As shown in Fig. 3.12, synthesis of one daughter strand, called the leading
strand, proceeds continuously from a single RNA primer in the 5’ to 31 direction, in
the same direction as the movement of the replication fork. Synthesis of the other
daughter strand, called the lagging strand, requires a slight delay before undergoing
replication and occurs discontinuously via Okazaki fragment in the direction opposite
to the direction in which the replication fork is moving. The lengths of Okazaki
fragments are generally between 100 to 200 nucleotides long in eukaryotes.
Single-strand binding
The leading strand ts
Helicases unwind the synthesized continuouBly
proteins stabilize the
parental double helix. in me 5— 3’ direction by
unwound parental DNA.
DMA polymerase.
DNA polymerase
Replication fork
The lagging strand is
RNA primer
synthesized discontinuously.
Primase Okazaki fragment Primase synthesizes a short
being made RNA primer, which is
extended by DNA polymefase
DNA---------- Id form an Okazaki fragment
polymerase
Parental DNA
Fig. 3.12. The DNA replication scheme. The helicase unzips the double-stranded DNA for replication,
making a forked structure. The primase synmesizes RNA primers that bind to the single-stranded
DNA to initiate ONA synthesis by the DMA pol. DNA pol synmetizes only in the 5' to 3* direction, so
it replicates the leading strand continuously. Synthesis of me lagging-strand is discontinuous via
Okazaki fragments
3.3. ONA Damage & Repair 121
The energy required to drive the reaction comes from cutting high energy
phosphate bonds on the NT PS usedi as the source of the nucleotides needed in the
reaction (Fig. 3.5).
The RNA primers of Okazaki fragments arc subsequently degraded and the gaps
are tilted by DNA pol and sealed by DMA ligase (Fig. 3.I2). The ONA ligase is an
enzyme that creates phosphod tester bonds between adjacent nucleotides between
Okazaki fragments.
prevent DNA replication. Ionizing radiation and certain drugs can block replication
by creating double-strand breaks in the DNA. Intercalating agents, c..g., ethidium
bromide, can cause abnormal insertions and deletions in the DNA sequence.
DMA Damage
DNA damage, if not repaired, has the potential to generate mutations in somatic
or germline cells, which ultimately can alter cellular phenotype, cause dysfunction
and/or disease. To prevent such deleterious effects and maintain genome stability,
cells have evolved a number of mechanisms to detect and repair the various types of
damage that can occur to DNA. no matter whether this damage is caused by errors
in. replication or by the environment (Tabic 3.6). Well-known mechanisms include
base-excision repair, nucleotide excision repair, mismatch repair, homologous
recombination, and non-homotogous end-joining (Pig. 3.21). Additionally, the cells
possess mechanisms in which damage is directly reversed most often by a single repair
protein without the incision of DNA backbone.
Table 3.6. Types of DNA damage and the mechanism of ils repair
paired correctly with the base in. the template strand. If an incorrect base has been
added, die enzyme removes and replaces it (Fig- 3.14 A). This is performed by the 3’
exonuclease activity of DNA pol. Once the incorrect nucleotide has been removed, it
can be replaced by the correct one.
A DNA proofreading
B Nfomalcn repair
f
adds ihe commi DNA ligase repairs
base and some adjacent bases. ! Lhe remaining nick
bases.
Fig. 3.14. DNA repair mechanisms..(A) proofreading activity of DNA pol. During replication, me DNA
polymerase enecks for incorrect oases in the new DNA strand and immediately replaces them witn
correct ones. (B) DNA mismaten repair. After replication, mismaten repair proteins searenfor incorrect
oases tn at were missed t?y DNA pol and replaces mem
and insertion and deletion mispairs generated during the replication. During
MMR. enzymes delect distortions caused by mismatched bases and subsequently
remove the incorrectly paired nucleotide and replace it with the correct nucleotide
(Fig 3.I.4 B).
Base-Excision Repair (BER) involves the recognition and removal of a single
damaged base. HER corrects DNA damage from oxidation, deamination, and
alkylation. These lesions cause little distortion to the DNA helix structure. HER
mechanism requires a family of enzymes called glycosylascs. The goal of DNA
glycosylascs is to locale quickly and efficiently the aberrant base amongst a huge
excess of the normal ones. Glycosylases cleave the bond between deoxyribose and a
modified or mismatched DNA base. At least II distinct mammalian DNA glycosylascs
arc known, each recognizing a few related lesions, frequently with some overlap in
specificities. DNA glycosylasc recognizes and removes the damaged base forming an
abasic site (AP site) which is repaired by AP endonuclease before the nucleotide gap
in the DNA. strand is filled by DNA pol (Fig. 3.15).
Nucleotide Excision Repair (NER) is a widespread mechanism used by mammals to
remove bulky DNA lesions such as those formed by UV light {Fig. 3.16), environmental
mutagens, etc. The modified nucleotides cause a significant distortion in the DNA
helix structure.
In NER, the damaged bases arc recognized and subsequently cleaved out within
a string of nucleotides by endonucleases and replaced with DNA as directed by the
undamaged template strand by DNA pol (Fig. 3.17).
Direct Reversal Repair is one of the lesser-known mechanisms of repair where
the damaged lesion is repaired directly by specialized proteins. In contrast to
excision repair mechanisms discussed above, direct reversal of DNA damage, docs
not involve the process of breaking Lhc phosphodicstcr backbone of the DNA and,
thus, docs not require a template. One of the examples of the reversible DNA
damage is the damage caused by alkylating agents reacting with DNA. Alkylating
agents arc carcinogens that generate a broad spectrum of DNA lesions. DNA
alkylation damage can arise from exogenous or endogenous sources. Elimination of
the damage by direct reversible repair is achieved by two major types of proteins: Or'
methyl guan inc methyl transferase (MGMT) and a Ip ha-kcto-glutaratc-depend cm
and iron-dependent oxygenases.
Methylation of guanine O6 position confers the greatest mutagenic and
carcinogenic potential. O^methylguanine changes guanine's hydrogen-bonding
pattern and can confuse the cell into pairing it with T. During replication, one of (he
strands ends up with A-T, where a G-C should have gone.
The suicide enzyme methylguamne methyltransferase (MGMT/MTasc) can
remove the abnormal methyl group by transferring it to the sulfhydryl group of a
cysteine in the active site (Fig. 3.18). Importantly, unlike most enzymes, this enzyme
is deactivated by the reaction, it can no longer participate in transferring methyl
groups.
3.3. DNA Damage & Repair 125
Damaged tease —
■ ■ > I lx I iIi i 3
. JJLSJLUL5JJUJL 6.
DMA glycosylase
APsite^-... "
i I ■ 3
3Ai±iAiiiiii5
AP endonuclease
DMA polymeiase
1 DMA ligase
Fig. 11 5. Schematic illustration ot ON A repair by base excision. BER is initiated by a DNA glycosylase
specifically recognizing and binding a base lesion, upon encountering a substrate base the
glycosylase flips me base out and generates an AP site, whicn is recognized by an AP endonuclease
tnat hydrolyzes the pnospnodiester bond 5r to the lesion. Polymerase fl fills in me nucleotide gap,
which is subsequently seated by the DNA ligase, restoring the original base sequence
O O 0 0
Damaged nudsotide ■—
d n ■■ o
?9aaPD9?DD9
’ ^...-------- Endonucleases
□ c □ □ □ □ □ aMl 3
■ millin’
51
...................................................................................
3'
DNA polymerase I
5'
1 DNA ligase
3’
3' 5’
Fig. 3.17. The schematic illustration of nucleotide excision DNA repair HER proteins recognize the
damaged DNA, excise the damaged nucleotides, and repair the DNA strand Dy re-syntnesis/iigaiion to
restore the integrity of the DNA molecule
3.3. DNA Damage & Repair 137
H—N H-—N
Q6MeG 'i G
H H
Fig. 3.18. Direct revere a! repair. OG mefflyl guanine (OGMeG) is mutagenic because polymerases
frequently misinsert T opposite 0EMeG in stead of C, therefore the metnytaiion alters hydrogen-
bonding patterns of guanine in duplex DMA. MGMT restores tne original guanine Dy transferring the
methyl from 0GMeG to its active site
Hereditary Diseases
Many genetic disorders arc caused by genetic defects of DNA repair mechanisms
Jk/w/cnwa p^wew/ftswm (XP) is the classical human disorder caused by defective
nucleotide excision repair of DNA damage, including pyrimidine dimers induced by
UV. Signsand symptoms of the disease may include: severe sunburn when exposed to
only small amounts of sunlight, irritation, rough-surfaced growths (solar keratoses),
and skin cancers. In the most severely affected cases, there is progressive neuronal
degeneration as well. Treatment is supportive. Radiation is contraindicated.
Other hereditary diseases characterized by genetic defects of DNA repair
mechanisms include Nijmegen breakage syndrome, Werner syndrome, I?loom
Syndrome, Fanconi anemia, Cockayne syndrome, trichothiodysirophy. These rare
3.4. TranscriptiDAz RNA Synthesis 129
The RNA is synthesized from a single strand or temp late/sense strand of a DNA
molecule. The opposite DNA strand is called a coding strand, because its nucleotide
sequence corresponds to the sequence of the RNA transcript produced. The section
of DNA that is transcribed into an RNA molecule is called a transcription unit
A transcription unit codes the sequence that is translated into protein.
130 Chapter 3. Gene expression ano protein synthesis
fig. 3.21. Eukaryotic core promoter elements. Some upstream regulatory elements that can participate
in transcription by RNA polymerase h are depicted rnr and a coding region with introns and exons and
poiy(A) tail. The first base on the DMA where transcription actually starts is iabeied +1. Sequences that
are upstream of me first base of me transcript are marked with negative numbers. Sequences that are
downstream of me first base of the transcript are marked with positive numbers
TF
TRIA 3 Stabilizes binding of TATA-binding protein (TBPI and TFIIB
TRIB 1 Binds TBP.. selects start site, recruits RNA pol II
TRID 12 Interacts with regulatory factors
TBP 1 Subunit of TFIID. specifically recognizes the TATA box
TRIE 2 Recruits TFIIH
TRIF 2 Binds RNA pol II and TRIB
TRIH 9 Unwinds promoter DNA, phosphorylation of RNA pal II
RNA poll II 12 Catalyzes RNA synthesis
When the complex, of TFs and RNA pol is assembled (Fig. 3.22 A), DNA is
unwound to expose the template strand for the synthesis of an RNA molecule.
3.4. Transcription: RNA Synthesis 131
Transcription
RNA polymerase ll start site
strand
5’ I I I I I
A A - I Q
’GA G-iA
31 l i ill ill i i
Fig. 122. Transcription: RNA syiitnesis. (A) eukaryotic transcription initiation. Simplified model of
protein complex assembly on DNA is shown; (B) RNA synthesis. The RNA poi moves stepwise along
the DNA, unwinding me DNA helix at its active site, it adds nucleotides one Dy one to me RNA chain
al me polymerization site using an exposed DNA strand as a template. The incoming nucleotides are
in me form of ribonucleoside triphosphates (ATP, uTP, CTP, and GTF'i. and the energy stored in their
phosphate-phosphate bonds provides the driving force for me polymerization reaction
mRNA Capping
Capping of the prc-mRNA is the addition of 7-mcthylgtianosinc (m7G) to the
S’-end (Fig. 3.23).
323. Cap structure, wnile ine pre-mRNA is stilt feeing synthesized, a 7-methytguanosine cap is
added to the 5-end of we growing transcript by capping enzyme
Capping is the first modification made to RNA.and lakes place co-transc optionally
in the nucleus when the first 25-30 nucleotides arc incorporated into the nascent
3.4. Transcription: RNA Synthesis 133
Fig. 324. Canonical consensus poiy(A) sequence. The canonical consensus has elements sucn as the
AAUAAA polyadenytation signal, cleavage site -15-30 nucleotide downstream of PAS, G/GU rich DSE
and a. U-ricti USE
In general poly(A) tails play crucial roles in maintaining mRNA stability and
turnover, transport of mRNA from nucleus to cytoplasm, and translation efficiency
of mRNA. Nearly all eukaryotic mRNAs have a poly (A) tail al their 3’-end, with the
exception of histone mRNAs, which lack a poly(A) tail.
In humans, pre-mRNAs can be polyadcnylatcd in several different ways due
to the existence of more than one polyadenylalion site, allowing a single gene to
encode multiple mRNA transcripts. More than half of the genes in the human
genome arc alternatively polyadenylated. Alternative polyadenylalion plays an
important role in cell growth, proliferation, and differentiation. Importantly,
alternative polyadenylation potentially can alter the dynamicsand properties of an
RNA transcript affecting stability, translation and/or subccllular localization. If
for example, the alternative site lies upstream of the usual stop codon, a truncated
RNA transcript that tacks one or more exons is produced. This eventually leads to the
alteration in protein sequence and function. Abnormal alternative polyadenylation is
134 Chapter 3. Sene expression and protein synthesis
a-Bialassemia HBA1
Infection and B-ce-ll differentiation IGHM CSTF2 leads to proximal PAS usage
immunological
STAR. sWraidogBiiic acuta ragulalory: Br-cAMP, bromoadBiiosina 3'.5'-cyclic moflopkc&phatB; PAS. poly-adenylarTofi signal;
HGRGrl4_nigh-gluCDSB-regulala<l gane 14: TCF7L2, transcription factor 7-lika 2: HBB, nemoglsDin p: UTR. untrarcslaiBd r&giort;
HBA1. hemoglobin a 1 RBX1. ring-box 1: DMKN. dDrmakina; PDXK. pyridoxal kinasB: PF'lE, psplidyipiopyl isomsrasB E;
GGNDl, cyclin 01: CPSF5. cfeavags and potyadenylalion spfldf icily factor 5; IGHM, immunogloiRjlin iwavy constant mu;
GSTF2. ctoavags stimulation factor 2: NF-ATC1. nuclear factor cl adivalC’d T-cells 1; GlMAPS, GTPasa. IMAP family member
5; lPEX_ immuiw dysfunctions. polyendocrinopalliy. wiferopaBiy, X-linked: FOXP3. lorknead box P3: SNCA. synuclein ll PD,
Parkinson's disease; PABPN1. pcJy A-binding protein nuclear 1; HTT. Huntington's tfisBaso; CNOTG. CCR4-NOT transcription
complex subunit 6; UTR, unti aiisiatBd region.
3.4. Transcription: RNA Synthesis 135
pre-m R NA transcript
A
2 miron Exon 3
I
Excm 1 I .__ E»0fi2 Exon 3 ~
Spliceosome
/ X
Alternative splicing
Exon I
/
| Exon 2 j Exon 3 ] |
\
Exon 1 | Exon 2 | Exon 4~
Translation
Protein A Prolein B
Fig. 325. RNA spicing. The spliceosome is assembled from small nuclear ribo nucleoproteins
{snRNPs}. snRNPs recognize conserved sequences in splice sites, remove the introns, and past the
exons togetner. (A) Sc nematic diagram of the steps of spliceosome assembly and intron removal,
introns are removed by RNA processing in wnicri the intron is looped out and cut away from the exons
by snRNPs, and tne exons are spliced togetner to produce tne translatable mRNA (B}. When different
coding regions of mRNA are spliced out different variations of matured mRNAs that encode different
proteins will eventually occur
3.4. Transcription: RNA Synthesis 137
Primer 5’-CUU-3’ is being used to replicate this piece of DNA. What strand this
primer will anneal to: the upper or the lower? Write what product of RNA replication
would be the first live nucleotides that had been added onto the primer by DNA pol.
Mark the 5’ and 31 ends.
1 L lien? is a short stretch of replicating DNA. The lop strand is the template. Assume
the are the RNA primers. Numbers *1,2, 3, 4* indicate parts of four Okazaki
fragments.
Which was made first, the Left or the Right fragment? Which primer will be the first
to be removed. Left, Middle, or Right?
When the primer in the middle is removed and filled in with DNA, the fragments
must then, be joined. Which enzyme joins the fragments? Where is the final,
connection made: a or b?
12. A. diagram of DNA replication is shown below; Redraw the diagram into copybook
and fill in the empty boxes in the figure.
5' 3'
3.4. Transcription: RNA Synthesis 139
13. A scheme of DMA replication is shown below. Rewrite the diagram into copybook and
fill in I he empty boxes in the figure.
Replication fork
RNA primer
14. A scheme of UNA replication is shown below. Redraw the diagram into copybook and
fill in I he empty boxes in the figure.
A A
The leading strand is
Single-strand binding
Helicases unwind the synthesized continuously
proteins stabilize the
parental double helix. in the 5 W direction by
unwound parental DNA
DNA polymerase.
i i i ii •1
The lagging strand is
synthesized discontinuously.
Okazaki fragment Pnmase synthesizes a short
RNA primer, which is
extended by DNA polymerase
to form an Okazaki fragment
15. A scheme of DNA replication is shown below. Indicate lhe sequence of events that
occur during DNA replication by placing number in the emptied semicircle.
DNA potymerase
Replication fork
The lagging strand is
RNA primer
synthesized discontinuously.
Primase Okazaki fragment Primase synthesizes a short
being made RNA primer, which is
extended by DNA polymerase
5 DNA------------ Lo Form an Okazaki Fragment
3'
potymerase
Parental DNA
16. A diagram of splicing is shown below. Rewrite the diagram into copybook and fill in the
boxes with the question mark in the figure.
pro-mRNA transcript
l
ST
1
m
> Exon 2 Exon 3
3.5. Genelic Code Properties 141
17. A scheme of transcription is shown below E Rewrite the diagram into copybook and fill in
the boxes with the question mark in the figure. Mark the 5'- and 3’-ends.
Nontempiate strand
Direction
□f synthesis —
■ ■ . i rrTTliQ
#111
s a a G
promoter J-l t-L Template strand
□ RNA polymerase
Second fetter
u C A G
uuu Pte
ucu UAU
Tyr
UGU
Cys
u
u uuc ucc Ser
UAC UGC c
UUA UCA UAA Stop UGA Stop A
Leu
UUG UCG UAG Stop UGG Trp G
► the genetic code has polarity. The code is always read in the 5’ -* 3’ direction:
► the- gene and the polypeptide it codes for arc collinear. The sequence of nucleotide
pairs in a gene specifics a coll incar sequence ofamino acids in its polypeptide product.
Ammoacyl-tRNA Synthesis
Amino acids arc the substrate for protein synthesis, but they cannot be used
directly. They must first bind iRNAinordcrtobe used in protein synthesis (Fig. 3.27).
lRNA is an adapter molecule typically 70 to 90 nucleotides in length (discussed in
Ch. 3.1 ). tRNA contains rare bases in addition to the common bases. The rare bases
are nucleotides, which arc unusual either by the nature of the base, which is often a
methvfated base, or even by the nature of the bond between the base and the sugar.
These modi float ions arc catalyzed by various specific enzymes after the synthesis of
tRNA. The 3’ terminus of all tRNA has a conserved CCA-3’ sequence to which amino
acids bind. The main funcLion of tRNA is the transfer of ami no acids during protein
synthesis.
3.5. Genelic Code Properties 143
n
n
Fig. 327. Alanine tRNA or tRNA3’3 (codon GCC). Anticodon (CGG) is indicated Dy me red circle. When
a FINA recognizes and binds to its corresponding codon in the ribosome, the tRNA transfers tne amino
acid to tne end of the growing amino acid chain
o o o o
I
c™o (Aminoacyl-tRNAj
I
HO— R
I
NH/ Aminoacyl-tRNA synthase
Rg. 3.28. Binding of an amino acin to its specific tRNA. Tne aminoacyt group is esterified to the 31
position of tne terminal adenylate residue
Twenty different aminoacyl-tRNA synthetases exist, one for each amino acid.
Therefore, each iRNA is specific of amino acid, i.c., it can. bind (or * accept M only that
particular amino acid. Thus, tRNA-dj denotes an RNA specific of alanine, capable of
binding and transferring alanine, iRNA^ denotes an RNA specific of phenylalanine,
etc.
Ribosome
Ribosomes arc organelles present in both prokaryotic and eukaryotic cells. The
main function of the ribosome is to synthesize proteins. Ribosomes can be found
floating within the cytoplasm or attached to the endoplasmic reticulum (ER).
Importantly, the location of the ribosomes in a cell determines what kind of protein
will be produced. The ribosomes that are floating freely throughout the cell, synthesize
proteins that reside in the cytosol. The bound ribosomes synthesize proteins of the
endomembrane system as well as proteins secreted from the cell. The ribosomes arc
made up of small and large subunits. In ribosomes, approximately two-thirds and
one-third of the mass consist of RNA and protein, respectively (discussed in Ch. 3d).
The ribosomal structure is highly conserved between all species. Interestingly, a
mammalian cell may contain as many as IO million ribosomes, but each ribosome has
only a temporary existence. Ribosomes can link up amino acids at a rate of 200 per
minute.
The ribosome has three main sites for tRNAs: the A-, P-, and E-sites (Tig. 3.29,
left panel). They span the space between the small and large subunits. The mRNA
is bound to the small ribosomal subunit. The large subunit contains the pc pt idyl
transferase site where the acceptor ends of two tRNAs come together to transfer the
nascent peptide to the incoming amino acid (Fig. 3.29, right panel).
Exit lumel
Large subutel
mRNA
tending
Small s_tunl
Fig. 329 Schematic models showing me binding sites ol me ribosome. Each ribosome has a binning
site for mRNA and three binding sites for tRNA molecules. The P site holds me lRNA carrying the
growing polypeptide chain. The A site carries the tRNA with the next amino acid. Discharged tRNAs
leave the ribosome at the E site. 28SRNA is me catalyst for peptide bond formation. The two subunits
assemble to translate mRNA and disassemble when translation is complete
146 Chapter 3. Sene expression and protein synthesis
Initiation of Translation
Initiation of translation is a complex process when initiator tRNA, 40S, and 60S
ribosomal subunits arc assembled by eukaryotic initiation factors (elFs) into an 80S
ribosome al the initiation codon of mRNA (Fig. 3.30). Translation docs not simply
begin al the 5’ end of the mRNA; instead it starts at specific initiation sites.1 mRNA
of eukaryotes has 5’-cap structure and 3’-polv(A) tail (discussed in Ch. 3.4), both
of which arc important for the initiation of translation, in eukaryotes, a cap-binding
protein and several other cl Fs assist the movement of the 40S small ribosomal subunit
to the 5’-cap.
Once at the cap, the initiation complex com prising mRNA, the 40S small ribosomal
subunit, cl Fs, Mcl-tRNA.Mcl(initiatormethionyl-tRNA> and nucleoside triphosphates
(GTP and ATP — sources of energy) tracks along the mRNA in the 5’-*3T direction,,
searching for the correct start codon. The AUG codon for methionine functions as
the start codon, in the vast majority of mRNAs. The initiation AUG triplet is usually
separated from the cap by 50-100 nt. The efficiency of recognition of the 5’-proximal
AUG is influenced by a Kozak consensus sequence 5’-GCCGCCRCCAUGG-3’,
where R is a purine (A or G). According to Kozak's rules, the nucleotides around
the AUG indicate whether it is the correct start codon. If the context of the 5’-proximal
AUG is poor, the efficiency of initiation is reduced, and those ribosomes, which fail
to initiate at this site, continue scanning and initiate at the next AUG (leaky scanning)
(Fig. 3.30 A).
Hydrolyzis of GTP provides the energy for the union of the 40S complex with
the 60S subunit to form the 80S initiation complex of eukaryotic cells. Initiation
factors arc then released and translation proceeds by elongation of the polypeptide
chain.
4Cc3
5’
AL<3 AUG
I—1
initiation
Efficient of translation
translation
Fig. 130. The model tor initialian of translation in eukaryotes. (A: classical (efficient translation) cap
dependent initiation mechanism: 40S ribosomal subunits associated with me elFs-GTP-tRNAiMei
complex scan me mRNA for initiation codons, and translation is initiated at the first AUG. Non-
ciassicai (leaky scanning) cap-dependent initiation mecnanism is as follows: the first AUG start codon
can De bypassed and translation can start at tne next AUG codon. (Bi scheme of eukaryotic translation
initiation. Translation begins when an initiator tRNA anticodon recognizes a codon on mRMA The large
ribosomal subunit ioins the small sub unit. The attachment of the aminoacyt-tRNA tome ribosome is an
energy-requiring process. GTP acts as an energy source during translation, at the start of elongation
and during the ribosome's translocation. ORF — open reading frame: uTR-untransiated region
149 Chapter 3. Sene expression and protein synthesis
Fig. 3.31. tRNA for methionine is of two types. Initiator 1PNA, tRNA.1*4, is used exclusively to initiate
protein chains, and eiongator tRNA, tRNAWcl, transfers mewionine to internal sites in a growing protein
chain
Elongation of Translation
The translation elongation cycle adds one amino acid at a time to a growing
polypeptide chain according to the sequence of codons found in the mRNA. During
chain elongation, each amino acid is added to the nascent polypeptide chain in a
three-step cycle:
► positioning the correct aminoacyl-tRNA in the A.site of the ribosome;
► formation of the peptide bond:
► translocation of the mRNA by one codon.
As is the case with initiation, a set of proteins, elongation factors (cEFsh arc
required to carry out the process (Table 3.9).
Following translation initiation, an SOS ribosome is poised on. mRNA with the
anticodon of Mei-tRNAj in the P site base-paired with the start codon (Fig. 3.32).
The ribosome is now ready to bind the second aminoacyl-tRNA al the A site, which
will be joined to the initiator methionine by the first peptide bond.
The aminoacyl-tRNA is escorted to the ribosome by an eE F-1, which is complexed
to GTP. When the correct a mi noacyl-tRN A is inserted into the A site of the ribosome,
the GTP is hydrolyzed. The cEF-l bound to GDP is released1.
The second step is the pepridyl-transferase reaction, which is catalyzed by the 28S
rRNA of the large subunit. Peptide bond between the now-adjacent first and second
amino acids is formed that connects one ami.no acid to another, c. g., the methionine
1 The requirement for hydrolysis of GTP before cEF-1 is released from the ribosome is the
rate-limiting step in elongation and provides a time for proofreading of the codon-anticodon
pairing before the peptide bond forms.
3.6. Translation: Prolan Synthesis 149
from tlx first tRNA is transferred onto the amino acid of the second tRNA in the A
site (Fig. 3.32).
Fig. 3.32. The scheme of eukaryotic trail sialion elongation. The initiating methionyl tRNA occupies
the P site, leaving an empty A site. The second aminoacyl tRNA (e.g. glycyl tRNA) is Brought to
the A site By eEF-1 (complexed with GTP). Following GTP hytiroiyas, eEFl {complexed wilh GDP)
leaves ine ribosome with second tRNA {glycyl ffiNA) inserted into the A site. A peptide bond is then
formed, resulting in the transfer of methionine to the aminoacyi tRNA at the A site. The ribosome
then moves three nucleotides along the mRNA. This movement translocates the Mei-Gly tRNA to the
P site and the uncharged tRNA to me E site, leaving an empty A site ready for addition of the next
amino acid. Translocation is mediated by eEF-2 coupled to GTP hydrolyzis. This process, elongation
and translocation, is repeated until all trie amino acids have been added and me elongation complex
reaches the termination codon
After rhe peptide bond is formed, the ribosome translocates again, thus causing
the uncharged tRNA io occupy the E site. The uncharged tRNA is then released to
the cytoplasm (Fig. 3.32) to pick up another amino acid. In addition, the A site is
now empty and ready to accept the next aminoacyl-tRNA.
Termination of Translation
Termination occurs when a stop codon in the mRNA (UAA< UAG, or UGA) enters
the A site (Fig. 3.33). No tRNAs recognize these codons; instead they arc recognized
by proteins called release factors (RF) (Table 3.9). The polypeptide is released, the
ribosome disassembles back into its two independent subunits and translation is
terminated.
150 Chapter 3. Sene expression and protein synthesis
Figi 3.31 The scheme off eukaryotic translation termination. Termination occurs when one of the three
slop codons reaches ine A site. Slop codons are recognized Dy RF, whicn fit neatly into ine P site
After many ribosomes have completed translation, the mRNA is degraded so the
nucleotides can be reused in another transcription reaction.
hi many cancers, the amounts ofc IPs arc increased. For example, there i.s a strong
relationship between levels of ex pression and cancer for cl F4E, whose overexpression
causes malignant transformation of human cells. The transforming activity of'clF4E
can be explained by its ability to promote translation of a subset of mRNAs encoding
proteins involved in regulating growth, proliferation, and apoptosis, most likely in a
combinatorial fashion. Translational control plays a role in learning and memory; it
is implicated in certain neurological disorders, such as fragile X mental retardation
(FMR) syndrome. The disorder is caused by changes in the expression level of
FMR protein (FMRP), which is mutated or produced in reduced amounts in this
disease. FM RP is an RNA-binding protein, which normally inhibits the translation
of mRNAs whose products have critical roles in synaptic plasticity. Mutation or
reduced expression of FM RP results in excessive synaptic plasticity leading to FMR
disorder.
Table 3.10. Example of poslfranslational modifications, meir target amino acid residue!s|- and tne
enzyme(s) or proteins involved
Glycation Lysine
Antitumor Antibiotics
Antitumor antibiotics arc not the same as antibiotics used to treat bacterial
infections. Antitumor antibiotics arc a type of anticancer drugs that block cell growth
by interfering with DNA inside cells. This slows or slops cancer cells from growing and
keeps them from multiplying.
Anthracyclines are antitumor antibiotics initially developed from compounds
produced by This class includes daunorubicin. doxorubicin, idarubicin,
and epirubicin as well as liposomal formulations of daunorubicin and doxorubicin.
They exon their cytotoxic effects through several proposed mechanisms:
► intercalation between DNA base pairs, resulting in disrupting DNA and RNA.
synthesis;
► inhibition of topoisomerase II. resulting in double-strand DNA breaks during
DNA replication.
Dactinomycin, also known as actinomycin D, is antitumor antibiotic used to treat
a number of types of cancer. It binds to DNA and inhibits RNA synthesis resulting in
impaired mRNA production.
Bleomycin is an antitumor antibiotic and acts by induction of DNA strand breaks
leading to inhibition of RNA synthesis.
Mitomycin C is an antitumor antibiotic and acts as a double-stranded DNA
alkylating agent. Mitomycin C covalently crosslinks DNA, inhibiting DNA synthesis
and cell proliferation.
Puromycin is an aminonudcosidc antibiotic that possesses antitumor activity.
Puromycin acts as a protein synthesis inhibitor. It does not inhibit the enzymatic
process, but instead competes by acting as an analog of the 3'-terminal end of
ami noacyl-1 RNA, disrupting synthesis and causing premature chain termination.
Vinblastine is an antitumor alkaloid isolated from plant Vinblastine
binds to the microtubular proteins of the mitotic spindle , leading to crystallization of
the microtubule and mitotic arrest or cell death.
154 Chapter 3. Sene expression and protein synthesis
Table 3.11. The moite of action of antibiotics that inhibit protein synthesis
Aminoglycosides 30S Blocks functioning of initiation complex and causes misreading of mRNA
Tetracyclines 30S Blocks tRNA binding to ribosome
Chloramphenicol 50S Blocks peptidyltransferase
Macrotides SOS Blocks translocation
Clindamycin SOS Blocks peptide bond formation
Linezolid 50S Blocks early step in ribosome formation
Telrthromyciit SOS Same as macrolides
Streptogramins SOS Causes premature release of peptide chain
1. In the diagram, label the three tRNA sites, codonsand anticodons, peptide and
mRNA. List the sequence of events that occur when the incoming tRNA sets
into its binding site. Redraw the diagram as it will appear immediately after the
next peptide bond is formed.
2. Working with an animal celt culture system, a researcher created random
point mutations in the genes for the RNA polymerases. Individual cells with
RNA polymerase mutations were isolated and used to generate a cell line
that expressed that particular mutation. There was only one problem; the
researcher did not know which RNA polymerase was affected in each, of the
cell lines. To help resolve this question, the researcher analyzed the levels of
different RNAs expressed in each of the cell lines. The obtained results arc
presented below:
156 Chapter 3. Sene expression and protein synthesis
Tpnstoe
Unmutated cells 100 100 100
Which type of RNA polymerase {I, II. or 111) appears to be mutated in each one
of the cell lines? Explain.
3. ?! particular gene codes for a mature mRNA contains 900 bases, which is
translated into a 40 kDa protein. A mutant form of the gene created by a
single point mutation yields a 30-base mRNA yielding a short peptide with
nonfunctional enzymatic activity. Why is the obtained product nonfunctional?
To answer die question please:
1) draw a diagram of protein synthesis without mutation;
2) explain what is the genetic code and list the properties of the genetic code;
3) list the sequence of events that occur when a mutated mRNA is translated at
the IOth amino acid;
4) redraw the diagram as it will appear immediately after the peptide bond is
formed between the 9th and I Oth amino acids.
4. Doxorubicin interacts with DNA by intercalation and inhibition of
macromolecular biosynthesis. It blocks topoisomerase II. When this drug can
be used? To answer the question please:
1) draw a diagram of macromolecular biosynthesis that is inhibited by the drug:
2) how docs doxorubicin affect the macromolecular biosynthesis?
5. A schematic diagram shows the translation process at the stage of incorporation
of the 6th amino acid into the growing p-chain of Hb. Determine what amino
acid will occupy the 6th position in p-protomers of Mb. To answer the question
please:
1) mark the 5’ and 3’ ends;
2) redraw the diagram as it will appear immediately after the peptide bond is
formed between 5th and 6th amino acids:
3} what type of Hb has this amino acid sequence?
Vai-Met
\
mRNA
/
6. Antibiotics are used in the treatment and prevention of bacterial infections.
Tetracyclines inhibit protein synthesis by binding to the ribosomal small subunit
(3 OS) of prokaryotes and blocking the ami noacyl-tRNA binding to A site. Why
docs tetracycline only affect bacteria? To answer the question, please explain:
3.8. ONA, RNA ana Protein Synthesis Inhibitors 157
I ) what is ribosome?
2) what is the difference between prokaryotic and eukaryotic ribosomes?
3) what is the function of ribosome in a cell?
7. Determine which amino acid should be attached to tRNAs with the following
anticodons:
1) 5’-UAC-3’;
2) 5’-GAU-3’;
3) 5’-AUU-3’;
4) 5’-AUG-3’.
8. Design mRNA sequence of a protein fragment: N-Ala-Pro-Mct-Thr-C.
9. In sickle cell hemoglobin, there is a Vai residue at position 6 of the chain,
instead of the Glu residue found in this position in. normal 14 bA. Can you
predict what change look place in the DNA codon for glutamate lo account for
its replacement by valine?
10. A portion of an mRNA molecule has the sequence
5,-AUGCCACGAGUUGAC-3\ What amino acid sequence does this code
for? To answer the question please:
1) explain what is the genetic code and list the properties of the genetic code:
2) draw a diagram of protein synthesis;
3) determine which iRNA should be attached to the mRNA:
4) what is the anticodon for the very first iRNA that will attach to mRNA?
11. The peptide hormone oxytocin contains 9 amino acids. What is the minimum
number of nucleotides needed to code for this peptide?
12. A portion of the coding strand of a gene was found lo have the sequence
S'-ATCjAGCXjACTrTCCjCCCATTA-S'. A mutation occurred in the gene,
making the sequence 5'-ATGAGCGACCTTCGCCCATTA-3\ What effect
would the mutation have on the amino acid sequence of the protein obtained
from this mutated gene? To answer the question please:
1) explain what the genetic code is;
2) identify what mutation occurred;
3) what amino acid sequence docs the non-mutated and mutated gene code for?
13. Glycosylation is the addition of to the protein.
A. Carbohydrate;
B Upid;
C. Fat;
D. Minerals.
14. Which of the following is not an example of post-translational modification?
A. Polyadenylation:
B. Myristoylation:
C. Alkylation;
D. Ubiquili nation.
15. Fill in
In a ribosome, the formation of the peptide bonds of the new peptide chain
occurs in the ________subunit, whereas matching the codons of the mRNA are
exposed on the surface of the subunit. During the peptide elongation
stage of translation, each incoming ammoacyl-lRNA binds lo the.
158 Chapter 3. Sene expression and protein synthesis
-site of the ribosome, whereas the growing peptide chain is held on the t RNA in
the -site.
16. Antibiotics arc used in the treatment and prevention of bacterial infections.
Macrolides, c.g., erythromycin, azytromycin, and clarithromycin, bind to
the 23S rRNA component of the SOS subunit of ribosome and interfere with
the assembly of SOS subunits. Elongation is prematurely terminated. Why do
macrolides on tv affect bacteria? To answer the question please explain:
I} what is ribosome;
2) what is the difference between prokaryotic and eukaryotic ribosomes;
3) what is the function of ribosome in a cell?
4} draw a scheme of a process that is inhibited by macrolides.
17. A scheme of eukaryotic ribosome is shown below. Redraw the scheme in the
copybook and fill in the boxes with the question mark. What is the function of
ribosome in a cell? What arc the three functional sites in the ribosome?
Small
ribosomal subunit
3.9. The Regulation of Gene Expression 159
Operon
P^moiwn^tor . Smictogl panes
Structural genes
Regulatory gene
DNAfp
* - *■ — ■
■ , ■, -,B,c
Transcription |
mRNA
Illi
Translation
O O A O
Protein Protein Protein Protein
A B C D
Fig. 3.35. Structure of an operon Three basic components of an operon: promoter — upstream
sequence to which RNA polymerase binds; operator — segment of ONA to which a
repressor protein binds (inhibits transcription by obstructing RM A polymerase); structural
genes — genes that are collectively regulated by the operon. Structural genes encode
products that serve as enzymes, and regulatory genes encode products that regulate gene
expression
If a repressor binds to the operator then the structural genes will not be
transcribed. Alternatively, activators may bind to the regulatory region, enhancing
transcription.
^lac Operon d
Struc-tLFai genes
Trans rapti on
Binds to ifte
operator to inhibit
bansenption
lad regulator
>Enzymes La import lactose
pro tetri and break it down
Fig. 3.36. The tec operon in £ rafi All of the opera's lactose metabolism genes (/acZ, tecYano tecA)
are vacated next to each other downstream of a single promoter. This promoter serves as a recognition
site for tne transcriptional machinery of the RNA polymerase complex. All genes in an operon are
transcribed into a single mRNA molecule, wnich is subsequently translated into individual protein
products. A tec/ regulator gene with its promoter is found just outside the lac operon. Tne regulatory
gene lad produces a tec repressor protein. When lactose is absent, the lac repressor protein binds to
tne operator, blocking lhe RNA polymerase from transcribing the tec structural genes. When lactose
is available, a lactose molecule binds tne tac repressor protein, preventing the repressor from binding
to tne operator sequence, and the genes are transcribed
j.c., the same transcriptional activator or repressor can regulate transcription of every
single gene that has that particular I)NA regulatory element associated with it.
A B
Octamer of core nistonos:
H2A, H2Br H3. H4 (each ons x2)
Histone Core
<------------Core DNA
Linker
Nucleosome
Nucleosome core
F------------- H
0.05 gm
Fig. 3.3T Nucleosome structure. |A) Nucleosome structure is tne basic unit of DNA packaging. It is
formed through trie interaction between DNA and nisione proteins. A segment of ONA is wound around
a core of 8 histone proteins (two each of histones H2A, H2B, H3: and H4i. The «taiis* of these histone
proteins stick out, where they can be modified by a number of different nistone acetyltransferases,
methyitransferases, and other epigenetic enzymes. Many of these enzymes modify very specific sites.
(B) The core DNA forms two loops around the octamer. The DNA that is between each histone octamer
is called the linker DNA. and can vary in length from 8 to 114 base pairs. This variation is species
specific, but variation in linker DNA length has also been associated with tne developmental stage of
the organism or specific regions of the genome. (C) Electron micrograph of chromatin: the beads on
a string
Short region of
Dima double helix
“Beads on a string"
1 nm
form of chromatin
30-nm cnromatin
fibre of packed out
nucleosomes
Section of
chromasome in an 300 nm
extended form
Condensed section
700 nm
of chromosome
Centromere
Enti/e mitotic
1.400 nm
chromosome
fig 3.38. Distinct levels of chromatin organization Mote me folding of me - beads on the string
structures into 30 nm fiber and further folding into higher-order loop structures
3.9. The Regulation of Gene Expression 1B5
expel histones away from DNA or facilitate the exchange of histone variants, thus
creating nucleosome-free regions of DNA for activation of transcription.
Transcription Factors
Much of the information required for regulation of gene expression at the
transcriptional level is encoded by enhancers, which arc short (-100-1000 bp)
3.9. The Regulation of Gene Expression 1&7
noncod ing DNA sequences located al various distances from the respective promoter.
Enhancers can be anywhere in the genome: upstream or downstream of the promoter,
within gene introns, in distal intcrgcnic regions or even within gene exons. Such
remarkable location flexibility is achieved through *DNA looping* which brings
together distal enhancers and promoters to regulate gene expression (Pig. 3.39). On
the nucleotide sequence level, enhancers contain clusters of short consensus binding
motifs recognized in a sequence-specific manner by regulatory proteins called specific
transcription factors (TFs, not to be confused with general transcription factors, see
Ch. 3.4).
Fig. 3.39 The regulation ol gene transcription. RNA polymerase ll (RNA Pol IL. light green) and its
associated general transcription factors bind to me promoter (green). The muitiproteirr Mediator
complex (Med+Cdkfl {cell division protein kinase 8) stabilizes promoter/enhancer ONA loops Dy
physically bridging transcription factors bound at enhancers (purple) with the RNA polymerase ll
transcription machinery. thereby coordinating transcription initiation events. Mediator also stimulates
deposition of the ring-shaped cohesin complex off genes by sliding tne complexes along DNA
5’ TGACGTCA3’
Tilt
ill!
3' ACTGCAGT 5’
Palindromic sequence
B
NH2- interacts with Dinar TFs DNA binding J Hormone binding - COOH
Fig. 3.41. Three distinct domains of steroid and thyroid hormone receptors
Lipophilic molecules, e.g., steroid hormones enter the cell by simple diffusion
across the plasma membrane. Thyroid a hormones enter the cell by facilitated diffusion
t discussed in Ch. 4). When a hormone binds to a receptor, number of events occurs:
> receptor activation;
»■ activated receptors bind to «hormone response elements*;
► activation or downregulation of transcription from those genes to which the
receptor is bound. Most commonly, receptor binding stimulates transcription.
170 Chapter 3. Sene expression and protein synthesis
Importantly; rhe production of too much or too little steroid or thyroid hormones
causes a number ofclinically important disorders, c. g_, thyroid hormone receptors are an
important regulator in different processes including development, growth, metabolism,
etc. The hyperthyroid condition results from an overproduction of thyroid hormones
resulting in a continual stimulation of thyroid receptors which is detrimental for the
patient.
It has to be noted that TFs often work cooperatively, the binding of one TF to DNA
may enhance the binding of a second TF to a nearby location. TF activity can also be
stimulated or inhibited through direct protein-protein interactions with other TFs.
Wild type mRNA 5' GCU GGA GCA CCA GGA CAA GAU GGA 3'
Wild-type polypeptide N Ala Gly Ala Pro Gly Gin Asp Gly C
Silent mutation
Note:
Miissense mutation these are all
substitutions
Noneeinse mutation
Fig. 3.42. Ttie scheme illustrating the different types of mutaliarrs. Some mutations do not change
the sequence of amino acids in a protein. Some swap one amino acid tor another. Others introduce
an early stop codon into the sequence causing the protein to De truncated. The red color indicates
mutation. The arrow indicates the insert of one nucleotide
► nonsense mutation occurs when the [3 NA change creates a pre mat urc STOP
codon, which truncates the poly peptide, and usually, nonfunctional protein
product is synthesized. Examples of diseases in which nonsense mutations
arc known to be among the causes include: cystic fibrosis (cystic fibrosis
transmembrane conductance regulator gcnc.h Duchenne muscular dystrophy
(dystrophin), beta thalassacmia (p-globin);
► insertion or deletion occurs when the addition or removal of a base alters the
reading frame of the gene. It may be small (one or a few base pairs within a
gene) or large (an entire gene, several genes, or a large section of a chromosome).
This change affects every codon beyond the point of mutation and thus may
dramatically change amino acid sequence. A Ira mesh i ft mutation may occur
when a number of inserted/ddeted nucleotides in a DNA sequence is not in.
multiples of three nucleotides. Frameshift mutations can lead to a premature
end to translation of the mRNA or the formation of an extended polypeptide.
The resulting protein is usually nonfunctional. Frameshift mutations arc known
to be a factor in Crohn's disease, familial hypercholesterolemia, cystic fibrosis,
colorectal cancer as well as other cancers;
► duplication is when extra, copies of a chromosomal region arc formed, resulting in
different copy numbers of genes within that area of the chromosome. This type
of mutation may alter the function of the resulting protein;
► inversion is a chromosome rearrangement in which a segment of a chromosome
is reversed end to end. An inversion occurs when a single chromosome undergoes
breakage and rearrangement within itself. An inversion docs not involve a loss of
genetic information, but simply rearranges the linear gene sequence;
3J 0. The Effects of Mutations 173
> translocations when segments of two chromosomes arc exchanged (may interrupt
gene sequences):
► loss of heterozygosity is a loss of one allele of a genetic locus. Il is a common
genetic event in cancer.
Table 3.1 Z The types of mutations
Fig. 3.43. The scheme illusiraTing the different types of mutations: duplication, deletion. inversion and
translocation of DNA. A region of a chromosome before and after mutation is indicated
hotspots al which mutations occur much more frequently. Most mutations that change
the amino acid sequence arc deleterious and will be eliminated by natural selection.
Few mutations arc advantageous; those that spread through the population eventually
replace the former sequence. When a new variant replaces the older version of the
gene, it is said to have become fixed in the population.
Some genes or protein sequences may accumulate mutations at a relatively constant
rate (c.g., I change per million years). If this rate of change is reliable, it is possible
to calculate the time of divergence according to the number of differences. E.g., if a.
gene, which mutates al a rate of I bp per 100,000 years has 6 bp different, divergence
occurred 600,000 years ago. This concept is called the molecular clock. The molecular
clock provides a valuable means of estimating evolutionary timescales from genetic
and biochemical data.
The sequences of genes or proteins can be compared among species and used to
build phylogenetic trees. Closely related species typically have few sequence differences,
while less related species tend to have more. For example, cytochrome C is a small
hemeprotein and it. is associated with the inner membrane of the mitochondrion. Il
is an essential component of the electron transport chain (see Ch. 4). The amino acid
sequence of cytochrome C has been analyzed in over 100 species. Fig. 3.44 shows
the sequence comparisons for cytochrome C between human and cither rat or yeast.
Alignment of cytochrome C sequence of human and rat has a high, level of homology
(top panel), whereas alignment of cytochrome C sequence of human and yeast has
a lower level of homology (bottom panel). Sequence similarity searches can identify
<■ ho motogou s* protoi n s or go nos by dcicc Li ng excess si m i la ri ty — statist ical ly sig n i lica n t
similarity that reflects common ancestry. Substitutions within the primary structure
of the protein are relatively constant, over lime and. therefore, cytochrome C can be
considered as a potentially useful molecular clock. The results shown in Fig. 3.44,
indicate that the evolutionary path leading to the human species diverged with that of
yeast far before the divergence from a rat.
Betow: Alignment of human and rat cyloc hrome c amino acid sequences using BLAST.
Sequence similarity between the two proteins is 01%
Below: Alignment of Human and yeast cytochrome c amino acid sequences using BLAST.
Sequence similarity between me two proteins is 64%
HumanCylC: GDvEKGKKlFlMKCSCX;HTvEKGGKHKTGPNLHGLFGRKTGQlAPGYSYTAANKNKGllwG 61
Alignment G 1-KG +F -tC QChTvEkGGHK GFNLHG-i-FGR -t-GQA GySyT AM K +1-W
Yeasl Cyt C: GS^KKGATlFkTRClQCHTVEkGGFHKVGPNLIHGiFGRhSGQAEGySyTDANIKKMVLVVD 66
Interestingly, traits can be gained and lost multiple times over the evolutionary
history of a species.
The frequency of new mutations in a single gene or organism over time is not
limited to a single type of mutation. The rate at which mutations accumulate is
a characteristic of each protein. A protein evolves by mutations, followed by either
elimination or fixation. The presence of two allelic variants in the population, is
called polymorphism. Importantly, mutations by themselves do not classify as
polymorphisms. Polymorphism is a DNA sequence variation that is common in the
population. In polymorphisms, there arc two or more equally acceptable alternatives
and to be classified as a polymorphism, the least common allele must have a frequency
of I % or more in the population. If the percentage of occurrence is smaller, a random
occurrence — mutation, takes place. Polymorphisms arc responsible for many of
the normal differences between people, eg., blood type (Ch. 7.9). Ah hough many
polymorphisms have no negative effects on a person’s health, some of these variations
may influence the risk of developing certain disorders.
Single nucleotide polymorphisms (SNPsJ are the most common type of genetic
variation among people. Each SNP represents a difference in a single DNA nucleotide.
For example, some DNA molecules in the same population may have a T-A base pair
al a particular nucleotide siter whereas other DNA molecules in the same population
may have a C-G base pair at the same site. This difference constitutes an SNP. The
SN P defines two alleles for which there could be three genotypes among individuals in
the population: homozygous chromosomes or heterozygous chromosomes with T-A
in one chromosome and C-G in the homologous chromosome.
SNPs may occur within coding sequences of genes, non-coding regions of genes,
or in the intcrgenic regions (regions between genes). SNPs within a coding sequence
do not necessarily change the amino acid sequence of the protein, due to degeneracy
of the genetic code. Importantly, SNPs that arc not within a coding region may affect
gene expression. About 3 million SNPs that are relatively common in the human
population have been identified, of which about I million arc typically used in a search
for SNPs that might be associated with complex diseases. Each DNA polymorphism
serves as a genetic marker for its own location in the chromosome. SNPs responsible
for a disease can occur in any region that can ultimately affect the expression,
structure, and activity of the protein. Examples arc SNPs in transcription factor
binding domains, in promoter regions, in areas involved in transcript processing, such
as SNPs al intron-exon boundaries, which may cause defective splicing, or in mRNA
processing signal sequences such as polyadenylation signal regions.
Most SNPs have no effect on health or development. However, in some cases SN Rs arc
very important in the study of human health, e.g., response to certain drugs, susceptibility
to environmental factors such as toxins, and risk of developing particular diseases. SNPs
arc often used to track the inheritance of disease genes within families. For example, the
major disease gene for breast cancer in women is the gene BRCA I.l. For women who cany
' Vertebrate genes and proteins have names (typically strings of words) and symbols, which
are short identifiers (typically 3 to S characters). For example, breast cancer type I. susceptibility
protein is a protein that in humans is encoded by the BRCA I gene. BRCA I is a human tumor
suppressor gene that is responsible forrepairing DNA The tyrosinase — TYRgenc is responsible
for the production of the enzyme tyrosinase, which is the key enzyme in the formation of
melanin pigment. These symbols are usually, but not always, coined by contraction oracronymic
176 Chapter 3. Sene expression and protein synthesis
a mutant allele of B RCA I, the lifetime risk for breast cancer is higher than for women who
arc not carriers. The importance of the genetic risk factor can be expressed quantitatively
as a relative risk, which equals the risk for disease in the individuals who carry the risk
factor as compared with the risk in those who do not. Another example is cytochrome-P
450. Cytochrome-P 450 (CYP) enzymes play a role in the synthesis of many molecules,
including steroid hormones, certain lipids and bile acids. Additionally, CYP enzymes
arc essential for the metabolism of many medications, and internal substances, such as
toxins. CYP enzymes account for 70-80% of enzymes involved in drug metabolism.
Polymorphisms in cytochrome P450 genes can affect the function of the enzymes. The
effects of polymorphisms arc most prominently seen in the breakdown of medications.
Depending on the gene and the polymorphism, drugscan be metabolized quickly or slowly.
If a cytochrome P450 enzyme metabolizes a drug slowly, the drug slays active longer and
less is needed to get the desired effect. A drug that is quickly metabolized is broken down
sooner and a higher dose might be needed to be effective. Therefore, polymorphisms in
CYP can lead to different drug responses or loxicitics.
Hereditary diseases arc diseases or disorders that arc inherited genetically.
Hereditary diseases arc passed on from one generation to another through defective
genes. The chromosomes in the humans arc responsible for passing the traits from the
parent to the offspring.
The most common hereditary diseases include Huntington’s disease, myotonic
dystrophy, neurofibromatosis, cystic fibrosis, Duchenne’s muscular dystrophy,
haemophilia, Marfan syndrome, a and p-thalassemia, fragile X syndrome, sickle cell
disease, etc. The following example illustrates how gend s defects causes disorders.
Albinism is one of the inherited disorders. Affected individuals typically have very-
fair skin and white or light-colored hair. Long-term sun exposure greatly increases the
risk of skin damage and skin cancers. All forms of albinism also cause problems writh
the development and function of the eyes.
A defect in one of several genes that produce or distribute melanin causes albinism
(Table 3.I3). As can be seen from the Table 3.I2, there are more than 300 different
mutations in 777? gene that arc associated with the disease. Il encodes a protein
tyrosinase of 529 amino acids. 7TJ? gene is located on chromosome llql4.3 and
consists of five exons (Fig. 3.45). The defect may result in the absence of melanin
production, or a reduced amount of melanin production. The defective gene passes
down from both parents to the child and leads to albinism.
Humans arc diploid organisms because they have two alleles at each genetic
locus, with one allele inherited from each parent. If the two alleles arc the same,
they arc homozygotes. If they are di Ik rent, they arc hcterozygoies. In the case of
hctcrozygotes, the individual can express cither one or a combination of the two
traits. Oculocutaneous albinism (OCA) is inherited in an autosomal recessive manner,
meaning that both copies of a gene in each cell have mutations. Most often, the
parents of an individual with an autosomal recessive condition each carry one copy of
the mutated gene, but they do not show signs and symptoms of the condition. Ocular
albinism (OA) is much less common and inherited in an X-linkcd pattern meaning
that the mutated gene that causes the disorder is located on the X chromosome, one
abbreviation of the name. .All human gene names and symbols can be searched online al the
«HGNC database of human gene names — HUGO Gene Nomenclature Committee*.
3J 0. The Effects of Mutations 177
of the two sex chromosomes.In males, one altered copy of (he gene is sufficient to
cause OA, because males have only one X chromosome.
Table 3.13. Mutations detected lor albinism associated genes*
-i
□CAI TYR 11q14-q21 303
□CA2 □CA2 15q11.2-q12 154
□CA3 TYRP1 9p23 16
OCA4 SLC45A2 5p13.3 78
□CAS ND 4q24 1
□CAO SLC24A5 15q21.1 2
□CA7 C1OORF11 1Oq22.2-q22.3 1
OA1 GPR143 Xp22.3 114
LYST CHS1 1q42_1-q42.2 53
HPS1 HPS2 1Qq23.1-q23.3 31
AP3B1 HPS2 5q14.1 20
HPS3 HPS3 3q24 7
HPS4 HPS4 22cen-q12.3 13
HPS5 HPS5 11p14 11
HPS6 HPS6 10q24.32 9
HPS7 DTNBP1 6p22.3 2
HPSfi BLOC1S3 19q13.32 2
HPS9 BLOC1S6 15421.1 1
OCA. oculocutaneous albinism. Tn® OCA1A is Hue most severa type with a complete lack of melanin picductic-n truoughout
life, while 1Fie milder forms OCAlB, OCA2, OCA3, and OCA4 slutw soma pigrront accumulation ever time. OA, ocular albinism
is a genelic confttiwi that primarily affects the eyas, OA reduces the coloring (pigmentation) of the iris, and the retina, which
is the ligm-sensifert tissue in the hack of the ey®. Il is clwacterizeo by severely impaired sharpness of vision and problems
with combining vision from Doth eyas to perceive depth (stereoscopic vision). OA Hoes not significantly affacl ths color of
the skin and hair. LYST, lysosomal 'trafficking regulator. HPS. Hemiansky-Pudlak syndrome, is a genetically heterogeneous
group of autosomal recessive disorders characterized principally by oculocutaneous afflinism (OCA), blooding tendency, and
progressive pulmonary fibrosis. AP3B1. adaptor related protein complex 3 subunit beta! TYRPi. tyrosinase-related protan 1.
is an enzyme in the melanin biosynthesis pathway, catalyzing She oxidation of 5.6-dinydroxyindblo-2-carbbxylic acid (DMCA)
monomers into melanin (see Ch. 7.).
1 Source: human gene mutation database. 05 Doc, 2013; JMD: not determined.
mjZOiSw AJTp2-yS GyS'JMFnc GrESl-bt Gly47GyS
wuzeTm AigE-2Th Gtfs:DCTfp GrrXiAig -EtyEiArg AM+'.&Sfli ASp+44Gly
amK&M:- Aip77Qn -E^^Aig GkiZtSLyS &y97ftg JVQJtf’Gifl ALp+4fiAST
waffifiTni AiCsTTGly ■
GyflS+V GkJ-l'JTwfTTi GiyJTvoi AJg«Htjty GmfiMs
jwg,nGTaffl wgTTTrp
j t Gy5SAfg GuZ2iLys leiBZ-As; Arg+CPLii rsrrr.
A-J2 i2LjS ASfCVnr GyiEE’yi Gki2EZ'Rm Hfifhug — ijSECk AipjfijAi- AflMCZ'Hffn Qu-32flMa
A^IiITnf
*jg2 ilTnr A^jiKTyr Gj-mEZ'^
A-zp i35Tyr ByMfiZyi GariSTtanm nsiOGri AnSSEPra ■SiiYKlKh.- wwtBSw GmSlKh
AJp2lTE*fl ALf i-HlAa- GyfiSE.7yf -ZlyluEdVC i«302Ajp am33iTw ATO4O3E&- ZVu.WhMJ
Aiy2l713y ‘ ‘ - --
■SpsEflAig - lOCMroi
ijh —• bfcsKKjn amS’JiCk ija'ETain- Jij-g^ZEiDu GiL*Z(lA!4< AM^EiGk.
A/p2r.rTi-p ASf2*jGiy GysSiEor HasSujug A£fl37l~nr BnSTSLyt aam2K«i GkM lET-witi AiMiOGtr
.AK|2?Kih A&p^SASn Gyt'llTyf ClySSG*. »tc2&£T-,r Air3.' i Tye GtrtiraTwiB AjjiHTip EkMrjWj-g Grfiat’&in
*jp233Trp ASjMiiEi^ GrSEToffn Giy4i>wg Ki23"ni ALfTjffiL-jiS G.yjJoGkj Anj+S«K GkMStArg Fn»6DS«
c
az; " Zj<.: ^lEaZLy: Qy3461affnAg-43&A5|l GAMAc-E^llTriMo rafffi
Excm 1
G4ji34€-¥3
■jlrHZPJVU.
EoSECfj.
EnrSEiAnj
<
Exon 4
HMlMASfl $a42*R-B
tw+C4Frc' Eom*2Fitc
lju2iEUct — ■"......... - TipflSSTom
■EjfWA/n tyau GIlu'ET tfiil
vu^— Sw33H3Jy HK3B3Tjf EnOSOPra -.Du^Pvai Ti-jJ+HK^
LAd)PJO 5tr4+Gjy _rp272Aj-g varTi'Fna ldu2BEFtid ~^-—
TwEEAia •irtSEJ'wg WQM.'HLyS Ti-psOOia.
L|Sl3H3b E^mEjZT<mi7 T.ytfTKyz - —
vit™"
SinF—
th LGu2®Eei- TjfSTCys rts3STTjr PnMSboj Tjf4l i»-h-
Ly5142w» E^ECTflffl TijtlflAX Lbj3'»2W9 voCTSPtk HfSXlAIfl WJGT7AM FTO+CfcLCu Tjrt/BCyt
LySMiMCI Efl'TillLdu TifMC/s MeE.'k "kQBlBuiu MaCTDK WM3K1FTW FlIM l2AM
lydJ+ETrr Ear75Pra TipliTiilTi Mafl32Tnr WsCTOTw FlD+lTbM
Ly533TTV nwiffiSw TipBDwg. RltiMtihj FTC-4'J I LUj
UaiTDUu TfpBZTwm PiwEsThr V3ME7Q^
MGEiffivai Ty1M9Cy£ SuMSwg M3m2.'Ftk
Wnr.Tw Ty f IBlCfS Bk3Sasi
HM1V3 TyfEGE+lj'
Fig. 345. Database of tyrosinase (Tr/7) gene mutations. The mutations identified in 0CA1 for each
exon are indicated. Nomenclature for tne description of mutations: Arg52Lys — Arginin al position 52
is reptacedi by tysine
178 Chapter 3. Sene expression and protein synthesis
Sickle cell disease is one oft he most common inherited blood disorders. The disease
is caused by a single point mutation of the p-globin gene (WBB), on chromosome
II. As a result, a sixth amino acid in. the chain is valine rather than glutamic acid
(Fig. 3.46). The disease is inherited in an autosomal recessive manner, which means
both copies of the gene in each cell have mutations, i.e., a baby born with sickle cell
disease inherits a gene for the disorder from both parents. When both parents have
the genetic defect, there is a 25% chance that each child will be born with sickle cell
disease. If a child inherits only one copy of the defective gone (from either parent),
there is a 50% chance that the child will carry* the sickle cell trait.
Wild-type haemoglobin DNA Mutant haemoglobin Dna
4-------- 4 ‘-QF
Fig. 3.46. Missense mutation and HDS. The 20A >T mutation in me HOB gene results in the production
of haemoglobin S (HBS)
Table 3.14. Examples of diseases resulting from duplications or deletions of chromosomal regions
The d
Charcot-Marie-Tooth disease type il Duplication 17p12
" in a small numbar of cases, people win Srmth-Maganfis syndrome tave irtnei itod tne deletion or mutation from an
unattncled molder who Harf me genetic cltartge only in her egg ceils. This phenomenon is called germline mosaicism.
” in a small percemage of cases, people with Williams syndrome inherit the chromo&omal deletion from a parent with the
condition.
Recombinant DNA
Recombinant DNA (or rDNA) is made by combining DNA from two or more
sources (Fig. 3.47). In practice, the process often involves combining the DNA of
different organisms. Recombinant DNA is an artificially made DNA strand that may
or may not occur naturally, but is engineered specifically for a purpose to be used in one
of the many applications of recombinant DNA.
Restriction enzyme is an endonuclease that recognizes a short, specific sequence
in DNA and cleaves DNA al these sites along the molecule. These regions arc called
recognition sequences and arc randomly distributed throughout the DNA. They arc
produced by bacteria in order to defend against bacterial viruses. Restriction enzymes
can be isolated from bacterial cells and used in the laboratory to manipulate fragments
of DNA. e.g., to cut DNA into smaller fragments. The cuts arc always made al specific
nucleotide sequences. Different restriction enzymes recognize and cut different DNA
sequences. The names of restriction enzymes arc derived from the genus, species, and
strain of the bacteria that produce them; for example, the enzyme EcoRI is produced
by E cofi strain RY 13 and specific recognition sequence is GAATTC, Bam HI is
produced bv strain II and specific recognition sequence is
GGATCC (Fig. 3.48), etc.
3.11. Recomninanl ONA Technology 181
Fig. 3.47. RecoinOinanl DNA technology, simplified scheme. Steps include: cutting the desired ONA
and vector Dy restriction enzymes, gene amplification Dy PCR; inserting tne gene into the vector,
transferring tne vectors into host organism, and obtaining the products of recombinant gene
182 Chapter 3. Sene expression and protein synthesis
L
5'-ATGGATCCAA-3' Bam H1 5-ATG-3r 5?-GATCCAA-3?
3’-TACCTaGGTT-5’ —“ 3-TADCTAG45' 3?-GTT-5’
I
5-GAATTC-3’ Eco R1 5-G-37 5-AATTC-3’
3'-CTTAAG-5: ----------- 3'-CTTAA-5' 3-G-5’
I
Fig. 3.48. Schematic representation of restriction enzymes Bam anti fcnffl cleavages. SamHI
recognizes the DMA sequence 5r-GGATCC-3r, and introduces a single-strand cut between tne G and G
nucleotides. This recognition site is a palindrome: the apposite strand also reads 5'-GGATCG-3’ and will
be cut in the same manner. EroRl recognizes tne DMA sequence 5'-GAATTC-3', and introduces a singte-
strand cut between the G and A nucleotides. Tnis recognition site is a palindrome: tne opposite strand
also reads 5'-GAATTG-3r and will be cut in the same manner. The cleavage sites of EccRl is shown
Gel Electrophoresis
Gel electrophoresis is a technique commonly used in laboratories to separate
charged molecules like DMA, RNA, and proteins according to their size (Fig. 349).
Samples
Marker ABC
1_200bp
I.ODObp
909 bp
BOO bp
700bp
BOO bp
SOObp
400bp
390 bp
200 bp
tOO bp
Fig. 3.49. Gel electrophoresis of DMA (left) and proteins (right). The drawing showing DMA hands
separated on a gel (left panel). The length of the DNA fragments (A, B, C) is compared to a marker
containing fragments of known length, illustration showing protein bands on a gel rrignt panel). The
size of me protein is compared to a marker containing fragments of known length
3.11. Recomninant ONA Tec neology 183
Double-stranded
DNA
Fig. 3.50. Three main stages of PCR incEude denaturation. annealing, ana elongation
At the end of the first cycle, each double-stranded DNA molecule consists of one
new and one old DNA strand. PCR then continues with additional cycles that repeat
the aforementioned steps. The newly synthesized DNA segments serve as tern plates
194 Chapter 3. Sene expression and protein synthesis
Fig. 3.51. PCR analysis. PCR products from assay controls and 12 clinical specimens. Samples in lanes
1,2, 4, and S are negative for the test viruses (internal control [iC] product visible oniy|. Samples in
lanes 9 and 10 are HSV**-1 positive, in lanes 11 and 12 are HSV-2 positive, and in lanes 6 and 7 are
CMV positive, and me sample in Eane 5 is VZV positive. Molecular mass (MW) markers (with selected
sizes in base pairs) are shown at both ends of the gel; neg — negative control (nuctease-free water).
‘ from Julian Druce et al. J. Clin. Microbiol. 2002; 40:1728-1732. * * — HSV — Herpes Simplex
Virus, CMV—Cytomegalovirus, VZV — Van cel la-zoster virus
DNA Ligation
DNA ligase is a specific type of enzyme that facilitates the joining of DNA strands
together by catalyzing the formation of a phosphodiester bond.
► If two DNA molecules have matching ends, they can be joined by DNA ligase:
► Restriction enzymes and DN.A ligase are often, used to insert genes and other
pieces of DNA into plasmids during DNA cloning.
The niost wc 11 - know n cxa mplc is t he R F L P d tic Lo (1-globi n gc nc mu talion (Fig. 3.^ 2).
Mst Ji Mst II
I Gene with sickle cell mutation |
Mutant ON A
CCTGAGG CCTGAGG
Fig. 3.52. RFLP resulting from p-Qlohin gene mulatiDiL in tie normal ce I, the sequence corresjpndino
to 5th to 7in am no acids of the p-gio&in peptide is CCTGAiGGAG mat is recognized by Msm Digestion
wiitn MsELI generates fragments 0.2 kb and 1.2 kb (upper panel), in tbe sickle cells there is A >T
mutation (discussed above), and the site is not recognized Dy Maty
(RNA) |
Southern Blot
(Restriction enzyme)
=-
Northern Blot
(Single stranded)
Western Blot
(Deanatured polypeptides)
XI I
«
E
□
a.
Ell *
I
□
-i—1
I I
u
a
a
e Agarose Acrylamide
0.5—50 Kbp 0.5-10 Kb 10-120 Kd
I
□
ft
ft
CC Sample contains specific Sample contains specific Sample contains specific
DNA restriction fragment RNA transcript Polypeptide
(e.g., mRNA)
Can measure fragment Can measure polypeptide
size and amount Can measure fragment size and amount
(single vs. repeated) size and amount (level of expression)
(level of expression)
1000 bp—
750 bp—*■
400 bp—
3. A missense mutation results in the presence of a different amino acid than was
encoded by the parental sequence. This type of mutation can have a drastic effect
or no effect at all depending on the importance of the amino acid and the type
of amino acid that replaces it. Some amino acids are structurally similar and
may be able to act as viable substitutes for each other. For example, changing
one acidic amino acid to another may not affect the final protein, buL changing
a polar amino acid to a nonpolar amino acid will likely disrupt the structure.
Please explain what mutations occur in //BB gene of abnormal hemoglobin and
their effect on the function of the protein.
5. The 7YB gene encoding for tyrosinase from two individuals was analyzed. It
has been found that one individual has a wild type TK/f and the other one has
the mutation. On the gel electrophoresis, the control unaffected and the mutant
have no difference in the mobility, i.e., no change in size. What type of mutation
had most likely occurred?
6. The starting sequence of a gene changed from jAUGTTCGACGTGp to
jAUGTTTTCGACGTG^. What type of mutation is this? To answer the
question, please explain:
1) what the mutation is?
2) what types of mutation do you know?
3) what arc the possible outcomes of mutations?
7. Scientistsstudying the genetics ofa congenital disease analyzed the chromosomes
of the patient and found a large portion of chromosome twenty-th rec in
188 Chapter 3. Sene expression and protein synthesis
3456709 10
364 bp—
291 bp—
201 bp—
90 bp—
pA/ps
11. What is the Zac opcron? What happens in terms of the Zac opcron when lactose
is absent from the growth medium?
12. You are working with a piece of DNA of the sequence:
3-ATAACrCGAGGGGCCTA-5'
You cut the above piece of DNA with a restriction enzyme that recognizes the
sequence 5’GAGCTC and cuts on. the 31 side of the A within this sequence.
Please, draw all products that you get after digestion. Label all 51 and 3’ ends.
13. Once transcription is complete, gene expression is controlled by...?
14. Why denaturation stage is important in PCR? To answer the question, please
explain:
3.11. Recomninanl ONA Technology 189
PhospMpids
imerior
Choline------
N+(CH3)3
Phosphate——CHZ
0
Glycerol -—m 0=P—O’
CHn—CH-CHP
6 o
Fig. 4.3. Membrane proteins: 1,2 — integral itransmembrane) proteins; 3,4,5,6 — surface proteins.
Part of the polypeptide chain of tne integral proteins is submerged into lipid bi layer. Those protein parts,
wtiicn interacts wim hydrocarbon chains of fatty acids, mainly consist of non-polar amino acids. Protein
parts, located in the polar «neads* area, nave lots of hydrophilic amino acid residues. Surface proteins
have different ways to attach to the membrane: 3 — connected to the integral proteins; 4 — bonded to
me polar - heads* of lipid Drlayer; 5 — ^anchored- in the membrane with me help of short hydrophobic
end domain; 6 — '’anchored* in the membrane with the help of covaienlty bonded acyl residues
Membrane proteins, which arc in contact with the hydrophobic pan of the lipid
bilaycr must be amphiphilic, holding a non-polar domain. The hydrophobic property
of this non-polar domain is guaranteed by the following;
► the amino acids residues contacting the lipid bilaycr mainly arc hydrophobic;
* a lol of membrane proteins arc covalently bound to the fatty acids residues
(acylated) that make them more hydrophobic.
Acyl residues bound to proteins provide their ^anchoring* to the membrane and
the possibility of the lateral dillusion. Along with that, membrane proteins undergo
posit ran slat ional modifications, such as glycosylation or phosphorylation. The
glycoproteins serve as receptors for compounds such as hormones, as cell attachment
and as cell — cell recognition sites. Viruses and bacteria also bind to these sites.
Membrane proteins take part in:
► selective transport of metabolites into the cell and back;
» hormonal signaling;
► formation of ^coated pits* which takes part in cndocytosis and cxocytosis;
► immune response:
► catalytic reactions:
► intercellular interactions, which provide the formation of tissues and organs.
194 Chaplet 4. Structure and functions of biological membranes. Cell signaling pathways
Oilier and inner layers of the same membrane have different lipid and
protein composition. This membrane structural specificity was characterized as
transmembrane asymmetry.
Fig. 4.5. Ca3* channel of trie endoplasmic reticulum, regulated by the inositol 1,4,5-hisphDsphate
(IP*). IP3 (inositol 1 AS-trispnosphate! is formed during nydrolyzis of membrane lipid PlP?
•fpnospnatiOylinositol 4,5-bispnosphate) Dy the enzyme phospholipase C. IP* binds to the specific sites
of Ca7, channel protomers in the ER membrane. Protein conformation changes and channel opens —
Caf • goes into cell cytosol by the gradient concentration
Membrane Membrane
outer inner
surface surface
Ca2+ — ATPase of the plasma
and endoplasmic reticulum
membranes helps to maintain
the low concentration of calcium
in the cell cytosol and also
calcium intracellular depot is
formed in the mitochondria
and endoplasmic reticulum.
Binding of two calcium ions In
Ca2*-ATPase sites, turned into
the cytosol, changes the charge
and conformation of the Ca2+-ATPase
conformation. The affinity
of the enzyme to ATP increases
and auto-phosphorylation is activated.
Binding the phosphate (P)
accompanied with the conformational
changes, Ga^-ATPase then closes
at inner side of the membrane
and opens from the outer side.
The affinity of the binding sites
to Ca2* ions decreases and they
dissociate from the enzyme.
Auto-de-phosphorylation is
activated by the Mg2+ ions (-).
Ca2+-ATPase lost its phosphate
and the affinity to the Mg2+ ions.
The enzyme conformation changes
and Ca^-ATPase gets back to its
initial state.
Membrane Membrane
outer inner
surface surface
Active symport - simultaneous
transfer of two substances in one
direction, one of which moves
against the concentration gradient
at the expense of the other moving
by the gradient concentration,
for example, Na*- dependent ( )
glucose transport into the gut ( )
Active antiport - transfer in
the opposite directions, one
of the substances moves against
the gradient concentration owing
to the other moving by the gradient
concentration, for example, Na41/ i
dependent Ca2+ transporter
in the saliva cells( )
© o o> @
Fig 4.8a Types of membrane receptors. Membrane receptors can De divided into tfiree groups:
Receptors: 1 — containing the subunit that binds a signal molecule and ionic channel, ex. acetylcnotine
receptor on synaptic membrane; 2 — snowing catalytic activity after binding a signal molecule, ex.
insulin receptor; 3,4 — transducing a signal to the adenylyl cyclase (AC; enzyme or phospholipase
0 (PLC) with me help of membrane G-proieins, ex. different epinephrine and acetylcholine receptors
and other primary messenger receptors
Active
PKA
Fig. 4.9. Adeiiylyl cyclase pailiway. PKA — protein kinase A, R — regulatory subunit, C— cataliyic
subunit
Binding of the as-GTP to the regulatory site of adcnylyl cyclase changes the
conformation of the enzyme and turns it to the active state, which leads to increased
formation of the second messenger — 3 \5’-cyclic adenosine monophosphate (cAM P)
from ATP:
200 Cnapief 4. Structure and functions of biological membranes. Cell signaling pathways
Asa result of this reaction, the cA MP concent ration in the cell drops consequently.
This triggers cleavage of the cAMP4R3 complex and increases the affinity of R- and
C-subunits, and inactive form of FKA (R3C3) thereby is formed.
One of the hormones, acting through the adcnylyl cyclase system. — epinephrine,
increases the heart rate, via stimulation of the cAMP rise in the cardiomyocytes.
Natural phosphodiesterase inhibitor — calle inc (coffee), decreases the rate of cAMP
cleavage, so its concentration rises in the cell. Therefore, taking high amount of
caffeine causes symptoms similar to the epinephrine action. There is list of drugs,
cardio stimulants, which arc inhibitors of phosphodiesterase.
Phosphoprotein phosphatase turns phosphorylated proteins and enzymes into their
dcphosphorylatcd form, so their conformation is changing, as the rate of the processes
they are involved in. As a result, the system regains the initial state and is ready to get
activated when the hormone would bind the receptor again. Thus, the balance between
the hormone blood concentration and the intensity of the target cells response is well
regulated.
mRNA
i
Specific proteins
I
Changes in uKiabolism
in target cells
Fig.4.10a Regulation of me genes expression by adenylyl cyclase system. FKA — protein kinase
A, R-regulatory subunits of FKA, C — catalytic subunits of FKA, ORES — CAMP response element
binding protein, CRE — CAMP response element, P — phosphate, AC — adenylyl cyclase
Hwimorw
Calmodulin
CalmoduSin
Calmodulin n
Ca7--calmodulin
-la pendent
PK inactive
Calmodulin
Ca7-calmodulin
dependent
F'K aclive
t-OH 4J®
ATP ADP
This signaling system includes the following proteins: calmodulin, protein kinase
C, Ca3' -depen de nt protein kinases, regulated Ca3' channels ofendoplasmic reticulum
(ER) membrane, Ca2 -ATPasesof the cell and mitochondrial membrane.
The binding of the activator (i.c.T its primary messenger) to the receptor (R) of
phosphatidylinositol 4\53 — bisphosphate (PIP.) signaling pathway initiates the
conformational changes in the receptor ( Fig. 4.1I). Therefore- the receptor affinity
to GW(-protein elevates. Then, binding of the complex primary messenger-receptor
to the Gp|f-GDP diminishesapif-subunit affinity to GDP along with the increasing
204 Cnapief 4. Structure and functions of biological membranes. Cell signaling pathways
affinity to GTP. Asa consequence, GDP is substituted for GTP in the binding site of
aPI< -subunit. This is followed by conformational changes in ap|(.-subunit and loss of
its affinity to py subunits, so the G?ir-proiein dissociation is taking place. The aJltc-
GTP subunit laterally drifts through the membrane to the enzyme phospholipase C.
Interaction of a^.-GTP with the binding site of phospholipase C changes the
conformation and activity of the enzyme, and the elevation of hydrolyzis rate of
specific membrane phospholipid — phosphatidylinositol 4,5-bisphosphate (P1P3) —
is taking place (Fig. 4.12).
(IP3)
charged «heads». DAG, sitting al the specific sites of protein kinase C, potentiates
its affinity to Ca2 even more. Eventually, the active state of PKC is formed al the
membrane inner surface: |PKC-Caj4-PS-DAG|. Now, being in that complex, it can
phosphorylate specific enzymes.
The PIPn system has a short-term activity, and right after the cell response the
phospholipase C, protein kinase C and Ca3'-calmodulin-dependent enzymes arc
getting inactivated. The aP[f-subunit in complex with GTP and phospholipase C
shows enzymatic (GTP-phosphatase) activity and hydrolyzes GTP to GDP. Being
bonded to GDP. ct?1(,-subun it loses its affinity to phospholipase C and gets back to its
inactive state, incorporating again into the complexafiy-GDP (Gprj£-protein).
Dissociation of aJlfC-GDP from PLC inactivates the enzyme, so the hydrolyzis of
PIP..terminates. Increased cytosol Ca2' concentration activates Ca2'-ATPascs in the
ER and plasma membranes, which pumps out calcium from the cytosol. The Na7
Ca2 - and H'/Ca2 -transporters arc also taking pan in this process, working by the
active antipon mechanism. Since Ca2’ concentration is decreasing, the dissociation
and inactivation of Ca2 '-calmodulin -depen de nt enzymes is taking place, as well as the
loss of the PKC affinity to the membrane I ipids and its activity loo.
The IP, and DAG, formed during the system activation, can interact again and
turn into the PIP-,.
Phosphorylated proteins and enzymes arc being transformed into their
dcphosphorylatcd state by the phosphoprotein phosphatase, which changes its
conformation and activity; thus, the whole signaling system returns to its initial state.
Fig. 411 Activation of the insulin receptor. Phosphodiesterase transforms cAMP into AMP and cGMP
into GMP. Fnospnoprotein phospnaiasedephosphoryiates specific phosphorylated proteins. GLUT4-
giucose transporter in me glucose-dependen t tissues (adipose and skeletal muscles). Tyrosino-protein
phosphatase depnosphoryiates p-subunits of the insulin receptor
4.3. TransmemDrarte Signal Transduction 207
OH
[MEK kinase]i ■ [ M E k kinase 1
7\—
ATP ADP
<e>
. MAP kinase ■ [ M AP kinase I
ATP^-J /
AC
ADP
Fig. 414 MAP-kinase pathway. Tne receptors of such kind are activated Dy epidermal growth factor
(EGF), nerve growth factor (NGF) and other growth factors. Grb2 — protein, interacting with growth
factor receptor (growth receptor binding protein); SOS (GEF) — GDP-GTP exchanging factor (Ras
subfamily of guanine nucleotide exchanging factors); Ras — G-protein (guanosine triphosphatase
family); Raf kinase — in its active state phosphorylates MEK-kinase; MEK kinase — MAP kinase
kinase 'mitogen-activated protein kinase kinase); WAP kinase - mitogen-activated protein kinase
4.3. TransmemDrarte Signal Transduction 209
Receptors will] guanylyl cyclase activity arc also catalytic receptors. Guanylyl cyclase
catalyzes the formation of cGMP from GTP, which is one of the most important
second messengers in the intracellular signal transduction (Fig. 4.15).
cyclase
E-OH
Fig. 4.15. M emu ran e bound guanylyl cyclase activity regulation. Membrane bound guanytyl cyclase —
transm em brane glycoprotein. Biding site of a signal moiecute is located in me external ligand-bounded
domain. The internal catalytic domain manifests tne catalytic activity in result of its activation
There arc two types of guanylyl cyclase receptors. One type is a mem brane
spa lining receptor in the plasma membrane with the external binding site for the
primary messenger atrial natriuretic peptide (AN Pl, regulating fluid homeostasis in
the organism. The other type of guanylyl cyclase receptor exists in the cytoplasm and
is a receptor for nitric oxide (NO), which is a neurotransmitter (neurohormone). NO
is a lipophilic gas that is able to diffuse into the cell. Thus, this receptor is an exception
to the rule that intracellular receptors arc gene transcription factors.
As primary messenger binds to the receptor, the guanylyl cyclase is getting activated
and catalyzes the transformation GTP to cyclic guanosine-3 \5’-monophosphalc
(cGMP) — a secondary messenger. The concentration of the cGMP in the cell is
getting higher. Molecules of cGMP can. bind reversibly to the regulatory sites of
the protein kinase G (PKG), which consisted of two subunits. Four molecules of
cG M P change conformation of an activity of the enzyme. Now active PKG catalyzes
phosphorylation of certain cytosol proteins and enzymes. cGMP — elevating drugs or
drugs releasing NO (such as glycerol trinitrate I arc used to treat a variety of disorders,
such as angina pectoris because the final response for NO is relaxation of the arteries,
erectile dysfunction (drugs inhibiting cGM P phosphodiesterase), heart failure (using
synthetic natriuretic peptide).
210 Cnapief 4. Structure and functions of biological membranes. Cell signaling pathways
Steroid Hormone
Cytosof
Shaperon
intracellular
receptor
Nucleus
Complex
Honrone-Receptor
binds to enhancer
and activates specific
genes transcription
mRNA
♦
Specific proteins
♦
Metabolic changes
in the targeted cells
y
Fig. 116. Hormonal signal transduction via intracellular receptors
212 Cnapief 4. Structure and functions of biological membranes. Cell signaling pathways
Review tests
L Choose the correct answer.
<rs-su burnt of the G-protein, bounded to GTP, activates:
A. Receptor.
B. Protein, kinase A.
C. Phosphodiesterase.
D. Adenylyl cyclase.
E. Protein, kinase C.
2. Match lhe figure with the letter.
Function:
A. Regulates the catalytic receptor activity.
B. Activates phospholipase C.
C. Transforms the protein kinase A into its active form.
D. increases Ca3' concentration in the cytosol.
E. Activates protein kinase C.
Secondary messenger:
1. cAMP.
2. Ca2'.
3. IP3.
3. Match the figure with the let ter.
Function:
A. Ability to laterally diffuse in the membrane bilayer.
B. Binds the enhancer in the complex with primary messenger.
C. Shows enzymatic activity when binds to primary messenger.
D. Can bind G-protein.
E. Interacts with phospholipase C during the signal transduction.
Receptor of:
1. Insulin.
2. Epinephrine.
3. Steroid hormone.
4. Choose the correct answers.
The conformational lability of the protein-transporters can be influenced by:
A. Cholesterol content in the membrane bi layer.
B. Membrane electric potential change.
C. Binding specific molecules.
D. The fatly acid composition of the lipid bilayer.
E. The amount of the transported substance.
5. Match lhe figure with the Idler.
A. Ca1' channels of the ER.
B. Ca2‘-ATPase.
C. GLUT 4.
D. N'a* dependent Ca2' transporter.
E. Na'/K* ATPasc.
1. Transport Na' by the gradient concentration.
2. Act l bro ugh the facilitated diffusion.
3. Transport Na' against the gradient concentration.
4.4. Signal Transduciion Dy intracellular Receptors 213
Situational problems
1. Card io myocytes contractile rate increases as Ca2+ levels in cytosol rise.
Releasing of Ca2 to the cytosol from endoplasmic reticulum is regulated by
cAMP-dependant transporters in ER membranes. Along with that, the cAMP
concentration in cytosol is regulated by two signal molecules.— epinephrine and
acetylcholine. Epinephrine, binding to j},-receptors, elevates the concentration
of cAMP in the cardiomyocytes and stimulates the cardiac output, and
acetylcholine diminishes cAMP concentration and, therefore, depress
myocardial contractility. Explain why, using the same signal transduction
pathway, these two primary messengers cause the di Here nt cellular responses in
the heart muscle. For that:
a) draw the scheme of signal transduction for epinephrine and acetylcholine;
b) point out the di (Terences in the pathways for these two primary messengers;
c) explain the opposite biological effects of these signal molecules.
2. Researchers have discovered significant changes in. the gene of some protein,
which is a substrate for the insulin receptor. What impact on the insulin signal
transduction would such changes have? For the answer:
a) draw the scheme- of insulin signal transduction in the target cell:
b) name proteins and enzymes, activated by insulin in the target cells;
c) explain the functions of these enzymes.
J. Steroid hormone calcitriol activates an intake of dietary calcium, increasing
the amount of Ca.2' transport proteins in the gut cells. Explain the calcitriol
mechanisms of action. For that:
aidraw a general scheme of a signal transduction by steroid hormones and
explain the process;
b) name the molecules, which synthesis is activated by a hormone in the nucleus
of a target cell;
c) explain the function of these molecules in the cytoplasm of the target cell.
4. Copy Table 4.1 to your notebooks and 11 1 I i t i n.
214 C napter 4. Structure and functions of biological memo fanes. Cail sig nah ng pathways
Tatle 4.1. Adenylyl cyclase and phosptalidylinosiwI 4,,5’-Disphosphale (PlPJI signaling pathways
5. The sense of smell — is one of the possible ways to de Leet a signal from the
environment in animals, including humans. It is now acknowledged that human
can delect about 10000 types of smells (odors). Receptors, receiving the scent,
can define molecules differed by only one carbon atom or by one functional
group. The molecule of an odorous substance — odorant, binds to granule
adsorbent in a mucus composite, covering the olfactory cells in the nasal
cavity. Complex odorant-adsorbent interacts with cell membrane receptor.
The final part of this signal transduction is the opening of cAMP-depcndcnt
Na -channels in the olfactory cell membrane. This results in excitation of the
olfactory neuron and transduction of action potential via olfactory nerve to the
central nervous system. Explain the specificity of all proteins and non-protein
molecules structure, which takes part in the odorant substance transduction.
For that:
a) copy Fig. 4.17 in your notebook and add the required compounds to the
scheme;
b) draw a general structure of the membrane receptor, which interacts with the
complex odorant-adsorbent, if known that such receptors are analogs to the
P^-cpincphrinc receptors;
c) name the protein, with one of the protomers is being able to move laterally
through the membrane and Lransducc the signal to the membrane enzyme;
describe its functions;
d) name the enzyme, which, in its active form increases the cAM P concentration
in the olfactory cell; write down the reaction that is catalyzed by this enzyme;
e) make a description of cAMP-depcndcnt Na' channels functioning:
4.4. Signal Transduciion Dy intracellular Receptors 215
f) explain, how docs the system get inactivated, after being activated by an
odorant and name the enzymes and proteins involved in this inactivation
process.
Adsorbent
granules
MuCuS
4 T ■*0 Q
•
> B1 _ Channel
0
Fig. 117. The scheme of an odorant molecule Din ding to an olfactory cell receptor
Chapter 5
CATABOLISM AND CELLULAR
BIOENERGETICS
5.1. Energy Producing and Energy Utilizing Reactions. The ATP-AD P Cycle.
Specific and Common Catabolic Pathways
52. Common Pathway for the Final Oxidation of Metabolic Fuels
53. Tricarboxylic Acid cycle (TCA cycle)
5.4. Mitochondrial Electron Transport Chain and Oxidative Phosphorylation.
Thermogenesis
5.5. Oxygen Toxicity
0 0
HO
‘N
f Adenine ]
OR OR:
[ Ribosg
The total quantity of ATP in the human body is about 0.2 moles, which is
approximately 100 g and is enough to support cellular demand only for few seconds.
Therefore, the cells need a con slant ATP supply. In adult humans, approximately 100
to 150 niotes (equivalent of 50 to 75 kg) of ATP arc synthesized daily. ATP is synthesized
mainly in oxidative metabolism and requires on average 27 moles of oxygen per day.
A human will typically use up his or her body weight of ATP over the course of the day.
In cellular energy metabolism, each ATP molecule is recycled (undergoes a process
of hydrolyzisand synthesis) 500-750 limes a day, which characterizes the intensity of
ATP metabolism in the body.
The ATP-ADP cycle as a process by which cells can store and release energy for
short-term use. The ATP-ADP cycle is the continuously ongoing energy recycling,
through oxidative phosphorylation of -flow energy> adenosine diphosphate (AD Pl to
*high energy* adenosine triphosphate (ATP), and the subsequent hydrolyzis of ATP
molecules back to A DP. in which energy stored in high-energy phosphate bond of
ATP is released and used in various cellular needs (Fig. 5.2).
AG = AH—T x AS.
where T is the absolute temperature.
According to the second law of thermodynamics, for systems reacting at fixed
temperature and pressure, there is a tendency to achieve a minimum of the Gibbs
free energy A chemical reaction will proceed spontaneously if the AG is negative and
is accompanied by a reduction of free energy. These reactions arc exergonic and
thermodynamically favorable. If an absolute value of AG is high, the reaction goes
to the end and is considered as non-reversible. If AG is positive, the reaction is not
thermodynamically favored and will not proceed spontaneously {endergonic reaction).
In this case, reaction can only proceed with energy inputs from other energy sources,
such as the Sun in photosynthesis, or anol her exergonic reaction. I n biological systems,
thermodynamically unfavored reactions can only proceed if they arc «couplcd» with
exergonic reactions. Such reactions arc energetically coupled. The majority of such
reactions proceed with the participation of ATP. which acts as a coupling factor.
Metabolic pathways The Sun is the source of all the energy in the living cells.
The Sun’s energy reaches Earth and is used for photosynthesis by plants and algae to
produce glucose, which can be used to make ATP or be stored as starches. Plants then
arc consumed by herbivorous animals and insects, which can break down the starch
for energy or store it as glycogen, fats, or proteins. Carnivorous animals can then cat
these animals so that they can also produce fat, glycogen, or proteins in a chain of
biochemical reactions — metabolic pathways (Fig. 5.3).
Metabolic pathways can be categorized into two major categories. Anabolic
pathways build complex molecules from simpler ones and need an input of energy.
Photosynthesis in plants, fatty acid synthesis, or biosynthesis of proteins from amino
acids arc examples of anabolic reactions. Energy from the Sun is fixed and stored
in chemical bonds of energy-rich complex molecules, such as glucose, amino acids
and fatty acids (metabolic fuels). Therefore, anabolism is the biosynthesis phase of
metabolism in which smaller simple precursor is converted to the large and complex
molecules of the cell.
Catabolic pathways involve the breakdown of complex molecules into simpler ones
and typically release energy. Metabolic fuels arc metabolized in a series of oxidation
reactions in specific catabolic pathways and energy is released (Fig. 5.3, Table 5.1).
Some of this energy is dissipated as heat and used to maintain a constant temperature
of human body. The rest energy can be used to synthesize ATP. the ultimate energy
currency in the cell. Therefore, catabolism is a degradation phase of metabolism in
which large molecules are converted into smaller and simpler molecules. Catabolic
pathways typically consist, of two phases: first, hydrolyzis reactions, in which the large
molecules are broken down to the smaller molecules with the release of energy (the
examples arc breaking down of polymers into monomers during the food digestion:
starch into glucose, proteins into amino acids or triglycerides into fatly acids): and
second, oxidation reactions that involve the removal of hydrogen or electrons from
the product molecules of the first phase reactions. Anabolic and catabolic pathways
are precisely coordinated through multiple feedback and control mechanisms that
include transcriptional regulation, enzyme modifications, allosteric interactions,
com part men ration and metabolic specialization of organs.
5.1. Energy Producing and Energy Uhlizing Reactions. TH e ATP - ADP Cycle. Specific... 21 g
E
PROTEINS STARCH .2
I
Acetyl~CoA
Oxaloacetate s. Citrate
I
>1
<0
Malate Isocitrate B
TCA cycle
2h
2.
2H Fumarate (i-Ketoglutarate
ATP ADP CO2*t^------------- 2H
2
Succinate - - SuocinnyWCoA
s
NADH c
E
NADH dehydrogenase ADP + Pi 0
2H £
(Complex I) ATP E
0
o
Fig. 5.3. Conversion of energy stored in metabolic fuels into chemical energy of ATP in a chain ol
biochemical reactions of food digestion, specific metabolic pathways, tricarboxylic acid cycle (TCA
cycle) and electron transport chain (ETC)
TaUle 5.1. ATP formation in me aerobic oxidation of glucose via glycolysis. me pyruvate dehydrogenase
complex reaction, me TCA cycle and oxidative phosphorylation (Lehniisger, Principles of Biochemistry)
Total 30-32
■ Calculated as 2.5 ATP per NADH and 1.5 ATP per FADH2. A iwgative value indicates consumption.
1 This number is eilher 3w5, depending on die mechanism used to shuttle NADH equivalents From the cytosol to the
rndochondriil matrix; tec me malate-aspartate shuttle antf me glycerol 3-ptiosphate shuttle.
Fig. 5.4. Structure of pyruvate dehydrogenase complex (PDH). licensed under the Creative Commons
222 Chapter 5. Catabolism ana ceituiar bioenergetics
A
Pyruvale
TPP, dehyckogenasa
lipoate, complex
S-CoA
. fad irreversibly
Pyruvate dehydrogenase converts
pyruvate to
complex (Ei + E2 + Eg}
CH3 ACfltyl-CoA,
a main fuel for
Pyruvate (Acetyl-CoAj the TCA cycle
A G n = -33.4 kJ/mol
Lip = Lipoamide
dehydrogenase complex
COO- COO“
H2O CoA-SH
CHg—C ■+■ O=C
. _ — COO" < J.
Citrate synthase
S-COA
ch2— coo- CH2~ COO"
[Oxaioacetate] C trate |
Acetyl-Co A
G’ = -32.2kJ/moi
H COO
[as-Aconitatel [ Isocrtfate-I
£ G“= T3.3k*Mnwi
Fig. 5.8. Formation of isocilrate in the second reaction of the TCA cycle
-
COO" COO- coo- COO"
1 1 1
. 2 fl COa GHj. CHn
r z
1
H— C— C. z V }
—N, ,Z „
I
H—C
fsotitraie Ub. °-
HQ—C—n defrydrogenase c=o
< 1 1
✓V ® Zvz © /v
Fig. 5 J. Oxidative decarboxylation of isocitrale and formation ot ti-keto glularate in the third reaction
of the TCA cycle
n-Ketoglutarale 1
-8
uehydfooenase C
complex
fa-Ketoglutarate) [ Succinyl-CoA )
Fig. 5.10. Conversion of a-ketogluiarate to succmyl-CoA and formation of NADH in me fourth reaction
of the TCA cycle
226 Chapter 5. Catano lism ana cellular bioenergetics
[n the fifth reaction of the TCA cycle, the succinyl-CoA is convened into
succinate (Fig. 5.11). The reaction is reversible and is catalyzed by succinyl-CoA
synthetase (SCS). SCS is the only enzyme- in the TCA cycle that catalyzes a reaction
in which a nucleoside triphosphate (GTP or ATP) is formed by substrate-level
phosphorylation. In the succinyl-CoA synthetase reaction, the high-energy bond
between HS-CoA and the succinyl group is hydrolyzed, and the energy released js
enough for synthesizing GTP from GDP + {Ph The GTP. from the energetic point
of view, is equivalent to ATP. In fact, GTP can transfer the (P) group to A DP to form
ATP. Since ATP can be produced from this reaction, without the participation of the
respiratory chain, this process is called Substrate Level Phosphorylation, in contrast
to the Oxidative Phosphorylation, where ATP synthesis uses energy generated in the
Electron Transport Chain.
COO’
CH?
COO’
Substrate-/eve/
p/rasp/Nyy/abon
COO" COO"
I Succinate I
H-—C-—H dehydrogenase H— C
I
H—G—H < "A ’ C —H
I -Ad FADHjj I
COO" COO"
(Succinate) (Fumarate]
Fig. 5.12. Sixth reaction ol TCA cycle - convers ion of s uccinate to fumarate by succinate dehydrogenase
The seventh stop of the TCA cycle, the reversible hydration of fumarate to malate
is catalyzed by fumarase ( Fig. 5.13). There arc two forms of fumarase, mitochondrial
and cytosolic. The mitochondrial isoenzyme is involved in Lhe TCA cycle, and the
cytosolic isoenzyme is involved in Lhe metabolism of amino acids.
h cc
II Fumarase
C*- OH
-OOC ^ \
H
£ G>=-3.B kJ/mol
The final, eighth reaction of TCA cycle is the reversible conversion of malate to
oxaloacctale that is catalyzed by malate dehydrogenase (MDH ). This reaction occurs
through the oxidation of hydroxyl group on malate and reduction of NAD* with
formation of NADH (Fig. 5.14). There arc two major isoforms of MDH in eukaryotic
cells. M DH, is found in the mitochondrial matrix, participating as a key enzyme in the
citric acid cycle that catalyzes the oxidation of malate. The other, MDH), is found in
the cytoplasm and participates in the ma late-aspartate shuttle.
COO" COO'
I MAD* NADH - H* I
HO—C—H O—*C
I
H~ C—H H— G—H
I
Malate
I I
dehydrogenase
GOO" COO"
L-MaJate ] [ Osalciacetate)
Fig. 5.14. The oxidation of malate Id oxaloacetate is coupled with NADH production and is catalyzed
by malate dehydrogenase (MDH)
228 Chapter 5. Catabolism ana ceituiar bioenergetics
The cellular signaling system determines whether the amphibolic pathway will
function as an anabolic or catabolic mode by enzyme-me dialed regulation at a
transcriptional and post-transcriptional levels. As many reactions in amphibolic
pathways are freely reversible or can be bypassed, irreversible steps that facilitate
their dual function, arc necessary. The pathway will use a different enzyme for each
direction for the irreversible steps, allowing independent regulation of catabolism and
anabolism.
Overall, at the end of glycolysis, we have two pyruvate molecules that have plenty
of extractable energy. From a single molecule of glucose two molecules of pyruvate
arc generated and then two molecules of acetyl-CoA are produced along with two
molecules of NA DEI and two molecules of CO,. Two molecules of acetyl-CoA enter
the TCA cycle. In a single turn of the TCA cycle, three molecules of NADH and one
molecule of FAD H, a re generated from a single molecule of acetyl-CoA. Additionally,
one molecule of ATP or GTP is generated in the substrate-level phosphorylation
reaction (fifth reaction of the TCA cycle). Electrons of reducing equivalents NADH
and FADH,arc then transferred to the electron transport chain, where more ATP
molecules arc synthesized in the oxidative phosphorylation reactions.
sh2+ko3*s+h.o,
where S is a metabolic substrate.
The energy released during the oxidation, reactions is partially spent on the
phosphorylation of ADP to form ATP. Overall, the body converts about 40% of
the energy released during oxidation into the energy of high-energy ATP bonds. A.
significant part of energy is dissipated in the form of heat.
The dehydrogenation reaction and the way by which the released energy is convened
to chemical bonds of ATP arc energetically coupled reactions. A set of complexes that
transfer electrons from electron donors to electron acceptors via consecutive redox
reactions is called an electron transport chain (ETC), or a respiratory chain (RC)
(Fig. 5J5 A and B). A process of synthesis of ATP from ADP in the ETC is called
oxidative phosphorylation
230 Chapter 5. Catabolism ana cellular bioenergetics
ATP
Matrix ADP+P,
itYier
mwctKh-Kliial
menbcjie
jniermerntrane fll-T
Sfiace ------------- 1--------
FtaUEtione Aniirr.yCtn A
AmyQI
Inteffnentwarte space
Fig. 5_15. A Electron transport chain (ETC) and oxidative phosphorylation - simplified diagram.
Fe-S — iron sulfur centers of proteins. nH+ indicates that number of protons are pumped from the
matrix to the cytosol side. The return of protons through me ATP syntnase pore drives ATP synthesis,
mnibitors of ETC complexes are framed and sites of their action are shown (T). B. More detailed
diagram of milocrmdriai electron transport chain: Com ptexes I, IIJII an d IV are components of electron
transport chain located in the inner membrane of mitochondria. Complexes I, ill and IV transport
protons (H'J across inner mitochondrial membrane and generate a transmembrane electrochemical
gradient lAjdTt a motive force for ATP synthase. Complex ll — succinate dehydrogenase does not
participate in generation of electrochemical gradient Cytochrome c(Cyt c) is a low molecular weight
heme-containing protein, possessing nigh mobility in the lipid layer of the mitochondrial membrane.
Coenzyme Q (Q) is a non-protein electron carrier. FeS proteins contain n on-heme iron and are part
of comptexes I, II and ill. ATP synthase sometimes is called complex V and catalyzes ATP synthase
5.4. Mitoctiondrial Electron Transport Chain and Oxidative PnospriDelation. Tnermogenesis 231
A B
CyS — S S - Cys
<*-(§}„ ..Xs)-Cys
CyS-(s)'' ’’(s)-GyS
CyS - S rS — CyS
+ e-
Fe3+ " Fe2+
- e-
fig. 5.16. Structures of iron-suilur center A — Fe-S center; B — Fe2-S2 center; C — Fe4-S4
center. Cysteine residues (Cys) are involved in binding of Fe-S clusters in the protein. D —
oxidation and reduction Fe. Iron atoms can be in oxidized (Fe3*) or reduced (Fe?+) states
Fig. 5.17. Chemical structure of coenzyme Q wim 5-10 isoprene repeats in the side cnain
Oh oh
NAD4-
FAD
Fig. 5.18. Coenzymes o! dehydrogenases. Structural formulas ol coenzymes NAD FAD and FMN
234 Chapter 5. Catabolism ana ceituiar bioenergetics
Flavin groups in FAD and FMN arc derived from riboflavin (vitamin U3)
(Fig- 5.18). The flavin group is capable of undergoing oxidation-reduction reactions
and can. accept electrons. Reduction is made with the addition of hydrogen atoms io
specific nitrogen atoms (Fig. 5.20).
Reduction
Oxidation
Reduction
Oxidation
f FAD or FMN]
Fig. 520. The redox reachoils of nicotinamide adenine dinucleotide (A) and flavin moieties of FAD
and FMN (B)
NADH-FMN-N.-N^-N.-N.-N^-N^-N.-CoQ,
where Nx is a label for iron sulfur clusters.
236 Chapter 5. Catabolism ana cellular bioenergetics
Succinate Fumarate
fad fadho
SDHA
3x[F&-SJ
Matrix
SDHB
SDHC SDHD
Inner mitochondrial
membrane
rn ' r. V'V'T1 IT Heme
inrermembrane space
Fig. 522. Mitochondrial Complex EL Schematic diagram of complex II m inner mitochondrial membrane.
SDH has two trans membrane subunits C. D and two units A, B, which are facing mitochondrial matrix
The process by which the electrons arc transferred from the ubiquinol to
cytochrome c is known as the Q cycle. This cycle actually consists of two half-cycles.
In the first half-cycle. a ubiquinol molecule (CoQHJ attaches onto complex III and
transfers the two electrons to the complex. One of these electrons moves onto the
Rieskc subunit, then onto cytochrome c^and finally onto cytochrome c. Cytochrome
c, unlike ubiquinone, can only carry a single electron at any given time. The other
electron that comes from CoQH, follows a different pathway and moves through the
heme groups of cytochrome b and onto ubiquinone (CoQ) to form a partially reduced
unstable species called a semiquinone (CoQH-).
238 Chapter 5. Catabolism ana cellular bioenergetics
2H+
Fig. 523. Mitochondrial Complex III Sen emetic diagram of complex in in inner mitochonariai membrane
and electron flows in me Q cycle. As a result of Q cycle. 2 protons are taken from the matrix, 4 protons
are released into me imermemdrane space and two electrons are passed to cytoenrome c. Qo ana
Qi — Ubiquinol (Qty and ubiquinone (Q) Dinging sites of complex III, respectively
The two protons that were originally attached to ubiquinol arc transferred into the
intermembrane space. In the second half-cycle of the Q cycle, another ubiquinol
attaches onto complex 111. Upon binding, the two protons arc moved into the
intcrmcmbranc space and the two electrons follow the same pathways as before. The
electron that travels through the Ricskc subunit eventually ends up reducing a second
cytochrome c while the other electron travels onio the semiquinonc to form a fully
reduced quinone — ubiquinol (the ubiquinone must take up two protons from the
matrix to form the ubiquinol). Therefore, a single Q cycle reduces two cytochrome
c molecules, forms a single ubiquinol molecule, pumps four protons into the
intcrmcmbranc space and takes up two protons from the matrix.
Complex (V (cytochrome c oxidase). Complex IV is the last electron acceptor in
the ETC and is involved in the reduction of O-, to H20. Complex [V is a mu hi meric
complex (Fig. 5.24.), subunits of which arc encoded both nuclear and mitochondrial
genome. Al least 20 subunits arc present in human complex IV. Three subunits of
Complex IV arc encoded by mitochondrial genome. Composition and stoichiometry
of subunits and prosthetic groups depend on tissues and cell types. Additionally, over
30 of accessory chaperone proteins arc required for the assembly of functional complex
IV. Complex IV catalyzes the transfer of electrons from reduced cytochrome c to the
final acceptor of electrons, On, in a process that is coupled to proton translocation
and formation of a proton gradient across the inner mitochondrial membrane. The
summary reaction can be expressed as:
5.4. Mitoctiondnal Electron Transport Chain and Oxidative PnospriDelation. Tnermogenesis 239
4CytCM
intermemtsrane space
4H*
Fig. 5.24. Mitochondrial Complex IV. Schematic diagram of complex IV in inner mitochondrial
membrane and electron flows
60
Comptexes I II III IV
Fig. 5.25. Redox potentials, and The corresponding free energy levels, of electron carriers in me
respiratory chain (complex HV). Blue arrows show electron flow and red arrows snow proton
pumping. The TCA cycle provides NADH, wnicn is reduced to NAD + H* in complex I. The TCA cycle
also provides FADH?I and tn is is reduced to FAD + H* in complex n. The glycerol 3-phosphate snuttie
{G3P] shuttle provides FADH.. to coenzyme Q (CoQ). Reduced oxygen, which recombines wim protons
to yield water, is the end product of respiration
The energy generated by the flow of electrons through the respiratory’ chain,
is used for the coupling with phosphorylation of ADP. These two processes arc
interdependent: oxidation cannot proceed in the absence of ADP. The ratio of
oxidation and phosphorylation is determined by the P/O ratio, which shows how
much Pi is used for the formation of ATP during the conversion of one grant-atom
ofO, to H,0 (the number of moles of phosphorylated ADP per 1/2 mole of oxygen).
The P/O ratio is called the coefficient of oxidative phosphorylation and depends on
the entry point of reducing equivalents in the electron transport chain. For example,
for substrates oxidized by NAD-depcndent dehydrogenase, P/O =2.5, since there
arc three sites in the respiratory chain, where electron transfer is associated with
ATP synthesis. Not all substrates transfer electrons and protons to NAD; some arc
oxidized by FA D-de pendent dehydrogenases, which transfer protons and electrons
directly eo ubiquinone, bypassing complex I. In this case, P/O = 1.5. In reality, the
phosphorylation coefficient is always less than the theoretical value. because part of the
energy that is released during the transport of electrons is not spent on ATP synthesis,
bin to transfer substances through the mitochondrial membrane or disappeared as
heat.
5.4. Mitocnoodfial Electron Transport Chain and Oxidative PhospriDelation. Thermogenesis 241
Fig. 5 26 Mitochondrial ATP synthase, or complex V. Schematic drawing of ATP synthase. ATP synthase
consists of two functional domains, Fj and Fr F, comprises different subunits (three u, three p, and
one y, a and c) and is situated in the mitocnondriai matrix. Fd contains subunits c, a. d, a, F6, OSCP and
the accessary subunits e, f, g and A6L F, subunits y, 6 and c constitute me central stalk of complex
V. Subunits D. d, FE and OSCP form the peripheral stalk. Protons pass from die intermembrane space
to me matrix through Fa, which transfers the energy created by the proton electrochemical gradient
to F., where ADP is phosphorylated to ATP. One p subunit is taken out to visualize the central stalk
2. Generation of ATP
Fig_ 527. Mechanism of ATP synthesis Uy ATP synthase (complex V}. Catalytic cycle of ATP synthase
involves 3 phases that occur in three different catalytic centers: 1 — binding of ADP and inorganic
phosphate (Pi); 2 — phosphorylation of ADP and generation of ATP; 3 — release of ATP
center prevents the transport of electrons from cytochrome c to oxygen, and as a result,
causes asphyxiation of cells, meaning that the cell can no' longer aerobically produce
ATP for energy and cell death, occurs rapidly. Cyanide is one of the most potent and
rapidly acting poisons known. The central nervous system is the primary target for
cyanide toxicity.
Nitric oxide (NO) also binds to the cytochrome c oxidase bi nuclear center, and
under physiological conditions, NO is thought to be an endogenous modulator of
oxidative phosphorylation.
II igher concentrations of molccular<JKygcn are needed to compensate for increasing
inhibitor concentrations. Oxygen therapy is often used in the emergency departments
as an. antidote for cyanide and carbon monoxide poisoning. Other ligands, such as
nitric oxide and hydrogen sulfide, can also inhibit cytochrome c oxidase by binding to
regulatory sites on the enzyme, reducing the rate of cellular respiration.
OLigomycin is an antibiotic that inhibits ATP synthase (complex V) by blocking its
proton channel ( F^ubunit), which is necessary for oxidative phosphorylation of ADP
to ATP. The inhibition of ATP synthesis would also slop electron transport chain.
Inhibitors of ETC complexes and the sites of their action arc depicted on the
Fig. 5.15, panel A.
ADP/ATP translocase is specifically inhibited by atraclylosi.de and bongkrckic
acid. The high affinity (Kd in the nano molar range) makes each inhibitor a deadly
poison by obstructing cellular respiration.
Hvpoenergetic state. Disruption of any stage of metabolism, leading to the
cessation of ATP synthesis, is fatal for the cell.
The state in which the synthesis of ATP is reduced is combined with the term
*hypoencrgctic*. The causes of hypocnergetic states can be fasting, deficiencies of
vitamins B,, PP+ Bp hypoxia. Hypoxia can occur with a lack of oxygen in the inhaled
air; in pulmonary diseases and impaired pulmonary ventilation; with circulatory
disorders caused by heart disease, spasm and vascular thrombosis, blood loss. The
causes of hypoxia can be hereditary or acquired defects of hemoglobin.
Genetic disorders caused by deficiencies of the TCA cycle and ETC. The TCA cycle is
crucial to the metabolism of living cells and any significant disruption of these reaction
systems causes an energy deficit in the cell. The TCA cycle enzymes arc all nuclcarly-
cncoded. Although very rare, inborn errors in the TCA cycle genes may be severe or
even be incompatible with life.
Conversion of pyruvate to Acetyl-CoA in PDH reaction and conversion of
a-keioglutaratc to succinyl-CoA in the fourth reaction of the TCA cycle requires
coenzyme thiamine (vitamin Bp. Thiamine is also important in the production of
NAD PH via the pentose phosphate pathway (section 6.4) and breakdown of branched
amino acids (section 8). Thiamine is a water-soluble vitamin that is not actively stored
in the body, and. therefore, must be obtained in the diet. Thiamine deficiency can
cause numerous disorders, such as Wernicke-KorsakofFs syndrome and beriberi
disease.
Aconitase deficiency is characterized by myopathy with severe exercise intolerance
in patients. Mutations in isocitratc dehydrogenase (1DH3) gene are associated with
various brain tumors and acute myeloid leukemia (AM L). Fumarate deficiency is
associated with severe neurological abnormalities, poor feeding, failure to thrive.
Recent studies suggest the importance of fumarase in DNA re pair path ways, suggesting
244 Chapter 5. Catabolism ana ceituiar bioenergetics
its tu mor-repress! ng. role. Citrate synthase deficiency may cause worsening of tumor
malignancy.
Genetic defects of ETC, Defects in ETC can be caused by mutations in. cither
mitochondrial or nuclear genes and cause mitochondrial diseases. Genetic diseases
affecting mitochondrial oxidative phosphorylation system are referred to as Oxphos
diseases.
Genetic defects affecting complex I include various neurodegenc native and
neuromuscular diseases, and Leigh syndrome.
Mutations in. genes that encode complex 11 subunits arc causative of various
disease, including Leigh syndrome, mitochondrial encephalopathy, optic atrophy,
numerous tumors (mostly benign), hereditary paraganglioma and hereditary
plicoc h romocy tom a.
Mutations in complex ill genes manifest as exercise intolerance, Ujomstad
syndrome and the GRACILE syndrome, which in neonates are lethal conditions.
Genetic defects in genes encoding cytochrome c oxidase subunits result in severe,
often fatal metabolic disorders. Cytochrome c oxidase deficiencies usually manifest in
early childhood and affect predominantly tissues with high-energy demands (brain,
heart, muscles). The vast majority of cytochrome c oxidase disorders arc linked to
mutations in nuclear-encodcd cytochrome c oxidase assembly proteins or chaperones.
Mitochondrial complex V deficiency can cause a wide variety of multi-organ
symptoms, particularly the nervous system and the heart. The disorder can be life-
ill rca ten ing in infancy or early childhood. Affected individuals may have feeding
problems, slow growth, low muscle tone (hypotonia), extreme fatigue (lethargy), and
developmental delay. They tend to develop elevated levels of lactic acid in the blood
(lactic acidosis), which can cause nausea, vomiting, weakness, and rapid breathing.
High levels of ammonia in the blood (hyperammonemia) can also occur in affected
individuals, and in some cases, result in abnormal brain function (encephalopathy)
and damage of oilier organs.
Ihermogenesis
The synthesis of ATP molecules consumes about 40-45% of the total energy of
the electrons transferred through the center of energy, about 25% is spent on work on
the transferor substances through the membrane. The rest of the energy is dissipated
in. the form of heat and is used by animals to maintain body temperature. Additional
heat generation can occur with the dissociation or uncoupling of respiration and
phosphorylation. Uncoupling of electron transport from oxidative phosphorylation
can generate additional heat, but will inhibit the ATP synthesis, without affecting ihc
respiratory chain.
The uncoupling of oxidative phosphorylation can be biologically beneficial, ft
allows to generate heal for maintaining body temperature in newborns, in winter
animals, and in all mammals in the process of adaptation to cold. The process is
called non-shivering ihcrmogencsis. In newborns, as well as winter-sleeping animals,
there is a special tissue that specializes in a heal production through the separation
of respiration and phosphorylalion — the brown faL, or brown adipose tissue (BAT).
The BAT cells contain lots of mitochondria. The mitochondria of BAT have a
5.5. Oxygen Toxicity 245
another molecule to complete its own orbital. The stepwise transfer of elect tons to O3
results in the formation of superoxide anion (Op, then hydrogen peroxide (H3O3)T
and finally hydroxyl free radical (OH ’):
e _e.2H* e.rr e.H*
O2 H2O2^ H2O + 0H' 2H2O
The end products of this reaction sequence arc two molecules of water. The
hydroxyl radical is the most dangerous free radical because it is involved in reactions
such as lipid peroxidation and generation of other toxic radicals. Hydrogen peroxide
itself is not a free radical but is convened be the Fenton-, or Haber-Weiss reactions
io the hydroxyl radical in the presence of Fe3' or Cu . which are prevalent in cells.
In mitochondrial electron transport chain highest ROS producers arc Complex I
and Complex III. Leakage of electrons at complex I and complex 111 leads to partial
reduction of oxygen to form superoxide. Superoxide (O’) is the anionic form of O3
and is a free radical. Respiratory complex I leaks superoxide into the matrix and
respiratory complex III leaks superoxide into both the matrix and the intcrmembrane
space. Because of its negative charge, superoxide is unable to cross the mitochondrial
membrane. However, hydrogen peroxide (H3O2)t a product of SOD2 reaction (see
below), is released from mitochondria to the cytosol in proportion to the proton
potential and can be ultimately convened by enzyme catalase into H/) and ()..
Another free radical is nitric oxide (NO), which is both essential to life and toxic.
Al low concentrations, nitric oxide acts as neurotransmitter or as a hormone that
causes vasodilation. It is synthesized from arginine by nitric oxide synthases. At high,
concentrations nitric oxide binds to oxygen to form reactive toxic species that contain
both nitrogen and oxygen (RNOS). These free radicals arc also involved in damage of
the cells in chronic inflammatory diseases.
Cell damage caused by reactive oxygen species. Overproduction of ROS (oxidative
stress) causes damage to proteins, nucleic acids, lipids and is implicated in various
disease states such as atherosclerosis, diabetes, cancer, ncurodegc notation, and
aging. Oxygen radicals are also produced in certain cells during inflammation caused
by bacterial infection. To combat microbial infections, phagocytes produce toxic
oxygen radicals in a process known as the respiratory burst. The phagocytes then kill
the engulfed bacteria by the toxic radicals. Ingestion of drugs and chemicals, smog,
radiation can lead to the formation of reactive oxygen species. Damage from reactive
oxygen species often occurs during perfusion of the tissues with solutions containing
high concentration of O, as happens in patients who had an ischemic episode in which,
localized O. levels arc lowered due to blockage of an artery- and then have undergone
procedures to remove the blockage. Myocardial ischemia/reperfusion injury develops
as a result of the accumulation of reduced coenzymes NADH and FADI-iJn the
mitochondria at the absence of oxygen due to blockage of blood flow. After removing
the blockage oxygen supply to the tissues is restored as electron flow in FTC. Thus,
electrons leak easily from the ETC and more reactive oxygen species arc formed.
High levels of mitochondrial ROS activate apoptosis/autophagy pathways capable
of inducing cell death. On the other hand, small amounts of ROS arc necessary for the
cell and serve as critical signaling molecules in cell proliferation and survival.
5.5. Oxygen Toxicity 247
Reactive oxygen species and lipid peroxidation. Free radical chain reactions, which
form lipid free radicalsand lipid peroxides in membranes, make major contributions
to ROS-induced injury in the cells.
The radicals of polyunsaturated fatty acids in phospholipids of plasma and
organelle membranes are subjected to lipid peroxidation (Fig. 5.28). These free radical
chain reactions arc initialed by removal of hydrogen from polyunsaturated fatty acid
by hydroxyl radical COH'), thereby forming a lipid radical (L ). The free radical chain
reaction is propagated by reaction with O3 forming the lipid peroxy I radical (LOO ‘)
and lipid peroxide (LOO ‘). One of the main end products is malondialdehyde and
can be detected in the blood. Malondialdehyde is an active metabolite and forms the
cross-li nk between the amino groups of proteins and phospholipids thus causing the
disruption of membranes.
Fig. 528. Meefianrsm of lipid peroxidation (Young IS. McEneny J (2001). -lipoprotein oxidation and
atherosclerosis-. BiODh&m Soc Trans 29. DOE 10.1042/bst02 90358)
DNA and proteins arc also subjected to oxidative damage. In proteins, the amino
acids methionine, cysteine, arginine and proline arc the most susceptible to oxidative
damage. The damaged proteins appear in many diseases, particularly associated with,
aging. The pigment lipofuscin consists of a mixture of oxidized crosslinked lipids and
damaged proteins formed by reaction between malondialdehyde and amino acids
radicals. These pigment spots appear on the skin of the hands of elderly individuals
and arc considered as a hallmark of aging. In patients with Parkinsori s disease, the
lipofuscin granules appear in degenerating neurons as Lewy bodies.
Cellular defenses against oxygen toxicity. The cells arc protected from oxidative
damage by several mechanisms. Al first dietary antioxidants, such as vitamins E, Cd
248 Chapter 5. Catano lism ana cellular bioenergetics
flavonoids and endogenous antioxidant, such as urate, can terminate the free radical
chain reactions. The enzymatic defense against reactive oxygen species includes the
enzymes superoxide dismutase, catalase , and glutathione peroxidase.
Mammalian mitochondria arc well equipped to scavenge the superoxide generated
either internally or externally and defend cells from ROS-induced toxicity. The
manganese superoxide dismutase (SOD2) is located in the mitochondrial matrix,
where it protects mitochondrial targets against the internally generated superoxide.
SOD2 catalyzes the reaction that converts two molecules of superoxide (O1 ‘ ) to one
molecule of oxygen (O,) and one molecule of hydrogen peroxide (H^O):
2 HO3-O34H3O3,
Review test
I. Match the number on the figure with the letter.
A
ATP
Matrix ADP + P,
Maiale
T
Oligomycin
Cytosol T_ nH*
Rotenone
Amytai
Components of ETC:
A. Cytocromoxidase.
B. Cytochrome b-cl complex.
C. NADH dehydrogenase.
D. Succinate dehydrogenase.
E. Fumarase.
5.5. Oxygen Toxicity 249
2. Using the scheme of test L match she number of the enzyme on the figure of EEC with
the letter denoting the coenzyme of this enzyme.
Coenzymes:
A. Heme.
B. TMN
C. Heme, Cu 3'.
D. NAD .
E. FAD.
J. Match the figure and the letter.
Lip = Lipoamide
dehydrogenase complex
Enzymes:
A. Pyruvate carboxylase.
B. D i hydro! ipoa mid dehydrogenase.
C. Pyruvate dehydrogenase.
D. Dihydrolipoyl transacetylase.
E. Malate dehydrogenase.
4. The catabolic pathways. Match the figure and the letter
A. Common catabolic pathways.
B. Krebs cycle.
C. Oxidative phosphorylation.
D. Digestion.
E. Oxidative decarboxylation.
250 Chapter 5. Catabolism ana cellular bioenergetics
5. Match the number of the reactions wilb the letter denoting the enzyme.
SCc-A
5.5. Oxygen Toxicity 251
Situational Problems
1. Oxidative decarboxylation of pyruvate and the TCA cycle in muscles arc
stimulated by increased aerobic exercise. These processes operate only when
O2 is present, although oxygen docs not participate directly in these processes.
Explain why oxidative decarboxylation of pyruvate is activated under aerobic
conditions. For the answer:
a) describe the overall reaction catalyzed by the pyruvate dehydrogenase
complex (PDH) and its regulation:
b) outline the intermediates and enzymes of the TCA cycle;
c) explain the relationship between the reactions of PDH and the TCA cycle
and the respiratory chain.
2. A 4-year-old girl was diagnosed with thiamine deficiency and the symptoms
include tachycardia, vomiting, convulsions. Laboratory examinations reveal
high levels of pyruvate, lactate and (x-kcloglutaraic. Explain which coenzyme
is formed from vitamin B and its role in oxidative decarboxylation of pyruvate.
For that:
a) describe the structure of pyruvate dehydrogenase complex (PDH) and the
cofactors that it requires:
b) discuss the symptoms which are connected with the thiamine deficiency and
its effects on PDH and a-kc login taraie dehydrogenase complex:
c) explain the changes in the levels of mentioned metabolites in the blood:
d) name the described disease.
3. It is known that Leber hereditary optic neuropathy (LHON) is an inherited
form of blindness due to the missense mutations of mitochondrially encoded
proteins of complexes I or HI of the ETC. Explain why disorder in the structure
of these components of ETC can impair electron transport and oxidative
phosphorylation. For the answer:
a) draw the scheme of ETC and describe the structure of its components:
b) explain how the electrochemical gradient is gene rated during the transport of
the electrons;
c) cxplainc what the respiratory conLrol of the ETC and P/O ratio is.
4. Mitochondrial DNA (miDNA) encodes some proteins of the ETC. Point
mutations of mitochondrial genes can impair the electron transport chain and
oxidative phosphorylation. One of the syndrome resulting from disorders of
miDNA-cncoding protein is ME LAS syndrome (mitochondria! myopathy,
enccphalomyopathy, lactic acidosis and stroke). The earliest manifestations are
the neurological and muscular abnormalities due to the greater dependence of
brain, heart and skeletal muscle on mitochondrial ATP synthesis. Explain why
252 Chapter 5. Catabolism ana cellular bioenergetics
the levels of lactate and pyruvate arc increased in ME LAS syndrome. For the
answer:
a) discuss the respiratory control in ETC and significance of the ratio NADH/
NAD";
b) describe the reactions of oxidative decarboxylation of pyruvate;
c) explain why the disorders in the structures of components of ETC leads to the
activation of the reaction, which is catalyzed by lactate dehydrogenase.
5. To control the damage that rats may cause in the buildings, fiuoracetate (FA)
is used. After entering a cell FA is converted into very toxic fluoroacctyl-CoA.
The effect of FA was studied in the experiments on intact isolated rat hearts.
It was found after perfusion with FA the rat hearts the concentrations of the
TCA cycle metabolites were decreased. Only the level of citrate was significantly
higher than in controls.
a) describe the significance of the TCA cycle as the common catabolic
pathway and explain why blocking of this cycle leads to the lethal effect of FA:
b) indicate the reaction where the FA blocks the TCA cycle and Lhe enzyme
which is inhibited by FA:
c) explain why concentration of citrate is increased.
6. Red blood cells transport oxygen to the peripheral tissues, but they do not
produce ATP by oxidative phosphorylation.
a) indicate the final electron acceptor in the ETC;
b) indicate the location of ETC in the cells and draw the scheme of the ETC;
c) describe the mechanism of oxidative phosphorylation and explain why this
way of ATP synthesis is not possible in RBC.
7. In the brown adipose tissue (BAT) in the newborns, transmembrane proton
gradient is used not only to produce ATP, but to keep the newborns warm.
The mitochondria of BAT have a unique protein in their inner membrane —
thermogenin, also called the uncoupling protein I (UCPI). Explain the role of
UCPI in thermoregulation. For the answer:
a) draw the scheme of the ETC and explain how the electrochemicaJ gradient is
generated during the electron transport;
b) describe the mechanisms of uncoupling of oxidative phosphorylation by
UCPI;
c) compare the changes in the transmembrane proton gradient after the action of
inhibitors of lhe respiratory chain (including rotenone, antimycin A, cyanide,
carbon monoxide) and the uncoupling agents.
Chapter 6
CARBOHYDRATE METABOLISM
ai/si
l+CCli, > II C— D
/I
IIO
■OH— C“I4
CH/MH
II—C— CHI
14
fl—c—OH
[ frucinno'.
CH/JII
|STARCH |
■ Glucono
GLUT 2 Kidney
Pancreatic-cell
Serosal surface oi intestinal
GLUT 3 Brain mucosa cells (neurons}
GLUT 4 Adipose tissue
Skeletal muscle
Heart muscle
GLUT 5 Intesti rial epitlie li um
Spermatozoa
Glucose
Glucose
/iransponer
E iciracejular (bssie 1|
Glucose
Jiran^ponef
Ejciraceiular
All receptors can be found both in die plasma membrane of the celt and in the
membranes of die vesicle in die cytoplasm. The number of receptors L 2. 3 and 5 in
die plasma membrane is al most constant a nd does not depend on the insulin level. In
die absence of insulin, GLUT 4 is almost completely in the cytosolic vesicles. Insulin
stimulates the movement of vesicles to the plasma membrane and their fusion as
a result of which the receptors arc embedded in the plasma membrane (Fig. 6.5).
When insulin concentration is low receptors are returned to the cytosol.
Fig. 6.5. Stimulation by insulin of glucose transport into muscle and adipose cells
ATP ADP
Hexokinase
There arc four isoenzymes that phosphorylate glucose (designated I to IV). They
arc encoded by four different genes and exhibit different Michaelis constant (Km)
values for glucose. Most hexokinase s have a low Km for glucose, and readily take
up glucose from the blood when blood glucose concentration is low (in the fasting
state). The liver isoenzyme of hexokinase called glucokinase has a high Km. The
brain hexokinase isoenzyme has a particularly low Km for glucose. The hexokinase
isoenzyme of myocytes has a high affinity for glucose. It is half-saturated at about
0.1 mM.
Differences in Km of glucokinase from Km of hexokinase correspond to the
conditions of liver enzymes functioning. In the postabsorptivc state, the blood glucose
concentration is about 5 mMol/L. At this concentration, the glucokinase reaction rate
is about one-fifth of the maximum rate, that is, the enzyme docs not work at full
capacity. During digestion, as a targe amount of glucose enters the portal vein and
further into the liver, its concentration in. the hepatocytes may exceed 10 mMol/L.
Accordingly, the rate of glucokinase reaction increases and a significant portion of
the glucose is retained in the liver. This prevents the excessive increase in glucose
concentration in the peripheral blood during digestion. Thus, the liver effectively
re moves the great a mounts ofglucose delivered by the portal blood after a carbohydrate-
rich meal (in the fed state) minimized hyperglycemia during the absorptive state.
Pathology of carbohydrates digestion can be the result of both defects of specific
enzymes that lake part in hydrolyzis of carbohydrates in the intestine and defects of
transport of monosaccharides through the absorptive cells of the in test inc.
260 Chapter 6. Carbohydrate metabolism
Any defect in a specific disaccharidasc activity of the intestinal mucosa causes the
passage of undigested carbohydrate into the colon. Asa consequence of the presence
of these osmotically active molecules, water is drawn from the mucosa into the colon,
causing osmotic diarrhea.
Lactose intolerance can be either the result of a primary deficiency of lactase
production in the small intestine or it can be secondary to an injury of the intestinal
mucosa where lactase is normally produced. Lactose intolerance may be acquired
and temporary. Il occurs with many gastrointestinal diseases, with some infectious
diseases, or after a stomach resection. The most characteristic manifestation of lactose
deficiency is diarrhea after milk intake. Un hydrolyzed lactose enters the lower pans
of the small intestine where it is fermented by intestinal Hora with the formation of
gases (flatulence) and acids. The osmotic activity of acids and gases causes an influx
of a large amount of water to the intestine and diarrhea arises. Flatulence is the reason
for the intestinal colic. Lactose deficiency disappears after the treatment of the main
disease. Temporary insufficiency of lactase in infants is especially dangerous because
their main food is milk. If the failure is not recognized in lime, severe dystrophy can
occur. More than three-quarters of the world’s adults a re lactose intolerant. Treatment
for this disorder is to reduce consumption of milk while eating yogurts and cheeses,
as well as green, vegetables such as broccoli, to ensure adequate calcium intake; to use
lactase-treated products; or to take lactase in pill form before eating.
Pre-paraTWy pfia&e
Phosphwyiaiion ol gkicose
and iis conversion ra
pfirnng
glycerafclehyde
■3-phosphare
Glucose e-prssphaie
o Hexckinase
Phosph oftejcsse
Ptwspno-
Second _. L- TruciDkiftase-l
priming (aJ I
reason K Aldolase
Triosephosphaie
O isomerase
SyccraBc^pic 3-pftxfftrtc
Com piste oxidation of glucose
OjT.iiic9cp:clo*K- phosphate
1,3-&sphospncgiycofae |2]-
Fira ATP-
tarnfc-g - 2 ADP
resown vj
(subsvae level *
phospnor yiairznj
aPhosptngiyGerae (2)
2-PtKKpmglycerae (2)
Phosptweno^yfuvaie |2)
Second ATP
- 2 ADJ’
L’ATP
IsdKiiaK-fewel
ptostfHaylateyij
Pyfuvaie |2)
t
Aca^-CoA
f
TCAGyde
COh, hUO, ATP
Glucose
ATP
Glucose-O-phosphate
112
Fructose-6-phasphaie
ATP
I3
Fructose-1,6-bisphosphale
4 Dihydroxyacelorte phosphate
Glyceraldehyde-3-p hospha le
1,3-Bisphosphnglycerale
3-Phosphog tycorale
■II
2-Phosphog lycerale
9
II
Phosphoenolpyruvale
Pyruvate
MAD+
Aerobic and anaerobic glycolytic pathways involve two stages. In the first stage
(Fig. 6.8. preparatory phase) conversion of glucose into fruetosc-l^-bisphosphaLc
(reactions 1-3) and then reversibly to two molecules of glyceraldehyde 3-phosphate
(reactions 4 and 5) proceeds with the consumption of two molecules of ATP lor each
molecule of glucose that is split.
in the second stage (ATP synthesis phase) the conversion of 2 moles of
glyceraldehyde 3-phosphate into 2 moles of pyruvate (steps: 6-10) is associated with
ATP formation. Al this stage, the dehydrogenation of glyceraldehyde-3-phosphate
and the formation of NADH + I I1 takes place (reaction 6).
FAD
Mitccnondrial
J KT
gly-D=r-3 3FDH
FADHj
NAD NADH + H
2H+ + 2e"
CHj—CH—
i z
OH
y
Lactate -dehydrogenase
------------------ S■ '
I
CHa—C—
o
NAD*" NADH+H
mitochondria and the activity of mitochondrial enzymes that ensure the complete
oxidation of glucose. Under these conditions, the anaerobic synthesis of ATP
increases dramatically and lactic acid accumulates in. muscles. After a night's sleep,
the concentration of lactate in the blood is 1-2 Mmol/L and after intense muscle
exercises, it can reach 20 Mmol/L. Anaerobic glycolysis is especially important for
short-term intensive work. For example, 20 m long run (for about 30 seconds) is fully
provided by anaerobic glycolysis. At the same time the rate of anaerobic glycolysis
decreases quite quickly and the rate of aerobic glycolysis increases. After 4-5 minutes
of running (distance is about 1-5 km), the energy is supplied equally by the aerobic
and anaerobic processes. Muscle activity at 10 km distance (about 30 minutes) is
almost completely provided by aerobic processes and during the first minute of
work anaerobic oxidation of glucose provides much more power than under further
muscular work. With longer muscle activity, more and more fatty acids arc used as
sources of energy rather than glucose.
Erythrocytes do not have mitochondria al all and their need for ATP arc met
completely by anaerobic glycolysis. Intensive glycolysis is also characteristic of
malignant tumour cells. This process is of less importance for the bean muscle, brain,
and kidneys.
Anaerobic conditions do not exist in livi ng tissues, so the term ^anaerobic* indicates
only that oxygen is not used in this process. Thus, the ratio of the share of aerobic
and anaerobic catabolism of glucose in energy production depends on the presence of
mitochondria in the cells, their number, as well as on the availability of oxygen.
Reactions associated with the synthesis of ATP occur in the second stage of
glycolysis. Anaerobic glycolysis is less energy efficient Lhan aerobic glycolysis.
ATP can be produced by reactions catalyzed by glyceraldehyde-3-phosphate
dehydrogenase (oxidative phosphorylation}. pyruvate kinase and phospboglyceratc
kinase (substrate-level phosphorylation) if cells have sufficiently high oxidative
capacity. Given the stoichiometric coefficient, the resulting value must be multiplied
by two. In the initial stages (reaction I and 3) 2 moles of ATP arc consumed. Thus,
aerobic glycolysis provides 6(8) moles of ATP per mole of glucose:
Wien glucose is oxidized completely to COT and H.,O, 36 or 38 moles of ATP arc
generated. In this case, pyruvate may enter mitochondria and be converted to acetyl -
CoA, which is oxidized by the TCA cycle generating additional ATP:
2 xThe energy yield from aerobic glycolysis + 2 X-Thc energy yield from the TCA cycle =
= 6 (8)+30=36 (38) moles of ATP.
ATP
s-1
Pnosphofruclokirta &e-1 1
Q AMP, F-2jB-6bP
9 ATP. citrate
Frucl.ose-1,6-bisP
I
Giyceri-jeiiyde-S P
NADH + H*
1.3-Ebphosphogtycerale
t - ATP
|
I
PEP
Pyruvate kinase
0 F-1.6 &SP
@ ATP
NAD* NADH
Lactate 2 Pyruvate
Fig. 6.12L Major sites of regulation in complete oxidation of glucose: 0 - activators: @ - i nnib itors
Review tests
I. Match the figure and the Idler.
CaLaboJic pathways of glucose:
1. Anaerobic glycolysis.
2. .Aerobic glycolysis.
3. Complete oxidation of glucose.
268 Chapter 6. Carbohydrate metabolism
2Pyfuvare SLaoiaie
2Ace[yS-GoA
C '
d ®—O—Ctfe—
Glucose
I
G iucasa-6-pnospnata
1
F rudose-O-pnosphate
EJ---- -1 Fructosa-1,5-Disphospnata
Gtyceratdebydo-S-pnosphate (2)
s— I
1.3-Bjsphospbogiycefa.te (2)
S----- 1
3-Phosprioglycefate (2)
I
2-Phospnogiycofate (2)
l
Phospnoeroipyruvato |2}
I
Pyruvate (2)
Situational Problems
I. A child was admitted to the clinic with diarrhea after feeding on milk. To
determine the diagnosis, a lactose tolerance test was conducted. The test
solution contained 50 grams of lactose and was taken on an empty stomach.
Blood glucose levels were measured regularly over the next two hours — 30,
60 and 90 min after test solution and it turned out that the blood glucose
concentration was increased slightly. Present the possible reasons for the results,
argue them. For this:
a) write a reaction of lactose digestion in the intestine, specify the enzyme:
b) explain why the blood glucose concentration was not increased;
c) indicate the molecular mechanisms of the monosaccharide transport to the
small intestine cells and answer whether the violation of their absorption can
lead to these symptoms.
270 Chapter 6. Carbohydrate metabolism
2. The patient’s blood glucose concentration was 160 mg/dL 30 minutes after
taking 100 g of sugar. The blood glucose level of the same patient 30 minutes
after eating 100 grams of bread was lower. Why? To answer
a) draw the structures of sucrose and starch:
bi describe lhe digestion of these carbohydrates, indicate the enzymes, the site
of their synthesis, and the parts of the gastrointestinal tract in which the
hydrolyzis of sucrose and starch occurs.
3. For breakfast, a student has a mu din with jam and milk. What carbohydrate
digestion productscan be absorbed into the blood? For answer:
a) name the polysaccharides and disaccharides that arc contained in the
student’s food, represent a scheme for their digestion, specify the enzymes;
b) describe the transport of digestion products from the intestinal cavity into the
cnicrocytcs at low and high concentrations in the intestinal cavity;
c) indicate the digestion product with an increased concentration in blood,
name its normal concentration, and its value an hour after the meal.
4. There is a sweet taste in the mouth with prolonged chewing food containing
starch. Why? For the answer:
a) list the main dietary carbohydrates:
b) name the enzymes which arc present in human saliva;
c) write the reaction scheme of carbohydrates digestion in the mouth.:
d) specify the products of digestion providing a sweet taste.
5. The sprinter finishes 100 m run: the stayer runs the tenth km. Point out the
differences in the energy supply of these runners. To solve lhe problem:
a) draw a diagram of glucose catabolism, which is an energy source for muscles
in the stayer:
b) indicate lhe energy yield of the glycolysis and the complete oxidation, of
glucose, the mechanisms of ATP synthesis;
c) write down the substrates involved in. the dehydrogenation reaction, specify
the path of hydrogen from one of the substrates to oxygen, in the ETC;
d) indicate the differences in lhe linal products and the energy yield of lhe
metabolic pathways providing ATP in the sprinter and stayer.
6. The lens of the eye is the light refracting medium of the eye, and it has no
mitochondria. Glucose is used as an energy source in the lens. Which metabolic
pathway of glucose supplies energy to the crystalline eye lens? To answer the
question:
a? write a diagram of the metabolic pathway that provides the eye’s lens with
ATP, specify enzymes and coenzymes;
b) mark the reactions associated with the ATP consumption and synthesis,
calculate the ATP yield during the process;
c) specify the mechanisms of ATP synthesis in the process:
d) name the tissues and cells in which the ATP synthesis is the same as in lhe
tens*
e) write the dehydrogenation reaction and the reaction of final product formation
occurring in lhe process;
6.3. Gluconeogenesis. Regulation of Glycolysis and Gluconeogenesis 271
D indicate the laic of the end product of the process and the consequences of i ts
accumulation.
7. The number of mitochondria and myoglobin in athletes’ skeletal muscles
increases. How will these people change the production of lactate in muscles
compared to untrained people with the same physical activity? For answer:
a) write the scheme of the metabolic pathway whose final product is lactate:
b) explain how and why the activity of the metabolic pathway differs in athletes
and untrained people.
8. If the food contains the predominant amount of peeled cereals or bread made
from high-grade (lour, H, hypovitaminosis may occur. What is the role of vitamin
B1 in carbohydrate metabolism in the body? For answer:
a) name the coenzyme that contains vitamin B( and enzymes requiring this
coenzyme to function;
b} write the pathway of the carbohydrates metabolism involving these enzymes,
and explain how the rate of the process changes under a lack of B j and why ;
c) indicate the reactions that include vitamin Br
9. Hypovitaminosis caused by Bs deficiency in diet occurs in many people in spring.
Drowsiness and increased fatigue arc the most characteristic symptoms of this
hypovitaminosis. Why can the B-deficiency lead to such states? For the answer:
a) specify the coenzyme derived from B5 and the metabolic pathway that
includes it:
b) draw the scheme of the metabolic pathway providing energy for muscle
activity and show reactions that include this coenzyme.
COO’
CO2 ATP ADP 4-P,
CHa
!“ X
1
« J . Biotin
Cbfe
in rx
1
cocr pyruvate carboxylase COO"
(Pyruvate) fOxatoacarate ]
ATP„ Glucose
adp -^As*
Fructose-6-P
ATP^
ADP ~'A'*
Frutfose-1,6-oisP
Giyceraicfenyde-S-P Dihydroxyacetone-P
NAD" NAD+ I
Glycerol
MALJH NADH
1 r3-bispliosphoglycerate
O
ADP 's>jiX’ ADP E
O
O
□
ATP ATP a
D
3-Pnosprogiycerale ■
I
2-Phosphoglycera.te
Fig. 6.13. Reactions of glycolysis and gluconeogenesis. Starting with pyruvate, most of the steps of
gluconeogenesis are simply reversals of those of glycolysis
Malate leaves the mitochondria for the cytosol through a passive antiport and
is then reconverted to oxa loace late. This reaction has two purposes: to provide an
important substrate for gluconeogenesis, and to provide OAA that can replenish the
TCA cycle intermediates that may become depleted.
274 Chapter 6. Carbohydrate metabolism
CH2OH CH2OH
Glucose Glycogen
j Glu-6-pnosphatase
I
F/uctoso-6-phosphate
I Fru-1.6-bisphosphatase
Fructose-!,6-Disphosphate
Malate
TCA cycle Iso citrate
l
Fumarate CO2
X Alpna-katogiutarate
Succinate
SuCCihyl-GoA
,/C C°2
Fig. 6.14. Gluconeogenesis from Ute most important gluconeogenesis substrates: glycerol. lactate, and
lite (x-amino acids
Amino acids derived from hydrolysis of tissue proteins arc the most important
sources of glucose during a fast. During the catabolism of many amino acids pyruvate
or oxaloacclatc arc formed as intermediate products and can be then involved into
the gluconeogenesis pathway. For example, alanine, the major gluconeogenic amino
acid, is converted toa-kctoacid pyruvate by alanine aminotransferase:
276 Chapter 6. Carbohydrate metabolism
CHg
Alanine
aminotransferase
H "C —NHg+
Other a-kctoacids derived from the metabolism ofglucogenic ami no acids can enter
the TCA cycle and formoxaloacctate (QAA), a direct prccursorofphospho-enol pyruvate
(PEP). Amino acids and glycerol arc mainly used in glucose synthesis when fasting or
at a low carbohydrate diet. Under these conditions gluconeogenesis provides glucose
for the brain, while other organs obtain energy from the fatty acid oxidation.
Lactate is released into the blood by exercising skeletal muscle, and by cells that
lack mitochondria, such as red blood cells. This lactate is taken up by the liver and
reconverted to glucose, which is released back into the circulation. The physiological
role of gluconeogenesis from lactate is significantly different. Lactate formed in
anaerobic glycolysis is not the end product of metabolism, hut its formation is a dead
end metabolic pathway. The only way to use lactic acid is to convert it back to pyruvate
by lactate dehydrogenase reaction:
The lactate as the source of the pyruvate is important both during usual human
activity and starvation. Conversion of lactate to pyruvate is the first stage of lactate
utilization. The lactate formed in intensively working muscles or in cells with
prevail i ng anaerobic catabolism of glucose enters the blood and then is mainly trapped
by the liver, where it is converted to pyruvate. In the liver. NADH/NAD+ ratio is
lower than in contracting muscle, therefore, lactate dehydrogenase reaction proceeds
reverse, i.c. , in the direction of pyruvate formation from lactate. Pyruvate is partially
convened to glucose in gluconeogenesis, and newly synthesized glucose enters the
blood and is taken by skeletal muscles (the Cory cycle or the glucose-lactate cycle) and
is partially oxidized in the liver to CO2 and H 2O (energy of its oxidation can be used for
ATP synthesis, needed forgluconcogenctic reactions). In the muscles, transaminase
transforms a part of pyruvate to alanine, which is transported to the liver and there
again forms pyruvate (glucose-alanine cycle) (Fig. 6.15).
Cory cycle allows the effective functioning of many extrahepatic cells at the expense
of the liver and partly of the renal cortex, c.g.:
► the lack of mitochondria makes RBCs completely dependent on anaerobic
glycolysis for ATP production. Then the lactate is partly disposed of by the liver
and renal cortex:
► skeletal muscle cell sand particularly fast-twitch fibers contracting under low
oxygen conditions, such as during intense exercise, produce much lactate. This
could lead to an intracellular accumulation of lactate, and a consequent reduction
in intracellular pH. In the liver, a large part of the muscle lactate is convened to
glucose, thereby allowing the muscle to use ATP for the contraction.
6.3. Gluconeogenesis. Regulation of Glycolysis and Gluconeogenesis 277
Gluco&e
AJ arire
I * I
Fig. 6.15. The Cori cycle and giucase-alanine cycle. Tne cycling of lactate and glucose between
p eripne rai tissues and liver is called tne Cori cycle and includes tne fol low ing steps: tne flow of lactate
from me contracting muscle and erytnrocytes witn tne blood flow to me liver; glucose synthesis from
lactate in tne liver; tne flow of glucose from tne liver tn rough tne bloodstream to me working muscle
and to the red mood cells; use of glucose as an energy substrate Dy tne contracting muscle and red
blood cells with tne formation of lactate. Alanine plays a special role in transporting amino groups to
tne liver via a pathway called tne glucose-aianine cycle
Thus, the Cori cycle performs two important functions: supplies lactate utilization
and prevents lactate accumulation and pH decrease (lactic acidosis).
During gluconeogenesis three enzymes require high-energy phosphate bonds:
> pyruvate carboxylase (I mole of ATP);
> phosphocnolpyruvate carboxykinase < I mole of GT P);
> phosphoglycerate kinase (1 mole of ATP).
As 2 moles of pyruvate are required for the synthesis of I mole of glucose, a
total of 6 high energy phosphate bonds arc cleaved (4 moles of ATP and 2 moles of
GTP). Thus, three ATP molecules arc consumed for each molecule of lactate duri ng
gluconeogenesis since two molecules of lactate arc necessary for the glucose formation.
The total equation of gluconeogenesis is described as follows:
The produced glucose can again enter the muscles, and there turn into lactic acid.
The total glycolysis reaction is described as follows:
These two equations give a result of the Cori cycle. The working muscles gel 2ATP
by splitting 6 ATP in the liver. All glucose present in the body, both from diet and
synthesized, is ultimately aerobically oxidized to carbon dioxide and water. Anaerobic
oxidation serves as an auxiliary pathway of energy production from glucose. It has a
local (e.g., in red blood cells) or situational (in a working muscle) significance. The
product of anaerobic glycolysis, lactic acid, is also eventually oxidized by an aerobic
pathway. In adults, about SO g of glucose can be synthesized per day, mainly in the
liver and also in the cortex and intestinal mucosa. The biological significance of
gluconeogenesis is not only to return lactate to the metabolic pool of carbohydrates,
but also Lo provide the brain with glucose under a low carbohydrate diet and starvation.
278 Chapter 6. Carbohydrate metabolism
Fig. 6J6. The three cycles called substrate cycles <potential futile cycles) are designated as 1.2 aid 3
The direction of carbon How of the second substrate cycle (Fig. 6.16, 2)
depends on the activity of phosphofructokinasc andl fructose-1 r6-bisphosphatc
phosphatase. After a high carbohydrate meal, an increase of the blood insulin
levels and a decrease of the blood glucagon, levels (i.c., an elevation of the blood
insulin/glucagon ratio) rise the concentration of fructose 2,6-bisphosphate. This
compound is not an intermediate of the glycolytic pathway, but a special allosteric
activator of phosphofrucloki.nasc-1 (PFK-1, the regulatory glycolytic enzyme)
and allosteric inhibitor of fructose-L6-bisphosphale phosphatase (the regulatory
gluconeogenic enzyme. Fructose-2,6-bisphosphatc is produced in tissues due to
phosphorylation of fruciosc-6-phosphate by the enzyme phosphofructokinasc-2/
fructose 2,6-bi.sphosphatase that has dual functions (i.c., it is bifunctional —
BFE, Fig 6.1.7)..
280 Chapter 6. Carbohydrate metabolism
fig. 6.17. Reaction catalyzed Dy bifunctional enzyme. ;t acts as a kinase after a meal when me insulin/
glucagon ratio is elevated and as a phosphatase during fasting when the insulinrtjlucagon ratio is low.
The activity of the enzyme is altered by phosphorylation and depnospnorylation. It is phosphorylated
Dy protein kinase A in die fasting state ^when it acts as a phosphatase} and depnospnoryiated in tnb
fed state i wtien it acts as kinase)
The activity of BFE is associated with the nutrition rhythm and is regulated by
hormones. Wien the instilin/glucagon ratio is high (after a meal, absorptive state),
insulin activates phosphop rotci n phosphatase, and the enzyme is dcphosphorylatcd
(BFE-OH); its phosphofrtictokin;isc-2 activity is enhanced, and it synthesizes
fructose 2,6-bisphosphalc from fructose 6-phosphalc and ATP. The levels of fructose
2,6-bisphosphalc arc elevated, PFK-l is activated and glycolysis is stimulated.
When the insulin/glucagon ratio is low (during fasting, postab sorptive state),
glucagon activates the adenylate cyclase system and causes the activation of protein
kinase A and the enzyme becomes phosphorylated (BFE-OPOsHn). Phosphorylation
enhances the phosphatase activity and inhibits the kinase activity of the bifunctional
enzyme, and fructose 2,6-bisphosphalc is converted back to fructose 6-phosphatc.
This reaction is not a simple reversal olThe reaction by which fructose 2,6-bisphosphatc.
is synthesized. Synthesis utilizes ATP, but the conversion of true lose 2,6-bisphosphatc
to fructose 6-ph.osphaLe produces inorganic phosphate (Pi.) rather than ATP. When
fructose 2,6-bisphosphatc levels arc low, the rate of glycolysis is decreased because
PFK-l is not activated: it leads to glycolysis slowdown and a switch of liver metabolism
to gluconeogenesis.
Fructose 2,6-bisphosphalc regulates both glycolysis and gluconeogenesis only in
liver.
The third substrate cycle is mainly regulated by pyruvate kinase, which is inactive
in phosphorylated form and active when dcphosphorylatcd (Fig. 6.16, 3). In the fed
state, insulin activates phosphoprotcin phosphatase, which d'ephosphorylaics and
6.3. Gluconeogenesis. Regulation of Glycolysis and Gluconeogenesis 281
At high ATP and NADH concentrations typical for the high-energy status of the
cell, the key enzymes of glycolysis, phosphofruclokinase, and pyruvate kinase, arc
inhibited causing inhibition of glycolysis. A high concentration of AMP activates
glycolytic enzyme phosphofruclokinase and inhibits gluconcogenctic enzyme
fructose-1,6-bi.sphosphate phosphatase, and ADP inhibits pyruvate carboxylase
stowing down gluconeogenesis. Thus, at the low energy status of the cell, glycolysis is
activated and gluconeogenesis is inhibited.
282 Chapter 6. Carbohydrate metabolism
Induction and repression of the cellular con cent ration of key enzymes is regulated
by hormones. Steroid hormones binding to intracellular receptors directly regulate
gene expression, increasing or decreasing the synthesis of key enzymes. Glucagon
and insulin signal transduction also affect the synthesis of key enzymes. But these
hormones change the activity of transcription factors that vary the synthesis of
glycolytic and gluconcogenctic regulatory enzymes. In the fed state, insulin induces
the synthesis of glucokinase, phospho fructokinasc and pyruvate kinase, which
leads to the activation of glycolysis and causes repression of phosphoenolpyruvate
carboxykinase and reduces the rate of gluconeogenesis. In the well-led state,
glucagon increases gene transcription and synthesis of key gluconcogenctic
enzymes, phosphoenol pyruvate carboxy kinase, fructose-1,6-bisphosphatasc, and
glucose-6-phosphatase, and gluconeogenesis is activated, in prolonged starvation,
steroid hormone cortisol causing the induction of the enzyme gluconeogenesis,
phosphocnolpyruvate carboxykinase, is of particular importance in stimulating
gluconeogenesis.
HOCH
HUGH
3 -zcse 6 phosphate
_______
ctfcbpog-
' s' NADP+
Glucose &-phosphaie rMl-s^
dehyrtio^enase ! l -
NADPH + H+
HOCH
I
HCOH
H-nH [6- Phospho-gluconate]
I
CHgOPOf"
NADP*
5--yiuCC'I Iai -?
B^hospnogfuconaie .
dehydrogenase ■- naDPH + H+
J* aw
CHsOH
i-o
rtCOH ____________
I-iqq^ p-RitjujosB 5 phosphate'
CHaOPOl"
Phospfliopefiiose |
isomerase I
GHO
Fig. 6.19. Oxidative reactions of the pentose phosphate pathway. The end products are ribose
5-phosphate. CO.. and NADPH
284 Chapter 6. Carbohydrate metabolism
Oxidative reactions of
pentose phosphate pathway
■"W"
Epimerase
I
Fruao&e 1,&■
bte^ho^aase
AMoiase
Triose phospnane
isixnerase
[ Xylulose S- phosphate J i G.yceraldehyde 3 phosphate 1
Fig. 6.20. Oxidative and nonoxitiative stages of Pentose Phosphate painway. The non oxi dative
reactions of the pentose phosphate pathway occur in all cell types synthesizing nucleotides
and nucleic acids. These reactions catalyze the interconversion of sugars containing three
to seven carbons. These reversible reactions permit ribulose 5-phosphate to be converted
either to ribose 5-phosphate (needed for nucleotide synthesis), or to intermediates of
glycolysis — fructose 6-phosphate and glyceraldehyde 3-phosphate
Pentose phosphate cycle. The oxidative phase of PPP and the phase of the return of
pentoses to hexoses (nonoxidaiivc portion of PPP in the opposite direction) constitute
together a pentose-phosphate cycle in which one molecule of glucose is completely
broken down in one turn of the cycle. The pentose phosphate cycle functions primarily
in the liver and the adipose tissue (Fig. 6.18 13). The total equation of the pentose
phosphate cycle:
Quantitative cnaraclenshcs:
1) the Diood lactate concentration al rest — 1 m Mol/l:
2) tne normal blood glucose concentiation is 80-100 mg/dL {3.3-5.5 mMoi f I);
3) tne energy yield of aerobic oxidation of glucose to CO3 and h30 is 38 (36) moles of ATP per 1 met
of glucose;
4) energy yield of anaerobic oxidation of glucose is 2 mol of ATP per 1 mol of glucose.
286 Chapter 6. Carbohydrate metabolism
Review tests
1. Match lhe figure and the letter.
Enzymes:
1. Phosphocnol pyruvate earboxykinase.
2. Pyruvate carboxylase.
3. Glycerol phosphate dehydrogenase.
Glycolysis and Gluconeogenesis in the liver:
I----------- -El
! 1.3-BispnosphoQlycefate J
1
I...... ............ G3
3-Phosphoglycerate__ )
(
I
2-PnospnoQiycerate
I
■phbsphoefldi-pyhjvate >>---•
(
E
rp^qaeetatg]
*[ Pyfuvata ] CD
2. Match lhe figure and the lellcr.
Property of an enzyme:
1. It contains biotin as a cofactor.
2. It is hydrolase.
3. It is oxidorcducrase.
Glycolysis and Gluconeogenesis in the liver:
(thediagram is presented in Lest I)
3. Match lhe figure and (he Idler.
Regulation of gluconeogenic enzy mes. The synthesis of an enzyme is:
1. Repressed by insulin.
2. Inhibited by fructose-2,6-bisphosphate.
3. inhibited with a decrease in Lhe ratio of ADP/ATP.
Glycolysis and Gluconeogenesis in the liver:
(the diagram is presented in test 11.
6.4. Pentose Phosphate Pathway 237
Situational Problems
1. A person, ate 200 g of carbohydrates and then did not cal anything fora day. What
process of carbon metabolism was stimulated in the liver 14 hours after the last
meal? For answer:
a) write a diagram of the process that speeds up in the liver after 14 hours after
eating:
b) indicate the regulatory reactions and their enzymes;
c) describe the mechanism of signal transduction by a hormone that regulates
this metabolic, pathway.
2. A well-trained athlete is running 5 km. What is the difference in glucose
metabolism in the liver and muscle by the end of the race? For the answer:
a) write a diagram of glucose metabolism in the liver in this case:;
b) draw a metabolic pathway of glucose in muscles;
c) name regulatory enzymes and describe the mechanism of their activation;
c) explain the significance of the metabolic pathways that occur in the liver and
muscle.
3. The ATP source for the glucose synthesis from lactate can be the conversion
of lactate to pyruvate k subsequent oxidation of it to carbon dioxide and water.
How many moles of lactate must be oxidized in the liver to CO2and H3O to
provide ATP' for the synthesis of 1 mole of glucose from lactate? For answer:
a) depict the diagrams of the lactate oxidation and gluconeogenesis from lactate;
b) indicate the reactions produced and consumed ATP;
c) name the hormones that regulate gluconeogenesis in the liver and explain
the mechanism hormonal signal transduction.
4. During prolonged fasting and intense exercise, the product of fat breakdown
in adipose tissue, glycerol, is one of the gluconeogenesis substrates. How can
glycerol be involved in gluconeogenesis? How many moles of glycerol and ATP
arc required to synthesize I mole of glucose? To answer:
288 Chapter 6. Carbohydrate metabolism
a) write a diagram for the glucose synthesis from glycerol and name the reactions
occurring with ATP consumption;
b) name the hormones that stimulate gluconeogenesis and describe the
mechanism of signal transduction bv these hormones;
c) explain the role of the bi functional enzyme in the regulation of gluconeoge
nesis.
5. During prolonged fasting, amino acids derived from muscle proteins and
connective tissue become the main substrate for gluconeogenesis. How docs
glutamate enter the gluconeogenesis? For answer
a> write a reaction of the glutamate conversion into a-kctoglutarate, name the
enzyme, indicate the energy effect of the reaction;
b) draw a diagram for the a-kctoglutarate conversion to oxaloacctatc and
consequent including of OAA to gluconeogenesis, specify the reactions
associated with. ATP consumption;
c) name the hormones that accelerate gluconeogenesis during fasting and
regulatory enzymes of the process; and describe the mechanism of signal
transduction by these hormones.
6. Students carry out a laboratory experiment with avidin (a protein in. egg white)
having a very high affinity for biotin enzymes. Which enzyme of glucose
metabolism would be inhibited by the addition of avidin to a cell homogenate?
For answer;
a) draw a reaction that would be blocked by the addition of avidin to a cell
homogenate.;
b) write a diagram of the carbohydrate metabolism in which this reaction takes
place.
7. Students carry out a laboratory experiment of the oxidative conversion of glucose
to ribose 5-phosphate on muscle and liver homogenates. The first carbon of
glucose is rad ioact ivcly labeled. Will the labeled atom appear in pentose? Which
tissue; liver or muscle will show a higher rate of the process? To solve the problem:
a) depict reactions of the oxidative stage of the pentose phosphate pathway using
chemical formulas;
b) indicate the significance of this process for the cells and for the whole organism;
c) name the regulatory enzyme and mechanism of its regulation.
8. In a patient with anemia, the presence of Heinz bodies in the RBCs was the
result of the hemoglobin subunits aggregation due to the oxidation of -SH
groups of Hb cysteine residues with active oxygen forms and the formation of
disulfide bonds. What metabolic disorders in the RBC can be the cause of this
clinical case? To solve the problem:
a) indicate which reactions maintain cysteine residues in a reduced state;
b) name the coenzyme involved in this process, write a diagram of the process in.
which the reduced form of this coenzyme is formed:
c) indicate the enzyme which deficiency may lead to a lack of the reduced
coenzyme and be the cause of the clinical case described above.
9. A patient was admitted to the hospital with suspicion of malaria. After the
examination was complete and the diagnosis confirmed, the patient was
6.5. Synthesis ana Degradation of Glycogen 289
Fig. 621. Glycogen structure. Glycogen is composed of glucosyl un its linked Dy cxl ,4-giycosid'ic don ds
and al ,6-glycosidic honds. Tne tranches occur more frequently in tne center of me molecule, and
less frequently in tne periphery
Glycogen is mainly deposited in both the liver and skeletal muscle and is stored in
large cytosolic granules. Glycogen granules are poorly soluble in water a nd do not affect
290 Chapter 6. Carbohydrate metabolism
the osmotic pressure in the cell. Some enzymes involved in glycogen metabolism arc
also associated with granules, which facilitate the enzyme interaction with substrates.
Synthesis of glycogen. Glycogen is synthesized in the absorptive state (1-2 hours
after carbohydrate ingestion), mainly in the liver and in muscles. Because of muscle
greater mass, muscle contains about three to four times as much glycogen as does
liver. The biosynthetic pathway is an energy-requiring pathway: the attachment of one
monomer to the polysaccharide chain is associated with the consumption of ATP and
UTP. As i n glycolysis, glucose is phosphorylated to glucose 6-phosphate, catalyzed by
hexokinase in muscle and glucokinase in liver. Glucose 6-phosphatc is isomerized to
glucose 1 -phosphate by phosphogluco mutase. Then glucose I-phosphate reacts with
uridine triphosphate t UTP) to form the active nucleotide uridine diphosphate glucose
and pyrophosphate (Tig. 6.22), catalyzed by UDP-glucose pyrophosphorylasc.
Fig. 622. Formation of an activated form of glucose, UDP-glucose, synthesized from glucose
1-phosphate and UTP by UDP-glucose pyrophosphorylase
■Giyccgsri core
UDP-^uccse
Glycogen synttiorLC
SuC'P-duccse
6uDP
f'
4:6-Transferase
{tM-anchnig enzyme;-
Qyccgm core
Glycogen synthase
I P'’*
t Glycogen pncsphoiylise
0 Glucose 1 pneepnaieiC' .
4^
cooffiooooo Glycogen core
4:4-Ti'3ftsrerase
a-1,6-Glucosidase
X. IGkicoseQ}
GJycogefl pfro&ptioiyiase
Degradation ccrr.ifjes
The terminal glucosyl residues from the outermost chains of the glycogen
molecule arc sequentially removed by glycogen phosphorylase until approximately
four glucose residues remain on cither side of 1,6 branch. Glycogen phosphorylase
cannot act on the glycosidic bonds of the four glucosyl residues closest to a branch
point. The debranching enzyme acts both as transferase and as an ahb-glucosidasc.
Transferase catalyzes the removal of a trisaccharide unit from one branch and adds
to the other by an a 1,4-glycosidic bond, exposing a single glucose residue joined by
ana 1,6-glycosidic linkage, a 1,6-Glucosidase hydrolyzes the a I, 6-glycosi die bond,
resulting in the release of a free glucose molecule. Glucose I-phosphate, the end
product of glycogen degradation, formed in the phosphorolytic cleavage of glycogen,
is then converted by phosphogluco mu Lase into glucose 6-phosphate to enter the
metabolic mainstream:
Regulation of glycogen metabolism in the liver. The primary stimulus for insulin and
glucagon secretion is the changing blood glucose concentration. After carbohydrate
meal, the blood glucose level elevates to 10-12 mmol/L increasing insulin level and
decreasing glucagon level. Thus, iiisulin/glucagon ratio increases and insulin effect
becomes predominant. Insulin influences the following:
► acceleration of glucose transport into cells of insulin-dependent tissues (muscle
and adipose tissue);
► change in enzyme activity by phosphorylation and dcphosphorylation (Fig. 6.26).
Membrane
Protein phosphatase
inactive
Cascade of
phosphorylation
reaction
i Glucose
AdOfiytllC Hiospiw
■EVdKC
fficsi erase?
Cyropiasfn
Glucose
i
’ GkKOftBUrU
'V
Praiein.
Fleguiaory Glucose 6-phosphate
kinase A
(riactreeji SiikHil-€AMP %
GlpZOQZF i
l
« AvxsptKqftaw- . &jrrtrusc-P •
r kmasc » Glucose 1 -phosphate
Ijnacimi'l Ki
Protein; ADP PfctDTl
3 i' Active protein <5 j
ptcs|.-^aLase phDfptnlafK'
w kinase A J
%
ATP .4
PhOtJpjKQkKC- ADP ■■ - *
' Sroftasc *•
HKKKC-P ■jnacb¥c$
nsdNc'l
Glycogen UDP-glucose *
Oycogcn
phcKphocyniw t:
ijrurtvc'i
K
Blood
dizxjse
Fig. 6^7. Regulation of glycogen synthesis anti degradation in me liver 1 — Glucagon and epinephrine
interact with specific membranereceptors. The hormone receptor complex activates adenylate cyclase
via G proteins which synthesizes CAMP from ATP. 2 — CAMP binds io P KA and activates the catalytic
subunits. 3 — PKA activates phosphorylase kinase by phosphorylation. 4 — Phosphorylase kinase
converts glycogen phosphorylase o to the active glycogen phosphorylase a by phosphorylation. 5 —
PKA phosphorylates glycogen synthase and decreases its activity. 6 — inhibition of glycogen synthase
and the activation of glycogen phosphorylase I eact to glycogen degradation to glucose 1 -phosphate
Epinenaluine
XaXKODtt
Cytoplasm
GByGag&n
Calmodulin - synthase
(ama dive)
dependent
protein fanase
Glycogen synthase
(active)
Endoplasmic Ca1*- calmodUsn
reticulum Glycogen
II
■i phosphorylase a — p
W.
(active)
Kinase
o
Glycogen
ptiosplwylase t>
(inactive)
Fig. 6.28. Regulation of glycogen synthesis and degradation in the liver by epinephrine. 1
Epinephrine binds to <i .-receptors in the liver. Epinepnrine-receptor complex transmits a signal via G
proteins to pnospnoiipase C. Pnospnolipase C hydrolyzes PlP2 to DAG and IPS.2 — IPS stimulates tne
release of Ca?* from the endoplasmic reticulum. 3 — Ca3, binds to me protein calmodulin. Calmodulin-
dependent protein kinase and phosphorylase kinase are activated. Both Ca3, and DAG activate protein
kinase C. -4 — These three kinases phosphorylate glycogen synthase at different sites and decrease
its activity. 5 — Phosphorylase kinase phosphorylates glycogen phosphorylase to the active form
activating glycogenolysis and inhibiting glycogen synthesis
At the transition from rest to vigorous muscular activity, the need) of skeletal
muscles for energy increases tenfold. In response to a signal from the central
nervous system epinephrine secreted from the adrenal medulla interacts with
receptors of muscle cell membranes and triggers the cascade mechanism (adenylate
cyclase, inositol phosphate, associated with calmodulin (Fig. 6,29, 2 and 3), which
provides fast activation of energy supplying reactions. Such cascades allow for large
amplification of the initial signal. For example, in the adenylate cyclase cascade, one
epinephrine molecule activates one adenylate cyclase molecule, then one molecule of
adenylate cyclase can synthesize many molecules ofcAMP and a signal is amplified.
In the same way, the signal is enhanced at all enzymatic stages. Thus, the cascade
mechanism ensures the inclusion of large amounts of glucose in catabolism in a short
time.
Epinephrine
Fig. 629. Activation of muscle glycogen phosphorylase by muscle contraction, neural impulses, ano
epinephrine. 1 - AMP produced from the degradation of ATP during muscular contraction aitostericaily
activates glycogen phosphorylase b. 2 — The neural impulses that initiate contraction release Ca3- from
the sarcoplasmic reticulum. The Cat, binds to calmodulin, wliich is a modifier protein mat activates
phospnorylase kinase. 3 — Phosphorylase kinase is also activated through phosphorylation by FKA.
The formation of CAMP ana the resultant activation of PKA are initiated toy the binding of epinephrine
to plasma membrane receptors
When the muscle returns to rest, epinephrine secretion, stops. Having been already
released epinephrine is destroyed and results in adenylate cyclase inactivation.
cAMP present in the cell is converted to AMP by phosphodiesterase, and therefore.
298 Chapter 6. Carbohydrate metabolism
protein kinases are inactivated; glycogen phosphorylase and glycogen synthetase arc
dcphosphorylatcd by phosphatases, and the system returns to a state where glycogen
mobilization is suppressed , but its synthesis can occur.
The cascade mechanism is triggered in muscles under intensive and severe muscle
activity. Under moderate load (mild muscle exercises), glycogen phosphorylase
practically remains in dcphosphorylatcd inactive state, hut the breakdown of
glycogen occurs nonetheless. This is due to the fact that phosphorylase-OH
can also be activated by covalent modification (Fig. 6.29, I). In the contracting
muscles, the AMP concentration increases due to reaction: 2ADPH - ATP
-I- AMP. AMP is an allosteric activator of muscle phosphorylase (AMP docs not
activate hepatic phosphorylase). Muscle phosphorylase is a homodi me r; each of
the protomers contains an AMP binding site and a phosphorylation site. Extremely
active phosphorylase is phosphorylated and is associated with AMP. Intermediate
phosphorylases arc less active and allow to perform moderate physical exercises.
For example, in experimental mutant mice with phosphorylase kinase deficiency,
the formation of phosphorylase-P is generally impossible. These mice do not dilTcr
from normal animals in normal motor activity (swimming in water for a long time}.
When frightened instead of rushing, such mice will have convulsions as a result of the
impossibility ofa rapid glycogen mobilization.
The significance of glycogen metabolism regulation. When a hormonal signal is
transmitted through intracellular mediators, its considerable amplification occurs:
therefore, activation ofglycogen phosphorylase via any signal transduction system in the
liver allows rapidly obtaining a large amount of glucose from glycogen. Strengthening
the hormonal signal in the muscles is of great importance for providing energy for
severe activity under stress conditions. At the transition of the postabsorptivc state to
the absorptive state or from muscle activity to rest, the system returns to its original
state. Adenylate cyclase and phospholipase C arc inactivated, cAMP is destroyed
by phosphodiesterase, and phosphoprotcin phosphatase causes the transition of
all intracellular enzymes of the ^cascade» to the dcphosphorylatcd form. Thus, the
regulation of synthesis and breakdown ofglycogen in the liver maintains the constancy
of the blood glucose concentration (13—5.5 mmol/l). The regulation ofglycogen
metabolism in muscles provides the energy for both intensive muscle work and energy
consumption at rest.
Typel Von Gierke’s disease Deficiency of glucose-^ Liver cells and renal tubule
phosphatase cells loaded with glycogen.
Hypoglycemia, lactic acidemia,
ketosis, hyperlipidemia.
Cory disease (type III) is quite common. Il is 1/4 ofal I cases of hepatic glycogenosis.
The accumulated glycogen is abnormal in structure, since the enzyme amylo-1,6-
glucosidasc (the debranching enzyme), which hydrolyzes glyoosidic bonds at branch
points, is defective. The low’ blood glucose love! manifests itself quickly because
glycogenolysis proceeds with an insignificant rate. Unlike type 1 glycogenosis, lactic
acidosis and hyperuricemia arc not observed. The patients with type 111 have a. milder
disease course.
Andersen’s disease (type IV) is an. extremely rare autosomal recessive disorder
caused by deficient activity of the glycogen-brane hi ng enzyme, amylo-1.4-1,6-
glucosykransfcrasc and results in accumulation of abnormal glycogen in the liver,
muscle, and other tissues. The structurally abnormal glycogen has few branch points,
as well as very long and sparse lateral branches that prevents glycogen to be degraded.
At the same time, hypoglycemia is moderately expressed. The disease develops
quickly, is aggravated by early cirrhosis of the liver and is practically not amenable to
treatment. Defect of the branch enzyme is found in the liver, in leukocytes, muscles,
and fibroblasts. Early and prevailing manifestations of the disease arc due to impaired
liver function.
Hers’s disease (type VI) is characterized by mild clinical manifestations and a benign
course. This glycogenosis is a consequence of the hepatic glycogen phosphorylase
deficiency. Glycogen of the normal structure accumulates in the hepatocytes. The
course of the disease is similar to glycogenesis type I. but the symptoms, hepatomegaly,
growth retardation, and mild episodes of fasti ng hypoglycemia during early childhood,
arc less pronounced. Reduced glycogen phosphorylase activity is also found in
leukocytes. Hers disease is a rare type of glycogenosis inherited in an autosomal
recessive manner.
Defect of kinase phosphorylase (type IX) is found in boys only, because the enzyme
deficiency is inherited in an X-1 inked manner.
The defect of protein kinase A (type X), as well as the defect of phosphorylase
kinase, manifests symptoms similar to Hers’s disease.
Muscle glycogenosis arc characterized by disorders of energy supply to skeletal
muscles. These diseases are manifested during physical activity and are accompanied
by pain and cramps in the muscles, weakness and fatigue.
MacArdle’s disease (type V) is an autosomal recessive myopathy caused by the
lack of muscle glycogen phosphorylase isozyme. Since the activity of this enzyme in
hepatocytes is normal. hypoglycemia is not observed (the enzyme structure in the liver
and muscles is encoded by different genes). Severe physical exertion is poorly tolerated
and may be accompanied by convulsions. However, lactate hyperproduction is not
observed during exercise. That emphasizes the importance of different energy sources
for muscle contraction, such as fatty acids (see Ch. 7). Although the disease is not sex-
linked, the high incidence of the disease is characteristic of men.
Phosphofructokinase deficiency is characteristic of type VII glycogenosis.
Patients can. perform moderate exercise. The course of the disease is similar to type V
glycogenosis, but the main manifestations arc less pronounced.
The defect of phosphoglyccrate mutase and the defect of the M-subunit of LDH
are characteristic of muscle glycogenosis. Manifestations of these pathologies are
302 Chapter 6. Carbohydrate metabolism
similar to Mac Ardle’s disease. The defect of phosphoglycerate mutase in the muscles
is described in one patient only.
Diseases are associated with disturbance of glycogen synthesis (e.g., deficiency of
glycogen synthase) and are accompanied by a decrease in its content in the tissues. The
most characteristic symptom of such diseases is acute hypoglycemia (since there are no
glycogen stores), especially after a night fasting. Hypoglycemia can lead to vomiting,
convulsions and loss of consciousness. Constant starvation of the brain leads to a delay
in menial development. The majority of these patients die in early childhood: frequent
feedings can significantly reduce the symptoms of the disease.
CH3CH£OH
E tnan:
O
Ch:C
H
Afraid my de;
NAD*
NADH + H +
; Acetate I
fip. 6.30. The pathway of ethanol metabolism in the liver. ADM, alcohol be hydrogenase: ALDH,
acetaldehyde dehydrogenase
The most significant pathway of ethanol metabolism in the liver is through the key
enzyme in alcohol metabolism, liver alcohol dehydrogenase (ADH). The ADH is a
cytosolic NAD4-requiring enzyme expressed at high concentrations in hepatocytes.
It oxidizes ethanol io acetaldehyde with, reduction, of NAD* to NADH (Fig. 6.30).
Approximately 90% of the acetaldehyde that is generated is further metabolized
to acetate in the liver. The major enzyme involved is a low Km mitochondrial
6.3. The Metabolism of Etnanoi tn the Liver 303
The pyruvate concentration in the cells and in the blood decreases, and the lactate
concentration, increases. As pyruvate is a precursor of the gluconeogenesis, the rate
of the pathway in the liver decreases and hypoglycemia develops. Especially acute
hypoglycemia occurs under lack of glycogen stores in the liver and muscles (after
taking alcohol on an empty stomach, after considerable muscle activity, in chronic
alcoholics, who have constantly decreased appetite). Hypoglycemia can cause a loss
of consciousness due to alcohol poisoning. With high doses and chronic alcohol
consumption, a lot of blood acetaldehyde damages cell membrane structures. In
particular, mitochondria arc damaged: the transmembrane potential and the efficiency
of coupling of respiration and phosphorylation arc reduced.
Review tests
I. Mulch the figure and the Idler.
The scheme of glycogen degradation:
Enzyme:
A. Phosphoglucomuiasc.
B. a1.6-glycosidasc.
C. Glycogen phosphorylase.
D. Phosphatase.
E. Ofigosaccharyl transfcrase.
304 Chapter 6. Carbohydrate metabolism
esecooccooc
*
■
■
I
Gl ucoso-6-phosphate
2. Match the figure and the letter.
The diagram of glycogen degradation:
Metabolite:
A. Glucose.
B. ATP
C H/O^
D. ADP.
E. Glucose I-phosphate.
eiexcaxcc
*
I
I
Glucose-6-phosphate
6.3. The Metabolism of Ethanol tn the Liver 305
ATP -h.
2 Pi Pi
— Glycogen phosphorylase-OH h/l\rC □lycogen synthase-OH 1
^-ATP
I
___________________________________ I
ADP-' * Glycogen phosphoiylase®^ Glycogen synthase® * * ADP
Pi Pi
Glycogen phosphorylase-OH p / X 4 Glycogen synthase-OH “
3- ,
ATP ATP
1 JL 3
Situational Problems
I. During an exam a student’s blood glucose was 7 mmol/l. Explain the reason Tor
the observed change in blood glucose if the student had breakfast 4 hours before
the exam. For answer:
a) indicate the normal blood glucose concentration:
b} name the hormone which concentration rises in the student’s blood in this
situation:
c) draw a chart of the process activated in the liver by this hormone and indicate
the regulatory enzyme:
d) draw the scheme ofhormonal signal transduction and describe the mechanism
of the hormone effect on the regulatory enzyme.
306 Chapter 6. Carbohydrate metabolism
2. A person performs intense exercise (for example, runs away from danger)
30 minutes after a carbohydrate-rich lunch. Why docs the glycogen synthesis
stop in skeletal muscles in th is situation? Why is glycogen breakdown stimulated?
To answer the questions:
a) write a diagram of glycogen synthesis, specify die reactions associated with
energy consumption when a glucose molecule is included in a growing
glycogen chain:
b) write the glycogen mobilization diagram and calculate the ATP amount that
can be produced if glucose I-phosphate is further oxidized to CO3 and H J)
in the muscles;
cl indicate the hormone with an increased level in blood under stress situations,
and how this hormone affects the activity of regulatory enzymes of glycogen
synthesis and breakdown.
3. Alkaloid caffeine contained in coffee seeds causes hyperglycemia and has a
stimulatory effect, although it does not interact with epinephrine receptors.
It is known that caffeine inhibits phosphodiesterase activity. Why can caffeine
decrease the blood glucose level? Explain the answer and draw the appropriate
schemes.
4. A patient had severe fasting hypoglycemia. The liver biopsy showed that
glycogen synthesis occurs, but that liver tissue accumulates molecules with short
side branches. What enzyme deficiency can cause this pathology? Explain your
answer by writing a diagram of the process that should occur normally. Indicate
the site of the disturbance on the diagram.
5. Popular ^energy* drinks arc nonalcoholic carbonated beverages. Advertising
emphasizes that they increase working capacity. Most beverages contain, caffeine
and its analogs, theobromine and theophylline (vegetable alkaloids), vitamins,
easily digestible sugar (glucose, sucrose), taurine, carnitine. These drinks arc
highly carbonated, which accelerates their absorption into the blood. Why do
energy drinks improve efficiency? For answer:
a) draw a diagram of the glycogen metabolism pathway with its the acceleration
possibly contributing to belter efficiency:
b) indicate the mode of caffeine action on the rate of this process, if it is known
that it inhibits phosphodiesterase;
c) explain why 3-4 hours after the taking of ^energetics* fatigue and drowsiness
occur.
6. In an experiment, students used liver tissue samples to study the ethanol
metabolism and the possibility of ethanol conversion into glucose. The ethanol
introduction in the investigated medium didn’t lead to glucose level increase.
Why is it impossible to convert ethanol to glucose? For the answer:
a) provide a scheme of gluconeogenesis, indicate the substrates of this process;
b) write the reaction of ethanol oxidation in the liver:
c) explain whether it is possible to use the metabolites of ethanol catabolism for
the glucose synthesis.
7. In experimental laboratory, the effect of epinephrine on glycogen metabolism
in a muscle tissue sample of the mu Lam mice line was studied. After adding
6.3. The Metabolism of Ethanol tn the Liver 307
All cells
Structural compounds
(as me compounds
Fatty acids R-COOH of lipids, fuel of other lipids — TaGs.
molecules
phospholipids)
0
Long — term storage
HbC-O-4-R.,
Triacyglycerots of energy, tnermo-
and mechanic Adipose cells
(TAG) R^-C-O-CH 0
protection
HjC -- 0—C "Rg. of a body
6 Structural components
0 H^C-O-A-R, of membranes,
Glycerophospholipids
X: chorine, monolayer Cell membranes,
ethanolamine, fu-A-o—Ah o on the surface lipoproteins, alveoli
inositol bisphosphale. of lipoproteins, of me lungs, bile
HjC-D-P-O-X
serine amphipathic compounds
Ah of bite micelles
| Ceram tdej
O A component of
Spliingophospholipkis the myelin sheath Central nervous system
(Sphingomyelin) O-P-O around neurons
1.
Chains
Sphingolipids:
I Ceramide ] intercellular Membranes of Che cells
Cerebrosides —
1 communication, and the myelin sheath
X-one monosaccharide.
Carbohydrate-X antigenic of the central
Gangl iosides-X-complex
determinants nervous system
sugars
The component
of me ceHs membranes
Cell membranes,
Steroids Cholesterol the precursor of bile lipoproteins, bite
salts and steroid
hormones
Regulators of the
Derivatives of 20 carbon Almost every cell
Eicosanoids cellular functions,
polyunsaturated fatty acids of the body
‘Local’ hormones
Palmitic (16:0) and stearic (18:0) acids arc the most common saturated fatty acids
in humans. The numbers 16 or IS denote the number of carbon atoms in the fatty
acids. Zero denotes that these fatly acids don't contain double bonds. Saturated fatty
acids account, for up to 40-45% of the fatty acids in human fat in adipose tissue.
Unsaturated fatty acids contain one or more double bonds. The most commonly used
systems designating the position of the double bonds in unsaturated fatty acid arc the
della (A) numbering system and cn-system (Table 7.2, Pig. 7.1). For example, 18:1A9
denotes that the fatty acid contains one double bond between the ninth and tenth
carbon atoms starting from the first carbon of the carboxyl group. The same fatly acid
is designated as 18:Iu>9 that denotes the distance of the double bond closest to the
<o-cnd (the lasL methyl group in an aliphatic chain of fatly acid).
310 Chapter 7. Lipid metaD ol ism
Carboxyl
group
Hydrocarbon
chain
(a) (b)
W)
Fig. 7.1 Structure ol saturated and unsaturated fatty acids: a) saturated fatly acid; b) monoenic fatty
acid wim one double bond in me cis conformation; c) orderly packaged saturated fatty acids in lipid
phase, solid at room temperature; di fatty acids with a lower melting point are fluid al body temperature
The double bonds arc usually in the cis conformation of the native fatly acids. Cis
conformation induces the kink in the fatty acid chain, preventing orderly packing
of the chains in lipid phases ( Fig. 7.1). The melting point of a fatty acid decreases
with the degree of saturation and increases with chain length. As a result, the double
bonds increase the fluidity of lipids, containing unsaturated fatty acids. Thus, the fatly
acids composition of membrane phospholipids determines the fluidity of the inner
hydrophobic layer of membranes at body temperature.
7.1. Structure ana Functions of me Major Human Lipids 311
Table 72. Tne main fatly acids of the humans. Designation of fatty acids by delta (A) and n/-systems:
16 denotes the number of carbon atoms in the fatty acid: 0 - fatty acid doesn't contain double bonds
(16:0 palmitic acid): 20:4A5,8,11J4-arachiconic acid contains 20 carbons and four double bonds at the
5* S* 11*14" carbon atoms starting from me 1st carbon of carboxyl group: ’ - essential fatty acid
Palmitic 16:0
Stearic 16:0
Palmitoleic 16:1 A9 9
Oleic 18:1 A 9 9
■3
a-Linolenic 18:3 A 9,12,15*
Fatly acids arc subdivided into nonessential and essential by their requirement to
humans. Noncssenlial laity acids arc saturated or nionocnic fatty acids (contain one
double bond) and synthesized in humans. These acids don’t have to be obligatorily
included in the diet. Polyunsaturated fatty acids arc essential and belong to two
series. One is w6 like linoleic (18:2 ci>6) and arachidonic (20:4co6) acids. Another is
w3 like a-linolcnic (I8:3 cd 3) acid (Fig. 7.2). Linoleic and n-linolcnic acids arc not
synthesized in mammalians as they lack enzymes necessary to insert double bonds
beyond the 9th carbon and w-carbon of fatty acids. These fatty acids must be in the
diet and can be obtained from plant oils or cold-water fish oil. The fish obtain these
fatly acids by consuming water plants. Linoleic acid can be converted to arachidonic
acid, whereas a-linolcnic — to cicosapentaenoic acid (20:5 A5,S,I I.14J7) in the body.
These 20 carbon fatty acids are the precursors of prostaglandins and other eicosanoids
which arc important in cell communication. Another important function of linoleic
acid is Lo form unusual lipid in the skin that helps to make the skin impermeable to
water. Dietary deficiency of essential fatty acids results in dermatitis.
Tri acylglycerols, more commonly known as fat, are the major lipids in human
body. Triacylglyccrol contains glycerol in which all three hydroxyl groups are cstcrificd
with fatty acids (Table 7.1). Most native triacylglyccrols have different fatty acids
cstcrificd to the three positions of the glycerol, the most unsaturated fatty acid is
usually bound with 2nd carbon of glycerol. The distribution of the fatty acids across
those three positions of the glycerol moiety of triacylglyccrol is influenced by many
factors., including diet and anatomical location of the stored molecule.
Triacylglyccrols arc stored in adipocytes of adipose tissue specialized lor long —
term energy storage. Adipose fat is very efficient fuel storage: triacylglyccrols contain
more calorics per gram than carbohydrate or protein (9,3kcal/g for fats as opposed to
4,0 kcal/g for glycogen), because they are the most reduced organic molecules in a
body and triacylglyccrols drop in adipocytes do not contain water. A normal man of
70 kg weight has about 15 kg stored triacylglyccrols in adipose tissue, which accounts
312 Chapter 7. Lipid metaD ol ism
for - 85% of his total stored calorics. Subcutaneous adipose tissue is also important in
temperature regulation because it provides a layer of insolation.
18
/uVuVVV\ 15 12 9
a-Linolsic acid (m3. 10:3^12,15)
COOH
20 17 14 11 3 5 COOH
Elcosapentaonoic acid (m3. 20:5A5,8,11,14,17 )
Fig. 72. The structures of the polyunsaturated fatty acids. denotes the position of me closest
double bond to m end (methyl end) of fatty acid; me same for qj3 and gj9 fatty acids: w6 linoleic (182)
and iu3 a-iinoienic n 8:3) acids are essential for humans
H +HCO1 -H3CO-H2O+CO3r
The next step is the emulsification of lipids. The step is required for the conversion
of big waler-insoluble fat particles to microscopic particles, so fat would become more
available for the action of digestive enzymes. Fat emulsification is carried out by bile
salts, which arc synthesized from cholesterol in the liver and arc secreted into the small
intestine from gallbladder where they are stored. The contraction of the gallbladder
and secretion of pancreatic lipase arc stimulated by the gut hormone cholecystokinin.
Bile salts, as amphipathic compounds, possess both, hydrophobic and hydrophilic
pans and act as detergents. They surround the lipid drop so that hydrophobic pans arc
immersed into lipid drop while hydrophilic parts arc on the surface of the drop. A big
lipid drop then divided into small droplets, as the surface tension lowers. Peristalsis
of duodenum and the presence of CO. arc also involved in small droplets formation.
Emulsification enormously increases the availability of lipid molecules for the action of
water-soluble enzyme — pancreatic lipase, because lipase acts on the oil-water interface.
The fat digestion is catalyzed by pancreatic lipase, which is secreted from pancreas.
The small protein — colipase — is needed for the activity of pancreatic lipase. Colipase
is secreted also from pancreas and is activated by trypsin through, partial cleavage.
Colipase stabilizes and activates pancreatic lipase improving the interaction of the
314 Chapter 7. Lipid metaD ol ism
enzyme with water-insoluble cm nisi tied fat drops. The pancreatic lipase hydrolyzes
triacylglyccrols by removing fatty acids from the 1st and 3d carbons of glycerol and
producing two fatty acids and 2-monoacylglyccroL
Dietary
TAG (lipidsj
Emulsified TAG
oooo Pancreas
oooo
Pancreatic lipase
Colipase ]-*
o
£
E
3
fl)
JZ
Mixed micelle
Resynttiesis of TAG
2 acyl-CoA
h2COH H3C O- CO Rf
fl)
J=
Hgp—OH F^c-O— CO— Rs
-I— 2 CoASH
□l
Formation of nascent
chylomicrons
Formation of mature
chylomicrons
Q
m
Fig. 7.3. Digestion and absorption ol dietary triacylglycerols. Packing of lipids into chylomicrons
7.2. Digestion ana ADsorplioa of Dietary Lipids. Transport of Lipids Dy Chylomicrons 315
Another two lipases exist in humans — lingual and gastric, but they digest mainly
emulsified fat of milk and arc important for infants. In adults these enzymes digest
not more than 15% of dietary triacylglycerols. Cholesterol esters present in dietary
lipids arc hydrolyzed by pancreatic cholesterol ester hydrolase (cholesterol esterase)
that produces cholesterol and fatty acid. The degradation ofglyccrophospholipids is
performed by phospholipases of pancreatic juice. Phospholipase A, is activated in the
intestine by trypsin and removes the fatty acid from position 2 of the glycerol, leaving
a Ivsophospholipid as a result.
The products of triacyl glycerols digestion — fatty acidsand 2-monoacylglyccrols —
arc packaged into mixed micelles, that help hydrophobic molecules pass through the
membranes of intestinal epithelial cells. Other dietary lipids — like cholesterol and
fat-soluble vitamins arc also packaged in the micelles stabilized by the bile salts. The
micelles arc water-soluble because the hydrophilic groups of bile salts, fatty acids,
2-monoacylglyccrols and cholesterol arc exposed to water a nd arc outside Lhe micelles,
but the hydrophobic are inside. The micelles travel to the microvilli of the intestinal
epithelial cells, where the products of lipids digestion arc absorbed but the bile salts
arc left behind in the In men of the gut and arc intensively resorbed, when reaching the
ileum (recycling of bile salts, Ch. 7.4).
The absorbed fatly acid is activated to acyl-CoA by acyl-CoA synthetase in
endoplasmic reticulum of the intestinal mucosal cells:
ATP
FACoA.
This is the first reaction of a fatty acid after entering a cell, like the reaction of
glucose phosphorylation by hexokinasc or glucokinase is always the first reaction of
intracellular metabolism of glucose. Like glucose 6-P, acvl-Co A cannot cross the
plasma membrane and is used only in intracellular metabolic pathways. The acyl-
CoA derived ITom the absorbed fatty acid reacts with 2-monoacylglycerol to form
at first diacylglycerol (DAG), then another fatly acyl-CoA binds to DAG to form
triacylglyccroL This process is called the resynthesis of triacyEglycerols:
Apc^raLein B43
Nascent
chyJonycrons
CoASH
'2 Monoacylgly-zerol] Triacylglycerol
High, insolubility in. water of the synthesized TAGs and other absorbed lipids
leads to their coalescence in blood, impeding the blood flow: this explains why the
lipids are transported by blood in water-soluble lipoproteins particles. Intestinal
cells package triacylgiyccrols and other hydrophobic molecules in lipoproLcins —
chylomicrons (CM). Lipoproteins arc composed of neutral lipid core surrounded
by a shell of compounds oriented so that their polar portion is exposed on the
316 Chapter 7. Lipid metaD ol ism
surface of the lipoproteins (Fig. 7.4). The protein compounds of lipoproteins arc
named apoproteins. They arc different in different types of lipoproteins (Table 7.3).
The major protein compound of chylomicrons is apoprotein B-48. This apoprotein
is coded by the same gone as apoprotein B — 100, which is synthesized in the liver
and serves as the major pro Lein compound ofVLDL. In the intestine, a stop codon is
generated during posttranscriptional modification ofmRNAand the protein which
is synthesized comprises only 48% of the size of the protein produced in the liver;
hence the designations are B-48 for apoprotein of chylomicron and B — I00 for
apoprotein ofVLDL. Besides apo B-48 apoprotein apo A-1 is synthesized in intestine
and included in the nascent chylomicrons. Triacylglycerols, which arc resynthesized
in the endoplasmic reticulum of intestinal cells, complex with the proteins to form
the chylomicrons. Triacylglyccrols are the major lipid compound of chylomicrons
(85%) and the function of chylomicrons is to deliver the dietary’ lipids to peripheral
tissues.
The absorbed lysophospholipids and cholesterol are converted into phospholipids
and cholesterol esters in rcacylation reactions and then, also, arc packaged in
chylomicrons. Short and medium-chain length fatty acids of milky fat arc not
convened to their CoA derivatives, but rather released into the portal circulation where
they are carried by serum albumin to the liver.
Proteins 2 10 11 22 50
Phospholipids 3 18 23 21 27
Cholesterol 2 7 8 8 5
Cholesterol 3 10 30 42 16
esiers
TAG 85 55 26 7 3
TAG — triacyigJyc&«M&; VlDL — vary low-dMisily lipopratflins; LOL — low-density lipoprelBins:; IDL — Intermediate density
lipoproteins; HOL — high-tfeasity lipopwtains; B — -4S structural apoprotein of chylomicrons: B —100 — structural
apoprotein of VLDL IDL; LDL; Cai — activator of lipoprotein lipase; E — mediates tha uptake nt chylomicron remnants and
iDl mainly try receprws on ins surfacB of hapafocytBs.
lipase synthesized in adipose tissue has a higher Km, than the isoenzyme synthesized
in muscle cells, therefore, the adipose lipoprotein lipase is more active after a meal,
when chylomicrons level increases in blood. Insulin, which secretion raises in the fed
stale, stimulates the synthesis and secretion of adipose lipoprotein lipase. This ensures
that dietary fat is directed mainly to adipose tissue in well-fed state. Lipoprotein lipase
in capillaries of the muscle cells hasa lower Km than in adipose tissue, thus, the muscle
cells can obtain fatty acids as fuel molecules from blood lipoproteins when they arc
needed for energy, even if the concentration of lipoproteins is low (fasting slate). After
realizing the triacylglycerols from chylomicrons, apoCi I returns back to HDL and
chylomicrons becomes chylomicron remnants, containing apoE. ApoE is recognized
by membrane receptors, particularly those on the surface of hepatocytes, allowing
apoE-bearing lipoproteins to enter these cells by cndocytosis for subsequent digestion
by lysosomes. Lysosomal enzymes degrade the chylomicron remnants and released
products of digestion can be reutilized by the hepatocytes (cholesterol, fat-soluble
vitamins, phospholipids, amino acids). In the capillaries of the liver, another lipase
is located - hepatic triacylglycerol lipase (HTGL). U LG L also digests triacyl glycerols
of lipoproteins, hut its substrates arc panicles already partially digested by lipoprotein
lipase. Hepatic iriacylglyccrol lipase facilitates the conversion of IDE into LDL.
Lymph
chylomicrons blood
Capillary walls
HDL
Chylomicrons
Chylomicrons
intestiaJ
epitheHal cell
Endocytic
Chylomicron
vesicle
remnants
Glycerol
Cholesterol
Receptors
Amino acids
Glicerol
A B
Cm
LDL{p)
VLDL(pre-|ij
HDL(a)
Increase of density
Fig 7.6. Analysis ol serum mood lipoproteins by ultracentrifugation (A) and Uy electrophoresis (B).
A (a) — 12 hours after a meal; A (D) — 1 nour after a fatty meal; B — electrophoresis of blood
lipoproteins in fed state ifatty meal); CM — cnyto microns; pre-p !i poproteins (VLDLI, p lipoproteins
(LDL); ci lipoproteins (HOL)
Review tests
I. Lipoproteins compounds. Match She figure with the letter.
A. Cholesterol.
B. Integral apopop rolcins (apo B-100 or apo B-48).
C. Phospholipids.
D. Peripheral apoproteins (ex. apoAL apoAII, apo EK
E. TAG.
7.3. Disorders of Li pi ds Digestion and Transport 321
A. Cholesterol.
B. Integra! apoproteins (apoB-l00orapoB-48).
C. Phospholipids.
D. Peripheral apoproteins (ck. apo A-1, apoa-ILapoE).
E. Cholesterol esters.
J. Lipoproteins of human serum blood 2 hours
after a meal. Match the figure with the letter
(u Itracentrifuga t ton method). Lipoproteins
A. HDL density
B. LDL. increse
C. CM
D. VLDL
E. CM nascent.
4. Lipoproteins of human serum blood I4 hours
after a meal. Match the figure with the letter
(u El raceulrtfuga I ion metho d). Lipoproteins
A. CM. density
B. VLDL. increse
C. HDL
D LDL
E. CM nascent.
Situational Problems
1. A 50-years-old man consumes daily about 90 g of fat of animal origin (fatly
meat, butler, cheese), another man of the same age — 50 g offal of animal origin
and 30 g of plant oil. Which food: is healthier? For the answer:
a) name the differences in fatty acid composition of these TAGs:
b) draw the structures of the TAG molecules specific to animal fai solid at room
temperature and to olive oil liquid at the same temperature;
c) explain which TAGs arc more useful.
2. During the meal, two different persons both got some dietary lipids, but the
content of tally acids was different. First one goL the lipids containing oleic,
linolenic, linoleic, arachidonic, palmitoleic fatty acids (named in order of
its decreasing content in the fat). The latter one got the lipids containing
322 Chapter 7. Lipid metaD ol ism
palmitoleic, stearic, oleic and linoleic acids. Which person got a fat with the
higher nutritional value? To answer the question:
a) describe all possible ways of fatty acids structure coding; give the examples;
b) list all the essential components of a lipid nature;
c) explain, which lai from those two, would be more beneficial for nutritional
health, and why;
d) name the fatty acids — the precursors of biologically active compounds.
3. Clinicians work on the case of a patient with chronic pancreatitis. This disease
leads to insufficient secretion of the pancreatic juice into the small intestine.
What changes in the dietary lipid processing that disease will cause? For the
answer:
a) draw the reactions of TAG digestion and the conditions they require;
b) list all possible consequences of lipid i ndigestion;
c) list the essential lipid compounds that would have a faulty absorption due to
this disease;
d) name the other detrimental causes of lipid indigestion and malabsorption.
4. A patient with food indigestion has been screened and diagnosed with
steatorrhea (the excretion of abnormal quantities of fat with the feces owing
Lo reduced absorption of fat by the intestine). What arc the causes and
consequences of the steatorrhea? For the answer:
a) draw the scheme of TAG digestion;
b) explain the role of the pancreas in dietary lipid digestion:
cl make a scheme of dietary’ TAG assimilation, up lo the step of chylomicron
formation;
d) list the dietary recommendation for that particular patient.
5. A patient with cholelithiasis (gallstone disease) has long-term failure of the
bile salts passage into the duodenum. The observation has shown an increased
clotting lime. What are the possible causes of that symptom? For the answer;
a) name the essential nutrients that could be at a deficit in that patient;
b) explain the possible causes of the bleeding disorder;
ci name the other symptoms that can develop in that patient.
6. Scrum blood of a patient with dislipoproteinemia type 1 has milky appearance
even in fasting. If serum stays at low temperature (4’) for several hours, the fatly
layer appears on its surface. What arc the possible causes of these symptoms?
Answer the questions and do the following tasks:
a) what compounds of scrum blood must be tested in. the patient, in a
biochemical lab?
b) what is the possible diagnosis of the disease?
c) draw the reaction which docs not occur property in the patient’s blood:
d) explain how the products of the previous reaction arc used in adipose tissue
and heart in a healthy person I hour after meal.
7.. Heparin is a complex polysaccharide, a component of proteoglycans. Isolated
heparin used as an anticoagulant, binds to the protein antithrombin and inhibits
clot formation. As lipoprotein lipase is bound to the capillary endothelium
through proteoglycan, heparin also can bind to LP lipase and dislodge it from the
7.4. Synthesis of Fatty Acids ana Triacylglycerols — Upogenesis, Hormonal Regulation. Obesity 323
cytosol Acetyl-CoA becomes available for fatty acids synthesis as the initial precursor.
Next reactions of fatty acids synthesis occur in the cytosol.
Fig. 7.7. Conversion of glucose catabolism products into palmitate me main tatty acid synthesized
in humans. Acetyl-CoA is transferred from mitochondrion to me cytosol in form of citrate. Then, the
released acetyt-CoA becomes the initial precursor of palmitic acid synthesis. The whole process is
stimulated Dy insulin inducing the synthesis of enzymes involved in conversion of glucose catabolism
products in fatty acids. T— The inducible enzymes: 1 — glucose 6-pnosphate dehydrogenase (the
enzyme of pentose pnospnaie pathway}; 2 — malic enzyme; 3 — acetyl-CoA carboxylase; 4 — fatty
acid synthase complex; 5 — citrate lyase
the end product. Palmitic acid is the main fatty acid synthesized in humans. Thus. the
previous name of fatly acid synthase was palmitate synthase.
Fig. 7.8. The general structure of the tatty acid synthase complex: 1 — SH cysteinyl sulfhydryl group
to which acetyl is attached; 2 — SH phospnopafitetneinyi group to which maionyt is attached
Fig. 7.9. The active sites of me fatty acid synthase complex. ACP — acyl carrier protein, Cys —
SH — radical of cysteine
Al first, an acetyl and malonyl arc transferred from acetyl-Co A and malonyl-CoA
to the SH-groups of fatty acid synthase complex by acetyl and malonyl transferases
(Fig. 7. IO, I, 2). Next, acetyl and malonyl moictics condense with the release of the
malonyl carboxyl group as CO.. A four-carbon (3-keto butyryl is formed (3). In the next
three mac l ions, p-keto group is reduced to hydroxyl group by NAD PH, then the water
is removed to form double bond, which is reduced by NADPH (4.5,6). In the result of
the first cycle, the original acetyl group is elongated by two carbons. This sequence of
reactions is repeated until the chain becomes 16 carbons in length. In total, 7 cycles arc
required for palmitoyl to being synthci ized. Then, palmitate is released from the fatty
acid synthase by reaction of hydrolyzis. The summary’ equation of pal mi Late synthesis is:
PaJmitoyl-E
Palmitate
Fi^TTO. Synthesis of palmitate by me fatty acid synthase complex. 1 - Transfer of acetyl from acetyl-
CoA to SH group of fatty acid synthase. 2 - Transfer of malonyl from malonyl-CoA to SH group of
me enzyme. 3 — Condensation of acetyl and malonyl groups wim the release of the malonyl carboxyl
group as CO?. 4 — Reduction of the P-keta group to hydroxyl group. 5 — Dehydration. 6 - Reduction
of the double bond. 7 — Buteryi is transffered to SH group of site ®. Then the cycle repeats until the
chain becomes 16 carbons in length and palmitate is released by reaction of hydrolyzis
Two carbon atoms — l6Lhand 15th of the palmitate arc derived front an acetyl-CoA.
The other carbons in the aliphatic chain arc derived from malonyl-CoA.
7.4. Synthesis of Fatty Acids ana Triacyigiycerois — Up agenesis, Hormonal Regulation. Obesity 327
The synthesized palmitate is used for the synthesis of other lipids, mainly
triacy(glycerols and glyccrophospholipids, or can be elongated by a series of reactions
in the endoplasmic reticulum. These reactions start from conversion of palmitate
(C 16:0) to palmitoyl-Co A, which is elongated by malonyl-Co A to stcaryl-CoA
(C 18:0). Very long-chain fatty acids (C22 to C24) can also be synthesized this way,
particularly in the brain. Desaturation of synthesized saturated fatty acids is possible,
but the main products arc palmitoleic (16:1 A9) and oleic (18:1 A9) acids (Fig. 7.11).
Oxygen (OjX cytochrome b5 and NADU arc required for desaturation reactions.
Polyunsaturated fatty acids of series tn 6 like linoleic (18:2 A 9,12 w6) and like
a-linolenic (18:3 A 9,12,15 co3) acids arc not synthesized in mammals as they lack of
enzymes necessary to insert double bonds between the 9th carbon and os-carbon of
fatty acids. Humans can obtain them only from dietary plant or fish oils (essential fatty
acids). In humans, linoleic and a-linolcnic acidscan be elongated to arachidonic and
eicosapentacnoic acids, which are the precursors for the synthesis of prostaglandins
and other eicosanoids.
The regulation of fatty acids synthesis. The hormone activating fatty acid synthesis
is insulin. Insulin, stimulates both glycolysis and pentose phosphate pathway of glucose
catabolism, acting through their regulatory enzymes bv increasing their activity and
synthesis (Fig. 7.7). This regulation makes the substrates for fatly acid synthesis more
available. Insulin, also induces the synthesis of acetyl-CoA carboxylase — the rate
limiting enzyme of fatty acids synthesis and regulates its activity.
The activity of the enzyme is regulated by:
► phosphoryiation/dephosphorylation;
► allosteric modification;
► induct ion/repression of its synthesis.
in fed state. acctyl-CoA carboxylase is dephospho rydated by in.su I in-stimulated
phosphatase (Fig. 7.12). The individual dcphosphorylatcd molecules of acetyl-
CoA carboxylase, consisting of four subunits, are polymerized and become active.
Polymerization is stimulated by citrate allostcrically activating the process. The level
of citrate is elevated in fed state in cytosol of hepatocytes and so the synthesis of fatty
328 Chapter 7. Lipid metaD ol ism
acids. During fasting, acetyl-CoA carboxylase becomes inactive and the synthesis of
fatty acids is inhibited. High secretion of glucagon or epinephrine (fasting or physical
activity) causes phosphorylation and inhibition of acelyI-CoA. carboxylase by protein
kinase A. Acetyl-CoA carboxylase is also phosphorylated by AMP-activated protein
kinase and becomes inactive. AM P-acrivaled protein kinase is activated in low energy
level of the cells. AMP is the product of ATP degradation and the most sensitive
indicator of low energy levels in the cells. This regulation helps the cell to survive when
the energy level is very tow by switching off noncssential biosynthetic pathways like
fatty acids synthesis.
4
Glucagon, Epinephrine
Palmitoyl-CoA, formed from palmitate, the end product of fatty acid synthesis,
inhibits acetyl-CoA. carboxylase, through depolymerization. This is feedback
inhibition of fatly acid synthesis by the high level of the end product.
Acetyl-CoA carboxylase synthesis is also regulated by insulin at the level of
transcription. An excess food consumption causes the over secretion of insulin, so the
rate of transcription of the gene coding acetyl-CoA carboxylase increases. Low-calorie
diet (low insulin secretion) leads to the suppression of the transcription of this gene.
Triacylgliycerol synthesis occurs in the liver and adipose tissue and requires glycerol
3-phosphate and fatty acids as substrates. The origination of glycerol 3-phospha to,
which provides glycerol for triacylglyccrol synthesis, and fatty acids required for
that arc different in these two tissues. In the liver, glycerol 3-phosphate is produced
from the reduction of dihydroxyacctone phosphate derived from glycolysis or by the
phosphorylation of glycerol delivered by blood (Fig. 7.I3). The phosphorylation of
glycerol, is catalyzed by glycerol kinase only in the liver. But adipose tissue lacks the last
enzyme, therefore, dihydroxyacctone phosphate can be produced only from glycolysis.
7.4. Synthesis of Fatty Acids ana Triacylglycerols — Upogenesis, Hormonal Regulation. ODesity 329
Gtvcolysis *
I
CHnOH
I
C—O O CHpOH
I
CHj—O—p — 0“ CHOH
CHsOH
D^rydroxyacelcne
[ijlyceroiJ
ph os prate
CHj tJ
[L-Xjlycera* 3-phoaphatej
2 R-COCoA I
Acyl transferase I
2 Go ASH-Xj
0
I Liver
Crtj—0 —C—R|
i Attachment di head
ch—o—c —b2 Dup (serine, choline,
elhanolamine, etc.)
Ri
I
Ch?— o—P — o-
? Q
I
CHn" Q — C ■— R |
I O"
I ?
[Phosphatidic dzid CH-O— C”Hn
I
Gh^O h I ?
[1,2-Diacylgfycerolj CHn— O— P — O
________ 0-________
, erophosphc^pid
Acyl transferase
I
CHt— O“ c— r,
I
Ch—O—C^R2
?
I O
I
Cbt^o—c — r3
[ Triacylglycerol]
In the liver, fatty acids synthesized de now from products of glucose catabolism arc
used for triacylglyccrols synthesis (Fig. 7.14). Two fatty acyl-CoA react with glycerol
3-phosphate to form phosphatidic acid and then TAG.
'Malonyl-CSA|
Fitp 7.14. CoirverSion of glucose into fatty acids and triacylgtycerols in the liver in fed state
In adipose tissue, the fatty acids required for the TAG synthesis mainly originate
from reaction catalyzed by LP lipase (Fig. 7.15). Lipoprotein lipase attached to the
capillary endothelial cells recognizes apo CH on the surface of VLDL as it docs with
chylomicron. This enzyme hydrolyzes triacylglyccrols in VLDL and chylomicrons to
glycerol and fatly acids. Fatty acid enters the adipose cells and is converted to acyl-
CoA. Then two acyl-CoA react with glycerol 3-phosphate to form phosphatidic acid.
Dcphosphorylation of phosphatidic acid produces diacylglyccrol. Finally, the third
fatty acyl-CoA reacts with diacylglyccrol to form triacylglycerol.
TAG synthesized in. the adipocytes is the main storage of fuels in human body. TAG
synthesized in the liver (endogenous fats) are packaged into the VLD L and delivered
io other tissues. The conversion of glucose to fatty acids and TAG in adipose cells is
possible, although the liver is the major site of fatty acid and TAG synthesis.
7.4. Synthesis of Fatty Acids ana Triacylglycerols — Upogenesis, Hormonal Regulation. Obesity 331
Fig. 7.15. Conversion of the fatty acids, delivered as TAG of chylomicrons and VLDL to Iriacylglycerols
stored in adipose tissue, insulin stimulates the synthesis and secretion of LP — tipase from adipose
cells to capillary walls and activates GLUT — 4 in plasma membranes, allowing glucose to enter the
cells. Thus, the synthesis of triacylgiycer-ots increases in the adipose ceils in fed state. FA — fatty
acids, OHAR — dihydroxyacetone phosphate
Transport of endogenous TAG from the liver to the peripheral tissues. The
triacylglycerols synthesized in cytosol of the hepatocytes, arc packaged into the
VLDL — ver? low density lipoprotein, together with phospholipids, cholesterol, and
proteins. The structure of VLDL particle is similar lo the structure of chylomicron
(Fig. 7.4). The major apoprotein of VLDL is apo B-100 wound through the surface
of VLDL particle. MTP — the microsomal triglyceride transfer protein, required for
chylomicron formation, is also required for VLDL formation. VLDL, formed! in the
liver as nascent panicles, arc secreted lo blood where they acquire apoC i I and apoE
from HDL to become mature VLDL particles (Fig. 7.I6). The function of VLDL,
containing 55% of triacylglycerols, is to deliver triacylglycerols synthesized in the liver
to other tissues, particular to adipose tissue and muscles.
In blood, LP lipase hydrolyzes triacylglycerols in VLDL and the content of
TAG in Lhe particles decreases. The released fatty acids arc transported lo the cells
mainly to adipocytesand hepatocytes. Glycerol is delivered to the liver. Thus, VLDL
becomes al first IDL (intermediate density lipoproteins), and then LDL (low density
lipoproteins). LDLs contain only abouL 7%- of TAG. The major compounds of LDL
arc cholesterol and cholesterol esters (Fig. 7.I6, Table 7.3). In Lhe liver TAG of VLDL
and chylomicrons arc also cleaved by hepatic triglyceride lipase (HTGL) which is also
attached to the capillary walls. The subsumes of this enzyme are lipoproteins already
partial!v digested by LP lipase. The hepatic triglyceride lipase facilitates conversion of
IDL into LDL
332 Chapter 7. Lipid metaD ol ism
Liver
(vlDl nascent j
(55% TAG, 15% C + CE)
ApoE
[ VLDL mature^
LP -Lipase
3 RCOOH
Glycerol
LP -Lipase
3 RCOOH
Glycerol
fig. 7.16. Metabolism of VLDL in blood HOL donates apoCli and apuE to nascent VLDL converting
inem to mature VLDL. Then LP lipase hydrolyzes TAG in VLDL. The amount of TAG in the particles
decreases and they become at first IDL (intermediate density lipoproteins} and then LDL (low density
lipoproteins}. C + CE cholesterol and cholesterol esters
[n case of LP lipase deficiency, the levels of VLDLs arc elevated as well as levels
of chylomicrons and TAG, and dislipoproteinemia type 1 develops. Patients with this
disease have exceedingly high plasma triacylglycerol concentration (over l()00mg/dl,
normal range <160 mg/dl). These patients suffer from eruptive xanthomas (yellowish
triacylglycerol deposits in the skin) and chronic pancreatitis.
Obesity. As the fatty acids in tri acyl glycerols of adipose tissue conic both from
chylomicrons and VLDL, humans produce fat stores both, from dietary fat delivered
by chylomicrons and dietary carbohydrates, which produce fat of VLDL. Therefore,
overeat i ng fat, carbohydrates and even proteins leads to excess accumulation of fat and
obesity.
Recent investigations have shown that the adipose tissue is not only the storage
of triacylglycerols but also is active endocrine organ secreting different hormones
and other regulatory factors (Fig. 7.17). The secretion of these regulatory factors
significantly changes in case of obesity. One of the first discovered factors was leptin —
the peptide released from adipocytes as their fat accumulation increases. Leptin, binds
to receptors in hypothalamus and stimulates the release of neuropeptide V into blood
that signal the cessation of eating. I?ut in some obese patients leptin, receptors become
less sensitive to leptin that leads to dysregulaiion of appetite and weight gain. Leptin
7.4. Synthesis of Fatty Acids ana Triacyigiycerois — Upogenesis, Hormonal Regulation. Odesity 333
Adiponectin LJP-Lipase
Angiotensin ll
Eicosanoids
FFA
IL - 6
Fig. 7.17. Secretion of honnooes and other Biologically active molecules By adipocytes. Leptin and
resistin secretion increase in case of obesity that cause the cellular resistance io insulin. Tne level of
free fatty acids (FFA) in D loo a is elevated. Secretion of adiponectin which increases the sensitivity of
the tissues to insulin decreases with the accumulation of fat in me adipocytes. IL — 6 interleukin —
stimulates acute phase protein synthesis
Fatly acids secreted From adipocytes arc transported by blood and taken up by the
liver. Some of fatty acids entered the hepatocytes can regulate the gene expression.
Faliy acids bind to the special receptors named PPARs — peroxisome proliferator-
act hated receptors (sec Ch. 7.6). These receptors arc members of a nuclear receptor
family and, when, activated, regulate the rate of transcription. This mechanism
resembles the action of steroid hormones, which arc also hydrophobic molecules, but
the receptors of hormones arc more selective.
The development ofobesity is a major problem worldwide and is associated with the
failure of energy balance mechanisms and regulation of food intake. Clinical obesity is
now clearly defined in terms of height and weight through the body mass index (UM I),
which is calculated as the weight in kilograms divided by the height in meters:
Weight (kg)
BMl =----------------- .
Height (m)2
some forms of cancer. As obesity is the primary risk for so many diseases, it has to be
treated intensively.
The normal quantitative characteristics of the TAG and fatty acids metabolism.
Blood:
TAG < 160 mg/dl:
Free fatty acids 1.0-25 nig/dl.
Review Test
1. Fatly acid synthesis. Match the figure with the letter:
HS
A. NAD PH.
B. Malonyl-CoA.
C. Acetvl-CoA.
D. NADP4.
7.4. Synthesis of Fatty Acids ana Triacylglycerols — Upogenesis, Hormonal Regulation. ODesity 335
2. Regulation of the key enzyme of the fatty acid synthesis. Match the figure with the letter:
(p
A. Cortisol.
B. Glucagon.
C. Phosphatase.
D. Pyruvate.
E. Insulin.
3. TAG synthesis from products of glucose ncthabolism. Match the figure with the letter:
[Glucogej
CHa-O— C— Ft 3 CHn—QH
Triacylgiycerol ] C3}
336 Chapter 7. Lipid metaD ol ism
A. TAG.
B. DAG.
C. Glycerol 3-phosphaic.
D. Dihydroxyacetonc phosphate.
E. Phosphatidic acid.
4. Conversion of glucose in fatly acids and Iriacylglycerok in the liver in fed state. Match
the figure with the letter:
H.3PO4
A. Malonyl-CoA.
B. Phosphatidic acid.
C. Glycerol-3P.
D. Monoacylglyccrol.
7.4. Synthesis of Fatty Acids artel Triacyigiycerois — Upogenesis, Hormonal Regulation. Obesity 337
Situational Problems
I . What are the numbers of carbon atoms derived from the first acelyl-CoA that
binds to fatly acid synthase complex in palmitate, the end product ofthe pathway?
For the answer:
a) name the initial substrates for fatty acid synthesis and their source;
b) draw die reactions of the first spiral of fatly acid synthesis;
b) mark the carbon atoms (*> derived from the first acc-tyl-CoA in the product of
the first spiral of fatty acid synthesis;
c) answer the question of the problem.
2. After gelling 300 g of carbohydrates with her meal, a student went to bed. What
metabolic pathways of fatty acids have been activated in the liver about an. hour
after having a meal? For the answer:
a) describe the glucose metabolism in the liver in this condition;
b) name the metabolic pathway of fatty acids which, has been activated:
c) draw the scheme of this pathway and explain the fate of the end products of
the pathway in the liver;
d) explain the regulation of the pathway.
3. A patient has got excess carbohydrate meal for the years and gain the weight. To
explain this:
a) draw the schemes of TAG synthesis in the liver;
b) describe the transport of TAG from the liver to adipose tissue;
c) describe the functions of insulin in the conversion of glucose to TAG in the
liver and adipose tissue.
4. Glucose containing l4C atoms was added to isolated hepatocytes in an experiment.
If the glucose was added in excess, the rale of triacylglyccrol synthesis increased:.
The determination of carbon atoms in the triacylglycerol molecule confirmed
that all carbon atoms in glycerol and fatty acids were 14C atoms. Explain the
results of the experiment. For that:
a) name the metabolites required for triacylglycerol synthesis, which arc
produced by glucose catabolism:
bldraw the scheme of glucose catabolism to metabolites required for
triacylglyccrol synthesis which would explain the appearance of “C atoms in
the triacylglyccrol molecule:
c)draw the scheme representing the conversion of extra glucose to
triacy I glycerols in. the liver.
5. One student has got 300 g of carbohydrates and 50 g of proteins with dinner,
another — 300 g of carbohydrates and 50 g of fats. Both d id not carry out any
physical activity. Explain the difference in the lipoprotein composition in their
blood one hour after the meal. For the answer:
a) name all types of lipoproteins and describe their structure and functions:
bldraw. the schemes explaining the difference in the lipoprotein formation in
both students:
c) name the methods used for lipoproteins analysis of scrum blood;
d) draw the scheme of electrophoresis of sc rum lipoproteins for both students.
338 Chapter 7. Lipid metaD ol ism
AM P system, rising the cAMP level in the adipocytes (Fig. 7.18). Protein kinase A is
activated by cAMP and phosphorylates hormone sensitive lipase (HSL), also called
adipose triacylglyccrol lipase. This enzyme cleaves a fatty acid from a triacyl glycerol.
Then other lipases complete hydrolyzis to glycerol and fatty acids, which are released
into the bloodstream. Fatty acids, as water insoluble molecules, bind to the major
serum, protein albumin and are transported to muscles and other tissues, where; they
are oxidized to CO, and water to produce energy. Glycerol travels to the liver and
serves as a substrate of gluconeogenesis during prolonged fasting.
Fi&7.18. Mobilization ol stored fats from adipose tissue. R — receptor: G — G-protein; AC — aaenyiate
cyclase
Recently, an important mechanism regulating the release of fatty acids from the
adipocytes has been described. It has been revealed that up to 40% of fatty acids
released from lipolysis arc rc-cstcrificd to triacvlglyccrols in the adipocytes. The
glycerol-3 phosphate required for triacylglycerol rosy n thesis in adipocytes is formed
by the shortened pathway of gluconeogenesis. The main substrates of gluconeogenesis
arc lactate and amino acids which arc taken from the blood. This mechanism controls
the amount of fatty acids, leaving the adipocytes during prolonged fasting.
340 Chapter 7. Lipid metaD ol ism
+ N (CH3)s
Carnitine il
Carnitine palmitoyl
transferase I (CPT I)
O ch2
CH
I 1
CHa—(CH^“C—O —CH—CH£—COOH + CoASH
[Acylcarnitine]
7.6. Utilization uf Fatly Acids For Energy Production - ^-oxidation. Disorders of p -oxidation... 341
Acylcarnilinc and free carnitine arc then exchanged across the inner mitochondrial
membrane by carnitine aeylcarnitinc translocase. Finally, the fatty acyl group is
transferred back to CoA by carnitine palmitoyltransferase II (CPT 11) on the matrix
side of the inner membrane. This process functions primarily in the transport of fatly
aeyl-CoAswith 12-18 carbons. Entry of shorter chain fatty acids into the mitochondria
is independent of the carnitine; they cross the inner mitochondrial membrane and arc
converted to CoA derivatives in the matrix.
Outer
mitochondrial
membrane
inner
mitochondrial
membrane
Matrix
p-oxidation ■*— Acyl-CoA CoASH
reaction of she sequence, the bond between a- and [3-carbons is cleaved by an enzyme
P-kelolhiolasc. The products are the acetyl-CoA and the fatty acyl-CoA that is two
carbons shorter than the original The shortened fatly acyl-CoA repeats these four
steps until all of its carbons are converted to acetyl-CoA. In the last spiral, cleavage
of the four-carbon acyl-CoA produces two acetyl-CoA. Thus, an even-chain fatly
acid such as palmitoyl-CoA, which has 16 carbons, repeats the 7 spiralsand produces
8 aceiyi-CoA, 7 FADH2 and 7 NAD FL
O
fi a
CH3W CH2“jCH2- - CHq- C~SCoA Acyl-CoA
4
Cn
Acyf-CoA
dehydrogenase
2 ATP
Enoyl-CoA
|^-h2o
Enoyl-CoA hydratase
.Q OH O
—
d
CH3wCH2—CH—CH2 C p-hyd roxyacyl-CoA
£
9
NAD’
ETC fS-hydroxyacyt-CoA
NADH+H’’ —■ 3 ATP dehydrogenase
O
1
CHjwCH; CHs -C~SCoA p-Ketoacyl-CoA
CoASH
p-Ketothiotase
CH3v^CH?—
The complete oxidation of fatty acid up to CO. and H,0 includes p-oxidation
pathway, oxidation of acetyl-CoA in the TCA cycle and oxidation of FADH, and
NADH in ETC. The theoretical energy yield from the oxidation of I mol of palmitoyl-
CoA to 8 mol of acetyl-CoA is, therefore. 35 mol of ATP: 2 for each of the 7 FADH.,
and 3 for each of the 7 NADH (7x2 +7x3= 35 ATP). The oxidation of 8 mol of
acetyl-CoA in the TCA cycle produces 96 mol of ATP (8x12). Thus, the complete
oxidation of palmiloyl-CoA io CO2 and H,0 produces 131 mol of ATP. To calculate
the energy yield from I mol of palmitate, two ATP need to be subtracted from the total
because two high energy bonds are cleaved when palmitate is activated to palmiloyl-
CoA (35+96-2= 129). The real production of ATP from the oxidation of palmitate is
approximately 106 mol due to the dissipation of energy.
Each of the four reactions of p-oxidation is catalyzed by several different enzymes
that have specificity for fatty acids of different chain length. The long-chain fatly acyl-
CoA starts to oxidize by long-chain fatty acyl-CoA dehydrogenase (LOAD). As the
length of the acyl chain becomes shorter, it is transferred to the enzyme that acts on
shorter acyl chains. Medium-chain fatly acyl-Co A dehydrogenase (MCAD) and short
chain fatty acyi-CoA dehydrogenase continue to oxidize fatly acyl to acetyl-CoA.
Regulation of the p-oxidation. Fatty acids arc used as fuel mainly when they arc
released from adipose tissue in response to hormones that signal fasting (glucagon) or
physical activity (epinephrine). The process of p-oxidation is regulated by the cells1
requirements for energy (N.ADH/NAD and ATP/ADP ratio), as it depends on. the
rate of NADH and FADH2 rcoxidation in the electron transport chain. Another type
of regulation occurs through the key enzyme carnitine: palmitoyl transferase I (CRT I).
The activity of this rale-limiting enzyme determines the rale of fatty acids transport into
the mitochondrial matrix where p-oxidadon occurs. Carnitine: palmitoyltransferase I
is inhibited by malonyl-CoA synthesized by acetyl-CoA carboxylase. Malonyl-CoA
is the first metabolite in the pathway of fatty acids synthesis activated by insulin.
Thus, if fatty acids synthesis starts in. a fed' state, malonyl-CoA is formed and inhibits
the regulatory enzyme of P-oxidation and fatty acids arc not oxidized. Glucagon
and epinephrine inactivate the enzyme synthesizing malonyl-CoA — the inhibitor
of P-oxidation (Ch. 7.4), so in fasting or physical activity, the rate of p-oxidation
increases. Low levels of citrate and high levels of acyl-CoA also stimulate P-oxidation
by action on the acetyl-CoA carboxylase. As the rate of P-oxidation depends on
substrate availability, it increases when fatty acid levels are elevated in blood under the
action of glucagon or epinephrine.
Disorders of p-oxidation. Recently, medium-chain fatly acyl-CoA dehydrogenase
(MCAD) deficiency has emerged as one of the most common of the inborn diseases.
The frequency of MCAD deficiency is 1 in 15 OCHJ. In MCAD deficiency, fatty acids
arc oxidized only until they reach medium chain IcngLh. As a result, p-oxidation is
not completed, and ATP synthesis reduces. Therefore, the body must rely mainly on
oxidation of blood glucose to meet its energy needs. The most frequent manifestation
of MCAD deficiency is low levels of glucose and ketone bodies in the blood during
fasting (intermittent hypokciotic hypoglycemia). In healthy individuals, P-oxidation
is needed to produce acetyl-CoA for ketogenesis and ATP for gluconeogenesis during
fasting. In MCAD deficiency both pathways arc compromised, thus, hypoglycemia
344 Chapter 7. Lipid metaD ol ism
and low levels of ketone bodies in the blood develops. The combined deficiency of
ketone bodies and glucose is especially bad for the brain as it depends on a mix of
ketone bodies and glucose during prolonged fasting. The management of MCAD
deficient patients includes the intake of a relatively high carbohydrate diet and the
avoidance of prolog fasting (more than 6-7 hours for adults).
Another cause of fl-oxidation defects is carnitine deficiency. Il can be caused by
impaired carnitine synthesis in the kidneys and liver. Carnitine deficiency in skeletal
muscle causes muscle weakness and cramps on exertion.
Mitochondrial p-oxidation oxidizes unbranchcd saturated fatty acids with even
number of carbon atoms and a chain length up to 20 carbons, fatty acids with other
structures arc oxidized through some other metabolic pathways.
► Oxidation of unsaturated fatty acids requires modifications of their double bonds
before p-o.xidation. Their complete oxidation produces 2 ATP less for each
double bond then the saturated fatty acid with the same number of carbons.
* a-Oxidation is a minor pathway in the peroxisomes, mitochondria, and ER.
It oxidizes ct-carbon of fatty acids releasing CO3 and is needed for catabolism
of methylated fatty acids. In Refsum disease, a recessively inherited defect of
peroxisomal a-oxidation causes the accumulation of branched-chain fatty
acid - phytanic acid which originates from chlorophyll in green vegetables
and accumulates in the fat. meat and milk of ruminants. Refsum disease leads
to slowly progressive peripheral neuropathy and blindness. As phytanic acid
is obtained only from diet, placing patients on a low — phytanic acid diet has
resulted in marked improvement.
► u>-Oxidation is the oxidation at the co-carbon. the fatty acid (the terminal methyl
group} by the enzymes in the endoplasmic reticulum. These enzymes need
cytochrome P450, CL and NAD PH. Fatty acids arc oxidized to dicarboxy lie
acids with 6 to 10 carbons. Dicarboxylic acids undergo P-oxidation or excrete in
urine as water-soluble products.
► Peroxisomal p-oxidation. Peroxisomes arc present mainly in the liver and
contain the enzymes for the oxidation of very-long-chain fatty acids with length,
of 20 carbons or more. This pathway is similar to mitochondrial p-oxidation,
but in the very first reaction peroxide (H2O2) is produced instead of FAD PL.
The sequence of the other reactions remains the same, releasing the NADH and
acetyl-CoA. Peroxisomal catalase inactivates hydrogen peroxide by conversion
to water and O2. The octanoyl-CoA that, is the end product of peroxisomal
oxidation leaves the peroxisome and the octanoyt radical is transferred through
I he inner mitochondrial membrane by the medium chain-length acylcarnitinc
transferase. In the mitochondrion, it enters the P-oxidation pathway and then is
oxidizes to CO, and water.
The function of the peroxisomal a- and p-oxidation and microsomal m-oxidation
is to decrease levels of water- insoluble fatty acids or xenobioiic compounds with a fatly
acid like structure that would become toxic to cells al high concentration.
Peroxisome pro I iterator-activated receptors (PPARs) play an important role in
lipid metabolism in the liver. The PPARs arc members of a nuclear receptor family
and, when activated, stimulate the gene transcription, in the liver, PPARs regulate
7.7. Synthesis artel Oxidation of Ketone Bodies. Ketonemia, Ketoacidosis S45
the activity of the genes that arc involved in fatly acid intake and oxidation. There
arc three major PPAR isoforms a, p and y. The major form in the liver is a. Fatty
acids are the endogenous ligand for PPARa. When the levels of fatty acids in blood
and hepatocytes are increased, there is increased gene transcription, for the enzymes
involved in fatly acid oxidation. The drugs ftbrates bind to PPARs in the liver activate
them and improve lipid me La holism. The ftbrates arc prescribed for the patients with
elevated triacylglyccrols because they increase the rate of TAG oxidation. This, in
turn, leads to a reduction, in scrum TAG levels.
Blood
Brain
Heart
Glucose
Muscles
Glucose
I
Acetyl-CoA Acetoa estate - jyhydroxyDutyratG
Oxalacetate
;
1
tca-
cyclo /
;
I
Acetoacstyl-CoA
Z Mitochondrion
Amino acids
Hepatocyte Cytosol
Fig. 721. Fatty acids, ketone bodies aiid glucose metabolism in fasting
in blood increases and they become an important energy source for many tissues,
including the brain, thereby sparing the use of glucose. But ketone bodies cannot be
oxidized in the erythrocytes because they tack mitochondria. Cells transport both
acctoacciatc and p- hydroxy butyrate from the blood in the cytosol and then into the
mitochondrial matrix. There p-hydroxybutyrate is oxidized by p-hydroxybutyrate
dehydrogenase back to acctoacciatc (Fig. 7.23). This reaction produces NADH which,
can be oxidized in ETC. Acctoacctate is activated by accepting CoA from succinyl-
CoA. The reaction is catalyzed by succinyl-CoA: aceloacetatc-CoA transferase. The
enzyme is absent in the hepatocytes, therefore, the liver produces ketone bodies, but
cannot utilize them. Next reaction is cleavage of acctoacctyl-CoA into two acctyl-
CoA. which enter the TCA cycle and arc oxidized producing ATP. The energy yield
from oxidation of acctoacciatc, in theory, is equivalent to the yield for oxidation
of 2 molecules of acetyl-CoA in the TCA. cycle minus the energy for activation of
acctoacciatc (2* 12-1-23ATP). Oxidation of p-hydroxybutyrate produces one
additional NADH. Therefore, the energy yield from oxidation of p-hydroxybutyrate is
26 molecules of ATP but due to the dissipation of energy in reality it is approximately
21,5 molecules of ATP.
Regulation of ketone body metabolism. The rate-limiting enzyme of ketone bodies
synthesis is 3-hydroxy-3methylglutaryi-CoA synthase (HMG-CoA-synthase). The
synthesis of this enzyme is stimulated by fasting, insulin deficiency and excess dietary
fat consumption. Fatly acids levels arc elevated in blood in these conditionsand a large
amount of fatty acids arc taken up by the liver. Fatty acids arc powerful inducers of
HMG-CoA synthase. The inhibitor of H MG-CoA synthase is free CoASH. The level
7.7. Syntnesis artel Oxidation of Ketone Bodies. Ketonemia, Ketoacidosis 347
O O
J^COASH
p-kstotnidase
o
I
CH-C
Cbk
1
C-O
SCoA O
K- CH3- C - SCdA
HMGJDoA
synthase
P* CoASH
S-HydfCwy-S-methyl
-glulafyl-CoA (HMG-CgA)
HMG-CoA
lyase
Acetyl-CoA
0
CH3 C-CHa- C -O’ (Acetoacgr^J
'/* NADH + H*
(Wiydraxybutyfata r _
Spontaneous
dehydrogenase
NAD*
CH3—
Fig. 7.22. Ketone Dodies synthesis. p-Hydroxybutyraie and acetoacetale are ketone Dodies wtiicfi
oxidation produces ATP; acetone is produced Dy nonenzymatic decarboxylation of acetoacetate in
case of nigh levels of ketone Dodies in blood
Oh o
✓
Ch3—C— Cnr C — O"
i D-fl-hydroxy tiu:yraiej
NAO+
|54iydraxytHjlyrate
dehydrogenase
NADrt -t-H+
a o
E I
CH3— C—CHn—C— 0"
t Ace La ace Sate )
Succinyl-CoA: Sucdnyl-CoA^-
AcelcacelJale-CoA
transferase Sucdnale
Q 0
1 1
Crt3 — C“ CH^- C SCoA
(Acetoapelyl-CoA ]
CoASH
Thiolase
O O
I
Cl-h-C -SCoA + CH.3- C - SCdA
2 x C Acetyl-CoA ]
OH OH
I _______ ~ I
ch3—c—ch?— cooh -*------- ► ch3—c—ch2— COO + H
|3 - hydroxybutyratc
Review test
1. TAG mobilization from the adipose tissue. Match the figure with the letter.
□
Praeii Kinase A
inactive
13
£3
Prosin Kinase A
aciiv2
QudBDOn
miissues
Adipocyte Blood
A. cAMP.
B. MAG.
C. Insulin.
D. Glucagon.
E. L.P lipase.
2. P-Oxidalion of fatty acids. Match the figure with the letter.
o
Enoyl-CaA
©
Oh O
ni 1
CH3W0+2— CH—CH?— C - SCcA |l hydic^j*zy: Cn-'
naef
naDh+h*
©
fl-KciMcyfrCaA
CcASH fr-KcfcfnLrx
1
i!-SCnA + CHj —C-SCoA
CHj^—
I
TCA-cyoc
350 Chapter 7. Lipid metaD ol ism
fi-kelotrialasc
Is* CoASH
{Acetoace tyf-CoAj
K—CHs— C - SCoA
P* CoASH
OH 0
I
CHa —C— Crtj— C — O
CH?
C«"O
4
SCoA
ch3-c—ci-k—c—o-
? L—£-----J
OH F
1
MADH + H*
fb~ p-hydrorybutyrate'
A. Acctoaceiatc.
B. Acetone.
C. Fatty acvl-CoA.
D. H MG-CoA.
E. Acetate.
7.7. Synthesis artel Oxidation of Ketone Bodies. Ketonemia, Ketoacidosis 351
CHa—C—Crtj—
f 2 1
L—NAD+
p* NADH + H*
0 0
1 1
Ch3— C—CMj—C — O"
Sucdnyl-CoA
p* Sucanale
o Q
1 1
ch3-c-- CHjt’- G’-’ SCoA.
AceLoacetyt-CoA]
CaASH
A. Acetyl-CoA.
0
1
r
GH3—C - SCoA + CH3—C - SCoA
□
I
B. p-hydroxybutyraic.
C. Acctoacctatc.
D. HMG-CoA.
E. Fatty acyl-CoA.
Situational problems
1. Following a physician recommendation, an overweight man, 40 years old,
decided to start a healthy lifestyle: daily running for an hour, fat consumption
reduction Lip to 20 g in a day, as well as a reduced amount of *fast carbohydrates*.
In result, he lost 2.5 kg of his body weight during the following two weeks. What
metabolic pathway must be accelerated to cause such fat accumulation drop in
this man? Answer the questions and do the following tasks:
a) what is an adult norm for fat and carbohydrates daily consumption?
b) draw the scheme, showing the influence of persistent physical exercising on
lai metabolism in adipocytes;
c) draw the scheme of the metabolic pathway, where products of lipolysis arc
used in skeletal muscles during persistent physical exercise.
2. How docs the fatty acids concentration changes in the blood during prolonged
fasting? To answer the question:
a) name the hormone which manages metabolism in. this condition;
b) draw the scheme of this hormone action on the adipose tissue:
352 Chapter 7. Lipid metaD ol ism
c) draw the scheme of the main metabolic pathway providing energy Tor the liver
during prolonged lasting.
1 After 12 days of starvation, a man reduced his weight by 4 kg. The doctor
prescribed him biochemical blood test to analyze the level of lipids in serum
blood. The results showed an elevated level of free fatty acidsand triacylgtyccrols.
Explain the results of the biochemical blood test. For that:
a> name the main, hormone which manages metabolism during starvation;
b) draw the scheme of this hormone action on the adipose tissue;
c) name the possible pathways of fatty acids usage in the liver during starvation
and write down the proper schemes.
4. A sportsman is doing a 10 km marathon for 1.5 hours. What changes in lipid
metabolism in adipose tissue and muscles arc triggered by long-term physical
activity? To answer the question:
a} name the hormone, controlling the lipid metabolism in adipocytes and write
down the scheme of the process;
b) explain. how some of the products of lipid mobilization, transported from
adipocytes to the bloodstream, would be utilized by skeletal, muscles and liver
in these conditions;
c) calculate the ATP yield in the result of palmitoleic acid oxidation.
5. A long-distance runner trained for a. year to achieve better results. As he trained,
he increased his mitochondrial capacity, as well as his oxygen delivery to the
tissues. Why did these physiological mechanisms of adaptation to intensive
physical activity allow him to win the competition? For the answer
a) name the hormone which controls the metabolism during physical activity
and the fuel molecules secreted to blood from adipose tissue during prolonged
physical activity;
b) draw the scheme of their oxidation in the muscles;
c) explain why the improved oxygon delivery’ to the tissues will affect this
pathway.
6. The popular those days energy drinks have caffeine, theophylline, and
carnitine among other compounds. Caffe inc and its analog theophylline
inhibit the enzyme phosphodiesterase (Ch. 4). Such drinks arc made fizzy
(carbonated) to escalate its absorption and the effect. What arc the effects of
these components on a lipid metabolism? Answer the questions and do the
following tasks:
a) wrhal metabolic cascade in the adipose tissue would be influenced by caffeine
and theophylline? Explain the mechanism of their action:
b) draw the scheme of metabolic pathway for which carnitine is required:
c) explain the name ^energizer* for this sort of drinks.
7. An experimental animal was tested for fatty acids concentration difference on
1st and 30th minute of the workout: in the inflow blood, feeding the actively
working muscle, versus the outflow blood, coming from this muscle. In what
case difference will be more prominent? To answer
a) draw the reactions of the metabolic pathway, responsible for the difference in
the fatty acid concentrations in these conditions;
7.7. Synthesis ana Oxidation of Ketone Bodies. Ketonemia, Ketoacidosis 353
(Fig. 7.25). The Greek letter subscript in the F series (PGFa) denotes the position
of hydroxy] group below the plane of the ring at carbon. 9, as does hydroxyl group
at carbon I I. The types of prostaglandin, as designated by the capital letter, have
different and sometimes antagonistic biological effects. The number of double bonds
in the side chains affects the potency of the prostaglandin but not the kind of effect
on the particular target tissue.
C2D2£mi,14
(precursor)
(jj6
Eicosatrienoic acid
(dihomo - y - linolenic acid} PGE!
CO0H
CxrSASB, 11,14, E7
(precursor)
£o3
[Eicosapentaonoic acid)
Fig. 7.25. Prostaglandins of the 1.2 aim 3 series aim their precursors
Tatle 7.4. The biological erieds of me main eicosanoids {derivatives of arachidonic acid)
PGEg Stomach, kidney, heart, endometrium, Suppress gastric acid secretion, reduce the risk
vascular endoth elial cells of gastric and duodenal ulcer
White blood cells Vasodilatation, induction of uterine contractions,
inhibition ol platelet aggregation
Mediator ol inflammation
TXA, Platelets, white blood cells Induces platelet aggregation and vasoconstriction
Mediator ol inflammation
The synthesis of the eicosanoids. Arachidonic acid, the main precursor of the
eicosanoids, yields the 2 series (2 double bonds in rhe side chains) of prostaglandins
(Fig. 7.25). The I. and 3 series of prostaglandins arc derived from cicosairienoic
acid — C 20:3 I AS, 11.14 cub) and cicosapentaenoie acid — C 20:5 (A 5,8,11,14,
17 m3). These acids arc cstc rifled to phospholipids located in the lipid bi layer of
the plasma membranes of the cell. The synthesis of the eicosanoids starts with the
rate-determining hydrolyzis of arachidonate (or another 20-carbon polyunsaturated
fatly acid) from the 2nd carbon of glycerol of membrane glyccrophospholipids. The
reaction is catalyzed by a receptor-activaicd phospholipase A3 (PLA2) (Fig. 7.26).
Molecules that act as primary signals for eicosanoid synthesis bind to the cell
membrane receptors and activate the phospholipase Ar Some primary messengers
activate inositol triphosphate system of signal transduction in the target cells.
The formed second messenger diacyiglycerol (DAG) activates membrane-bound
phospholipase A, which hydrolyzes phosphatidylcholine located in the membrane.
Released arachidonic acid or another 20-carbon polyunsaturated fatty acid arc the.
7.8. Structure Metabolism and Eiologicai Effects of Eicosanoids 357
0 CHL—O^C— r,
,-Uo-Ah
Ahj— O—®—Chriine
Membrane phospholipid
Phospholipase A2
Q Glucocorticoids
cyt
Epoxides -P4b0
Lipoxygenase
Arachidonic acid
(Cxr-tAS.fiJ1,14;)
Cyclooxygenase
—I Aspirin and other NSAJDs
Prostaglandins
The eicosanoids have a wide variety of physiologic effects ( Fable 7.4) which arc
initiated though the interaction of the eicosanoid with a specific receptor on the
plasma membrane of a target cell. These receptors arc connected with adenylate
cyclase or guanylatc cyclase systems or cause an increase in the level of calcium in
the cytosol of the target cells acting through inositol triphosphate system. Some
eicosanoids modulate the degree of systems act ivatio n in response to other stimuli. If
the eicosanoid binds to the stimulatory subunit connected with the receptor the effect
of the stimulus is amp I i heated. If it activates inhibitory subunit, the cellular response
is reduced. Thus, the eicosanoids contribute to the regulation of the cellular functions
by hormones and other primary messengers.
Many eicosanoids have antagonistic biological activities. Prostacyclin tPGlj
and thromboxane (TXAp arc potent antagonist regulators of platelet aggregation,
thrombus formation and smooth muscle tone. Normal vascular endothelium
7.8. Structure Metabolism and Eiologicai Effects of Eicosanoids 359
Fig. 721. Regulation of vascular smooth muscle tonus Dy prostacyclin ir intact vascular endothelial
cells produce PGi?which inhibits platelet aggregation and causes vasodilation. Nitric oxide released by
me enuotneiiai ceils nas the same effect on tonus of smooth muscle ceils
Daily consumptions of low-dose aspirin inhibit the cyclooxygenase and block the
production of thromboxane TXA2 thus decreasing the risk of thrombus formation and
preventing the acute myocardial infarction (Fig. 7.29). A lot of researches report about
the effect of the diet that include cold-water fish with a high content of polyunsaturated
fatty acids ofco-3 family (C:20 A 5,8 J 1,14,17) on prevention of heart attack. This diet
increases the content of to-3 fatty acids in membrane phospholipids. In this case, the
cicosapentacnoic acid becomes the initial substrate for thromboxane synthesis and
more TXA, is produced relative to TXA,. TXA.S is less effective in stimulating platelet
aggregation than its counterpart TXA, But the discussion about benefit oftu-3 fatty
acids is still continued.
As the prostaglandins arc important in mediating the inflammatory response, drugs
that block prostaglandin synthesis should provide relief from inflammation symptoms.
The cyclooxygenase enzyme is inhibited by all nonsteroidal anti-inflammatory drugs
(NSAIDs) such as aspirin. Acetyl group from aspirin covalently binds to the enzyme,
irreversibly inactivating it.
Fi^ 7.29. Inactivation of cyclooxygenase hy aspirin. Acetyl group of aspirin binds by covalent hond to
Hydroxyl in the active site of cyclooxygenase causing its irreversible inhiDition
Review test
I. Match the figure with the letter:
A. TxA,.
B. TxA^.
C PGEr
D. PgCr
E. arachidonic acid.
2. Match the figure with the letter. Inhibitors of the enzymes:
Membrane
pnospnoiipids
L*—Phospholipase
Aracnidonic acid
Leucotriens Prostaglandins
362 Chapter 7. Lipid metaD ol ism
A. Glucocorticoids.
B. Aspirin.
C. Vitamin D.
D. Vitamin E:-E glucagon.
3. Choose lhe correct answer. The Ley enzyme of proslagtan din synthesis is:
A. Lipoxygenase.
B. Cyclooxygenase.
C. Peroxidase.
D. Phospholipase Ar
E. Adenylate cyclase.
4. Choose lhe correct answer. The key enzyme of leukotriene synthesis is:
A. Lipoxygenase.
B. Cyclooxygenase.
C. Catalase.
D. Phosphodiesterase.
E. Adenylate cyclase.
Situational problems
1. Rare side effect of aspirin is the development of acute asthmatic bronchospasm
after taking a pill ofaspirin. Why can this coni plication can develop? For that do
the following tasks and answer the question:
a) explain the mechanism ofaspirin action;
b) draw the scheme needed for the explanation ofaspirin action on the metabolic
pathway;
c) explain using the scheme the possible cause of this side effect of aspirin;
d) why the administration of glucocorticoid analog dexamethasone significantly
reduced the symptoms?
2. Aspirin ceases the symptoms of inflammation and reduces the rate of blood
coagulation. How the daily consumption of low-dose aspirin (cardio aspirin.)
can prevent acute myocardial infarction?
For the answer:
a) name the enzyme inhibited by aspirin;
b) explain the mechanism ofaspirin action on the enzyme;
c) name the eicosanoids which synthesis is inhibited by aspirin:
d) explain why the rate of blood coagulation is reduced despite lhe aspirin
inhibiting simultaneous synthesis of the both, eicosanoids act ingas antagonists
in blood coagulation.
both playa role in the generation of lipid-signaling molecules. Glyccro phospholip ids
also form the phospholipid monolayer on the surface of blood lipoproteins.
Glyeerophospholipids (phosphoacylglycerols) arc the derivatives of phosphal id ic
acid (PA) containing glycerol two fatty acids and a phosphate group on the Ehird
carbon (Fig. 730). The initial steps in the synthesis ofglycerophosphol ip ids arc similar
to those of triacylglyccrol synthesis (Ch. 7.13). Glycerol 3 — phosphate derived from
di hydroxy acetone phosphate the product of glycolysis subsequently interacts with two
acyl-CoA to form phosphatidic acid. Then different polar «hcad» groups are added to
the phosphate.
"dl
zCH3‘
Pnospnatidyt-
— CK-2—ch2—nh-3+
ethaoDlamine
- r Jffianoiamine J
Pncspnatkiyiinositoi
I 4^4>ispnospoate
Priosproiipase D
PalmitoykCoA + serine'
CoA.SH
CO2
H-NADPH + 2H+
P NADP*"
Dinydrospnigosine
Acyl-CoA
t
[Ceramide]
COASH
I - Pnospnatidytchoiine
p DAG
Ceramide Choline
t------------------- -------------------- *1
O CH,
O" ch3
c—c
(CH2)n
ch3_____
Q~Spnin-goirye in
Fig. 7.33. Synthesis of sphingomyelin. Ceramide is spningosine with a fatty acid attached to its amino
group Dy an amide linkage (blue Dox). Sphingomyelin contains pnospnocnoiine the polar «nead»
of sphingomyelin
Fig. 7.34. Synthesis of glycosphingol ipids from ceramide. Glc — glucose: Gal — galactose; Gal MAC —
M-aceiyigaiactosamine; WAN a — N-acetylneuraminic acid; UDP — sugars, active forms of sugars;
CMP-NANA — active form of N-acetylneuraminic acid
Type 0
Ry. 7.35. Structure of blood group determinants. These structures are the same except mat type A has
N-acetyigaiactosamine (GatNAc), type B has galactose (Gai) and type 0 has neither. Fuc — fucose;
CicNAc — W-acetyiglucosamine
Table 7J5. Examples of sphingoliptaosis - the Inherited diseases connected wim deficiency of enzymes
involved in catabolism of sphingolipids
Gaucher's disease fi-Glucosidase Cer-Gfc (Glu cosyice ramide) Most common lysosomal
(glucocerebrosi- storage disease.
dase) Enlarged liver and spleen,
erosion of long bones,
mental retardation
] n. case of the enzyme deficiency, its substrate is accumulated in the cells and finally
destroys it. As most of sphingolipids a re in the nervous tissue, the mental retardation is
specific for many types of sph ingo lipidosis.
Situational problem
L Phosphatidylcholine molecules arc the main lipid compounds of all cell
membranes. Phosphatidylcholine, is also an important compound of the lungs
alveoli surfactant hut its fatty acids composition differs from phosphatidylcholine
present in the cell membranes. Explain the difference in fatly acid composition
of these two types of phosphatidylcholine molecules. For the answer:
a) draw the structural formula of phosphatidylcholine of the lungs surfactant
and give the full name of this structure:
b) explain the functions of phosphatidylcholine in. the membranes of the cells
and in the lungs surfactant;
c) compare the fatty acids composition in both types of phosphatidylcholine and
explain how it correlates with their functions;
d) explain the mechanism of respiratory distress syndrome development in
premature infants;
c) indicate the possible way of treatment of these infants.
370 Chapter 7. Lipid metaD ol ism
Fql 7.36. Structure of cholesterol and cholesterol ester. A — formula and model of cnotestefoi. B —
formula of cholesterol ester. R is usually the radical of linoleic acid
Cholesterol is obtained from the diet or synthesized in most cells of the body buL
mainly in the liver (:= 70%) and intestine (Fig. 7.37). Smaller amounts of cholesterol
arc synthesized in the adrenal cortex and gonads being the precursor of steroid
hormones.
7.10. Cholesterol Functions and Metabolism. Regulation of Cholesterol Synthesis 371
________ _________
Usage and excretion Ceils
Absorption of dietary cholesterol. Most people in modern societies get close io 0.5-
1.0 g of cholesterol per day with food. Cholesterol is present only in the food of animal
origin. Plants contain phytosterols instead, like ergosterol in fungi and P-sitostcrol
in higher plants. These steroids are poorly absorbed from dietary sources and not
involved in mammalians’ metabolism. A vegan diet is essentially cholesterol free. Part
of food cholesterol is present as cholesterol esters. Cholesterol and cholesterol esters
being water-insoluble molecules arc emulsified by bile salts. Cholesterol esters arc
digested by the enzyme cholesterol esterase. This enzyme secreted by the pancreas
cleaves cholesterol ester to free cholesterol and fatty acid. These products together
with the products of TAG digestion are absorbed as mixed micelles in the intestine.
Approximately only half of dietary cholesterol entering the gut per day is absorbed.
Cholesterol absorption by intestinal cells is an important regulatory point in human
cholesterol metabolism. Although dietary steraoids arc absorbed by diffusion from
the intestinal lumen to enterocytcs, there is a special mechanism to remove excess
cholesterol and plant sterol from the enterocytcs. The transport of sterols out of
the enteiocyies occurs by the adenosine triphosphate (ATP) — binding cassette
proteins (ABC). These proteins use ATP energy for the transport of unwanted sterols
from the enterocytcs back into the gut lumen and cholesterol and other sterols are
eliminated from the body mainly in the feces. Patients with rare autosomal recessive
disease phytosterolemia have a defect in the function of ABC proteins and high
372 Chapter 7. Lipid metaD ol ism
levels of cholesterol and phytosterol in the blood. This accounts for atherosclerotic
cardiovascular disease in the patients. Some drugs that block cholesterol absorption by
intestinal cells arc used for the treatment of hypercholesterolemia.
In the intestinal epithelial cell, most of the absorbed cholesterol is converted again,
to cholesterol esters by enzyme ACAT (acyl-CoA — cholesterol acyltransferasc). This
enzyme catalyzes the transferor a fatty acyl from CoA to the hydroxyl group on carbon
3 of cholesterol (Fig. 7.36). Then free cholesterol and cholesterol esters together with
triacylglyccrols and other absorbed dietary lipids arc packaged into chylomicrons.
Most of TAG in chylomicrons is removed by LP-lipase in the blood capillaries of
peripheral tissues and cholesterol-rich remnant particles thus formed arc taken up
by the liver. The compounds of chylomicron remnants arc hydrolyzed by lysosomal
enzymes and cholesterol becomes available for usage by hepatocytes. Thus, most of
the dietary TAG goes to extra hepatic tissues but most of the dietary cholesterol is
delivered to the liver.
Cholesterol synthesis. Endogenous cholesterol synthesis amounts up to 500-
1000 mg/24h, depending on the dietary supply. The complex cyclic structure
of cholesterol is synthesized from simple initial precursor — acetyl-CoA. Other
precursors required for cholesterol synthesis arc NADPH as donor of hydrogen for
reduction reactions and ATP as an energy source. The precursors can be obtained
from the glucose catabolic pathways: acetyl-CoA from aerobic glycolysis and NADPH
from pentose phosphate pathway. Other sources of acctyl-CoA arc ^-oxidation of fatly
acids in ease of high content of fats in the diet and the oxidation of kctogenic amino
acids such as leucine and iso I cue inc.
The pathway of cholesterol synthesis is one of the longest in. human cells
and involves at least 30 enzymes. The synthesis of I mole of cholesterol requires
IS moles of acctyl-CoA, 36 moles of ATP and 16 moles of NADPH. All reactions
of cholesterol synthesis occur within the cytoplasm, although some of them occur
in the endoplasmic reticulum. The whole pathway is subdivided into several stages
(Fig. 7.38).
The first is the synthesis of mevalonic acid (mevalonate) from acetyl-CoA. In
this pathway two molecules of acetyl-CoA condense, forming acetoacetyl-CoA,
which then condenses with the third molecule of acetyl-CoA to yield the 6-carbon
molecule p-hydroxy-p-nicthylglutaryl-CoA (HMG-CoA). These reactions, catalyzed
by acctoaceiyl-CoA thiolase and HMG-CoA synthase- arc the same as the ones in
the synthesis of ketone bodies, although the latter process occurs only within the
mitochondria. In the next reaction, catalyzed by HMG-CoA reductase. HMG-CoA
is reduced to mevalonate. Hydrogen atoms required for this reducing reaction are
donated by two molecules of NADPH. H MG -CoA reductase is the key enzyme of the
pathway of cholesterol synthesis.
The second stage is the conversion of mevalonate into two activated isoprenes
A3-isopentenyl pyrophosphate and dimethylallyl pyrophosphate. Mevalonic acid
(mevalonate) al first is phosphorylated in reactions requiring kinases and ATP.
Subsequent decarboxylation leads to the formation of isopcntcnyl pyrophosphate and
di methyl allyl pyrophosphate which, arc the isomeric 5-carbon isoprene units. Three
ATP molecules arc required for this sequence of reactions.
7.10. Cholesterol Functions and Metabolism. Regulation of Cholesterol Synthesis 373
c>
CHa—€ SCcA
[Acetyf-CoAj
C- CHa— C—SCoA
[’■* CoASH
O
1 1
i3 — C — Chk^— C1—SCoA
1 1
L- Ch3—C-*—SCoA
HMG-CoA
synthasie 1 CoASH
OH 0
[ I 1
“O-* C— Cbfc—C“CtV“ C—SCoA
ch3
|! hydroxy |!. ne:hy. 'i'Jlaryi Co A HMS Go A
HMG-CoA |
c 2NADFH + 2H'
reductese « * 2NADP*
OH Ir*1 Coash
I I
“O— O— CH?— C — Ona— CHnOH
r+ADr
~ Squalene;
NADP NADPH + H+
Fig. 7.38. The pathway of cholesterol synthesis. C„- — Geranyl pyrophosphate, Cl3 — Farnesyf
pyrophosphate
1 n. the fol low i ng sequence ofreactions, the double bonds in these 5-carbon isoprenes
allow them to condense with the formation of a 10-carbon chain compound, known
as geranyl pyrophosphate. Pyrophosphate groups of these isoprene units are the energy
source for the reaction. Geranyl pyrophosphate then undergoes the condensation
with the third molecule of isopcntenyl pyrophosphate with, the formation of 15 carbon
molecule brnesyl pyrophosphate. After this, two molecules of famesyl pyrophosphate
react, both pyrophosphate groups are removed and 30 carbon, molecule squalene is
formed.
374 Chapter 7. Lipid metaD ol ism
The last stage of the synthesis is the conversion of squalene to die cholesterol
One atom of oxygen is included in squalene by monooxygenase with the formation
of 22-epoxide. Then the linear structure of squalene epoxide is converted to the
cyclic, structure of lanosterol, a sterol with the four-ring structure characteristic of
the steroid nucleus. The sequence of complex reactions then leads to the formation
of cholesterol
Transport of synthesized cholesterol from the liver. Cholesterol synthesized in liver
and del ivered by chylomicron remnants forms cholesterol pool. In the liver cholesterol
is used for the formation of membranes of hepatocytes, for bile acids synthesis and a
fraction of cholesterol is converted to cholesterol esters by acyl-CoA — cholesterol
acyl transferase (ACAT).
Cholesterol leaves the liver by one of the following routes. The hepatocytes package
cholesterol esters together with cholesterol, triacylglyccrols, phospholipids and the
major apoprotein B-100 into nascent very low-density lipoproteins (VLDL). These
particles then arc secreted into the bloodstream by cxocytosis. The nascent VLDLs
acquire apoCll and apoE from circulating high-density lipoproteins (HDL) in the
blood and become the mature VLDL transporting cholesterol and TAG to peripheral
tissues. Another fraction of cholesterol is secreted as bile acidsand biliary cholesterol
to the bile and that is the main route of excessive cholesterol excretion from the liver
(Fig. 7.37).
Regulation of the cholesterol synthesis. In healthy individuals, there is an inverse
relationship between dietary cholesterol intake and the rate of cholesterol biosynthesis.
This regulation ensures a relatively constant daily supply of cholesterol. The rate of
the cholesterol synthesis in the liver is regulated through the key enzyme HMG-CoA
reductase catalyzing the conversion, of H MG-CoA to mevalonate. The regulation, of
HMG-CoA reductase activity is controlled in multiple ways.
Transcriptional control is one of them (Fig. 7.39). HMG-CoA reductase has a life
span of approximately 4 hours and a change in its rate of synthesis or degradation can.
affect cholesterol synthesis rather rapidly that looks like feedback inhibition. When
cholesterol levels in the hepatocytes are high, it binds to the transcription factors that
leads to a decrease in. transcription of the HMG-CoA reductase gene and thus less
enzyme is produced, and less cholesterol is synthesized. When cholesterol level in the
hepatocyte lowers, the rate of transcription increases. Asa result, synthesis of HMG-
CoA reductase increases, and production of cholesterol rises. Transcription of HMG-
CoA reductase gene is also regulated by thyroid hormone acting as inducer and bile
salts repressing the gene.
Proteolytic degradation of HMG-CoA reductase is another mechanism of
regulation. High levels of cholesterol and bile sails in the hepatocytes cause the
conformational changes in the membrane domain of HMG-CoA reductase that
makes the regulatory enzyme more susceptible to the action of proteolytic enzymes
and the rate of cholesterol synthesis decreases as a result.
Covalent modification of HMG-CoA reductase as a result of hormonal regulation.
HMG-CoA reductase activity is regulated by phospho rylai ion/dcphosphorylation
(Fig. 7.40). In lasting state, hormone glucagon stimulates the phosphorylation of the
key enzyme and phosphorylated enzyme becomes inactive. Therefore, in fasting state
7.10. Cholesterol Funciions and Metabolism. Regulation of Cnolesteroi Syntnesis 375
Fig. 7.39. The regulation of cholesterol synthesis by transcriptional control ot the key enzyme HMG-CoA
reductase, if cholesterol levels in me cell are low SREBP (steroid response element binding protein)
can bind to SCAF1 (SREBP cleavage-activating protein). The SCAP/SREBP complex transfers from the
EFl to me Golgi apparatus, where SREBP is cleaved by a protease, releasing the active transcription
factors (TF), wtiicn are translocated to me nucleus and activate the SRE (steroid response element)
thus inducing me transcription of the HMG-CoA reductase gene and cholesterol synthesis increases,
if cholesterol levels are high it binds to SCAP/SREBP complex and activate binding of this complex
with another protein — insig (insulin induced gene). This large complex cannot be transferred through
membrane of me Golgi apparatus, transcription factors are not released and me gene is repressed;
cholesterol synthesis decreases
in fed state, elevated insulin levels activate the enzyme phosphatase that
stimulates dephospho ryiation of HMG-CoA reductase, it becomes active and the
rate of cholesterol synthesis increases. The enzyme that phosphorylates HMG-CoA
reductase is the cell * energy sensor*. This enzyme is adenosine monophosphate
(AM P)-activated protein kinase (AMPK) which itself is regulated by AMP-activated
protein kinase kinase (AMP is the product of ATP cleavage). Cholesterol synthesis
is highly ATP depended process, thus, cholesterol synthesis decreases when .ATP
levels in the liver cells arc low (AMP level is high) and increases when ATP levels arc
high. All effects regulating the synthesis and the activity of HMG-CoA reductase arc
summarized in Fig. 7.4L
376 Chapter 7. Lipid metaD ol ism
MEVALONATE *- Cholesterol
mRNA
/ndt/crjon Repression
r--------------------- "'i
* Cholesterol
• Bile salts
Gene
Fig. 741. The summary of regulatory effects on HMG-CoA reductase synthesis and activity
7J1. BHe Salts, Functions, Synthesis, ano Role in me Cholesterol Metabolism 377
Conjugation of bile salts. The main function of the bile salts is the emulsification
of dietary fats in the lumen of the intestine for their digestion and formation of
mixed micelles required for the absorption of products of digestion. Bile salts arc the
detergents which activity depends on the amphiphilic properties of the molecules.
. ct-hycruxycf olesterol I
Fig. 742. Synthesis of the primary Hile acids — cholic and chenocholic. 7u-hydroxyiase is the key
enzyme cf bite acids synthesis. The synthesis of this enzyme is regulated at transcriptional level.
Cholesterol and thyroid hormone KJ increase the rate of enzyme synthesis and bite salts and
estrogens suppress it
7J1. BHe Salts, Functions, Synthesis, ano Role in me Cholesterol Metabolism 379
Conjugation reaction increases the amphiphilic properties of the bile salts that makes
them better detergents. At first, the carboxyl group al the end of the side chain of
the bile salt is converted to active-CoA derivative and then it is conjugated with
glycine or taurine (Fig. 7.43). Owing to the functional groups of glycine and taurine,
the conjugated bile salts arc more ionized in the contents of the intestinal lumen,
therefore, are more potent detergents. The conjugated bile salts arc laurocholic,
glycocholic, taurochcnocholic and glycochcnocholic. The synthesized bile salts
are secreted into the bile. Bile consists of a mixture of organic and inorganic
compounds. Bile salts and phosphatidylcholine arc the main organic compounds of
the bile. Bile can cither pass directly from the liver where it is synthesized into the
duodenum through the common bile duct or can be stored in the gallbladder when
not immediately needed for digestion. Bile is released from the gallbladder into the
intestine after a fatty meal. The gal I bladder con tract ion is stimulated by the intestinal
hormone cholecystokinin. In the intestine, the bile sails emulsify the dietary lipids
that arc needed for the activity of pancreatic lipase and form mixed micelles for the
absorption. The enzymes of intestinal bacteria cleave off the taurine and glycine from
primary conjugated bile salts and remove a hydroxyl group al position. 7. Asa result,
the primary conjugated bile salts are converted to secondary bile salts — dcoxycholic
and lithocholic (Fig. 7.44). The dcconjugatcd and de hydroxylated bile salts are less
soluble and therefore are less readily absorbed from the intestinal lumen than the
primary conjugated bile salts. About 95% of bile salts secreted to the intestine arc
resorbed and return to the liver via the enterohepatic circulation (Fig. 7.45) but the
bile salts mainly excreted with feces are the secondary bile salts. Only 5% of the bile
salts entering the gut arc excreted with feces every day.
The resorbed secondary bile salts arc rcconjugatcd in the liverbut not hydroxylated
and secreted again into the bile. Because of the enterohepatic circulation, both pri mary
and secondary’ bile salts arc present in the bile. Each bile salt molecule is recycled live
to eight limes every day, and it remains in the system about one week before it is finally
excreted.
Gallstones disease. Cholesterol is the most water-insoluble constituent of the
bite; and to be kept in solution, it must be included into bile micelles. Bile salts
and phosphatidylcholine secreted from the liver into gall bladder serve as detergents
and form these micelles. Cholesterol is soluble in the bile as micelle’s compound
only if the ratio of the bile compounds is 12,5:2,5:1 (Bile salts: Phosphatidylcholine:
Cholesterol). If the secretion of cholesterol into the bile increases, the micelles
arc destructed, cholesterol loses solubility and start to sediment in the gall bladder
forming the stones. The stones consisting of only cholesterol {white cholesterol
stones) or cholesterol aggregated with bilirubin, calcium and other compounds
(mixed brown stones) can be formed in the gall — bladder. The tendency for
cholesterol stones formation is inherited, occurs more frequently in females than
in males, and is associated with obesity. The bile salts c he nod coxy cholate and its
isomer ursodeoxy cholate arc available for oral use to dissolve gallstones at the initial
steps of their formation.
380 Chapter 7. Lipid metaD ol ism
Fig. 7.44. The secondary hile sails formed in the intestine by bacteria I enzymes
7.12. Cholesterol Transport Dy Blood Lipoproteins 381
Gall-bladder
Intestine
circulation
v
Feces (0.2-0.8 g/day)
Fig. 7.46. Transport of exogenous cholesterol from me intestine to peripheral tissues. C — cholesterol:
CE cholesterol esters; CMn—chylomicron nascent: CMm —cnylomicron mature; CMr — chylomicron
remnants
382 Chapter 7. Lipid metaD ol ism
Blood HDL
Liver
Fatly acids, glycerol receptors
Fig. 747. Transport of endogenous cholesterol to peripheral tissues. Cholesterol and cholesterol,
esters synthesized in the liver are released to bloodstream in nascent VLDL. in blood nascent VLDL
acquires apoCll and apoE from nigti density lipoproteins (HDL) and are transformed to the mature
VLDL. under the action of LP-lipase VLDL is converted at first into IDL and then into LDL particles
enriched Dy cholesterol. LDL are taken up Uy the celts through special LDL — receptors
Anothcrcnzyme hepatic lipase located in the liver is also involved in the conversion
of IDL into LDL by hydrolyzis of TAG in IDL. When the amount of TAG in the
particles becomes lower apoCll and partly apoE are transferred back to HDL. The
only apoprotein in LDL is a solitary apo 13-100 molecule.
Approximately half of the LDLs arc transported back to the liver, where they are
internalized up by the hepatocytes through the receptor-mediated endoevloses. The
remaining LDL particles deliver cholesterol to the other tissues where it is used for
the membrane synthesis, production of steroid hormones (adrenocortical and gonad
cells) and vitamin D synthesis. Pan of free cholesterol molecules is converted to the
cholesterol esters by the enzyme fatly acyl-CoA: cholesterol acyttransfcrase (ACAT)
in. cytoplasm of the cells using cholesterol as the precursor for the synthesis of steroid
hormones.
LDL is taken up by the cell through the special receptor on the plasma membrane
which recognizes apoi3-IOO and apo L. Besides LDL, this receptor also binds
VLDL, IDL and chylomicron remnants — all panicles containing cholesterol and
its esters. The amount of LDL receptors on the surface of a hepatocyte is about
50 000-70 000. LDL receptor is coded by the gene located on the short arm of
chromosome 19: the gene is very long and contains 18 exons. The protein encoded
by this gene composed of six different regions (Fig. 7.48). The region at the N end is
7.12. Cholesterol Transport Dy Blood Lipoproteins 383
Fig. 749. LDL uptake By the receptor-mediated endacytosis; recycling ol LDL receptors
Other receptors, such as the LDL receptor-related proteins (LRP) and the
macrophage scavenger receptor (SR) have broad specificity. These receptors
can bind and internalized LDL with modified chemical structure. Two types of
macrophage scavenger receptors are known asSR-Al and SR-A2. The main function
of these receptors is to clear the blood from lipoproteins with modified structure.
The macrophages arc usually located near the endothelial surface of vascular
endothelial cell.
Reverse cholesterol transport by the blood lipoproteins. HDL carries out two main
functions. The first, HDL has a unique function as a donor of apo CI I and apo E
(Tig. 7.50} to VLD L and chylomicron. Apo CH is the activator of LP lipase, and apo
E is a ligand for binding of several types of lipoproteins to the LI>L receptor. HDL
particles arc formed as nascent HDLs, mainly by the liver and less in the intestine.
These particles have the discoid shape and contain more variety of apoproteins than
any other lipoproteins (apo Al, apo AIL apo CL apo CH, apo E) and very low levels
of triacylglycerols, cholesterol and cholesterol esters.
7.12. Cholesterol Transport Dy Blood Lipoproteins 395
Nascent HDL contacts in the blood with nascent VLDL nascent chylomicron and
apoproteins Cl I and E arc transferred from HDL to VL.D Land chylomicron which are
converted to mature particles.
The other main function of HDL is to remove excess cholesterol from the
cholesterol — laden cells and to return it to the liver. a process known as ■ reverse
cholesterol transports. The reverse cholesterol transport requires the transfer of
cholesterol from the cell to the HDL particle. Cells contain the protein ABC! (ATP-
bindin* cassette protein I) which uses the ATP energy to move the cholesterol from
the inner layer of the cell membrane to the outer. H DL particles bind to the outer cell
surface through apoproLcin apo Al and the cholesterol di (fuses from the cell membrane
towards the HDL particle. To trap the cholesterol within the HDL core, cholesterol
must be converted to cholesterol ester by the enzyme LCAT (lecithin cholesterol
acyl transferase) attached Lo H DL surface and catalyzing the reaction:
Fig. 7.50. Function of HDL in cholesterol metabolism: the reverse cholesterol transport to the liver
A1, E, Cil — apoproteins; HDLn — HDL nascent; HDL3 — HDL enriched with cholesterol esters.
LCAT — lecithin cholesterol acyf transferase, CE — cnoiesterot esters. C — cholesterol, CETP —
cholesterol ester transfer protein, HTGl — hepatic triacylglycerol lipase, SR — B1 — scavenger
receptor; ABCl — ATP-hinding cassette protein I
The ratio in healthy adult people is <3,5. This coefficient represents the ratio of
LDL cholesterol and HDL cholesterol. The higher is the coefficient, the higher is the
risk of atherosclerosis.
Hypercholesterolemia can result from a variety of genetic and acquired reasons,
all of which may lead to an accumulation of LDL cholesterol in the blood and
atherosclerosis. Familial (genetic) hypercholesterolemia develops when the cells lack
functional LDL receptors (receptor-negative). Familial hypercholesterolemia is the
result of mutations in the gene encoded L DL receptor. This disease is one of the most
common inherited diseases (heicrozygoic — one per 500 people, homozygote — one
per I million people). H etc rozygotes have approximately half of the normal amount
of LDL receptors on. the cell surface. Thus, the total cholesterol levels in blood
increase twice (350-400 mg/dl) because cells cannot take up LDL al a normal rate.
Homozygotes produce almost no LDL receptors and cholesterol levels rise up to 700-
7.13. Hypereno lester oiemia. Biocrtem ieal Aspects of Affler oscie rosis. C n oiest erot-Lo wer i ng Ag ents 387
800 mg/dl). More than 350 types of mutations arc known for LDL receptor gene. The
consequences of mutations depend on the location of the protein damage structure.
If mutation changes the structure of LDL — binding region on N-cnd, the receptor
does not bind LDL, if mutation changes the structure of receptor close to C-end, the
receptor binds LDL but cannot be cndocytosed. The result of these mutations is that
blood levels of LDL (and cholesterol) arc elevated because the cells cannot take up
these panicles at a normal rate. Iking impaired in their ability to acquire cholesterol
from LDL, the liver cells increase the rate of endogenous synthesis of cholesterol.
Cholesterol pool in the body increases together with cholesterol levels in the blood.
These mutations are so common because the heterozygous die from the complications
of accelerated atherosclerosis after the end of the reproductive ago when the mutation
has already been transmitted to their offsprings. Thus, there is not much selection
against them.
Another genetic form of hypercholesterolemia develops in the patients who have
unusual apoprotein (a) in LDL panicles. These particles arc called lipoprotein little
*a* to differentiate them from the apoprotein A found in HDL. The patients having
apoprotein (a) have hypercholesterolemia and high risk of atherosclerosis, coronary
artery disease, myocardial infarction and stroke.
Non-genetic types of hypercholesterolemia are the result of:
* excess consumption of fatty animal meal containing plenty of cholesterol;
► excess consumption of carbo hydrates (hypercaloric food};
► chemical modification of LDL and LDL — receptors by glycation of proteins;
► oxidative modification of LDL (cigarette smoking);
► low levels of HDL.
Excess consumption of cholesterol with food leads to its accumulation in the
cells, and intracellular cholesterol inhibits the replenishment of LDL receptors by
down regulation of their expression. L DL particles arc not taken up by the cells with
the normal rale and circulate in the blood longer increasing the cholesterol levels in
blood. Overeating of carbohydrates also causes hypercholesterolemia, as all substrates
required! for cholesterol synthesis originate from glucose catabolic pathways (acetyl-
CoA, NAD PH and ATP). Excess synthesis of TAG and cholesterol leads to increased
synthesis of VLDLs which arc converted to LDL.
Prolong hyperglycemia in patients with diabetes mcllitus type 2 increases the rate
of noncnzymalic attachment of glucose through NIL group to various proteins in the
body (glycation of proteins). Glycation involves all proteins, including LDL receptors
and apoB-IOOof LDL particles changing their conformation. In healthy people, the
process of proteins glycation occurs at a low rate, but in patients with diabetes me ll it us
it increases. LDL receptors and their ligands become noncomplemcntary and less
circulating LDL arc internalized into the cells through specific receptor — mediated
uptake. That results in more active capture of modified LDL particles by scavenger
receptors of macrophages converting them to «foam cells*. The macrophages
overloaded with cholesterol and modified lipids which arc accumulated in cytoplasm
of the cells arc named «thc foam cells*.
Oxidative modification of LDL particles is a result of the peroxidation of
polyunsaturated fatty acids radicals (Ch. 5) of phospholipids and cholesterol esters
in LDL. Nonenzymalic peroxidation of polyunsaturated fatty acids can be initiated
388 Chapter 7. Lipid metaD ol ism
by free radicals formed during cigarette smoking. Oxi datively modified particles
arc more available for nonspecific uptake of LDL by the macrophage nonspecific
<■ scavenger* receptors. The scavenger receptors arc not down regulated as the LDL
receptors that allow the macrophages to lake up oxidatively modi lied LDL particles
long after intracellular cholesterol levels arc elevated (Fig. 7.51). The formed foam cells
can migrate through the space between endothelial cells and initiate the process of
atherosclerotic plagues formation.
Scaversger receptors
fig.7-51-The foam cells formation. ROS - reactive oxygen species and nitric oxide initiate peroxidation
of polyunsaturated fatty acids the compounds of LDL Vitamin E C and fl-carotene act as antioxidants
and irtfiitiit peroxidation of lipids, thus stabilizing structure of LDL* (modified LDL)
The foam cel Is accumulate in the subindmal space ofarteries and cause atherosclerotic
lesion within the wrall of an artery by the formation of laity streaks. The damaged
endothelial cells start to secrete adhesion molecules that stimulate the circulating
monocytes to accumulate under the endothelium (Fig. 7.52). This movement of
monocytes and macrophages to the injury tissue resembles the inflammatory response.
That loads to suggestion that atherosclerosis is the inflammatory disorder and can be
treated with anti-inflammatory drugs. The deformation of the endothelium exposes the
compounds of the underlying extracellular matrix to the blood and these areas serve as
sites for adhesion and aggregation of platelets. Activated platelets secrete cytokines and
thromboxanes that stimulate this process and increase the risk lor thrombus formation
locally. I f thrombus grows, it can occlude the blood vessel and cause an acute infarction
of the tissue.
Cholesterol-lowering agents. Hypercholesterolemia leads to the development of
atherosclerosis and its complications such as heart attack and stroke. As these are
the most abundant causes of the death, among the people, the very intensive research
develops to prevent and to treat hypcrchole sterol cm iaand atherosclerosis. Healthy food
is important factor in reducing the risk of atherosclerosis. Food of plant origin does
not con rain cholesterol and is enriched writ h native antioxidants such as vitamins E and
C (ascorbic acid), fl-caroLcnc. These vitamins inhibit peroxidation of polyunsaturated
7.13. Hypereno lester oiemia. Biocnem ieal Aspects of Atner oscie rosis. C n oiest erot-Lo wer i ng Ag ents 339
Fig. 7.52. Amero sclerotic plaque formation Oxidatively modified LOL particles are taken up Dy the
macrophage ^scavenger* receptors: tne macrophages overloaded Dy cholesterol are converted to
the foam ceils. These cells are accumulated in su Dendotneii urn. Tne deformation of the endotneiiirm
exposes me compounds of tne underlying extracellular matrix to the Diood and this area serves as site
for adhesion and aggregation of platelets. The risk forinromous formation increases locally
Tatty acids of cholesterol esters of LDL and prevent the formation of the oxidatively
modified LDL particles. Thai reduces the rate of conversion of macrophages to the
foam cells and initiation of the atherosclerotic plaques formation. Fiber, the compound
of whole-grain rye Hour, is not digested in the intestine and absorbs fraction of diclary
cholesterol: then the cholesterol together with fiber is excreted with feces. So low
calorie and low cholesterol food containing native antioxidants and enriched with
fiber is useful for lhe prevention of hypercholesterolemia and atherosclerosis.
* The first step in. treatment of the patient with hypercholesterolemia is a diet
with a low level of cholesterol (<300mg daily). If this diet cannot reduce the
blood cholesterol to the desired levels, the daily cholesterol consumption must
be reduced to <200 mg. Low carbohydrate diet reduces the availability of acctyl-
CoA — the initial substrate for cholesterol synthesis and also lowers the secretion
of insulin activating H MG-CoA reductase.
► The resins, such as cholestyramine and colestipol, which bind the bite salts
within the lumen of intestine and interrupt the cntcrohcpatic circulation, arc
used as cholesterol-lowering drugs. These drugs act as a bile sails sequestra nt
because lhe re sin-bound bile salts arc excreted with the feces and do not recycle
to the liver. This, in turn, stimulates the hepatocytes to uptake more LDL from
the blood because more cholesterol is required for the synthesis of bile salts.
As a consequence, the blood levels of LDL cholesterol arc decreased. The use
of the bile salts scqucstranis cause many gastrointestinal side effects and new
cholesterol absorption blocker is introduced. Ezctimibe reduces the absorption
of dietary cholesterol from the lumen of the gui. The net result is the reduction
390 Chapter 7. Lipid metaD ol ism
of cholesterol pool in hepatocytes. This effect causes the induction of the LDL
receptor synthesis by the liver cells and the capacity of the liver to uptake more
LI>L from the circulation increases. These events lead to lower levels of LDL
cholesterol in the blood.
► The drugs statins the potent inhibitors of HMG-CoA reductase arc used if
the levels of the cholesterol in blood arc very high as in patients with familial
hypercholesterolemia. The statins have the structure similar to the structure of
HMG-CoA — the substrate of the HMG-CoA reductase. Therefore, the statin
binds to the active site of the HMG-CoA reductase and acts as competitive
inhibitor of the key enzy me, thus, reducing the rate of cholesterol synthesis in
the liver cell. Low levels of cholesterol in the cells stimulates the expression of
LDL receptors. As the number of receptors rises on the cell surface, the uptake
of LDL by the cells increases. As a rcsulL, the blood level of LDL cholesterol
decreases.
Review test
L Cholesterol synthesis. Match the figure with the letter.
□
I
CH3— C—S&aA
[Acetyj-CcA] O
I
CHz— C — 5C0A
CaASH
CH3—C*—CH2—
[Aceioacgiyi CaAj; o
CH-3—C—SCoA
CaASH
0
SCaA
□CHtOH
7.13. Hypereno lester oiemia. Biocrtem ieal Aspects of Affler oscie rosis. C n oiest erot-Lo wer i ng Ag ents 391
A. Mevalonate.
B. HMG-CoA.
C. HMG-CoA reductase.
D. HMG-CoA synthase.
E. Acetoacclyl-CoA.
2. Bile salts synthesis. Match the figure with the letter.
A. Cholic acid.
B. Glicocholic acid.
C. Taurocholic acid.
D. Liiocholic acid.
E. Deoxycholic acid.
Situational problems
1. 65 years old woman with low physical activity got the excess carbohydrates
(about 400 g daily) for several years. The laboratory value of total cholesterol in
scrum blood was 260 mg/dl. Why the excess consumption of carbohydrates can
cause hypercholesterolemia? To answer the question:
a) compare the cholesterol concentration in the scrum blood of the patient with
the normal range:
b) draw the schemes of the metabolic pathways confirming that overeating of
carbohydrates can result in hypercholesterolemia;
c) draw the scheme of the cholesterol synthesis regulation;
d) give your prescriptions for the diet and treatment of the pau ent.
2. The patient of 48 years old has survived a heart attack. The laboratory lest of his
scrum blood were: total cholesterol 440 mg/dl (reference range is <200 mg/dl).
392 Chapter 7. Lipid metaD ol ism
hospital his physician told him that his in fare Lion is the result of a long history
of smoking. Explain the conclusion of the specialist. For that:
a) explain the functions of lipoproteins in cholesterol transport using the
schemes:
bi describe the structure of LDL receptor, regulation of its synthesis in the liver
and function in. cholesterol metabolism;
cl name all possible causes of the hypercholesterolemia;
d) explain the mechanism of prolonged smoking on. the hypercholesterolemia
and atherosclerosis development.
7. LDL/HDL ratio is important for the regulation of cholesterol levels in
blood. Disbalance of these lipoproteins leads to hypercholesterolemia and
atherosclerosis development. What are the functions of these lipoproteins in
cholesterol metabolism? For the answer draw:
a) the scheme, representing LDL function in cholesterol metabolism;
b) the scheme, representing HDL function in cholesterol metabolism;
c) the reaction catalyzed by LCAT and explain the role of this reaction in HDL
functioning;
d) the formula for the atherogenic coefficient and indicate its normal range.
8. Hypercholesterolemia is a frequent complication of diabetes mellitus in patients
with prolonged hyperglycemia. Why a high level of glucose in blood causes
hypercholesterolemia and atherosclerosis? For the answer explain:
a) how glucose can interact with the proteins and the consequences of this
reaction for the proteins;
b) glycation of which proteins results in hypercholesterolemia;
c) possible causes and complications of hypercholesterolemia.
9. 65 years old man, who considered him self healthy was examined prophy tactically.
The laboratory value of total cholesterol was 240 mg/dl The atherogenic
coefficient was 3.8. A .specialist prescribed to increase physical activity and
correct food composition. He was advised to decrease the red meat consumption
and to change his diet for vegetables, enriched with cellulose vitamins C and E.
Explain the specialist's prescriptions. For that:
a) name all causes of acquired hypercholesterolemia:
b) explain, why the red meat must be excluded from the diet of the patient;
c) explain, why a diet enriched with cellulose vitamins C and E can influence on
the cholesterol levels in blood;
d) explain the formation of the foam cells and their role in atherosclerosis
development.
Chapter 8
NITROGEN METABOLISM
8.1 Digestion of Proteins and Amino Acid Absorption. Essential Amino Acids
82. Transamination and Deamination of Amino Acids
8.1 Ammonia Detoxification. Urea Cycle. Urea Cycle Disorders, Hyperammonemia
8.4. Synthesis of Nonessential Amino Acids
8.5. The Main Steps of Amino Acid Degradation; Glucogenic, Ketogenicand both Gluco
and Ketogenic Amino Acids
8.6. Special Pathways of Some Amino acids: Phenylalanine Metabolism, Tetra hydrofolate,
Vitamin B^and s Adenosylmethionine
8.7. Decarboxylation of Some Amino Acids, Synthsis of Neurotransmitters and Hormones
8J. Special Products Derived from Amino Acids
a-amino group is separated from the carbon skeleton and shunted into the pathways
of amino group metabolism.
zl/l 20zfm/no acids present in linntan body profems arc c/assi/ied info 4 groups:
► non-essential amino acids which are synthesized in the body in the adequate
amount: Ala, Asp, Asn, Glu, Gin. Pro, Gly, Ser;
► essential amino acids which are not synthesized in the body: Vai, Leu., lie. Met,
Phe, Trp, Lys, Thr;
► partially essential amino acids which arc synthesized i n insufficient quantities for
body needs: Arg, His;
► conditionally essential amino acids which arc synthesized from essential amino
acids: Cys. Tyr (from Met and Phe).
The quantities of aig/nine generated by the urea cycle (see below) arc adequate only
for the adult and arc insufficient to support growth.
Nitrogen balance
Amino acids include 95% of all body nitrogen. The organism's nitrogen balance
is therefore primarily determined by protein metabolism. Attrogen balance is tke
correlafion between nitrogen intake and nitrogen lost Ay tAe Aody. Nitrogen is lost
mainly as urea in the urine, as well as in feces and saliva, by desquamated skin, hair,
and nails.
hi adults, the nitrogen balance is generally in equilibrium — i.e., the quantity of
protein nitrogen taken in and excreted per 24h is approximately equal.
I f some of the nitrogen taken in is more than tost, then the balance is postfive. This
is the ease duri ng growth. pregnancy, tissue repair after injury, recovery from illness: in
other wo rds, the situations when the protein synthesis exceeds the protein degradation.
A^arire balances (if nitrogen taken in is less than lost) arc rare and usually occur due
to disease: tuberculosis, cancer: negative balance occurs in starvation and may be in
very old individuals.
Lost cofjrcifn; is quantitatively determined by protein degraded in the complete
protein starvation. So under the condition of complete protein starvation 25 g of tissue
proteins arc degraded in 24 h.
Protein minimum is determined by the amount of protein needs for nitrogen
equilibrium, it is 30-50 g (-40 g) of proteins per day. But this quantity is not enough
for physical activity, capacity for work and health.
Protein optimum is determined by the amount of protein necessary for normal
growth and development: this quantity is 100-120 g per day for adult.
Anfririotts (alimeataryj value of protein depends on amino acid composition
and ability for assimilation: if protein contains the essential amino acids in. the right
proportion required by humans, it has AigA biological va&e.
Animal proteins arc high-value ones: they contain essential amino acids in
adequate quantity and proportion, and arc more digestible than plant proteins which
are low-value (Table 8.1). Plant proteins generally have a lower content of some
essential amino acids, such as lysine, tryptophane and methionine. Soy protein is
one of the best plant proteins, but nevertheless, the most prominent difference is
the proportion of the essential sulfur-containing amino acid methionine. Egg white
396 Chapter 8. Nitrogen metabolism
protein has approximately three limes more methionine than is found in soy protein.
Vegetable proteins arc usually badly digested and some plants (for example, legume
seeds and cereals) involve protease inhibitors, which arc peptides that form stable
inactive complexes with some of the pancreatic enzymes causing protein maWiges/jon.
Protein-energy malnutrition can lead to severe disorders occurring in growth
children, usually under 5 years old of age in developing areas of Asia, Africa, and
South America. Two extreme forms arc recognized: marasmus and Jbrasftjr&rAor. In
kwashiorkor energy intake may be adequate, but there is a deficiency in both the
quantity and the quality of protein. Kwashiorkor is characterized by edema and swollen
belly (as a result of low levels of proteins in blood), liver enlargement, the change in
skin and hair color Ito a rust color, caused but insufficient synthesis of melanins),
fatigue, diarrhea, loss of muscle mass, damaged immune system, which can lead to
more frequent and severe infections, and irritability. Nutrient deficiency is the main
cause of marasmus. Marasmus is a state of extreme emaciation; it is the outcome of
prolonged negative energy balance. It occurs in children who don't ingest enough
calories, proteins, carbohydrates, and other important nutrients. The main symptom
of marasmus is underweight. Children with this condition have lost a lot of muscle
mass and subcutaneous fat and stunted growth. They suffer from chronic diarrhea,
respiratory infections, and intellectual disability.
Table 8.1. Biological value and Uigeslibilily of food proteins
The Biological Value (8V) is a scale of measurement used to determine what percentage
of a given nutrient source is utilized by the body: in short — BV refers to how well and
how quickly your body can actually use the protein you consume). The highest BV of any
food source is 100%. Egg white protein is considered to have one of the best amino acids
profiles for human nutrition
Certain adult people in developed countries arc also al risk. This also includes
vegetarians and vegans who follow an imbalanced diet, as well as institutionalized
older people and hospitalized patients.
Some individuals experience distress when eating gluten-containing foods, and it
is commonly referred to as gluten sensitivity (GS> or gluten intolerance. Gluten is
a family of proteins found grains like wheal, rye, spelt and barley; it is protein that
grain plants use to store nutrients for the next generation of plants. Gluten intolerance
8.1. Digestion of Proteins ana Amino Acid Absorption. Essential Amino Acids 397
estimates between 0.5 and B percent of the population. Symptoms which occur
shortly after consuming gluten involve diarrhea and constipation, bloating, abdominal
pain, nausea and fatigue. People who have gluten intolerance have to avoid any food
with gluten in it, which includes any food that contains: wheat, and any derivatives of
wheat, barley, rye, brewer’s yeast (that is usually derived from beer): pasta, bread and
pastries, many baked goods, noodles, crackers, cereals, pancakes, waffles, and crepes,
biscuits etc.
Protein digestion
In humans, the degradation, of ingested proteins to their constituent amino acids
occurs in the gastrointestinal tract. Enzymes participating in protein digestion arc
proteinases (the same — peptidases, proteases, proteolytic enzymes), which mostly
released to gastro-intestinal tract in. inactive form (zymogens) and activated by partial
proteolysis in the lumen of stomach and intestine. Activated peptidases specifically
hydrolyze peptide bonds formed by appropriate amino acids. Endopeptidases
(c.g., — pepsin, trypsin) cleave the internal peptide bonds in the polypeptide chain;
exopeptidases (e.g., — carboxypeptidase, aminopeptidase) remove either N- or
C-terminal amino acid (Fig. 8.1).
Fig. 8.1. Digestion of proteins in the gastrointestinal trad. Marked in blue proenzymes are inactive
precursors of pepsin, trypsin, enymotrypsin, elastase and carboxypeptidase
398 Chapter 8. Nitrogen metabolism
Proteins arc not digested in the oral cavity. The entry of dietary proteins into the
j/owtic/j stimulates the gastric mucosa to secrete the hormone gastrin, which in turn
stimulates the secretion of hydrochloric acid by the parietal cells and pepsinogen by
the chief cells of the gastric glands. The acidic gast ric juice (pH 1.0 to 2.5) is both,
.antiseptic, killing most bacteria and other foreign cells, and a denaturing agent,
unfolding globular proteins and rendering their internal peptide bonds more accessible
to enzymatic hydrolyzis. An acidic medium is important for pepsin, which optimum
pH activity spans within 1-2. Pepsinogen (Afr 40,554), an inactive precursor, or
zymogen, is convened to active pepsin C.Wr 34,614) by the HCI and enzymatic action
of pepsin itself (autocatalysis), the activation occurs by partial proteolysis (Fig. 8.2).
(Autoactivation)
Masking
sequence
In the stomach, pepsin hydrolyzes ingested proteins at peptide bonds on. the
carboxy]-terminal side of the oromaric a/n/m? ac/J residues (Phe. Trp, and Tyr), as well
as peptide bonds between Glu or Asp with any amino acid, cleaving long polypeptide
chains into a mixture of smaller peptides.
Intrinsic factor is a glycoprotein secreted by parietal cells of the gastric mucosa.
In humans, it has an important role in the absorption of vitamin B]2 (cobalamin) in.
the intestine, and failure to produce intrinsic (actor results in the condition
Lactic acid is not present in stomach juice. But lactate may be formed in the reduced
or complete absence of free hydrochloric acid as a result of accelerated reproduction of
Lactobariffus, or in malignant tumors of the stomach.
8.1. Digestion of Proteins ana Amino Acid Absorption. Essential Amino Acids 399
HCI and pepsin arc able to damage gastric epithelium cells. In healthy people, it
docs not occur due to the presence of protective factors of the mucus tunic, such as:
► formation of mucus on the surface composed of hctcropoiysaccharides which
can't be split by peptidases;
► secretion of HCO^ by epithelial cells creating the less aggressive medium at
parietal layer with pH 5.0-6.0 in which pepsin is inactive;
► besides, damaged epithelium cells possess the ability to rapid regeneration.
The digestion of proteins now continues in the small intestine. The arrival of
amino acids into the upper part of the intestine (duodenum) causes the release of
the hormone cholecystokinin into the blood, which stimulates the secretion of several
pancreatic enzymes with activity optima at pH 7 to 8.
Trypsinogen, chymotrypsinogen, procarboxypeptidases A and B. and proelastase
arc the zymogens of trypsin, chymotrypsin, carboxy peptidases A and B. and elastase
which synthesized and secreted by the exocrine cells of the pancreas. Trypsinogen
is converted to its active form, trypsin by enteropeptidase, a proteolytic enzyme
secreted by intestinal cells. Free trypsin then catalyzes the conversion of additional
try psinogen to trypsin. Trypsin also activates chymotrypsinogen, procarboxypeptidases,
and proelastase tFig. 8.3; 8.4). All activations occur by partial proteolysis (splitting-ofT
peptide from zymogen, more often from N-termini, with rearrangement of the protein
structure that completes the active site turning inactive proteins into active ones).
The synthesis of the enzymes as inactive precursors prevents the cleavage of
ow n proteins of the stomach and intestinal mucosa, and exocrine cells of pancreas
from destructive proteolytic attack. The pancreas further protects itself against self
digestion by making a specific inhibitor, a protein called pancreatic trypsin inhibitor
that effectively prevents premature production of active proteolytic enzymes within
the pancreatic cells. Stomach ulcers are a type of peptic ulcer disease when pepsinogen
Trypsinogen
Procarboxy Carboxy
Praelastase Elastase
peptidase peptidase
- - —
/-----------“---------- i
Chymotry-
Chymotrypsin Prolipase Lipase
psinogen
Fig. 8.4. Action of the digestive proteases. Pepsin, trypsin, chymotrypsin, and elastase are
endopeptidases; they hydrotyze peptide bonds within chains. The others are exopeptidases;
aminopeptidases remove the amino acid at the N-terminius and the carbaxypeptitfases remove the
amino acid at the C-terminus. For each proteolytic enzyme, the amino acid residues involved in the
peptide bond that is cleaved are listed beside the R group to the right of the enzyme name
8.1. Digestion of Proteins ana Amino Acid Absorption. Essential Amino Acids 401
Portal vein
Fig. 8.5. Trans epitnelial amino acid transport Sodium-dependent carriers transport both Na1 and an
amino acid into tne intestinal epitnefial cell from me intestinal lumen. Na* is pumped out on the serosal
side {across tne nasoiaterai membrane) in exchange of by tne Nai,K'-ATPase. On me serosal side,
the amino acid is carried by a facilitated transporter down its concentration gradient into tne blood.
This process is an example of secondary active transport
402 Chapter 8. Nitrogen metabolism
Dipcptidcs and tripeptides enter the brush border of die intestinal mucosal cells,
where they arc hydrolyzed to free amino acids, which arc then transported into the
hepatic portal vein. Relatively large peptides maybe absorbed intact, cither by uptake
into mucosal epithelial cells (tra.nsccll.ular). Many such peptides arc large enough to
stimulate antibody formation. - this is the basis of allergic reactions to food compounds.
y-Glutamyl cycle (Fig. 8.6). y-Giuiamyt cyc/e functions in inlenstine, kidney,
pancreas, liver and spleen: but brain and other tissues do not contain a high
quantity of this system. y-Glutamyl transpeptidase (y-GT) plays the key role in the
system. This enzyme catalyzes the transfer of y-glutamyl group from glutathione
(y-glutamylccsteinylglycinc) or another y-glutamyl peptide to transported amino
acid andthc following transport of the complex into the cell. Glutathione is found in.
all animat tissues. For a single amino acid transport into the cell by y-glutamyl cycle,
three ATP molecules are expended.
Fig 8.6. y-Glutamyl cycle, in cells at tne intestine and kidney, amino acids can be transported across
me cell membrane by reacting with giutatnione ij-giutamyi-cysteinyi-giycine) to form ay-glutamyl
amino acid. The amino acid is released into tne cell, and glutathione is resynthesized. However,
the major role of inis cycle is glutathione syntnesis, and many tissues lack tne transpeptidase and
5-oxoprotinase activities
y-GT activity in blood scrum is 30-50 ME/L (pMol/min* mg) for men and 25 —
35 ME/L for women. Determination of y-GT activity in blood scrum is used for
diagnosis of liver and heart disorders, as well as it is the marker in cancer of pancreas,
liver and prostatic gland. The test is also may be used to reveal persons who at risk of
alcoholism and for control of the treatment of chronic alcoholics.
8.2 Transamination ana Deamination of Amino Acids 403
Transamination reactions
The first step in the catabolism of most L-amino acids is the removal of the
a-amino groups, catalyzed by enzymes called aminotransferases or transaminases.
During transamination, the a-amino group of an. amino acid is transferred to
a-carbon of ci-ketoacid (Fig. 8.7).
COO’ COO"
l
c=o COO HgN*— C— H coo-
1 I
ch2 + HgbT—C — H ■ b. ch2 + C—O
1 ArrancMr an st erase I
ch2 R tPLP) R
COO" COO"
Fig. 8.7. Transamination reactioii The amino group from one amino acid is transferred to anomer. Pairs
of amino acids and tneir corresponding u-keto acids are involved in these reactions. u-Ketoglutarate
and glutamate are usually one of the pairs. Tne reactions, which are readiEy reversible, use pyridoxal
priospnaie (PIP) as a cofactor. Tne enzymes are called transaminases or aminotransferases
COO" COO"
COO" COO"
Fig. 8.8. Alanine aim aspartate transamination Dy alanine transaminase (ALT) ano aspartate
transaminase (AST)
Fig. 8.9. Serum transaminase activity (AST and ALT) in myocardial infarction
u/L
Norm
Ritz coefficient is the ratio of AST. ALT in blood serum (11.33 ± 0.42). ALT is
predominantly localized in the cytosol of the liver cells and cardiomyocytes. AST
is found in the cytosolic and mitochondrial fraction of the liver, cardiac muscle
and the majority of organs. Increased activity of mitochondrial AST is found in
acute liver damage and myocardial infarction accompanied by necrosis and cell
membrane distraction.
406 Chapter 8. Nitrogen metabolism
In viral hepatitis (see above Fig. 8.10) the level of ALT increases up to 600—
800 unit/L (5-30 uniis/L in the normal range); AST elevates till 300—400 unit/L (8-
40 nnits/L in the normal range). Thus, the Ritz ratio decreases to 0,6 in hepatitis. In
myocardial infarction (sec above Fig. 8.9), AST increases to 400—500. buL ALT — till
150-200, so the coefficient increases.
Ritz ratio is very’ important for not only diagnosis but to carry differential
diagnostics, for estimation of the effect of treatment and to make a prognosis.
CH2—CH—COO nh;
Histidase
NHg
IMHj OH q NH
] 1 1
HO—CHa--CH—COO" CH3—CH--CH—COO"
L PLP L PLP
h2o H20
Serine ^-HsO Threonine ^-HsO
dehydratase dehydratase
^nh;
nh;
o 0
Indirect deamination
Most amino acids arc unable to be deaminated in one stage as glutamic acid
(L-a mi no acid oxidases present in tissues have insufficient activity). Amino groups of
these amino acids arc transferred to cr-kctoglutaratc with glutamate formation, which
then undergoes direct oxidative deamination. The mechanism of deamination in twro
stages is referred to as indirect deamination (Fig. 8.12).
Indirect deamination occurs with the participation of two enzymes: aminotransferase
(coenzyme pyridoxvl pyrophosphate) and glutamate dehydrogenase ( coenzyme NAD+).
Indirect deamination is the main pathway ofdeamination of the majority of amino acids.
The effect of transamination reactions is to collect the amino groups from many
di fie rent amino acids in the form of L-glutamate. The glutamate then functions as an
amino group donor for excretion pathways that lead to the elimination of nitrogenous
waste products. Ammonia formed as a result of deamination reactions has to be
transported to the liver for the following detoxification.
Another type of amino acid deamination is indirect nonoxidative deamination
with IMP-AMP cycle characteristic for muscular tissue and brain, where glutamate
408 Chapter 8. Nitrogen metabolism
COO"
I
ci-Kelo add n
Glutamate
de hyd rogc nasc has lower activ ily (F i,g. 8.13). A m i no g roup of amino aci ds is t rans ferred
to i M P (inosine monophosphate) by two consecutive reactions of transamination with
the formation of AMP. which is deaminated hydrolytically with ammonia releasing.
Nitrogen-free residue of amino acid is a-kclo acid which may be involved into:
► oxidation reactions to CO2and H2O;
► transamination reaction lor non-essential amino acids synthesis:
» anap I erotic reactions to* supply TCA ITom the loss of metabolites or other
substances synthesis;
* gluconeogenesis;
► ketogenesis.
The quantitative characteristics of the metabolism of amino acids learnt in this section
Y-GT activity in blood serum:
Men - 30-50 U/L
Women — 25—35 U/L
ALT and AST activity sei blood scrum: 5-40 U/L
Ritz ratio: 1.33+0.42
Review tests
I. Organs, involved into digestion, secret enzymes:
Stomach pH Food
1.5-2.0 proteins
I®
Polypeptides
Enzymes:
A. Aminopeptidase.
B. Trypsin.
C. Amylase.
D. Lipase.
E. Pepsin.
410 Chapter 8. Nitrogen metabolism
2. To digest proteins, the following enzymes are synthesized in cells of stomach, pancreas
and small intestine:
Stomach pF - Food
1.5-2.0 proteins
I®
Polypeptides
■ Amino acjds
Absorption
Compounds:
A. Dipeptidase.
B. Chemotrypsin.
C. Lipase.
D. HCI.
E. Amylase.
Situational problems
1. Kwashiorkor, also known as ^edematous malnutrition* because of its association
with edema (fluid retention), is a nutritional disorder most often seen in
regions experiencing famine, h’s most common in sub-Saharan Africa and
other countries where people routinely have a. limited supply of food. It is a
form of malnutrition caused by a lack of protein in the diet. People who have
kwashiorkor typically have an extremely emaciated appearance in all body parts
except their ankles, feet, and belly, which swell with fluid. The symptoms of
kwashiorkor include: change in skin and hair color (to a rust color) and texture,
fatigue, diarrhea, loss of muscle mass, failure to grow or gain weight, irritability,
enlarged liver (hepatomegaly). What causes the symptoms of kwashiorkor1? For
the answer:
a) specify what proteins everybody needs for nourishing diet:
b) explain the meaning of * nitrogen balance*. Specify the nitrogen balance;
observed in protein starvation and in growing children:
c) enumerate biologically active compounds formed in the human body from
amino acids;
d) point out the pathways which, arc accelerated or slowdown in protein
starvation.
2. The young man with weakness, insomnia, and weight reduction visited a doctor.
During the examination, the doctor found out that young man decided to
be vegetarian and during several last months followed plant diet without any
consultation with the doctor. What arc the possible reasons for the described
.symptoms'? For the answer:
8.2 Transamination ana Deamination of Amino Acids 411
a) give the definition to the * nitrogen balance* and whether or not it is changed
in the patient;
b) point out the cause of changed nitrogen balance;
c) point out compounds do contain, high-value, proteins;
d) specify the metabolic pathways of these compound in the human body:
e) answer the main question of the task.
3. To research different diet influences on human health, volunteers were subjected
to particular experiment. It is known that potatoes practically do not contain
tryptophane and methionine, and the total amount of protein in this vegetable
is 2%. Presume, why prolonged artificial diet containing potatoes and cocktail
includingTrc-Arg-Lys-Pro-lle peptide can induce damage to metabolism in the
human body? For the answer:
a) write down the formula of this peptide;
b) explain, whether this peptide being digested in stomach or not;
c) enumerate peptidases which can take pan in given peptide digestion in small
intestine; name proenzymes of these enzymes, mechanism of activation and
activators;
d) specify the group due to the possibility to be synthesized in the body these
amino acids belong to. Explain whether nitrogen balance is changed or not
in animals on this diet;
c) answer the main question of the task.
4. A weight-reducing diet heavily promoted some years ago required the daily
intake of liquid protein* (soup of hydrolyzed gelatin), water, and an assortment
of vitamins. All other food and drink were to be avoided. People on this diet
typically lost 10 to I4 pounds in the first week. Opponents argued that the weight
loss was almost entirely due to water loss and would be regained very soon after
a normal diet was resumed. What is the biochemical basis for this argument? For
the answer:
a) a number of people on this diet died. Explain some of the dangers inherent in
the diet, and how it can lead to death;
b) enumerate essential components which arc shortened in such diet:
c) present particular schemes to confirm your answer.
5. The inflammation associated with atrophic gastritis and a damage to the
stomach lining causes digestive problems; nutrient deficiencies arc observed as
well. Patients suffer from weight loss, vomiting, lack of appetite, nausea, iron
deficiency anemia, pain in the stomach. Why these symptoms occur in damaged
protein digestion in the stomach? For the answer:
a) point out the value of gastric juice pH in healthy people;
b) specify functions of HCI;
c) describe wrhich peptidase takes part in protein digestion in stomach: peculia
rity of its activation and specificity.
6. Amino acid alanine containing N15 isotope in a-amino group was administrated
to mouse with food. It was found that the N15has rapidly appeared in a-amino
groups of aspartate, glutamate and other amino acids in the liver (with the
exception of lysine and threonine). Explain, why does it occur? For the answer:
Chapter 8. Nitrogen metabolism
c) draw the graph indicating the change ofthese enzymes activity in this disorder;
d) write down reactions catalyzed by these enzymes, give full names of direct
and reversible reactions, specify coenzyme;
e) point out the Ritz ratio in this disorder.
II. The most highly sensitive lest in viral hepatitis is y-Glutamyl transpeptidase
(y-GT) increased activity in blood, wrhich level rises 10—15 times more than
the norm (30-50 ME/L). What is the diagnostic value of this enzyme? For the
answer
a) present the main properties of enzymes which activity determination in
patient blood are widely used in. enzyme diagnostics;
b) explain the y-Glutamyl transpeptidase (y-GT) system and present an
appropriate scheme;
c) givc examples of other enzymes which activity is determined in the liver
disorders.
H H 1+
The intensity rate of amino acid’s deamination in tissues is high but low ammonia
levels in bloodstream is the result of active binding of ammonia to non-toxic
compounds (glutamine and alanine) transporting it to the liver and kidneys.
414 Chapter 8. Nitrogen metabolism
k )
Fig 8.14. Summary of ammonia sources and pathways of its metabolism in different tissues
COO- COO-
I
H—C— NHq ATP ADP + Pi
I
H—C— NHt
I
CH2 + NH:3 < 2. I
ch2 + h2o
(Jh2
Glutamine
synthetase
I
C-0
COO" nh2
Glutamate Glutamine
COO’ COO"
f J— C
1 NH3
1
H-€“" NHg
I
ch2
+ H^O 1
CHn + NH3
1
c—o
Glutaminase 1
1 CHe
nh2 COO-
Glutamine Gtulamate
nh5+ h -nh4
Glutaminase in the kidneys releases ammonia, which is excreted as ammonia softs
in exchange for one and two valence cations. This mechanism is one of the main of
maintenance of acid-base balance and preservation of important cations lor die body,
yf/nmon/a cxcat/km wf/A urine is ft 5— /. 2 hour
In metabolic acidosis, there is an increase in glutamine processing by the kidneys
due to induced glutaminase activity. Produced ammonia is used to neutralize acidic
products and forms salts with metabolic acids | mainly NH^CL (NH facilitating
their excretion in the urine. .Ammonia salts excretion can reach IO g/24h in acidosis.
This mechanism of ammonia excretion mainmins acid-base balance and ■Vo4’
and X\/br Auman body: the loss of the same amount of Na' could lead to decreasing of
osmotic pressureof interstitial fluid and blood, and this could result in water depletion.
In the intestinal cells, the same enzyme (glutaminase) hydrolytically liberates
amide nitrogen from glutamine:
Muscle, intestine A. D.
urine
Ammonia salts
I
Urea
Figu &1& Pathways of amino acid nitrogen, and ammonia metabolism. A — elimination of nitrogen
from muscles and intestine in the composition of alanine and giuiamine; B — elimination of nitrogen
from drain and muscles in the form of glutamine; C — ammonia excretion as ammonia salts Dy kidney;
D - inclusion of amino acid nitrogen into urea in the liver; CP — creatine phosphate
Liver Muscte
Glucose
Glucose Glucose
2 NAD"
Muscle proteins
2 NADH
Gluconeogenesis
2 NADH
Glycolysis I
Amino acids
BLOOD
2 Pyruvate 2 Pyruvate
Biosymnelw aminolransiefase
pain ways
2 Alanine
2 Alanine 2 Alanine
Fig. 8.16.The giucose/aianine cycle, within ine muscle, amino acid degradation leads to me transfer
of NHj-group to n-ketogrutaraie and pyruvate. Trie formed alanine travels to me liver, wnere the
cartoons of alanine are used for gluconeogenesis and me alanine nitrogen is used for urea synthesis.
This coufd occur during exercise, when the muscle uses blood-borne glucose
These products must find their way to the liver where pyruvate and lactate arc
converted into glucose by gluconeogenesis, which is returned to the muscles, and
a m mo nia i s co nvc tied to urea for exo ret io n. T he g lucosc -a lan i nc cvc le, i n co ncert w it h
the Cori cycle, accomplishes this transaction. The energetic burden ofgluconeogenesis
is thus imposed on the liver rather than the muscle, and all available ATP in muscle is
devoted to muscle contraction.
Glutamate Giutamme
defiytfiageiiase synth erase
(GDW) COD’
2 ATP 2ADP + Pf
NHJ
J
+ HCOj ------- —-<£.---------- -
I
HjN —C—O—P— o-
I
CPS-I
OH
Cartoamoylphosphate
COO"
I
+ HqN—C—H
I
ocxr (CHa)a
I -Pi |
NH? + -C —H ■------------ ---- ----------- NH
I I Ornithine
transcarbamoylase A q
C—'O iCH2)a
I
OPO2.H2 NH2 NH2
The second amino group now enters from aspartate (generated in mitochondria by
transamination and transported into the cytosol) by a condensation reaction between the
amino group of aspartate and carbonyl group of citrulline, forming
This cytosolic reaction, catalyzed by argininosuccinate synthetase, requires ATP:
COO" COO"
HaN—C—H %N-—C—H
1
(CH2h COQ- (CH2h COO"
1 ATP AMP-PP; 1
ch2
NH
1
C=iO
11
+ p
’H-jjN— C — H
....... 5 1
.A.
Argjnirtosuccinase
NH
|
C—N — C — H
synthetase 1
nh2 coo nh2 COO
NH CHj CH
Argimnosuccinas-e
tyase
COO"
This is the only reversible step in the urea cycle. In the last reaction of the urea
cycle, the cytosolic enzyme arginase cleaves arginine to yield urea and ornithine:
420 Chapter 8. Nitrogen metabolism
COO"
H.N—C—H
I
(CH2h COO" NH2
*h2o I
NH + HN—C —H + C=O
Arginase
I
I I
C = NH (CHzh nh2
I
nh2 NHz
Ornithine is transported into the mitochondrion to initiate another round of the urea
cycle.
COO"
H^hl*C-H
CHa
COCr
l ■ Aspartate
Genesis of urea atoms: amino groups of urea arc derived from ammonia, formed
by amino acid deamination orcntcring from blood (mainly portal veins), and aspartic
acid:
Omit hi no cycle consumes 4 maciocrgic bonds of 3 ATP per turn. Anyway, the
process of urea synthesis has possibility to compensate energy inputs (Fig. 8.19):
► when aspartate is regenerated al the stage of malate dehydrogenation (fumarate -*
malate) - NADH formed provides 3 ATP molecules by oxidation in electron
transport chain:
► glutamate oxidative deamination yields NADH and provides 3 ATP, as well.
Fig. 8.19. The Krebs bicycle, indicating the common steps between the TCA and urea cycles
The urea cycle and the citric acid cycle arc independent cycles but arc linked.
Aspartate formed in mitochondria by transamination between oxaloacetate and
422 Chapter 8. Nitrogen metabolism
COO"
I
+H.3N—C — H
H—C — H Glutamale
Acefiyi-'CoA
H—C—H
COO-
N-Aceiylgluiamate
sy-ftiJiase Arginine
CoA-SH
O COO"
I I
che c-h
N-AcetyfaJutamate
; I
H —C — H
I
COCF
2 ATP 1 2 ADP + P, O 0
nh4+
1 I
rtCO3+ + HgN---- C---- O---- P----- O’
Carbamoyl phosphate
syflihasc l cr
Carbamoyl phosphate
Argi nosucci nate lyase Elevated arginosuccinic acid in urine Lethargy; seizures: vomiting; poor
feeding: hyperventilation; hepatomegaly
Glycine
Glutamine
The quantitative characteristics of the metabolism of amino acids leant! in this section
Ammonia concentration in blood sennit 0.04-0.07 mg/OL {25-40 pmol/L)
Urea blood serum concentration; 15-50 rng/dt (2.5-84 mMol/L)
Urea excretion: 25-40 g/24b
Ammonia sails excretion: 0.5-15 g/24h
Review tests
1. The following compounds lake part tn I he reaction:
COO co—nh2
H-—y—H
H — C— H
H— C— NH?
I
coo" (2 COO"
a
Components:
A. Glulaminc.
B. ATP.
C. Glutamate.
D. Glu La mine synthetase.
E. NH3
8.3. Ammonia Detoxification. Urea Cycle, urea Cycle Disorders, Hyperammonemia 427
CO—'NHz COOH
H—i— H
HgO g)
H—H
I o.
H—C—H H—C—H
Compounds:
A. Glutamate.
B. H2O.
C. Glutamine.
D. Glutaminase.
E. NHr
3. Ammonia sources in the human body:
CD ® ®
Processes:
A. Protein putrefaction.
B. Glucose metabolism.
C. Nucleotide metabolism.
D. Amino acid deamination.
E. Urea catabolism.
4. In tissues ammonia is fixed with the formation of:
Compounds:
A. Glutamine.
B. Alanine.
C. Urea.
D. .Ammonia salts.
E. Glutamate.
428 Chapter 8. Nitrogen metabolism
Tissues:
A. Kidney.
B. Liver.
C. Brain.
D. Muscles, intestine.
E. Brain, muscles and other tissues.
H—c — H—C—H
H—C— NHn
r
C h H—C=NHn
I I I
COO" COO" COO"
8.3. Ammonia Detoxification. Urea Cycle, urea Cycle Disorders, Hyperammonemia 429
Enzymes:
A. Arginine succinate lyase.
B. Arginine succinate synthetase.
C. Carbamoyl phosphate synthetase.
D. Arginase.
E. Ornithine carbamoyl transferase.
Situational problems
1. Normal human blood plasma contains all the amino acids required for the
synthesis of body proteins, but not in equal concentrations. Alanine and
glutamine content is much higher than any other amino acid. Suggest, why?
Confirm your answer with the appropriate schemes.
2. In a study, cats were fasted overnight then given a single meal complete in all
amino acids except arginine. Within 2 hours, blood ammonia levels increased
from a normal level of 94 pmol/L Lo 470 pmol/L, and the cals showed
the c Li nical sy m ptorn s of am mo n ia toxic i ty. A coni rol g rou p fed a co mp I etc amino
acid diet., or an amino acid diet, in which arginine was replaced by ornithine
showed no unusual clinical symptoms. What caused the ammonia levels to rise
in the experimental group? Is arginine an essential amino acid in cals? Why or
why not? For the answer:
a) point out the role of fasting in the experiment:
b) explain, why the absence of arginine can lead to ammonia toxicity:
c) explain, the possibility of arginine substitution by ornithine:
d) present the particular schemes to confirm vour answer.
3. A 15-year-old boy presented with a history of episodic vomiting associated
with subacute encephalopathy from the age of one year. There were 2-3 months
of asymptomatic periods between the episodes. He was born of non-
consanguincous parents at lull term had a normal perinatal period with no
evidence ofbirth asphyxia. During the hospital stay he had 2 episodes ofconvulsive
seizures. Investigations revealed high blood ammonia (288 pg/dL, normal
level is 40-80 pg/dL), markedly elevated levels of citrulline (2200 p moles/L,
normal is 1-55 pmolcs/L), and low blood urea (5 mg/dL, normal is around 7 to
20 mg/dL). He was treated with ant iconvulsivcs and sodium benzoate, advised
a restricted protein diet. What is the reason for the presented symptoms? Why
blood urea concentration is reduced? For the answer:
a) specify the compound 90% of body nitrogen is excreted in;
b) write down the process damaged in this patient:
c) point out the enzyme deficient in this clinical case:
d) explain the mechanisms of ammonia toxicity;
e) explain the expediency of prescribed diet and sodium benzoate.
4. Patient came down with the flu and suffered from dizziness, nausea, convulsive
attacks. Ammonia blood concentration is I. mg/dl. In consideration of influenza,
virus influence on the liver, what mechanisms of pathological symptoms
development? For the answer:
a) specify normal ranges of the blood ammonia level;
b) write down damaged metabolic process a nd point out the site of £ he metabolic
block in flu;
430 Chapter 8. Nitrogen metabolism
b) write down the reaction catalyzed by th is enzyme and explain its physiological
significance;
c) point out the reasons for acidosis development and increased loss of sodium
and potassium in given disorder:
9. it is noted recurrent vomiting, weakness, sleepiness, and convulsive attacks, as
well, in patients with an. inherited disorder of ornithine cycle after food intake.
What are the molecular reasons for observable symptoms? For the answer:
a) write down the scheme of process dam aged in these patients, specify enzymes,
defect of which can cause such disturbances of metabolism;
b) explain the biological function of this process;
c) enumerate substances the content of which is increased! in the blood of these
patients;
d) describe the lox icily of one of them on the nervous system.
Alanine: Pyruvate
ALT (PLP)
Gtu ci-Ketoglutarate
ATP AMP+PPj
HgN+
Glutamine Glutamate
0 NH2
Aspartate Asparagine
432 Chapter 8. Nitrogen metabolism
NH,
P
The synthesis of partially essential amino acids Arg and His docs not respond to the
body needs, especially in growth. Some amount of arginine is formed in the ornithine
cycle. Histidine can be synthesized from ribose and ATP.
Conditionally essential amino acids Tyr and Cys arc formed from the essential:
phenylalanine is converted to tyrosine under the action of phenylalanine hydroxylase.
Methionine is the donor of sulfur required for cysteine; serine provides the carbon
skeleton and a-amino group, as well (Ch. 8.6).
colX
a) Amino acids Pyruvate Oxaloacetatc
434 Chapter 8. Nitrogen metabolism
Enzyme pyruvate carboxylase (coenzyme — biotin) is found in the liver and muscles,
Valine
c) Propionyl-CoA------- Succinyl-CoA
I so leucine
These reactions occur in many tissues where pyruvate carboxylase is not found.
CHj
+HqN—-C—COO
H CO2
Tyrosine metabolism
Tyrosine is used lor protein synthesis as other amino acids: some enzymes involve
tyrosine in their active site (for example, insulin receptor, which is (p/ww pro/em
h'nase); tyrosine is very important as the source for a variety of important metabolites,
as well.
436 Chapter 8. Nitrogen metabolism
In adrenal medulla, brain, and some sympathetic nerve fibers tyrosine isconvertcd
io catechol a mines - various naturally occurring amines: dopamine, epinephrine
(adrenaline), and norepinephrine (noradrenaline) that function as neurotransmitters
and hormones within the human body (Fig. 8.25). Tyrosine hydroxylase (specific
monooxygenase) catalyzes the conversion of tyrosine to DOPA; the reaction
requires tetrahydrobiopterin, O2and Fc1' (the process is analogical to phenylalanine
hydroxylation: see above Fig. 8.24). Dopamine is produced by DO PA de carboxylation
(pyridoxal phosphate is the coenzyme). The following reactions yield
norepinephrine (hydroxylation) and epinephrine (methylation). The particular
catecholamine that is synthesized by a nerve cell, or neuron, depends on which
enzymes are present in that cell. Dopamine, a neurotransmitter widely distributed
CHa—CH—COO-
NHg
[Tyrosine]
Tyrosine hydroxylase
{totranydrofciopterin, Fe2\ O?)
CH2— CH—£>OCr
NHa
DOPA
DOPA decarboxylase
- CO2
[pyridoxaj-pnospnata)
Dopamine
Dopamine Hydroxylase
it h srariy drobiOLitorin;
ascorbate, Qu , O?)
— CHz—NHa
Noradrenaline
Methyltransferase
(S^adenosylmethionine)
Adrenaline
Fig. 8.25. Catecholamines are derived from the amino acid tyrosine
8.6. Special Pathways of Some Amino Acids: Phenylalanine MetaDoiism, Tetranyttrofolate... 437
in the central nervous systemT including the basal ganglia of the brain that collectively
control muscle tone, inhibit movement, and control ircmour. Norepinephrine is a
neurotransmitter of sympathetic nervous system released in response to stress. The
general function of norepinephrine is to mobilize the brain and body for action.
Norepinephrine is important for attentiveness, emotions, sleeping, dreaming, and
learning. Epinephrine (adrenaline) is the hormone of intensive physical activity and
stress; it regulates basal metabolic rate and strengthens cardiac muscle contraction,
as well.
In thyroid gland, tyrosine is the precursor of iodothyronines (thyroxin and
triiodothyronine}, these hormones arc iodinated tyrosine residues.
In melanocytes (skin pigment cells, hair follicles and retina) tyrosine is the precursor
of melanin — high molecular weight hydrophobic pigment (Fig. 8.26). The synthesis
of melanin is a niultistep process. The first two steps in the synthesis of melanin arc
catalyzed by tyrosinase (DOPA oxidase), a fopper-co/j/amiXgoxidase, which converts
DOPA todopaquinonc.
Tyrosine
Tyrosine Hydroxylase .
Tyrosinase D0PA
(02; Oi*]
(DOPA oxidase} l
z DOPA quinone
Fig. 8.26. Tyrosine is me precursor of meraniiiSu DOPA quinone is converted to further products,
eventualFy forming eumelanin and pneomelanin — pigments of Dfack-Drown and yeltow-red color
Degradation of phenylalanine and tyrosine takes place in the liver (Fig. 8.27).
Homogentisic acid is an. intermediary product of tyrosine catabolism and converted
to acid with the cleavage of the aromatic ring. Homogentisate
dioxygenase requires vitamin C and Fe2' (or heme, sometimes Cu2') for catalysis. The
last step in the pathway produces both/wmera/c (which can be used for gluconeogenesis)
and the ketone body ace/Mce/a/c.
438 Chapter 8. Nitrogen metabolism
CH
Aminotransferase Dioxygenase
CH—NHg
GOO coo-
I
Tyrosine P-Hydroxypnenylpyruvato H— C— H
C-0
COO" CHa
H-A—H
Acetoacetate
+ Qa I + h2c
OH C»0
Homogentisate
dioxygenase
I +
H—C —H
(n+C;Fe/+)
I
CH2-COO" C=0 COOT
T
OH
I
CH
CH
CH
Sh
Homogantisic acid I
cocr COO'
Fumarytacetoacetate Fumarate
Fig. 827. Phenylalanine and tyrosine degradation Note, that phenylalanine is not present in tne
scheme (Phe is oxidized to Tyr — see above)
DihydrotHCiplerin Oxidated
Protein synlresrs
Tyraane CaSocfiolamines
Pneflylalartine Thyronne
Hydrcnyla&e Melanin
NADU + H
Fhenyllaclic sod
Hwtuciion
Those who suffer from this form of hyperphenylalaninemia may have a deficiency of
tyrosine (which is created from phenylalanine by PAH), in which case treatment is
supple mental ion of tyrosine to account for this deficiency.
Levels of dopamine can be used to distinguish between these two types.
Teirahydrobioptcrin is required to convert Phe to Tyr and is required to convert Tyr
to L-DOPA via the enzyme tyrosine hydroxylase. L-DOPA, in turn, is converted
io dopamine. Low levels of dopamine lead to high levels of prolactin. By contrast,
in classical PKU (without dihydrobioptcrin involvement), prolactin levels would be
relatively normal.
Screening newborns for genetic diseases can be highly cost-effective, especially in
the case of PKU. The tests (no longer relying on urine odor) arc relatively inexpensive,
and the detection and early treatment of PKU in infants (eight to ten cases per
100,000 newborns) saves millions of dollars in later health care costs each year. More
importantly, the emotional trauma avoided by early detection with these simple tests
is inestimable.
Albinism includes a spectrum of clinical symptoms characterized by hypomela nosis
due to heritable defects in eye and skin melanocytes (tyrosine hydroxylase converting
DOPA io dopaquinone is affected: Fig. 8.28). Hair bulbs from these patients fail to
convert added tyrosine to pigment melanin and melanocytes contain unpig me med
mclanosomes. Patients suffer decreased visual acuity and photophobia. Prolonged
insolation can lead to burnsand skin cancer.
Another inheritable disease of phenylalanine and tyrosine catabolism is
alkaptonuria, in which the defective enzyme is homogentisate dioxygenase (Fig. 8.28).
Less serious than PKU. this condition produces few ill effects, although large amounts
of homogentisate arc excreted and its oxidation turns the urine black. Accumulation
of homogentisic acid can. occur in cartilage, casing arthritis. There’s no specific
treatment for alkaptonuria. Treatments arc focused on preventing and relieving
possible complications: arthritis, heart disease, and kidney stones.
Phenylalanine
BH I Pheny
i hydroxylase
Homogentisic acid Tyrosine
Tyrosine
* hydroxylase
Norepinephrine
SAMe
Epinephrine
The major sites of serine synthesis arc the liver and kidney.
1 n human body, serine is involved into protein synthesis r first of all, and is found in
many enzyme active sites, as well (serine proteases, for example). Serine can be used
for synthesis of sphingosine-containing lipids, phosphatidylscrinc, glycerol, cysteine,
and glucose. Serine is degraded lo form pyruvate (sec above).
Glycine is a non-essential amino acid: serine (three carbons) is the precursor of
glycine (two carbons) through removal of a carbon atom by serine hydroxymethyl
transferase:
442 Chapter 8. Nitrogen metabolism
nh5
HO
L-Serine
!
Serine hydraxymeUiyl
Iransferase (Pl_P]
M^nyiens-Hd-foiaie
Glycine
The coenzyme in the reaction is tetrahydrofolate (TH I7. H4-folate) which binds
P-carb on of serine forming methylene-11^-folate. The reaction is reversible and glycine
is converted to serine by enzymatic addition of a hydroxymethyl group that requires
the coenzymes methylene-tetrahydrofolate and pyridoxal phosphate.
Glycine is abundant in. proteins, for example, collagen involves 33% of this amino
acid. Glycine is the precursor of glutathione, creatine, heme, purine nucleotides,
coenzymes etc.
Glycine undergoes the cleavage to COr NH‘, and a methylene group (-CH,-).
This readily reversible reaction, catalyzed by glycine cleavage enzyme (called glycine
synthase. as well), requires tetra hydrofol ate, which accepts the meth dene group. 1 n this
oxidative cleavage pathway the two carbon, atoms of glycine do not enter the citric
acid cycle. One carbon is lost as CO and the other becomes the methylene group of
A/5, Ar"-methyl one-tetrahydrofo late, a one-carbon group donor in certain biosynthetic
pathways:
NHq
I
ch2xc^o-
I
0
Glydne
H*-folate
Glycine
synthase
NE,.N10-CH2-H4-fulate
(mettiylena-HrloJate)
CO2+NHj+NADH+ Hh
8.6. Special Pathways of Some Amino Acids: Phenylalanine MetaDoiism, Tetrafiytf rofoiate... 443
Fig. 829. Reduction of lol ate to letratiyflrofolate: A — folic acid inctudes pteridine ring attached
to para-ami nobenzoic acid iPABA}, and glutamate; B - tetra hydrofolate (H4-foEate); 0 — Dy liver
enzymes (fa late reductase and dinydrofoiate reductase, coenzyme — NADPH: fotate is successively
reduced to dihydrofolate and tetrahydrofoiate
Fig. 8.30. A key feature of H+-folate is that it can carry a variety of one-cartoon groups
NAD* NADH + H+
Fb/afe deficiency is relatively common, and can be caused by imbalanced diet (low
in vegetables, fruits and meal), malabsorption of folic acid, hepatitis, cirrhosis and
other liver lesions inducing decreased activity of folate reductase. Folate deficiency
leads to damaged nucleotide biosynthesis and thus cell proliferation. As the precursors
for blood cells divide particularly rapidly, disturbances of the blood picture can occur,
with increased amounts of abnormal precursors for megalocytes
8.6. Special Pathways of Some Amino Acids: Phenylalanine Metabolism, Tetrabydrofolate... 445
antimatabolite
Folic
acid
Methionine +
R R - CHg
S-adeno sy im etnioni ne
(SAMe)
< J
Methylfransferase
S-adenosyl nomocy stei ne
(SAH)
Transfer of the methyl group from S-adcnosyl meth io nine to an acceptor yields
S-adenosylhomocysteine, which is subsequently broken down to homocysteine and
adenosine. The detachment of methyl-group and its transfer to acceptor is catalyzed
by methyltransferase (Fig. 8.34).
Synthesis:
adrenaline,
choirne,
creatine,
camrline. etc.
Detoxification
Homocysteine ol toxic metabolites
and medians#
Adenosine compounds
Serine
PLP
Cysteine
Homoserme
Transmethylation reactions
Creatine synthesis
Muscle celts and nerve tissue use a phosphorylated form of creatine to store energy
(Fig. 835). Of the approximate 120 grams of creatine that exists in the human body,
95% is localized in the skeletal muscle, and only a few grams arc localized in the
brain. Creatine phosphate (macroergic compound) is particularly important since
it replenishes ATP <a cellular unit of energy) without relying on oxygen. Normal
metabolism cannot produce energy as quickly as a muscle cell can use it, so an extra
storage source is needed. The phosphate group can be quickly transferred to ADP
448 Chapter 8. Nitrogen metabolism
to regenerate the ATP necessary tor muscle cont raction. The role of creatine in the
brain is essential. Energy synthesis in the brain via the cicatine/phosphocreatine/CK
system is thought to be involved in signal transduction in the central and peripheral
nerves, and in maintenanceof membrane potential.
Creatine phosphate can be broken down into creatinine, which is then excreted in
the urine.
NHZ* NH2+
I
C NH2
IC—NH2
NH argmne amfinine NH N — CH-,
1
CHS —
V
J. 1 SA Me SAH 1
CH3 A CHa
ATP
o.
ADP
Creative
COO" COO coo- kisia&e
Creatinine clearance tests are used Lo measure kidney function: it maybe not within
the normal ranges due to kidney disease or damage . Creatinine excretion may be raised
in excessive food intake, intensive physical activity and increasing muscle bulk, as well
as diabetes mcllitus, hypothyroidism, acromegaly (pituitary' gland pathology), and
febrile slate. Urine creatinine excretion is reduced in starvation, vegetarian nutrition,
muscle dystrophy, hyperthyroidism, and anemia as well.
Creatine synthesis requires three amino acids: arginine, glycine and methionine. The
p roccss beg ins i n kid ney wi th the form ation ofgua nid i noacctale. T he n guanid i noaceta to
is transported into the liver where methylated by SAMc and converted to creatine.
Creatine is transported to muscles and the brain from the liver. Creatine kinase (CK)
catalyzes the reaction of creatine phosphate synthesis.
Phosphatidykholi ne synthesis
Phosphatidylcholine (lecithin) can be synthesized (Fig. 8.36) by the methylation
pathway that converts phosphatidylctha nolam inc to phosphatidylcholine, principally
in the liver. The methyl donor is S-adenosylniethionine. Phosphatidylcholine is
transported lo tissues by LDL. Phosphatidylcholine plays a particular function in the
metabolism of HDLand cholesterol reverse transport (Ch. 7.10).
8.6. Special Pathways of Some Amino Acids: Phenylalanine MetaDoiism, Tetranytfrofolate... 449
[EthanolamiFej
Phosphatidyl- y Pnosphatidylserine \
etnanOlamcne-serine 1
transferase A
O Phosphatidyl-
[ Sec ine) \ ■ 1 serine
* CH2—0—C—R1 cfecarboxyksse
O J
1 2
CH—0— C—R2
O
a
CH2—0 — P — 0 — CH?—■C4a— NHg \
O" COa
Phosphatidylemanoiamina
3SAM
Methyltransfera st?
3SAH
□ — GHZ— CH2“+N(CH3)3
Phospnatidyicnoiine
Carnitine synthesis
Carnitine is the transporter of the acyl group into the mitochondria. Skeletal
muscles have 97% ofall carnitine in the body, they are dependent on carnitine provided
from endogenous synthesis (occurs in the liver and kidney, but not in skeletal or heart
muscles) or diet (meat and fish products). Carnitine levels were correlated with the
intake of essentia I amino acids, methionine, and lysine (as substrates of its endogenous
synthesis). Carnitine synthesis occurs by methylation of y-aminobutyric acid with
SAMc as methyl group donor; hydroxylation reaction ofy-buti robetai ne requires Fc2'
and vitamin C as reducing agent:
450 Chapter 8. Nitrogen metabolism
( Lysine )
T
y-Aminobutirate
3SAMs
3SAH
3 Methyl transferase
y-Butirobetaine
Carnitine
r- ch—coo’ —:-----
■Amino • f*—ch2—nh2 + cot
acid
' * decarboxylase
nh3 (PlP) Amine
Amino acid
Products of this reaction arc amines with expressed biological activity: many
important neurotransmitters arc primary or secondary amines, derived from amino
acids. In addition, sonic polyamines that form complexes with I) NA (c.g., spermine,
spermidine) are derived from the amino acid ornithine, a component of the urea cycle.
Catecholamines synthesis and function see above in chapter 8.6.
1. Glutamate decarboxylation gives rise to y-aminobutyrate (GABA) which is the
main inhibitory neurotransmitter:
CO2
► GABA content in brain is ten limes more than other neurotransmitters. GABA
increases permeability of postsy naptic membranes for K *■ that induce deceleration
of nerve impulse, it increases respiration of nerve tissue and improves blood
supply of brain, as well. GABA inactivation occurs either by transamination and
conversion to succinate (TCA metabolite), or oxidation by monoamine oxidase
(MAO).
» GABA underproduction is associated with epileptic seizures.
► GABA analogs arc widely used in the treatment of epilepsy, damages of
cerebral circulation, mental retardation, depression, and hypertension. Levels
of GABA can. also be increased by administering inhibitors of the GABA-
degrading enzyme — (SABA aminotransferase, which is used as anticonvulsants
(e.g.T convulex).
2. Another important neurotransmitter, serotonin, is derived from tryptophan in
a two-step pathway (note that first reaction catalyzed by //rp/op/™
require tetrahydrobiopterin as the cofactor and molecular oxygen):
CO
CO2
* con tracts smooth muscle in lungs, and induces asphyxia (shortness of breath);
• acts as neuralransmliter (Fig. 8.37).
system
J . _________ V J . Chorine
Serine decartojcylase Ethanolamine
(PLP) meffiyhransfera&e
0
rt3C HaC 1
H3C—N4-— Ch?—ch?—-Oh H.3C—n *■—Ch?—Ch?—O-— C — CHS
H3C H^C
ChoSfW AcetylchoBme
+
Choline
O acetyltransferase CoASh
CoA—s—c —“ ch3
Ace?yl-CoA
► Acetylcholine is the ncu rot ransm it ter that transmits signals between motor
nervesand skeletal muscles.
► Myasthenia is a chronic autoimmune neuromuscular disease that causes weakness
in the skeletal muscles, which arc responsible for breathing and moving parts
of the body, including the arms and legs. In most individuals with myasthenia
gravis, this is caused by antibodies to the acetylcholine receptor.
8.7. Decarboxylation of Some Ammo Acids, Synthesis of Neurotransmitters and Hormones 453
'AceLyl Coa)
Acetyicholffw
In tre synapse, ACh is rapidly
Proven down by me enzyme
acetylcholinesterase {ACME)
Cholinergic
f receptor
fadh2 +H2O
-nh3
R— CHwaNH
Monoamine
Amins oxidase Imine Atdfifiyde H Carbonic On
acid
SAMs
The quantitative characteristics of the metabolism of amino acids learnt in this section
Creatinine contents in urine: 8.8—17.6 mMol/L
Creatinine excretion: 1—2 g/24 h (directly proporlional to muscle bulk
454 Chapter 8. Nitrogen metabolism
Review tests
1. To CCP metabolites the following arc? converted:
[ Glucose ]
Pyruvate < 1
Ma'a'.e-
CAC
Fumarate a-K&togJutarate ^Glutamate
Succinate Succmyl-CcA
Amino acids:
A. Lysine.
B. Threonine.
C. Asparagine.
D. Glycine.
E. Histidine.
2. From glycolysis and CCP metabolites the following arc synthesized:
Glucose
Phospnoglycerate^ —-11 |
*
Sucotfiate SuccinykCOA
8.7. Decarboxylation of Some Ammo Acids, Synthesis of Neurotransmitters and Hormones 455
Amino acids:
A. Alanine.
B. Glutamate.
C. Cysteine.
D. Asparagine.
E. Serine.
.1. Coenzymes I l^-folatc derivatives are used for:
Processes:
A. Purine nucleotide synthesis (C2).
B. Methionine regeneration.
C. Purine nucleotide synthesis (Ca).
D. Pyrimidine nucleotide synthesis (TTP).
E. Methylation reactions.
4. SA Me synthesis:
ch3 CH3
s
11
ch2
NHj
COOH
Situational problems
1. Glucose synthesis was investigated in isolated hepatocytes. Different amino acids
were added to the cell culture and the rate of glucose formation was checking
in. In control experiment, the rate of gluconeogenesis was 0.15 pmol/min of
glucose forma lion. Wien alanine, proline and glutamic acid were introduced to
cell culture, the rale of gluconeogenesis was increased to 0.17-0.18 pmol/min,
but the adding of lysine or leucine had no effect. Why the introduction of certain
amino acids increases the rate of *n.cw glucose* formation? For the answer:
a) write down the process taking alanine as the substrate:
b) specify the group to which used in experiment amino acids arc related to.
c) write dowrn the scheme of gluconeogenesis originated from these amino acids.
2. In volunteer group starving more than 2 days, blood glucose concentration
reduced till 60 mg/dl. Il was revealed that glucose in blood increases to 130 mg/dl
after glutamate and aspartate injection. What cause mentioned changes? For the
answer:
a) write down the first reaction in catabolism ofGlu and Asp;
b) specify the group due to the fate of nitrogen-free residue they belong to;
c) write down the scheme of these amino acid nitrogen-free residues usage in
reduced blood glucose levels.
3. A 2-year-old child was taken to the hospital. His mother said that he vomited
frequently, especially after feedings. The child’s weigh Land physical development
were below normal. His hair, although dark, contained patches of while. A urine
sample treated with ferric chloride (FcCI3) gave a green color characteristic of
the presence of pho nylpyru vale. Quantitative analysis of urine samples revealed
the presence of phenylalanine 7.0 pMol/L (0.01 in health), phe nylpyru vale
8.7. Decarboxylation of Some Ammo Acids, Synthesis of Neurotransmitters and Hormones 457
4.8 pMol/L (Din health) and phenyl lactate (Din health). Suggest, which enzyme
might be deficient i n this child? Why does the abnormal pathway come into play
under those conditions?
a) designate the disorder;
b) write down the reaction of damaged pathway; explain the appearance of
phenylalanine in the urine in the large amounts;
c) write down the scheme of an alternative metabolic pathway of the substrate;
d) explain the presence of white patches in the boy’s hair;
e) propose treatment.
4. The mother of a 4-month-old female baby attended in the well-baby clinic with
the complaint of black staining of the diaper after few minutes of urination.
The baby was born, of a non-consanguineous marriage, healthy and breast fed.
Mother noticed that stain first at the age of two and half months. The urine,
when kept in a test tube for two hours, turned black. What is the reason of
present symptom? What compound was revealed in increased concentration in
laboratory examination of urine? For the answer:
a) name compounds which, impart dark color to the urine of patient, point out
the disorder this symptom is observed in;
b) write down the scheme of metabolic pathway in which named! component is
intermediary metabolite, specify enzymesand cofactors;
c) specify the reaction in the scheme decreased rate of which causes the disorder;
d) presume. wrhat type of mutation can. lead to this pathology.
5. Amino acid methionine is used as medicine due to its lipotropic effect (^removes*
fat excess from the liver) in initial stages of liver cirrhosis, toxic liver lesions,
and chronic alcoholism. Methionine treatment reduces cholesterol contents
in blood and i ncreases the level of phospholipids. What arc mechanisms of the
therapeutic effect of methionine? For the answer
a) specify methionine functions in the human body:
b) write down the reaction of methionine activation;
c) name particles which arc formed to transport synthesized lipids from the liver
into the blood and then to tissues; describe their composition and metabolism;
d) point out the compound, which is necessary to build chosen lipid transport
forms. Specify the role of methionine in its synthesis. Present an appropriate
scheme;
c) specify, which particles have antiatherogenic activity and reduce the risk of
atherosclerosis development.
6. The doctor prescribed vitamin Bq (U(„ Iblic acid) in complex with Bpto infant
suffering megaloblastic anemia to stimulate erythropoiesis. What is the action
mechanism of these vitamins? is prescription expedience? Why severe liver
disorders can lead to B,,deficiency symptoms? For the answer:
a) write down the scheme of vitamin Byconversion to coenzyme, specify
enzymes, name amino acids which arc donors of one-carbon group:
b) enumerate derivatives of this coenzyme;
c) point out the processes these derivatives take place in;
d) answer the main question of the problem.
458 Chapter 8. Nitrogen metabolism
3 types of modifications
I. 4 methyl groups, CH3
II. 2 vinyl groups, CH=CH3
ill. 2 propkmate groups, CH^-CHs-COOH
The two starting materials in heme synthesis arc succinyl-CoA, derived from
the citric acid cycle in mitochondria and glycine. The product of the condensation
reaction between succinyl-Co A and glycine isa-aminolevulinate (ALA). This reaction
sequence is catalyzed by ALA synthase (pyridoxal phosphate-de pendent enzyme).
Synthesis of A LA occurs in. mitochondria (Fig. 8.40).
In the cytosol two molecules of ALA arc condensed by the enzyme ALA
dehydratase to form two molecules of water and one of porphobilinogen (PEG). ALA
dehydratase is a zinc-con lai ni ng enzyme and is sensitive to inhibition by lead, as can
occur in lead poisoning. The formation of a cyclic tctrapyrrolc — i.e., a porphyrin —
occurs by condensation of four molecules of PEG to form uroporphyrinogen HI by
the action of uroporphyrinogen HI synthase. Uroporphyrinogen has the pyrrole
rings connected by methylene bridges Uroporphyrinogen III is converted
460 Chapter 8. Nitrogen metabolism
ALA dehydraiase
- Liver (-10%):
MITOCHONDRIA where cytochrome P450 is
synthesized. Synthesis is
Heme Rofpnobdmogen
up-regulated in response to
Hydf DwyiTl ei hylbilane
Fw.ocHslaiMS |fof) | syntfiase drug/alcohol metabolism
Coproporpriyrinogon ill
1 1 Uroporphyrinogen
decartozylase
— Copropaphyrinogen III
- Cytosol:
the intermediate 4 steps
Note: matire RBCs don’t have a
mitochondria and cannot make heme
Fig. 8.4(1 Heme synthesis pathway. Ta produce one molecule of heme, 8 molecules each cf glycine
and succinyl-CoA are required. A series of porphyrinogens are generated in sequence
io co pro porphyrinogen HI and then to protoporphyrin [X. Several steps arc involved in
this conversion. The final step in heme synthesis involves (he incorporation of ferrous
iron into protoporphyrin IX in a reaction catalyzed by ferrochelatase (heme synthase),
another mitochondrial enzyme.
85% of heme synthesis occurs in erythroid precursor cells in the bone marrow and
the majority of the remainder in hepatocytes. The porphyrinogens described above are
colorless, containing six extra hydrogen atoms as compared with the corresponding
colored porphyrins. However, the porphyrinogens arc readily auto-oxidized to their
respective colored porphyrins. These oxidations arc catalyzed by light and by the
porphyrins that are formed.
ALA synthase is the key regulatory enzyme in the biosynthesis of heme. Heme acts
as a negative regulator of the biosynthesis of ALA synthase: thus, the rate of synthesis
of ALA synthase increases greatly in the absence of heme and is diminished in its
presence. Fe*+ is an inductor of A LA synthesis in. reticulocytes.
weakness or paralysis, red or brown urine, mental changes, such as anxiety, confusion,
hallucinations, disorientation or paranoia. CuAanrous /rorpAyrior can result in
permanent skin damage. The oxidation products, the corresponding porphyrin
derivatives, cause photosensitivity (Fig. 8.41), a reaction to visible light of about
400 nm. The porphyrins, when exposed to light of this wavelength, arc thought to
become ^excited- and then react with molecular oxygen to form oxygen radicals. These
latter species injure lysosomesand other organelles. Damaged lysosomes release their
degradative enzymes, causing variable degrees of skin damage. The skin, blisters can
become infected and when the skin heals alter cutaneous porphyria, it may have an
abnormal appearance and coloring, be fragile, or leave scars.
I
tMconjugaied bilirubin UrotJifinaqeft
Blood
Hepatic sinusoid
LMcortfugaDea bilirubin
iianiporlod
Hepatocyte icqugJKd to
gtKWDnic and
UdDirKjgen
foddered in
bancral
Friases
Conju^aied bilirubin - Urobilinogen
Small intestine
Fig. 8.43. Heme degradation Heine is degraded to Li limb in carried in me blood try albumin, conjugated
to form tn e di glucuronide in tn e liver, and excreted in me bile, in tne intestine bilirubin is converted to
urobilinogen by fecal microflora
Bilirubin is only sparingly soluble in water, but its solubility in plasma is increased
by noncovalent binding to albumin. Each molecule of albumin appears to have
one high-affinity site and one low-affinity site for bilirubin. In 100 mL of plasma,
approximately 25 mg of bilirubin can be Lightly bound to albumin at its high affinity
site. Bilirubin in excess of this quantity can be bound only loosely and thus can easily
be detached and diffuse into tissues. A number of compounds such as antibiotics and
other drugs compete with bilirubin for the high-affinity binding site on albumin. Thus,
these compounds can displace bilirubin from albumin and have significant clinical
effects.
in the liver, the bilirubin is removed from albumin and taken up at the sinusoidal
surface of the hepatocytes by a carrier-mediated saturable system. Once bilirubin
enters the hepatocytes, it can bind to certain cytosolic proteins, which help to keep it
solubilized prior to conjugation. They may also help to prevent the efflux of bilirubin
back into the blood stream.
464 Chapter 8. Nitrogen metabolism
Bilirubin is hydrophobic and would persist in cells (c.g.. bound to lipids) if not
rendered water-soluble. Conjugation of bilirubin with glucuronic acid occurs in the
liver. Hepatocytes convert bilirubin to a hydrophilic form (water solublek which is
readily excreted in the bile, by adding glucuronic acid molecules to it. This process is
called conjugation. The conjugation of bilirubin is catalyzed by a specific glucuronyl
transferase. The enzyme is mainly located in the endoplasmic reticulum, uses UDP-
glucuronic acid as the glucuronyl donor, and is referred to as bilirubin-UGT. Bilirubin
monoglucuronide is an intermediate and is subsequently converted to the diglucuronide
referred to as conjugated bilirubin t Fig. 8.44}.
’Conjugation
BdifLibin
monoglucuronidg .
Bilrfufin
diglucuronidfl
Fig. 8.44. Formation of bilirubin monoglucuronide and bilirubin diglucuronide (direct) in hepatocytes.
UDPQA - UOP-glucuronic acid
Most of the bilirubi n excreted in the bile of mammals is in the form of bilirubin
diglucuronide. Bilirubin-UGT activity can be. induced by a number of clinically useful
drugs, including phenobarbital.
Secretion of conjugated bilirubin into the bite occurs by an active transport
mechanism, which is probably rate-limiting for the entire process of hepatic bilirubin
metabolism. The hepatic transport of conjugated bilirubin into the bile is inducible by
those same drugs that arc capable of inducing the conjugation of bilirubin.
As the conjugated bilirubin reaches the terminal ileum and the large intestine, the
glucuronides arc removed by specific bacterial enzymes (glucuronidases), and the
pigment is subsequently reduced by the fecal flora to a group of colorless lelrapyrrolic
compounds called urobilinogen. In the terminal ileum and large intestine, a small
fraction ofthe urobilinogen is reabsorbed and rccxcrctcd through die liver to constitute
the e/f/cro/jcpij/fc MrofcilfHogef? cycle. A small part of urobilin is excreted by kidney
(1,-2 mg/24 h). Under abnormal conditions, particularly when excessive bile pigment
is formed or liver disease interferes with this im rahepa tic cycle, increased urobilinogen
may also be excreted in the urine. Normally, most of the colorless urobilinogen formed
8.8. Special Products Derived from Amino Acids 465
in the colon by the fecal flora arc oxidized there to urobilin (or stercobilin - colored
com pounds) and arc excreted in the feces. The darkening of feces upon standing in
air is due to the oxidation of residual urobilinogen to urobilin.
A method for quantitatively assaying the bilirubin content of the scrum was first
devised by van den Bergh by application of Ehrlich's test for bilirubin. The Ehrlich
reaction is based on the coupling of Ehrlich’s diazo reagent and bilirubin to produce
a reddish-purple azo compound. The form of bilirubin that would react without the
addition of methanol was thus termed -direct-reacting * To that form of bilirubin
which could be measured only after the addition of methanol, the term "indirect-
reacting* was applied. The indirect bilirubin is - free* (unconjugated) bilirubin its route
to the liver from the reticuloendothelial tissues, where the bilirubin was originally
produced by the breakdown of heme porphyrins. Conjugated bilirubin, being water-
soluble, can react directly with the diazo reagent, so that she :■ direct bilirubin-.- is
actually a bilirubin conjugate (bilirubin glucuronide). Because of its hydrophobicity,
only unconjugated bilirubin can cross the blood-brain barrier into the eeniral nervous
system: thus, encephalopathy due to hyperbilirubinemia (kernicterus ) can occur only
in connection with unconjugated bilirubin. On the other hand, because of its water
solubility, only conjugated bilirubin can appear in urine (choluria).
Type of bilirubin elevated Unconjugated bilirubin Bota conjugated & C-zoiugaled bilirubin
unconjugated bilirub- -
Urine
’ r Conj. Bi&ubin Absent ++ + * +■
,z UrobSncaen +++ + early, Obst -dec. Absent
Bite salt “ Abaani + ++
De novo synthesis
o o
Oh Oh Oh Oh O O
Gin Glu
Amidotransferase
Fig. 8.46. The origin of purine ring atoms. Pay attention: nitrogen atom in NH?-group of
5-pnospnoriDosyl-t-amine is N,. in purine ring. Nr N^ — amide nitrogen of Gin; C4, Cr Nr — Glycine;
Cy, CE—metnenyl-H4-foiate(8};Jformyl-M(-foiate{2); Cs — COZ; N, — Aspartate
H
“OOC- - CHy—C—- COO"
Fig. 8.47. AMP and GMP synthesis: 1 — adenylosuccinate synthetase; 2 — adenylosuccinate lyase;
3 - IMP dehydrogenase; 4 — GMP synthase -iXMP glutamine amidotransferases)
M3N 3
E pnotphace
O
SyffltldM II
ncHz
Carbamoyl Aspsrlk acid Carbamoyl aspartic acid (CAA| Dtfiydroorolic add IDHC'AI
prtosprtale (CAP)
WAD"-
DifiydroorotaEe
aEe
«e
deliytfrogenase ■.*•
NADH + H+-
CO2
V®
■j ■: I: z ,-lic acid
decarboxylase
LIMP OMP
NADPH * H” NADP*
dUDP
Ribonucleotide
reductase
jx“NS,Nl0-Me1hilene H4 Folate
© [ THymidylaEe syttdiase
P* H2 folate
TMP
Fig. 8.48. Conversion of carbamoyl phosphate ana aspartate to UMP. Not mentioned enzymes in me
figure: 7 — NMP kinase: 8 — NDP kinase (ubiquitous enzyme): 11 — phosphatase. Pay attention —
cseoxyribonucseatides are syntnesizecf in diphosphate form, OllMP is me substrate fordTMP formation
8.8. Special Products Derived from Amino Acids 471
Fig. 8.49. Formation of fleoxYribonucteolides. Reduction requires tniorefloxin (a small protein with
molecular weight 12 kD}, which is oxidized during me reduction of the ribose 2r-hydroxyl group of
the nucleotide dipn ospnate. rib ortuc leotide reductase, and NAD Ph. The immediate reductant, reduced
thio redoxim is produced by NADPn and thio redoxin reductase
Salvage synthesis
Free purine and pyrimidine bases arc constantly released in the cel! during the
metabolic degradation of nucleotides. Free purines arc, in large part, salvaged and
reused to make nucleotides, in a pathway much simpler than the de novo synthesis
described earlier. One of the primary salvage pathways consists of a single reaction
catalyzed by adenine phosphoribosyl transferase, in which free adenine reacts with
PRPP to yield the corresponding adenine nucleotide:
Free guanine and hypoxanthine {the deamination product of adenine) arc salvaged
in the same way by hypoxanthine-guanine phosphoribosyhransferase:
they arc extremely hostile and show compulsive. self-destructive tendencies: they
mutilate themselves by biting off their fingers, toes, and lips.
The devastating effects of Leach-Nyhan syndrome illustrate the importance of the
salvage pathways. Hypoxanthine and guanine arise constantly from the breakdown
of nucleic acids. In the absence of hypoxanthinc-guaninc phospho ribosy II ransferase,
PR PP levels rise, and purines arc overproduced by the de novo pathway, resulting m
Ajgft levels of uric acid production andgouf-b'ke damage fa tissue. The brain is especially
dependent on the salvage pathways, and this may account for the central nervous
system damage in children with Lesch-Nyhan syndrome.
ATCase T-i ”
ATP..... I
Carbamoyl aspartate
De novo pathway |
(steps 3-6) t
UMP
UDP
I
CTP synthetase
G7P...............
Ribose 5-pnospnate
PRPP synthetase
e--
I
5-pnospnor ibosyl 1 -pyjophospnais
(PRPP)
Glutamine phosphorytKssjl
e-1.....
amid'Otransferase
5-pnospncxibosyi 1-amine
t Adenylosuccinate
XMP * * Adenylosuccinate
1 ■ 1
! \ *
r GMP AMP **
jr
1 1
I I
GDP ADP
1 I
I
L GTP ATP
Fig. 8.51. The regulation of purine synthesis. PRPP synthetase has two distinct allosteric sites, one for
ADP, the other for GDP. Glutamine pnosphorioosyi aminotransferase contains adenine nucleotide and
guanine nucleotide-binding sites; me monophosphates are the most important, although the di- and
tri-pbosphates will also bind to and inhibit the enzyme. Adenylosuccinate synmetase is inhibited by
AMP: IMP dehydrogenase is inhibited by GMP
IIL 7'Ae PRPP synthase reaction. The rate of PRPP synthesis depends on the
availability of ribose 5-phosphate and on the activity of PRPP' synthase, an
enzyme sensitive to feedback inhibition by AMP, ADP, GMP, and GDP.
Purines degradation
Purine nucleotides arc degraded by a pathway in which they lose their phosphate
through the action of5'-nucleotidase. Adenylate yields adenosine, which is deaminated
to inosine by adenosine deaminase. and inosine is hydrolyzed to hypoxanthine (its
purine base) and ribose. Hypoxanthine is oxidized successively to xanthine and then
8.8. Special Products Derived from Amino Acids 475
_____________ AMP
Atiteflylaie deaminarse
Nucleotidases
NH<+
Xanthine Urate
The end product of purine catabolism in humans is uric acid. Both undissocialcd
uric acid and the monosodium salt (primary form in the blood) arc only sparingly
soluble and present in biologic fluids at the limit of its solubility: concentration of
urates in blood plasma is 0.1-0.3 mM/L; excretion with urine is 0.3-1.29 g/24h. A
very high concentration of urate in the blood (Ajpmrwemw) leads to a fairly com mon
group of diseases referred to as gout. Crystallization of urates in soft tissues and joins
forms deposits called tophi (tophus), which causes acute and chronic gouty arthritis
(Fig. 8.53). The joints become inflamed, painful and arthritic owing to the abnormal
deposition of sodium urate crystals. Gout occurs predominantly in males.
Gout is effectively treated by a combination of nutritional and drug therapies. Food
especially rich in nucleotides and nucleic acids such as liver or glandular products,
meal and fish, is withheld from the diet. Major alleviation of the symptoms is provided
by the drug alfopurinoL which inhibits xanthine oxidase (ewwpr/ftfve m/tibi/ion). Wien
xanthine oxidase is inhibited the excreted products of purine metabolism are xanthine
476 Chapter 8. Nitrogen metabolism
and hypoxanthine, which are more water-soluble than uric acid and less likely to form
crystalline deposits.
The limited solubility is not ordinarily a problem in urine unless the urine is
very acid or has high |Ca3'|. Urate salts coprccipitatc with calcium salts and can
form stones in kidneys: nephrolithiasis, which maybe reduced by alkalinization of
urine.
Xanthine oxidase deficiency due either to a genetic defect or to severe liver damage
in severe cases may exhibit xanthinuria and xanthine lithiasis.
.Adenosine deaminase deficiency leads to an accumulation of toxic purine
degradation by products, most potently affecting lymphocytes. Whilst most notable
effects arc on lymphocytes, other manifestations include skeletal abnormalities,
ncurodcvclopmcntal effects and pulmonary manifestations. Affected patients
present in early infancy, usually with persistent infection, or with pulmonary
insufficiency.
Purine nucleoside phosphorylase deficiency is one of several disorders that damage
the immune system and cause severe combined immunodeficiency (SCID). People with
SCID lack virtually all immune protection from foreign invaders such as bacteria,
viruses, and fungi. Affected individuals arc prone to repeated and persistent infections
that can be very serious or life-threatening. These infections arc often caused by
^opportunistic* organisms that ordinarily do not cause illness in people with a normal
immune system. Infants with SCID typically grow much more slowly than healthy
children and experience pneumonia, chronic diarrhea, and widespread skin rashes.
Without successful treatment to restore immune function, children with SCI D usually
do not survive past early childhood.
8.8. Special Products Derived from Amino Acids 477
Catabolism of pyrimidines
Human cells degrade pyrimidine nucleotides to their component bases. These
reactions, like those of purine nucleotides, occur through dcphosphorytaiion,
deamination, and gtycosidic bond cleavages. CMP, UMP and dTMP arc convened
into cytosine, uracil and thymine, ribose-1.-phosphate and deoxyribose-1-phosphate.
The Pyrimidine catabolism pathway generally lead to NH' production and: thus to urea
synthesis (Fig. 8.54).
Fig. 8.54. Pyrimidiiie nitrogenous liases degradation. Cytosine is deaminated into uracil: reaction
is catalyzed Dy cytidine deaminase, uracil and thymine are converted into dinydrouracii and
dihydromymine Dy dinydrouracii dehydrogenase: one NADPH -t- H* is oxidized into NADP*.
Dinydrouracii and dihydromymine are converted into p-atanine and fkamino isobutyrate i cyclized
molecule is converted into linear Dy cleaving the covalent Doud at particular place}
Nucleosides and nucleotides are widely used in clinical medicine and science
These derivatives:
* inhibit a particular enzyme of nucleotide or nucleic acid synthesis;
* arc synthetic analogous and inserted into DNA or KN A synthesis damaging the
complementation between, nitrogenous bases and polymerization of nucleic acid
chains.
The fluoropyrimidinc - 5-fluorouracil (5-FU) is an anti metabolite drug that is
widely used for the treatment of cancer, particularly for colorectal cancer. 5-FU exerts
its an ticancere fleets through inhibition of thymidylate synthase (TS)and incorporation
of its metabolites into RNA and DNA (Fig. 8.55: Fig. 8.56).
Synthesis
dUMP---------------------------- * dTMP
Thymidylaie synlfiase
Acyclovir is widely used in the treatment ofherpes virus infections, particularly herpes
simplex virus (H SV) and varied la-zoster virus (VZV). Acyclovir (9-[2-hydroxymethyl |
guanine) triphosphate) competitively inhibits viral DNA polymerase by acting as
an analog to deoxyguanos inc triphosphate (dGTP). Incorporation of acyclovir
triphosphate into DNA results in chain termination since the absence ofa 3’ hydroxyl
group prevents the attachment of additional nucleosides.
Azido th vmidine (AZT) is an antiretroviral medication used to prevent and treat
H1V/AIDS. AZT is a thymidine analog. AZT works by selectively inhibiting HIV’s
reverse transcriptase (Fig. 8.57), the enzyme that the virus uses to make a DNA copy
of its RNA. Reverse transcription is necessary for the production, of HIV's double
stranded DNA, which would be subsequently integrated into the genetic material of
the infected cell (where it is called a provirus). Cellular enzymes convert AZT into the
effective 5’-triphosphate form. The termination of HIV's forming DNA chains is the
specific factor in the inhibitory effect.
The quantitative characteristics of the nwtaboGsm of amino acids learnt in this section
Blood uric acid concentration: 0.15-0.46 mMd/L
Uric acid excretion: 40D-600 mg/dl. (0.3—1.29 g/24h)
Review tests
I. The origin of C and N atoms in purine base:
480 Chapter 8. Nitrogen metabolism
dUMP
dTMP
Enzyme:
A. Thymidylate synthase.
B. Thymidine kinase.
C. Serine hydroxymethyl transferase.
D. Di hydro folate reductase.
E. Ribonucleotide reductase.
Situational problems
I. The girl visited a. doctor with complaints of erythema, edema and itching which
was appeared on open parts of skin after the rest on the beach. Doctor found
out that the patient had taken the drug including sulfonamide not tong ago.
5-aminolevulinatc and porphobilinogen were found in blood, the urine was
8.8. Special Products Derived from Amino Acids 481
dyed to red. What could cause photo dermatosis in patient and what disorder
this girl sutlers from'? For the answer:
a) write down the first reactions and the following scheme of metabolic pathway
which intermediate products were found in the blood:
b) whaL enzyme synthesis was induced by sulfonamides, point out the mechanism
of its regulation;
c) explain the molecular mechanisms of occurred symptoms.
2. Two newborns with jaundice were prescribed to phototherapy. One newborn
showed the improvment in his state and symptoms of jaundice had been
disappeared. But phototherapy was useless lor the second baby and he was
prescribed phenobarbital. Unfortunately, this therapy was also inefficient,
moreover, encephalopathy symptoms became revealed. How to explain doctor
recommendations? What can cause jaundice in the second newborn? To answer
the question:
a) explain the cause of ^physio logical* jaundice in newborns:
b) specify the changes in contents of bilirubin in blood, stercobilin and urobilin
respectively in feces and urine in newborns:
c) explain the mechanism of phototherapy and phenobarbital: curative effect.
Write down the reaction which rate is increased by phenobarbital;
d) explain the results of the treatment.
3. A patient with high temperature 38.5 and clinically apparent icteric skin
and mucous tunics was delivered to the admitting office of hospital. Blood
concentration of both direct and indirect bilirubin was increased. Direct
bilirubin is found in urine, but urine urobilin and feces stercobilin were reduced.
What type ofjaundice patient suffered from? To answer the question:
a) present the scheme of indirect bilirubin formation;
b} write down the reaction of bilirubin conjugation:
c) point out the properties of direct and indirect bilirubin, explain the reasons
for indirect (Linconjugaicd) bilirubin toxicity;
d) specify enzymes, the activity ofwhich is of particular importance for diagnosis
for the liver damage. Describe the main principles of enzymod iagnostics.
4. A 67-ycar-old male attended the clinic with a painful 1-st metatarsophalangeal
joint, which had progressively worsened in pain, mobility and deformity. In
examination it was revealed redness, edema and increased local temperature.
Patient diet involves much amount of meat. Biochemical blood analysis revealed
uric acid concentration 0.51 mMol/L (normal for male 0.17-0.42 mMol/L).
What is the expected diagnosis in this case? For the answer:
a) write down the scheme of uric acid formation;
b) specify the mechanisms of presented symptoms development;
c) represent your diet recommendations to the patient;
d) propose the remedy prescribed by the doctor in case of this disorder.
5. Goul can be caused by supcractivalion of PRPP synthase, or partial deficiency
of hypoxanthine-guanine phospho ribosyl transferase. Why the change in these
enzyme activities can induce the development of this disorder? For the answer:
a) write down the scheme of the reactions catalyzed by these enzymes;
482 Chapter 8. Nitrogen metabolism
External internal
©
Environmental I Centra nervous system
signals
-X^ .___
Pituitary
Tropic hormones
i
Endocrine Hands
Metabolism of carbohydrate, lipid, amino acid insulin, glucagon, epinephrine, cortisol, somatotropin
Synthesis and secretion ol endocrine gland pituitary tropic hormones, liberins. and hypothalamic
hormones statins
Ligand-binding
domain
Extracellular side
Cell membrane
Cytoplasm
Cytoplasmic
domains (I to VII) domain
I MI J Mineralocorticoid
[ — I"" Progestins
Estrogens
ran j Vitamin D
Thyroid hormone
Fig. 9.3. Ligand bin ns to noma in - C-terminal region (on the right). DMA binds to central nomain.
Proteins associated wim tne promoter region binds to variable domain (on tne left)
the variable region of the receptor, is near the N-tcrminusof the polypeptide chain, it
binds to other proteins, that arc complexed with their D NA-binding sequences in the
regulatory region, of the gene controlling transcription.
The concentration of receptors within the cell or on its surface and their affinity to
this hormone arc normally regulated in various ways, and can also change in diseases or
when using hormones or their agonists as medicines. For example, when p-adrcnergic
agonists arc exposed to cells for several minutes in response lo a new addition of the
agonist, activation of adcnytvl cyclase slops and the biological response disappears.
Such, a decrease in the sensitivity of the receptor to the hormone (desens itizalion)
may occur as a result of a change in the number of receptors by a down-regulation
mechanism. The hormone binds to the receptor, the hormone-receptor complex
enters the cell through endocytosis (internalizes), where some receptors undergo
proteolytic cleavage under the action of lysosomal enzymes, and some arc inactivated,
separating from other membrane components. This leads to a decrease in the number of
receptors on the plasma membrane. For example, in the case of insulin, glucagon and
catecholamines, this can happen within a few minutes or hours. When the hormone
concentration decreases, the receptors return lo the cell surface, and the sensitivity
to the hormone is restored. Receptor activity, j.e.. its affinity for the hormone, may
also change as a result of the covalent modification, mainly by phosphorylation. The
concentration of intracellular receptors can also be regulated by the mechanism of
induction and repression.
According to the location of receptors and the nature of the signal used to mediate
hormonal action within the cell (mechanism of signal transduction). hormones can
be. divided into 2 groups (Fig. 9.4). The hormones of the first group arc hydrophilic
and cannot diffuse throughout lipid bi layer of cell membrane, therefore, they interact,
with plasma membrane receptors (peptide hormones, epinephrine, eicosanoids and
cytokines). The hormones of the second group are hydrophobic, they penetrate in
target cellsand interact with intracellular receptors.
To deliver a hormone message to the cell the hormone (primary messenger) binds
to a receptor and causes a change in the receptor conformation. This change initiates
a sequence of biochemical events that include the conjugation of some molecules with
others (signal transduction). Thus, a signal is generated that regulates the cellular
response by changing activity or amount of enzymes and other proteins (Fig. 9.5).
Depending on. the mechanism of hormonal signal transduction to the cells, the rale of
metabolic reactions varies:
► as a result of changes in the activity of enzymes;
► as a result, ofchanges in. the amount of enzymes.
The synthesis of hormones can occur cither in specific cells of specialized
endocrine glands, or in cells that perform additional functions. Many protein
hormones, such as growth hormone (GH). PTH, prolactin, insulin and glucagon,
arc produced in specialized cells using standard protein synthesis mechanisms
common to all cells. These secretory cells contain secretory granules to store large
amounts of the hormone and release the hormone in response to certain signals.
9.1. Role of Hormones in Regulation of Metabolism and Functions 489
Fig. 9.4.Two general mechanisms of hormone action The peptide and amine hormones act throughout
surface receptors and steroid and thyroid normones act throughout intracellular receptors
Fig. 9.5. The main stages of the hortooitai signal transduction to target cells
ladle 9.4. Biological 1uisclions of principal hormones of tde anterior pituitary gland
Thyra id-stim ulati ng ho rmo ne (TS H), o r Thy retro pi n Stimu I at e s synthesis and se cretio n of thyro id
or thyrotropic hormone) hormones
Prolactin (PR L. luteotrop ic hormon e o r Iuteotropin j Stimu lates m ilk secretio n an d mammary g rawth
Prolactin is synthesized in lac tot rop hie cells of the anterior pituitary as a
prohormonc. The number of J actol rop hie cells increases sharply during pregnancy
under estrogens action. Prolactin receptors arc present in many tissues: in the liver,
kidneys, adrenal glands, testes, ovaries, uterus, and other tissues. The prolactin main
physiological function is the stimulation of lactation. Prolactin induces the synthesis
of a-lac tai bum in and casein, activates the synthesis of phospholipids and TAG.
Prolactin, affects growth processes to a much lesser extent than growth hormone. In
men, prolactin increases the sensitivity of Leydig cells to luteinizing hormone, thus
maintaining the necessary level of testosterone synthesis; in the kidneys, prolactin
reduces the water excretion, affects the reabsorption of Na* and K ions; prolactin also
enhances humoral and cellular immunity. The synthesis and secretion of prolactin
is stimulated by thyrotropin-releasing hormone; serotonin, oxytocin, acetylcholine,
and dopamine have an inhibitory effect. Like most hormones, prolactin is secreted
into the blood occasionally al intervals of 30-90 minutes. The maximum secretion is
observed 6-8 hours a her a falling asleep. I n women the blood prolactin concentration
is 8—10 ng/ml, and in men that is 5-8 ng/ml. The half-life of prolactin in scrum is
15—20 minutes. The placenta produces a hormone (placental lactogen), homologous
in amino acid composition to growth hormone and prolactin. All 3 hormones have
common antigenic determinants and have growth-stimulating and lactogenic activity.
There is a hypothesis that the genes of these hormones arose as a result of the
duplication of a single precursor gene.
494 Chapter 9. Molecular eoaocrmoiogy
enlarging the width and thickness of bones, and the growth of other tissues, including
connective tissue, muscles and internal organs; enhances the total amount of R.NA,
DNA and the total number of celts. On the whole, the overall effect of G H on protein
turnover is an anabolic one.
In. adipose tissue, GH increases lipolysis leading to increased circulating free fatly
acids. In muscles, these fatty acids are preferentially served as fuels, and glucose uptake
is indirectly suppressed! with glycolytic pathway decreased. In liver, the GH stimulates
production and release of ICF-I. accelerates hepatic gluconeogenesis, while it has
modest effects on. liver glucose uptake, utilization, or storage. Il also enhances the
oxidation of fatty acids in the liver and consequently stimulates ketogenesis with
increased blood ketone body level in fasting (when insulin level is tow).
Thus, GH exerts diverse effects on tissues. Many of the physiological effects of
GH arc still unknown. Now targets ofGHR signaling arc steadily emerging, and the
metabolic actions of GH may not be as clear as was initially believed. Understanding
the role of GH in physiological and pathological states could contribute to the
development of new therapeutic strategics.
Thyrotropin, LH, and FSH arc glycoproteins. Thyrotropin (TSH) is synthesized
in thyrotrophic cells of the anterior pituitary' gland. The thyrotropin secretion is
stimulated by thyrotropin-releasing hormone, and is mainly inhibited by the thyroid
hormones level increase. The peak TSH secretion is observed in the hours immediately
preceding sleep, followed by a decrease during the night.
The main biological function of thyrotropin is to stimulate the synthesis and
secretion of the thyroid hormones, thyroxine (T4), and Tr from the epithelial cells
making up the follicles of the thyroid gland. Thyrotropin binding with plasma
membrane receptors activates adenylyl cyclase. Thyrotropin has two types of effects
on the thyroid gland. First effects manifest rapidly (within a few minutes) and i nclude
stimulation of the thyroid hormone's synthesis and secretion. The second group of
effects that has several days exhibited includes stimulation of the synthesis of proteins,
phospholipids and nucleic acids and an. increase in the size and number of thyroid
cells. A numberofimmunoglobulinsG, interacting with thyrotropin receptors, mimic
the effects of the hormone. Similar immunoglobulins arc found in most patients with
hyperthyroidism.
The group of hormones related to glycoproteins also includes the gonadotropic
hormones of the pituitary gland of LH and FSH and the placental chorionic
gonadotropin (hCG). FSH and LH play the important biological roles in the
reproductive system in mates and females. Also, hCG, which is initially produced by
the human trophoblast upon implantation in the uterus, mimics the actions of LH and
is essential for maintenance of pregnancy in its early stages.
Frequently several active hormones can be carved out of the same prohormonc.
Macromolecule POMC (proopiomelanocortin) is an example of multiple hormones
encoded by a single gene. The POMC gene encodes a large polypeptide that is
progressively cleaved into at least nine biologically active pepLidcs (Fig. 9.6). POMC
is synthesized in the anterior and intermediate lobes of the pituitary and in some
other tissues (intestines, placenta). The polypeptide chain consists of 265 amino acid
496 Chapter 9. Molecular endocrmoiogy
residues. The processing of this large precursor molecule in different cells proceed
differently with the formation of a different set of peptides. After removal of the
N-terminal signal peptide, the polypeptide chain is split into two fragments: ACTH
(39 a.L)and [3-lipotropin (42-134a.k.). Additional proteolysis leads to the formation
ofa-and p-MSH (melanocyte-stimulating hormone), endorphins, corticotropin-like
hormone intermediate lobe peptide (CLIP} and y-lipotropin. P-MSH and CLIP of
the intermediate fraction in humans arc practically not formed, since in. adults the
intermediate fraction is not developed. B-lipotropin, y-lipotropin and p-endorphin
were found in the human pituitary gland. The function of all products of the POMC
proteolysis is not sufficiently studied.
Fig. 9.6. Processing of proopiomelanocortin (POMC). The protein POMC is produced Dy the anterior
pituitary in response to corticotropin releasing hormone (CRH). The N-terminai region is removed,
and the remainder is cleaved to form adrenocorticotropic hormone (ACTH) and 0-lipotropin (B-LPH).
Additional cleavages produce the melanocyte stimulating hormones (a- and P-MSH), endorphins,
corticotropin-like intermediate lobe peptide (CLIP), and y-fipotropin fr-LPH)
In the human, the posterior pituitary gland stores and secretes two important
nonapepiides, oxytocin and a nt id in retie hormone, into the circulation. These hormones
arc synthesized in hypothalamic neuronal cell bodies, together with a ncurophysin
iliat accompanies the hormone from the cell body through the axon to the nerve
ending awaiting an appropriate signal to release the complex into the bloodstream.
Oxytocin stimulates smooth muscle contraction in the uterus and bieast; it functions
in parturition (childbirth) and lactation. Antidiuretic hormone (ADH), also known as
vasopressin (VP), controls water balance (this hormone is covered in Ch. 9.9}.
Fig. 9X Structure of insulin. Mature insulin is farmed from pre proinsulin Dy proteolytic processing.
Removal of tne signal sequence ano formation of three disulfide Dorias produces proinsulin. Removal
of the C peptide from prornsulin produces mature insulin, composed of A and B chains
498 Chapter 9. Molecular enaocrinoiogy
Fig. 9.8. insulin Biosynthesis scheme in p-cells of Langerhans islets. ER — endoplasmic reticulum.
1 — formation of the signal peptide; 2 — synthesis of preproinsulin; 3 — cleavage of me signal
peptide; 4 — transport of proinsulin lo Ihe Golgi apparatus; 5 — the conversion of proinsulin to
insulin and C-peptide and tne insulin and C-peptioe incorporation in secretary granules; 6 — insulin
and C-peptide secretion
Proinsulin (86 amino acid residues) enters the (iolei apparatus, where it is
cleaved by specific proteases al several silos lo form insulin (51 amino acid residues)
and C-peptide consisting of 31 amino acid residues. Equimolar amounts of insulin
and C-peptide arc incorporated in secretory granules of the pancreatic beta cells. In
granules, insulin combines with zinc to form d imers and hexamers. The kidney is the
principal site of C-peptidc degradation. Because little degradation of the C-pc pt ide
occurs in the liver, its concentration in blood is useful for estimating the rate of
insulin secretion and evaluation of beta-cell function.
The secretion of insulin is primarily regulated by blood glucose concentration.
The GLLFT-2 proteins on the membrane of pancreatic p-cclls have a relatively high.
Km for glucose (- 20 mM) and, therefore, provide an active glucose transport from
the blood into the p-cctls of the pancreas. Entering the pancreatic cells, glucose is
phosphorylated and further oxidi zed. This causes an increase in the ATP/ADP ratio in
the cell, which initiates closure of the ATP-dependent K1 channels. This depolarization
causes voltage-gated Ca2' channels to open, initiating docking and secretion of insulin
from secretory vesicles. After secretion into the blood, insulin oligomers disintegrate.
9.3. Structure, Synthesis and Basic Action of Principal Hormones Regulating Fuel Metabolism 499
The half-life of insulin in blood plasma is 3 -10 minutes, C-peptide is about 30 minutes.
Insulin is inactivated by the enzyme insulinase, mainly in the liver and to a lesser extent
in the kidneys. The main stimulus of insulin synthesis and secretion is glucose. Insulin
secretion is also enhanced by sonic amino acids (especially arginine and lysine),
ketone bodies and fatty acids. Epinephrine, somatostatin, and some gastrointestinal
peptides inhibit insulin secretion.
Insulin is the main anabolic hormone. It participates in the regulation of
metabolism, transport of glucose, amino acids and ions, and also protein synthesis.
Insulin affects the processes of replication and transcription, thus participating
in the regulation of cell differentiation, proliferation and cell transformation. The
participation of insulin in the regulation of metabolism is discussed in. the relevant
units (see also Ch. 4.3 and Pig. 4.12). The effect of insulin on key metabolic enzymes
is presented in Tabic. 9.5.
Activation
Pyruvate dehydrogenase
Phosphorylase kinase
Acetyt-CoA carboxylase
HMG-CoA-reductase
Induction
Citrate lyase
Malic enzyme
Pyruvate kinase
AcelyFCoA carboxylase
Repression
Fructose bisphosphatase
H2O2
Colloid OH
Ta I*
Fig. 9.9, Synthesis of the tftyroid hormones (I3 and IJ. Synthesis of tn e protein thyroglobulin (Tgb) in
thyroid follicular cells. Active transport of iodide by the sodrum/iodide symporter into the thyroid gland
follicular ceils. Oxidation of iodide by peroxidase, s 0 di nation cf Tyrosyl residues within and formation of
monoiodotyrosyl (MiT) and diiodotyrosyl (DlTj within the protein thyroglobulin. Transfer and coupling
of iodotyrosines within thyroglobulin to form thyroxine (Tj and triiodothyronine {Tj. Storage of
thyroglobulin as the colloid in the lumen of The thyroid follicle. Endocytosis of the colloid back into
the thyroid epithelial cell. Proteolysis of thyroglobulin with concomitant release of T+ and I3 as well
as free and io dothyronines. Secretion of T# and T3 into the blood. De iodination of iodotyrosines within
the thyroid follicular cells for reutil izati on of the liberated iodine. R£R — rough endoplasmic reticulum
Hypothalamus
Pituitary
Thyroid
Fig. 9.10. Feedback regulation of tnyroid hormone secretion. TRH stimulates (+) the release of TSH,
wnicn stimulates {+) tne release of T3 and Tr T4 is converted to T, in the liver and other cells. T3 and
T( inhibit (-) the release of TSH and of TRH
► mineralocorticoids (aldosterone);
► glucocorticoids (cortisol);
► eaten riol (active form of vitamin Ds).
In (he adrenal cortex that is anatomically divided into three zones the three
di fie rem major classes of the steroid hormones, or corticosteroids, arc produced:
mineralocorticoids arc formed in zona glomcrulosa: glucocorticoids are synthesized
in the zona fasciculate; and androgens arc formed in zona reticularis. Glucoconicoids,
C,, steroids, play an important rote in adaptation to stress. They have a variety of
effects, and the most important is the stimulation of gluconeogenesis. The main
human glucocorticoid is cortisol. Mineralocorticoids, steroids, are required to
maintain Na* and KJ levels in living cells. The most active this group hormone is
aldosterone. Androgens arc C|9 steroids. The adrenal cortex generates androgen
precursors, the most active is dehydroepiandrosterone (IJHEA) and the weakest
is androstenedione. The more potent androgen testosterone is synthesized in the
adrenal glands in a small amount. The adrenal androgens are converted into more
active androgens outside the adrenal gland, in the adrenal glands, trace amounts
of testosterone can be converted to estradiol. Normally, the adrenal production of
Cl9steroids playsan insignificant role.
A com mon precursor ofal I corticosteroids is cholesterol. The sources of cholesterol
for the corticosteroids synthesis arc cholesterol esters that enter the cell as part of LDL
or deposited in. the cell. The release of cholesterol from its esters and the synthesis of
corticosteroids arc stimulated by corticotropin. More than 40 metabolites with different
structure and biological activity arc formed in corticosteroid pathway. The final
product depends on the set of enzymes in the cell and the sequence of hydroxylation
reactions (Fig. 9.11). The main corticosteroids with pronounced hormonal activity arc
cortisol, aldosterone and androgens.
At the first stage ofcorticostc.ro id synthesis cholesterol is converted to pregnenolone
by cleaving the 6-carbon fragment from the cholesterol side-chain and oxidizing the
carbon atom Cjn. Then pregnenolone can be converted both io progesterone (C3I.)T
the precursor of cortisol and aldosterone, and to CN steroids, androgen precursors.
The primary hydroxylation of progesterone by 17-hydroxylase, and then 21- and
11-hydroxylase leads to the synthesis of cortisol. Reactions of aldosterone formation
include hydroxylation of progesterone, first with 21-hydroxylase, and then with
11-hydroxylase. The final steroid product is dependent on the set of enzymes in the
cell and the sequence of hydroxylation reactions.
Cortisol synthesis reactions occur in different compartments of the adrenal cortex
cells (Fig. 9.12).
The cortisol synthesis begins with the conversion of pregnenolone to progesterone.
This reaction proceeds in the cytosol of cells, where pregnenolone is transported from
mitochondria. The reaction is catalyzed by 3-(]-hydroxy steroid dehydrogenase. In
the membranes of endoplasmic reticulum 17-a-hydroxylase converts progesterone to
17-hydroxy-progesterone. The same enzyme catalyzes the conversion of pregnenolone
to 17-hydroxyprcgncnotone. A 17,20-lyases cleaves the two-carbon side chain
from 17-hydroxyprcgnenolone to form the -steroid, dehydroepiandrosterone.
9.3. Structure, Synthesis and Basic Action of Principal Hormones Regulating Fuel Metabolism 505
HO
Fig. 9.12. Steps of cortisol synthesis in ceiL The release of cholesterol from its esters and the synthesis
of corticosteroids are stimufatea by corticotropnin. The reactions of cortisol synthesis occur in
different cell compartments of me adrenal cortex
Fig. 913. Regulation of cortisol secretion. Various factors act on hypotnaiamusto stimulate tne release
of CRH. CHR stimulates (+) tne release of ACTH from me anterior pituitary, vriiich stimulates (+) tne
release of cortisol from adrenal cortex. Cortisol inhibits (-) tne release of CRH and ACTH
due to oxidation of fatty acids, as well as ketone bodies formed in the liver from fatty
acids.
Liver, adipose tissue and muscles are the main organs that provide changes
in metabolism in accordance with the rhythm of nutrition. Many hormones affect
fuel metabolism including those that influence absorption, transport, and oxidation
of fuel molecules. Insulin is the major anabolic hormone: glucagon is the major
counterregulatory hormone. Epinephrine, cortisol, thyroid hormones, and growih-
hornione also have contrain.su la r activity. Insulin and contra instil ar hormones provide
a balance between the needs and capabilities of the body to obtain energy necessary for
normal functioning and growth.
Muscle activity slows down the storage processes during digestion, because a
part of the products of digestion coming from the intestine is directly consumed in
muscles. In the postabsorptivc stale, muscular activity stimulates the mobilization of
nutrient storage, mainly fats. Epinephrine plays an important role in the regulation
of metabolic changes in the body associated with the muscle work-to-rcst transition.
Metabolic changes of the major fuel molecules in the absorptive state arc mainly due
to the high insulin-glucagon index (Tig. 9.14).
cell
Fig. 9.14. Major metabolic pathways in absorptive state: 1 — glycogen biosynthesis in the liver; 2 —
glycolysis; 3 — TAG biosynthesis in me liver; 4 —TAG biosynthesis in adipose tissue; 5— muscle
glycogen biosynthesis; 6 — protein biosynthesis in various tissues, including the liver: FA — Fatty
acids
9.4. Hormonal Regulation of Fuel MetaDolism at Normal Nutrition Rnytnm 511
Blood Liver
I Glucose
Cl .i I Insulin
f Glucagon
X Fattv .acidsAcetvI-CoA
5J
Ketone bodies
I ©
Ketone bodies
®I
TAG-----------
Adipose
cell
Urina
Muscles
In the first few hours after a meal, the blood glucose level is diminished slightly,
and tissues receive glucose released from liver glycogen. Liver glycogen mobilization
is accelerated by glucagon. Glycogen stores in the liver arc depleted for the 24-
hour fast. As glycogen stores arc exhausted, the main source of glucose becomes
gluconeogenesis, which begins to accelerate 4-6 hours after the last meal. Substrates
for the glucose synthesis arc lactate, glycerol and amino acids. The rate of fatty
acid biosynthesis decreases due to phosphorylation and inactivation of acetyl-CoA
carboxylase, and the rate offJ-oxidation increases. In adipose tissue, the rate of TAG
synthesis decreases and lipolysis is stimulated. Stimulation of lipolysis is the result
of the activation of the hormone-sensitive TAG-lipase by glucagon. As lipolysis
increases during postabsorptivc state, fatty acids are released from adipose tissue and
travel to various organs. In parts these fatty acids arc taken by the liver for C-oxi dal ion.
9.4. Hormonal Regulation of Fuel MetaDolism at Normal Nutrition Rnytnm 513
Acetyl-Co A. produced in the liver, is convened to ketone bodies. Fatty acids become
important sources of energy in the Liver, musclesand adipose tissue. Thus, in the post
absorption state, the blood glucose concentration is maintained at 60-100 mg/dL
(3.5-5.5 mmol/Lh and the level of fatty acidsand ketone bodies increases.
Review Tests
Match the figure and the letter.
1. Stages of the insulin biosynthesis and maturation. Com pern on ts:
A. N-terminal amino acid.
B. N-signal peptide.
C. Preproinsulin.
D. Proinsulin.
E. C-pep tide.
Pituitary
Thyroid
9.4. Hormonal Regulation of Fuel MetaDolism at Normal Nutrition Rnytnm 515
Situational Problems
I. A student, halfan hour after the dinner, containing about 150 g of carbohydrates,
20 g oflat, and 40 g of protein, is silling in a chair and reading. What metabolic
changes occur in this state? For answer:
a) name the hormone level in blood that is elevated during resting:
describe the steps of its synthesis, name the secretion stimuli and target tissues;
b) present a diagram of the hormonal signal transduction to target cells:
c) draw diagrams reflecting the student's metabolic changes of glucose and fats
in adipocytes:
d) enumerate the regulatory enzymes of fatty acid synthesis from glucose that
have their activity and amount increased under the in fluence ofthe mentioned
hormone.
516 Chapter 9. Molecular eoaocrmoiogy
c) draw a diagram of the fatty acid metabolic pathway that has its rate increased
under these conditions;
d) describe the process regulation mentioned in c), write the regulatory reaction
of this process, name the key enzyme, its activatorsand inhibitors;
e) name all metabolic pathways of glucose that provide the precursors formation
for the fatty acids synthesis, and list all needed components formed from
glucose.
6. With the presence of fast-food chains, many people have the opportunity to
«havc a quick bite* while their daily physical activity is decreased. Explain why
such a diet leads to obesity more quickly under hypodynamic conditions. For
answer:
a) draw the chans of metabolic pathways in adipocytes with their activation
leading to increased weight:
b) give the origin of substrates, enzymes, ways of using final products:
cidraw an appropriate chart and describe the stages of sequential hormonal
signal transduction to adipocytes and explain the biological effects of the
hormone.
7. After visiting a gym, a student had 250-300 g of carbohydrates for lunch and
decided to rest. How will the student’s metabolism of carbohydrates and fats in
the adipose tissue change 2 hours after meal?
a) draw a graph displaying the student’s blood glucose concentration over time
during the absorptive state; explain the curve;
b) name the hormone that has its level elevated in the student’s blood in the
absorptive state and describe the hormonal signal transduction to adipose
tissue;
c) list all metabolic pathways of glucose utilization and catabolism products in
adipose tissue and explain their physiological significance;
d) draw’ a chart of the fat metabolic pathway in adipoesies and explain how the
hormone stimulates the process.
S. A patient suffering from infectious polyarthritis, has been receiving prednisone
for a long time as treatment. Prednisone is a structural analogue of cortisol. As
the patient fell better, he deliberately stopped taking this d rug. Soon the patient’s
state look a dramatical turn for the worse. Screening showed a decrease in the
patient's blood glucose concentration, the blood pressure dropped, and the
content of 17-kctostcroids in the urine was diminished. Why did the patient’s
state worsen after he quitted this drug? For the answer:
a) draw a flowchart of hormone synthesis and secretion for the hormone with
inhibited production caused by the prolonged administration of prednisone:
b) predict an improvement in the patient’s state if he is given corticotropin;
c) name the reasons for the blood glucose concentration, and 17-kctostcroids
decrease: and blood pressure decline; write the metabolic pathway with its
decreased rate in the liver when prednisone administration was stopped;
d) substantiate the prednisone use to treat arthritis.
9. A 46-year-old woman visited a doctor with complaints of sweating, rapid
heartbeat, general weakness and fatigue. The patient also told the doctor that
3-4 hours after eating, she has been experiencing acute bouts of hunger, with
clouded consciousness, speech disorders, spatial and temporal disorientation.
518 Chapter 9. Molecular eoaocrmoiogy
Moreover her body weight. has been constantly increased for the past few
months. Screening showed the blood glucose level was 2.8 mol/L, the insulin/
glucagon ratio is 0.7 (norm >0.4), the C-peptide level was elevated. What allows
to suspect insulinoma (endogenous hyperinsulinism) in this woman? Please,
answer the following questions:
a) what hormone is high secreted in this condition?
b} what is the possible cause of this disease?
c) why do people with such condition gain weight? Draw a diagram of the
metabolic pathway reflecting the cause of excess weight.
d) what is the role of this hormone in the regulation of this process?
e) what treatment is needed lor this disease?
10. Symptoms of insulinoma, a hormone-producing tumor of the 0-cclls of the
islets of Langerhans of the gastric gland, manifest themselves in increased
blood insulin levels, bouts of hypoglycemia, accompanied by general weakness,
fatigue, sweating, tachycardia, loss of consciousness, and acute hunger. Explain
the causes of the disease symptoms. For that:
a) name the normal blood glucose concentration:
b) draw a chart of an insulin receptor and describe the steps of insulin signal
transduction to the target cells;
c) recount the features of glucose transport into cells of different tissues, name
the insulin-dependent tissues;
d) explain bouts of hypoglycemia in insulinoma, and draw the metabolic
pathways of glucose metabolism with increased speed in the liver that arc
associated with this condition.
11. A 45-year-old man complained of rapid weight loss, tachycardia, increased
sweating, occasional elevated blood pressure and increased excitability.
Additional screening revealed a tumor in the medulla of one of the adrenal
glands. The patient was diagnosed with pheochromocytoma. What molecular
mechanisms caused the development of the condition showing the sc symptoms?
For answer:
a) draw a chan of the catecholamines synthesis in adrenal glands;
b) name the hormone that has its synthesis increased in. this disease, list its
target organs, stimuli of synthesis and secretion, target tissues and receptors;
c) draw a chart of signal transduction of this hormone to adipocytes and the
metabolic pathway of fat with i ncreased rate in. the ^patient’s adipocytes;
c) list the physiological effects of this hormone and explain the causes of the
disease symptoms.
and the insuJin/glucagon ratio is reduced. Cortisol secretion follows its basal circadian
rhythm and it's level remains almost unchanged until prolonged hypoglycemia results
in increased blood cortisol concentration in the late stages of fasti ng. These hormonal
signals designate the predominance of triacylglyccrols mobilization and protein
degradation and exhaustion of glycogen, stores. Fatty acids now become the primary
fuel for muscle and liver.
In this state, contra insular hormones regulate the flux ofsubstrates be tween the liver,
adipose tissue, musclesand the brain to maintain the blood glucose: concentration due
to gluconeogenesis to ensure glucose-dependent tissues brain, red blood cells): and to
mobilize other fuel molecules, primarily fats, to provide energy to all other tissues. The
manifestation of these changes allows us to conditionally distinguish three phases of
starvation. Due to the switch of metabolism to fuel mobilization, even after 5-6 weeks
of fasting, the blood glucose glucose levels arc still in the range of 65 mg/dl. The main
changes during lasting occur in the liver, adipose tissue and muscles {Fig. 9.I.6).
Blood Liver
tGhjeose Glycogen
I Insulin
ffGhJcagon
[Cortisod Glucose
Glycerol
Brain
Lactale
Ketone bodies
.Amino acids
Adipose
cell
Kidney
Muscles
Fig. 9J6. Metabolic Changes of me main energy sources during fasting: 1 — decrease in me insulin/
glucagon ratio; 2 — glycogen mobilization; 3,4 — glucose transport to the Drain and red blood cells;
5 — TAG mobilization; 6 — FA transport io the muscles; 7 — synthesis of ketone bodies; 8 — FA
transport to the liver; 9—Amino acid transport to the liver; 10 — gluconeogenesis from amino acid;
11 — lactate transport to the liver; 12—glycerol transport to me liver. Dotted lines indicate processes
mat are slowing
520 Chapter 9. Molecular enaocrinoiogy
Phases of fasting. Fasting may be short-term — during the day (first phase) T
continue for a week (second phase) or several weeks (third phase). During fasting, a
number of changes in fuel metabolism occur.
During the first phase, the blood glucose concentration decreases to approximately
60 mg/dl, leading to a decrease in the blood insulin level by about 10-15 times
compared with the absorptive state, and a rise in the blood glucagon, level.
The changes in hormonal status and the action of intracellular regulatory
mechanisms result in glycogen stores in the body to be almost completely depleted
by a 24-hour fasting, and the rate of fat mobilization and the rate of gluconeogenesis
increase. The liver glycogen is converted back to free glucose to maintain normal
blood glucose levels between meals, thus, only short-term fasting is provided by
the liver glycogen mobilization. The only process that provides tissue with glucose
during long-term fasting is gluconeogenesis. Gluconeogenesis begins eo accelerate
4-6 hours after the last meal and becomes the only source of glucose during
prolonged fasting period, maintaining the blood glucose at lower limit of the normal
in subsequent periods of fasting. Amino acids, glycerol and lactate arc the main
substrates of gluconeogenesis.
During the second phase, a decrease in the blood glucose level keeps low insulin/
glucagon ratio and leads to increased secretion of other counted nsular hormones,
cortisol and growth hormone. During the first days of lasting tissues consume
less glucose than they use in postabsorptive slate. The only source of glucose is
gluconeogenesis from amino acids and glycerol. Although body proteins arc not
primarily a reserve form of fuel as glycogen or fats, during the first few days of fasting
muscle proteins arc quickly degraded into amino acids which become the main
carbon source for glucose synthesis. The energy needs of the muscles and most
other organs arc met principally by fatty acids and ketone bodies, because the blood
insulin concentration is very low and glucose does not enter muscle cells. The rate
of TAG mobilization continues to rise, the blood fatty acid concentration increases
approximately twice compared with the postabsorptive stale. The rate of ketone bodies
formation in the liver increases significantly driven by low instilin/ghicagon ratio and,
accordingly, their blood concentration increases too (Fig. 9.17).
After 3-5 days of fasting the concentration of ketone bodies in the blood reach 2
to 3 mMol, and exhaled air and sweat of the fasting person smells of acetone. Under
these conditions, only insulin-in dependent cells, and especially brain cells, become
glucose consumers. However, ketone bodies begin to provide for a notable portion of
the metabolic needs for brain and other nervous tissues and, consequently, these tissues
oxidize less glucose too. The only source of glucose is gluconeogenesis that continues
due to amino acids derived from the breakdown of tissue proteins and glycerol from
adipose triacylglyccrols. After a week of fasting metabolic rale is generally reduced and
oxygen consumption decreases by about 40%.
During the third phase, the metabolic rate continues to stow down. The rate of
protein breakdown decreases, and after several weeks of fasting, it stabilizes at about
20 g per day. When this amount of proteins is broken down, about 5 g of urea per day
is formed and secreted (at normal nutrition rhythm — 20-25 g). The nitrogen balance
9.6. Metabolic Changes During Muscle Activity 521
Fig. 9.1T Changes in blood fuels during fasting. The plasma levels of fatty acids and ketone Dodies
increase in starvation, whereas mat of glucose decreases
in all phases of fasting is negative. The rate of gluconeogenesis from amino acids is
reduced. In this phase, the brain continues to use both ketone bodies and glucose, bin
less actively due to a decrease in the rate ofgluconeogenesis and a decrease i n the blood
glucose concentration. A decrease in the level of gluconeogenesis from amino acids is
necessary to preserve protein, since severe protein loss (loss of 1/3 of all proteins,
including heart muscle mass) causes a malfunction of the major organs and can lead
to death. Continued slow protein loss during fasting of an extremely obese person can
lead lo death, from protein depletion even before TAG stores are depleted. The rate of
oxidation of ketone bodies in the muscles is reduced, and they almost exclusively use
fatty acids. Almost all of the body's energy needs are met by oxidizing fatty acidsand
ketone bodies until TAGs are depleted. The duration of starvation depends on how
long ketone bodies can be synthesized and oxidized to produce ATP. The oxidation of
the ketone body requires oxaloacctatc and other components of the TCA cycle, which
are formed from glucose and amino acids at a normal rhythm of nutrition, and when
starvation only from amino acids.
the glucose synthesis from lactate, amino acidsand glycerol, as well as by mobilizing
other types of fuel, which can serve as an alternative (fatty acids, ketone bodies). The
endocrine system in these conditions is important primarily for the maintenance or
replenish me nt of fuel in the muscles.
Du ring short-term muscular activity, the main sources of energy that ensure muscle
functioning arc endogenous .ATP stores in muscles, creatine phosphate and glucose
from glycogen. The total ATP content in the muscle is approximately 5 pmol per I g
of its mass that is enough only for about I. s muscular activity. A high-energy molecule
creatine phosphate is reversibly formed from creatine and ATP under the action of
creatine kinase (Ch. 8). The level of creatine phosphate in resting muscle is several
fold higher than that of ATP. This stores provides intensive muscle work for 2-5 s
due to rapid ATP rcsynthesis by substrate-level phosphorylation using the high-energy
creatine phosphate and ensures a person to run 15-50 meters. As creatine phosphate
stores are depleted, glucose from muscle glycogen becomes a major source of energy.
For short-term maximum efforts, energy is released from the fuel anaerobically, before
regulation of the blood circulation can provide the necessary oxygen. Epinephrine and
norepinephrine, released from the adrenal medulla and sympathetic nerve endings in
response to central activation of the sy mpathetic nervous system, stimulate glycogen
degradation. The breakdown of glycogen lo glucose and the glucose oxidation to
lactate provide the required ATP. Calcium released from the sarcoplasmic reticulum
in response to nerve stimulation not only causes muscle contraction, but also activates
glycogen phosphorylase. Lactate released from working muscles is converted to glucose
in. the liver and can be exported back to the muscles in the Cory cycle. More long-
duration muscle activity requires adequate oxygen delivery and the capacity for the
muscle to utilize the oxygen delivered. During exercises overall oxygen consumption
may increase 10- to 15-fold in a well-trained athlete. The relative intensity of the
anaerobic and aerobic glycolysis in this ease changes. The rate of anaerobic glycolysis
decreases while the rate of aerobic glycolysis increases.
Glucose is also an important fuel in the early stages of moderately intense exercise.
As physical exercises continue, skeletal muscles significantly deplete their own glycogen
stores and begin to compete with the brain for the glucose available in the blood (for
example, glucose released from the liver}. Hut during prolonged physical exercises, the
muscles begin actively to use fatly acids as fuel molecules. As the duration of physical
exercise increases and the level of insulin decreases in combination with an increase in
the contra-insular hormones level, the lipolysis enhances that releases fatty acids from
adipose tissue. Maintaining low insulin levels allows laity acids lo circulate in higher
concentrations available for tissue use, sparing glucose. Fatty acids arc transported
lo the working muscles and liver. In muscles, they arc actively used for the oxidation
and synthesis of ATP. In the liver, fatty acids are oxidized to acetyl-CoA, which, arc
the precursors for the ketone body synthesis during prolonged physical load. Ketone
bodies provide useful fuel for various tissues, including the brain, while reducing the
blood glucose level.
The combined effects of the fall in insulin secretion and the rise in glucagon
secretion arc complemented by the contribution of catechol a mines, epinephrine and
norepinephrine, and in the liver they increase glycogenolysis and gluconeogenesis,
9.7. Metabolic Changes in Hypo- ana Hypersecretion of Hormones 523
absolute insulin deficiency occurs and severe metabolic disorders develop. In most
eases 1DDM usually begins before age 20, affecting children, adolescents and young
people, bin can occur al any (starting from I-year-old) age.
NIDDM develops as a result of various causes involving impaired conversion of
proinsulin to insulin, regulation of insulin secretion, increased insulin catabolism,
damage to insulin signal transmission mechanisms in target cells (for example, insulin
receptor defect, damage to intracellular insulin signal mediators, etc.), the formation
of anti bodies to insulin receptors (and the concentration of insulin in the blood may be
normal or even elevated). The factors determining the development and clinical course
of the disease include obesity, improper diet, sedentary lifestyle, stress. NIDDM
typically arises later in life than docs the insulin-dependent form a fleeting people,
usually over40 years old. develops gradually, the symptoms arc moderately expressed.
Acute complications arc rare.
in diabetes, as a rule, the insulin/glucagon raLio is reduced. At the same time, the
stimulation of glycogen and TAG synthesis is reduced, and mobilization of the energy
reserves is intensified. Even after a meal, the liver, muscles, and adipose tissue function
in a postabsorptivc mode. Moreover, products of digestion, as well as their meta bo I i Les,
instead of being deposited in the form of glycogen and fat. circulate in the blood.
All forms of diabetes arc characterized by an increase in the blood glucose
concentration — hyperglycemia, both after meals and on an empty stomach, as well as
glycosuria. In. normal individual a fasting blood glucose concentration is maintained
al around 90 mg/dL of plasma (5mM). After a meal, the blood glucose levels return
to baseline values within two hours, and the peak value docs not rise above 140 mg/
dL(7.8 mMol/L). In diabetes blood glucose levels rise much higher and maybe 300-
500 mg/dl and remains high at the post-adsorptive state (reduced glucose tolerance).
A decrease in glucose tolerance is also observed in cases of latent diabetes mcllitus.
In these cases, people have no complaints and clinical symptoms characteristic of
diabetes, and the fasting blood glucose concentration corresponds to the upper
limit of normal. However, the use of provocative tests (for example, sugar load)
reveals a decrease in glucose tolerance (Fig. 9.18). A fasting blood sample is taken
to establish a baseline glucose level. Then a patient drinks a glass of concentrated
glucose solution (75 g of glucose arc dissolved in 250 to 300 ml of water). To confirm
suspected diabetes, blood is drawn again after two hours and the blood glucose level
is measured.
The increase in blood glucose concentration, in IDDM is caused by several
factors. With a decrease in the insulin-glucagon index, the effects of contra insular
hormones increase, the number of glucose transporters (G LU T-4) on the membranes
of insulin-dependent cells (adipose: tissue and muscles) decreases. Consequently,
glucose consumption by these cells is reduced. In muscles and liver glycogen synthase
is phosphorylated and inactive; and glucose is not deposited as glycogen. In liver and
adipose tissue, the glycolytic enzymes and pyruvate dehydrogenase arc inactive and,
consequently, the conversion ofglucose into acetyl-CoA required for the TAG synthesis
declines, thus the rale of lipogencsis decreases. In addition, contrain.sular hormones,
primarily glucagon, activates gluconeogenesis from amino acids, glycerol and lac talc.
Thus increased hepatic production of glucose is combined with diminished peripheral
528 Chapter 9. Molecular enaocrmoiogy
Fig. 9.13.Glucose intolerance test Bload glucose curves of a normal and a diabetic individuai after oral
administration of glucose solution. A criterion of normality is me return of tne curve to me initial value
witnin 2 nours. 1 — in a rteaitny person; 2 — in a patient wim diabetes
tissue due to the reducing storage processes and low activity of LP-lipase, and enter
the liver, where they arc converted imo triacylglycerols, which are transported from
the liver as part of VLDL. Nonenzymatic glycosylation of apoproteins and receptors
disrupts the interaction of receptors with lipoproteins and reduces their entry into
cells, which also leads to hyperlipidemia.
Insulin deficiency results in the protein synthesis rale slowdown and acceleration
of body protein breakdown and in patients with poorly controlled diabetes can leads
to an increase in the blood amino acid concentration. Amino acids enter the liver and
undergo deamination. The resulting ammonia is included the ornithine cycle, which
leads to a rise in. urea production, and in severe insulin deficiency an increase in. blood
urea concentration and, accordingly, in the urine may be experienced. The nitrogen-
free glycogenic amino acid residues arc included in gluconeogenesis, which further
enhances hyperglycemia.
Polyuria and polydipsia arc common diabetes signsand symptoms. The capability
of kidney to excrete high concentration of glucose and ketone bodies is limited.
Excretion of large amounts of osmotic active molecules such as glucose and ketone
bodies requires much more water excretion. Patients with DM excrete urea 2—3 Limes
more than a healthy human and the loss of water is accompanied by excessive thirst.
For example, urine excretion in some cases reaches 8-9 liters per day, but more often
it docs not exceed 3-4 liters. The water loss causes constant thirst and an increase in
water consumption. In severe forms of diabetes dehydration can occur. As a result of
the excretion of large urine quantities, blood volume decreases: cells receive water
from the extracellular fluid; the extracellular fluid becomes hyperosmolar and «pump*
water out of the cells; external signsofdehydration develop — dry mucous membranes,
loose and wrinkled skin, sunken eyes, blood pressure decreases and oxygen supply to
the tissues reduces.
Acute complications of diabetes (comatose states) arc resulted from the changes
in carbohydrates, fats and proteins metabolism. Diabetic coma manifests as a sharp
impairment of all body functions, accompanied by loss of consciousness. Acidosis
and tissue dehydration are the main precursors of diabetic coma (Fig. 9.19). In
decompensation of diabetes, a disorder of water and electrolyte metabolism develops.
It is caused by hyperglycemia, accompanied by an increase in osmotic pressure in
the bloodstream. To maintain the osmolarity, a compensatory movement of the fluid
from the cells and the extracellular space into the bloodstream begins. This leads to
tissue loss of water and electrolytes, primarily Na'.. K , Cl . and HCOq ions. As a
result, severe cellular dehydration and a deficiency of intracellular ions (mainly KJ
develop, accompanied by general dehydration. This leads to a decrease in peripheral
blood circulation, a decrease in cerebral and renal blood How and hypoxia. A diabetic
coma develops slowly over several days, but can sometimes arise in a few hours. The
first signs may be nausea, vomiting, lethargy. Patient’s blood pressure is reduced.
1 n diabetes me Hilus comatose states can manifest in three main forms: kctoacidotic,
hyperosmolar and lactic acidotic.
Diabetic Ketoacidosis (DKA) may be caused by the cessation or reduction of
insulin administration, late diagnosis of the disease, infectious diseases, stress state.
The metabolism in DKA simulates that in starvation with marked insulin deficiency
but the intensity of lipolysis, gluconeogenesis and ketogenesis is very high in DKA,
530 Chapter 9. Molecular endocrinology
Fig. 9.19. Mechanisms ol development of diabetic soma. Metabolic changes of diabetic coma arise
from insulin deficiency with increased levels of counterregulatory hormones like catecholamines,
glucagon, cortisol and growth hormone. The changes of hormonal ration result in increased glucose
concentration because of inadequate glucose utilization by insulin sensitive tissues like muscles,
adipose tissue and liver; increased glycogenolysis; and increased gluconeogenesis by the liver from
amino acids, increased lipolysis leads to increased free fatty acids undergoing p-oxidation io generate
ketone bodies. High intensity of lipolysis, gluconeogenesis and ketogenesis leads to profound acid
base and electrolyte disturbances. Osmotic diuresis results in loss of large amounts of water, sodium,
potassium, magnesium and phosphate
9.8. MetaDolic Changes in Diabetes Meiliius 531
In hyperosmolar coma the major criterion is extremely high blood glucose level
associated with markedly elevated scrum osmolarity (> 340 mOsm/L, if the patient
is comatose) with minimal or no ketoacidosis, polyuria* polydipsia, and always
manifested severe dehydration. It is associated predominantly with type 2 diabetes.
But* it is important to realize that any of these metabolic emergencies can occur in any
types of I) M, irrespective of the age or gender of the patient.
Hypotonia, a decrease in peripheral circulation and tissue hypoxia, that leads to
metabolism shifting towards anaerobic glycolysis, with increased the blood lactate
concentration (lactic acidosis), arc predominant in lactic acidosis.
Pure forms of the comatose states described arc practically not found. Their
development may be caused by various factors, for example, infectious diseases,
injuries, surgical interventions, toxic compounds, etc.
Late complications of diabetes niellitus are associated with long-term damage and
failure of various organ systems and a consequence of prolonged hyperglycemia and
often lead to early disability of patients. Late diabetic complications include a variety
of clinical pictures, mainly related to the involvement of the arterial wall both of large
vessels (macroangiopathy) and small vessels (microangiopathy), and of the peripheral
nervous system (neuropathy). Hyperglycemia causes damage to blood vessels and
impaired function of various tissues and organs.
The main mechanism of tissue damage in diabetes is glycosylation of proteins and
the associated dysfunction of tissue cells and changes in the rheological properties
of blood and hemodynamics (fluidity, viscosity). Some compounds normally
contain carbohydrate components (glycoproteins, proteoglycans, glycolipids). These
compounds arc synthesized by enzymatic reactions (enzymatic glycosylation).
However, non-enzy malic interaction of glucose aldehyde group with free amino
groups of proteins (non-enzymatic glycosylation) can also occur in. the human
body. In tissues of healthy people, this reaction proceeds slow and accelerates with
hyperglycemia.
One of the first signs of diabetes is a 2-3 times increase in glycosylated
hemoglobin. Throughout the lifetime of the erythrocytes* glucose freely penetrates
their membranes. Then in non-enzy malic reaction it irreversibly binds to hemoglobin,
mainly to 0-chains to produce a glycosylated hemoglobin HbA|r. Small amounts of
HbA can be also found in healthy people. In chronic hyperglycemia, the percentage
of HbAk in relation to the total amount of hemoglobin increases.
The degree of proteins glycosylation depends on the rale of protein turnover. As
proteins age they arc more likely to acquire molecular damage. Proteins with, slower
rates of turnover arc, therefore, al greater risk of suffering a deleterious modification.
Long-lived proteins accumulate more alterations and include extracellular proteins,
matrix, basement membranes and crystalline (lens of the eye). Thickening of the
basement membranes is one of the early and permanent signs of diabetes, manifested
as diabetic angiopathies.
Diabetic macroangiopathies represent the changes manifested in a decrease in
arterial elasticity, damage to large and medium vessels of the brain and heart, lower
extremities and include such com plications as coronary artery disease, cerebrovascular
disease, peripheral artery disease, non-sign ifleant carotid stenosis and poly vascular
532 Chapter 9. Molecular enaocrinoiogy
Review Tests
1. Match the figure an d the letter. Dynamics of change:
A. In a healthy person after a meal-
B. Under the insulin action.
C. Under the glucagon action.
D. Ina patient with diabetes.
E. In a patient with an increase in. glucose tolerance.
[ insulin deficiency j
I
I
21
I
.31
I
Turea level in the urine
Hyperglycemia
p-cell damage
I
GJ
I
(2.1
I
d
534 Chapter 9. Molecular eoaocrmoiogy
Situational problems
LA student has fasted for a day to lose weight. How did the carbohydrate
metabolism change when the absorptive state was switched to 24 hrs fasting? For
answer the question:
a) compare the changes in carbohydrate metabolism in the student's liver after
the last meal and by the end of the first day of fasting, and draw appropriate
charts;
b) list the peptide hormones with an increased level in the student’s blood
during the absorptive state and by the end of the first day of fasting, name the
site of their synthesis, secretion stimuli, target organs and draw the diagram
of mechanism of signal transduction of these hormones in the target cells;
c) name the regulatory enzymes of the metabolic pathways with increased
activity and amount under the action of these hormones.
2. A girl decided to lose weight and has been fasting for three days. What metabolic
changes occurred in the girl’s organism by the end of the 3-day fast? For answer
a) draw appropriate charts of metabolic pathways reflecting the changes in fat
metabolism in the liver and adipose tissue during the prolonged fasting;
b} name the hormones that accelerate these processes, and draw the charts of
signal transduction of these hormones to the target cells;
c) enumerate the regulatory enzymes with changed activity under the influence
of the mentioned hormones.
3. Tourists miscalculated food supplies and have been starving for 2 days before
reaching a settlement. How has their fuel metabolism changed in this case? For
answer:
a) explain how the concentration of glucose in the blood of tourists changed by
the end of the second day of fasting:
b) enumerate the metabolic pathways that maintain the blood glucose
concentration on the first day of fasting;
c) name the hormones that regulate the blood glucose levels during this period?
d) draw a chart of the signal transduction of these hormones.
4. Tourists were cast away on a deserted island and were starving for a week. None
of them developed a hypoglycemic coma. By chance, a captain of a cruise liner
came to their rescue and delivered them to a medical center on land where they
were examined. The blood glucose concentration in everyone did not fall below
65 mg/d L. Why was the blood glucose concentration of tourists within the lower
limit of the norm? To answer:
a) explain the molecular mechanisms that maintain the blood glucose levels
during long-term starvation;
b) draw charts for the metabolic pathways with increased activity under these
conditions; specify regulatory enzymes;
c) enumerate the hormones that stimulate these metabolic pathways;
d) present a diagram of signal transduction of these hormones on hepatocytes.
5. There is an expression; diabetes mellitus is ^hunger among abundance.* What
metabolic changes in diabetes confirm the validity of this statement? For
answer:
9.8. MetaDoi ic Changes in Diabetes Melliius 535
which terminate in the posterior pituitary where ADH is stored, and then secreted into
the bloodstream with appropriate stimulation.
The release of ADH from its storage vesicles in the neurohypophysis (posterior
pituitary; pars nervosa) is primarily caused by increase the plasma osmolality
(osmotically active solute concentration in millimolcs/kg of plasma waler) and
increase the osmotic pressure of the extracellular fluid (Fig. 9.20).
Urine ceil
Fig. 9.20. Secretion and action of ADH. ADH is produced Dy neurons in tne hypothalamus and stored
bound to its neurophysin (NPIIJ in tne posterior pituitary. ADH is synthesized and released in response
to increased sodium level and decreased plasma volume. ADH Dinos to cell membrane receptors,
stimulating tne activation of protein kinase A {PKA)_ PKA phosphorylates proteins that stimulate
synthesis of aquaporin, a protein that forms channels in the renai tubular membrane. Water, resorbed
through these channels from the urine, enters the blood
538 Chapter 9. Molecular endocrinology
The most important target cells for ADH arc the cells of the distaJ tubules and the
collecting tubules of the kidneys. The cells of these ducts arc relatively impermeable
to free water, and in. the absence of ADH, the urine is not concentrated and can be
excreted in quantities exceeding 20 liters per day (the norm is 1-1.5 liters per day).
The biological effects of ADH arc mediated through two types of receptors, V(
and V2.
The major action of vasopressin binding to V.-reccptor on renal tubular cells is to
promote waler reabsorption through the luminal membrane of the epithelial cells of
the cortical and medullary segments of the kidney collecting ducts ( Fig. 9.21).
Kinase A
I
Water
reabsorption
The complex ADH — V.,-receptor causes activation of the adcnylyl cyclase., which
produces cAMP. cAMP activates protein kinase (PKA), which phosphorylates
proteins that stimulate the expression of the membrane protein gene, aquaporin-2.
Aquaporin-2 moves to the apical membrane, integrates into it and forms waler
channels through which water molecules diffuse freely into cells of renal tubules,
and then enter the interstitial space. As a result, water is reabsorbed from the renal
tubules.
Receptors V'^rc localized in smooth muscle membranes. The interaction of ADH
with the V]-receptor leads to the activation of phospholipase C, which hydrolyzes
phosphatidylinositol 4,5-bisphosphate forming diacyl glycerol (DAG) and inositol
1,4,5-trisphosphatc (IP,). IPjstimulates the release of Ca2k from the endoplasmic
reticulum and the vascular smooth muscle layer is contracted resulting in the volume
of the vascular bed decrease and the pressure rise.
9.9. Actions of Hormones Regulating Sodium and Water Balance 539
LimEfi cd cwiicaJ
collecting dusr
This acts as a signal for osmoreceptors of the hypothalamus and ADH secretion
from the nerve endings of the anterior pituitary, which stimulates the reabsorption
of water from the collecting tubules. Havi ng a potent direct vasoconstrictor action
on the resistance vessels of the body angiotensin 11 increases blood pressure and also
increases thirst. Excess water intake causes the body to retain waler to a greater extent
than normal. An increase of fluid volume and an increase in blood pressure lead to the
elimination of the stimulus that caused the activation of the renin-angiotensin system
and the secretion of aldosterone and, as a result, lead to the restoration of the blood
volume.
The reduction of perfusion pressure in the renal glomeruli can also occur due to
narrowing (stenosis) of the renal artery or nephrosclerosis. In this case, the entire
renin-angiotensin system is also included. But since the initial volume and blood
pressure are normal, the inclusion of the system leads to an increase in blood pressure
above the norm and the development of so-called renal hypertension.
542 Chapter 9. Molecular eoaocrmoiogy
1,000 to 1,500 mg, mainly in dairy' products. Blood inorganic phosphate level is about
3.5 mg/dL. About 55% is present as Tree ions, about 35% is complexed with calcium
or other cations, and 10% is protein-bound.
Parathyroid hormone, cafcitriol and calcitonin are the main regulators of Ca2 and
phosphate exchange.
Parathyroid hormone is synthesized in the parathyroid glands in the form of
preprohornionc that is converted to mature 84-amino acid, single-chain peptide
hormone (PTH) by partial proteolysis. PTH is secreted in. response to a decrease in
extracellular ionized calcium or an increase in serum phosphate concentration. The
hormone causes the concentration of free scrum Ca2 to increase to a level just above
the normal range (above a critical set point). The latter acts as a * negative feedback*
signal to the chief cells of the parathyroid gland, inhibiting fun her release of PTH
until the free Ca2' level in the blood again falls below a specific threshold or set point
(Fig. 9.26). Aller secretion into the blood, intact PTH is inactivated by proteolysis.
Parathyroid
gland
PTH
Fig. 9.26. Effects ol PTH. • 1: PTH stimulates the mobilization of Ca?i from bone: (2) PTH stimulates the
reabsorption of Ca?* in the distal tubules of the kidneys: (3) PTH activates the formation Gtl.SSfOHjPg
in the kidneys, which leads to stimulation of the absorption of Ca3- in the intestine: {4) increased
concentration of Ca74 ions inhibits the secretion of parathyroid hormone
The main target organs for the hormone arc bones and kidneys. The hormone
initiates a cascade of events associated with osteoblast adenylate cyclase that stimulate
the metabolic activity of osteoclasts. Ca2’, is mobilized front the bone and phosphate
enters the blood. In the distal tubules of the kidneys Ca2', reabsorption increases and
phosphate reabsorption decreases, resulting in the restoration of normal calcium ion
levels in the extracellular fluid.
546 Chapter 9. Molecular endocrinology
JlT I H
Or
I
J—t
r'A;
p (7-ctetiydfDchoie&iaror|
ho!^3jx
Skirt J-
(axrtecaiLil&.'aliviTa.-wi Pg))
"r
Liver
fgl-hydroajf' caJcjierof (25 OHDj ]
KiHrtEy
Review Tests
Match the number and the letter.
1. Regulation of hormone secretion. Molecule:
A. Natl.
B. Aquaporin.
C. Ncurophysin.
D. ADH.
E. V, receptor.
2. The mechanism of aldosterone action.
Induced proteins:
A. Proteins forming the Na' and K‘
channels.
B. Na', K-ATP-ase.
C. ATP synthase.
D. Citrate lyase.
E. Citrate synthase.
Situational Problems
1. On a hot day a tourist in the wilderness could not find a source of drinking waler
for a long time. Finally, he reached a village and quenched his thirst. How will
his water and salt balance change after that? For answer:
a) name the hormones that regulate the water and salt balance;
b) draw a chart explaining the occurrence of thirst;
c) using the chart, compare the state of the tourist’s water and salt metabolism
before he drank water and after.
2. A 4-month-old child has signs of rickets. Digestive disorders were not noted.
The child has had adequate sun exposure, he has been receiving vitamin D-, for
2 months, but the manifestations of rickets did not diminish. Why did the doctor
prescribe calcitriol to treat rickets in this child? For answer:
a) draw a chan of hormone synthesis derived from vitamin D5 and list the
possible causes of rickets in children;
b) enumerate the target organs of the hormone and draw a chart of the hormonal
signal transduction;
c) name the cause of rickets in this child and explain the doctors prescription.
3. A 45-year-old woman was admitted to hospital because of frequent headaches,
muscle weakness, increased thirst. Increased blood pressure, impaired blood
electrolyte composition and significantly reduced renin levels were determined
during the examination. A hormone-producing tumor in the right adrenal gland
was detected during a computed tomography scan, allowing to diagnose pri mar}'
hype raldosteron ism (Conn’s syndrome). What are the causes of hypertension in
this patient? For answer;
al list the biological functions of the main adrenal cortex hormones, name the
precursor for their synthesis;
b) name the main mineralocorticoid, the site of its synthesis and the target cell;
c) draw a diagram of the hormonal signal transduction to the target cells;
d) present a chart explaining the regulation of the hormone’s synthesis and
secretion;
c) explain the consequences of the hormone hyperproduction.
4. A 39-ycar-old patient was presented writh persistent arterial hypertension
(200/120 mm Hgh which was not eliminated by conventional antihypertensive
drugs. An ultrasound of the renal arteries revealed signs of stenosis of the right
renal artery. Why docs renal artery’ stenosis lead to an increase in blood pressure.
For explanation:
a) present a chart of water and salt balance regulation, indicate the main stimuli
of system activation and the final effects;
b) name the compound, with its increased synthesis caused by a drop in renal
arteries perfusion pressure;
c) describe the consequences arising from an increase in the blood concentration
of this substance, using the scheme presented in paragraph a).
5. A 23-ycar-old man was admitted to a hospital fora planned surgical operation.
When performing the surgical operation to remove a tumor from the upper
section of the anterior pituitary, an isthmus of the posterior pituitary was
552 Chapter 9. Molecular eoaocrmoiogy
Ccqugahon
rPoiarity
iSzkitnIrty in'water
isadfh NADP^.H*
Cytadhrarre Cytochrwne
P450 reductase P45O
Fig, 1OJL General slructure of the P45Q enzymes. 0? Di nets to the P450 Fe-heme in the active site and
is activated to a reactive form by accepting electrons. The electrons are donated Dy the cytochrome
P450 reductase, whicn contains an FAD together with an FMN or (Fe-S)-center to facilitate the transfer
of single electrons from NADPH to O?. For example, for CYP2E1 (CYP2E1 is also referred to as me
microsomal ethanol oxidizing system, MEOS), RH is ethanol (CH3CHj£)H}, and ROH is acetaldehyde
(CH3COH)
The latter drug’s concentration in the blood would then rise. Moreover., many
substances or drugs impair or inhibit the activity of the CYP3A4 enzyme, thereby
impairing the body's ability to metabolize a drug.
The cholesterol-lowering agents known as the statins CH MG-CoA reductase
inhibitors) require CYP3A4 for degradation. Appropriate drug treatment and dosing
take into account the normal deg rad alive pathway of the drug. However, grapefruit
juice is a potent inhibitor of CYP3A4-mcdiaicd drug metabolism. Evidence suggests
that if a statin is regularly taken with grape fruit juice, its level in the blood may increase
as much as IS-fold, This marked increase in plasma concent ration could increase the
muscle and liver toxicity of the statin in question. Because side effects of the statins
appear to be dose-related.
The cytochrome P450 isozymes all have certain features in common. The
term «P450* is derived from the spectrophotometric peak al the wavelength of the
absorption maximum of the enzyme (450 nm) when it is in the reduced state and
complexed with carbon monoxide. Most CYPs require a protein partner to deliver one
or more electrons to reduce the iron (and eventually molecular oxygen).
All of them:
I ) contain cytochrome P450, oxidize the substrate, and reduce oxygen;
2) have a flav in-containing reductase subunit (hat uses NAD PEL and not NADH,
as a substrate;
3) found in the smooth endoplasmic reticulum and arc referred to as microsomal
enzymes (lor example, CYP2E1 is also referred to as the microsomal ethanol
oxidizing system, MEOS);
10.1. Mechanism of Toxic Compounds Detoxification 557
Sonic alcohols, arylamines, and phenols arc sulfated. The sulfate donor in these
and other biologic sulfation reactions (e.g.s sulfation of steroids, glycosaminoglycans,
glycolipids, and glycoproteins) is adenosine 3 -phosphate-5’-phosphosulfate (PAPS):
this compound is called «active sulfate* (Fig. 10.4):
R + GSH - R-S-G.
The enzymes catalyzing these reactions arc called glutathione S-transferases and
varieties of them arc present in high amounts in liver cytosol and in lower amounts
in other human tissues. If the potentially toxic xcnobiotics were not conjugated to
GSH, they would be free to combine covalently with DMA, RNA, or cell protein
and could thus lead to serious cell damage. GSH is, therefore, an important defense
mechanism against certain toxic compounds, such as some drugs and carcinogens.
If the levels of GSH in a tissue such as liver arc lowered, then that tissue can be
shown to be more susccpiiblc to injury by various chemicals that would normally be
conjugated to GSH.
10.1. Mechanism of Toxic Compounds Detoxification 559
A
N CHj COO"
H
Y -gkJtamyt-s-y&tejny-glycine
Ghjlalhione
Dibromomelf^ane
*GSH
H -CO2
h2n — — C —COOH H2N—(CH2)s— nh2
! E
Omitine nh2 Pulrescine
The amino acid tryptophan undergoes a scries of reaction to form skatole and
indole:
COOH
Tryptophan Indole
Phenylalanine and tyrosine arc converted to cresol and phenol:
OH
4-1/2^2
Monooxygenase
(hydroxylase) H
Indole Indoxyl
The other type of conjugate formation is the biosynthesis of sulfate esters with the
help of phosphoadenosine phospho sulfate (PAPS), the -active sulfate:
OSChH
Adenosine — Ribose — ©— O—SO3H
Saks of indoxylsulfuric acid arc excreted with urine in trace amounts and
referred to as “animal indicant". Increased animal indicant in the urine reflects the
intensification of putrefaction in the large intestine.
Amide formation with glycine and glutamine plays a considerable role in
conjugation. For example, benzoic acid conjugation with glycine forms the more
soluble and less toxic hippuric acid (.V-bcnzoylglycinc), the measurement of which
excretion is used to determine the liver function:
The conjugates arc eliminated from the liver either by the Bihary route — i.c.T by
receptor-mediated excretion into the bile or by the miai router via the blood and
kidney by filtration.
Fi^ 10.6. Some cnemicai modifications: A — bar Disrate oxidation: B — aspirin hydrolyzes
10.4. MetaDolism and Detoxification of Ethanol 563
► The second stage is the conjugation of drugs (either native or modified by first
stage) with glycine, acetate, glycuronatc, sulfate, glutathione, etc. (Fig. 10.7);
Fig. 10.7. Conjugation reactions ol some drugs wim glucuronic acid: A — acetaminophen conjugation;
H — salicylic acid conjugation
rno2-rnh2.
Some medicine doses require increasing because of their activity goes down in
permanent intake that occurs due to the induction of enzymes of monooxygenase
system and conjugation reactions. Exactly these circumstances lay in drug addiction
and some poisons. Broad substrate specificity of detoxification system enzymes has a
particular significance as well.
IMADH +H NADH + H
H H
I I
I- 0 C O H
I I
H H
Etnanoi
H H NAD* NADH H
I 1
I 1
H—C--C— —H H—C
1 1 ADH 1
H H H
Fig. 10.10 Ethanol oxidation By MEOS: ADH — alcohol denyarogenase; MEOS — microsomal esnanoi
oxidizing system; ALDH — aldehyde dehydrogenase
Oxidized substrates may be all iron-containing hemo prole ins: Hb, ETC
cytochromes, cytochromes of monooxygenase system of endoplasmic reticulum.
568 Chapter 10. Tissue metabolism. Liver detoxification function
Niirosammcs R.N-N=O formed in the body from nitrous acid (HNO,) and
second amines (R.NH) are sufficiently strong mutagens inducing alkylation (for
example methylation) of DNA nitrogenous bases. Nitrites convert cytosine residue
to uracil in DNA chain, as well, and the complementary pair GC becomes UC:
Cytosine uracil
During the course of replication of mutant DNA U forms complementary pair UA
which converted to AT in the next replication. If mutation occurs in proto-oncogcnc of
protein responsible for regulation of cell cycle, the next replication can lead to damage
of its structure, uncontrolled cell division and tumor development.
Nitrates and intermediary products of their conversions decrease the activity of
some enzymes of antioxidant system that leads to accumulation of reactive oxygen
species (ROS) and lipid peroxidation activation.
Aromatic amines. Aromatic amines arc compounds which are used in. the production
of aniline colors and resins. One of them is 2-naplnytamine which converted to
carcinogen 2-amino-l-naphtol in the liver. However, it conjugates with PAPS (or
Ul)P-glucuronic acid) and is converted to neutral compound in hepatocytes. By
excretion some conjugates arc hydrolyzed and that loads to formation of carcinogen
again which can induce the development of urinary bladder cancer (Fig. 10.12).
Aflatoxins arc metabolites of some mold fungi, for example Aspergillus Jlavus
which arc developed in wrong storage of cereal products, groats and nuts Aflatoxin
I? I is converted to epoxide as a result of chemical modification in the liver which is
carcinogen and induces the development of liver cancer (Fig. 10.13).
DNA/RNA
CYP450
Aflatoxin B-a.9-epoxide
Situational problems
1. Some amount of benzol during technical processing in a shoe factory was
evaporated. The workers inspired it with the air. Wliat mechanisms are employed
by the liver to gel rid of toxic compounds? To answer the question:
a) point, out the phases of toxic compound detoxification:
b) write down rcactionsofhcnzofdctoxilication, specify enzymesand coenzymes.
2. Epidemiological evidence suggests that diets high in meat and fat and low in
fermentable carbohydrate increase the colorectal cancer risk. One mechanism
that could explain the association with meal is increased colonic protein
metabolism due to increased protein, intake from high meat diets. Products
of colonic protein degradation and metabolism include ammonia, phenols,
indoles and amines which have been shown to exert toxic effects in. vitro and in
animal models. Why metabolites from colonic protein metabolism contribute to
this increase in genotoxicity during high meat intakes? To answer the question:
al write down the formulae of amino acids these toxic compound arc formed
from, and reactions of these toxic compounds formation;
570 Chapter 10. Tissue metabolism. Liver detoxification function
Paracetamol or Panadol
The main ptarmacologkaB^
active metaMite
Hut phcnacctin production was discontinued due to its side effects and
oncological disorders induced in prolonged usage. Paracetamol is less toxic and
rarely causes complications in. therapeutic doses; it’s faster metabolized in the
fiver and excreted with, urine. Why is paracetamol less toxic and faster excreted
from the body? To answer the question:
a) examine these medicine formulae and explain the less toxicity of paracetamol;
b) write down reactions of paracetamol detoxification:
c) specify the oxidative product of hemoglobin formed in prolonged treatment
by phcnacctin or in poisoning by large doses of paracetamol.
11. Isoniazidum is widely used in t u be re u los i s prolonged therapy. Why this medic inc
induces side effects and complications (for example, neuritis, damages of optic
nerve)? To answer the question pay attention to reactions linked with this
medicine:
572 Chapter 10. Tissue metabolism. Liver detoxification function
a) specify compound formulae of which arc present in these reactions; point out
biological importance of these compounds;
b) specify enzyme catalyzing the second reaction and its class;
c) point out the biochemical derangements which, can occur in patient body as a
result of reactions with isoniazid:
d) name vitamin, which, have to be administrated in prolonged treatment by
isoniazid.
INDEX
Electron transport chain (ETC) 174. 229,, Feedback inhibition 95, 328, 374
421 Ferrochelatase (heme synLhasc) 460. 461
Electrophoresis SO. 182, 264 Fibrous 29
Embryonic hemoglobin (Sec Fetal hemo Flavonoid(s) 248
globin) .35, 36 5-lluo ro uraci I. (5 - F U ) 478
Endergonic reaction 218 FMN 60,75
Endopeptidase 397 Foam cell(s) 387-389, 393
Enhancer 167, 210 Folate reductase 443. 444
Enoyl. Co A hydratase 341 Folic acid (vitamin Be or vitamin Bg) 99,
Entero hepatic circulation 154. 379. 389 443-445
Entero hepatic urobilinogen cycle 464 Follicle-stimulating hormone < FSH > 485,
Enteropeptidase 399 492, 493
Enzyme 57 Formyl-H4-folate 444. 468
activity 65—67, 92 Fructose 96,. 279-281
classification 78 Fructose 6-phosphate 266, 274
clinical applications 100 Fructose -1.6-bisphosphatc phosphatase
induced fit model 65 279,281
lock-and-key model 64, 65 Fructose-1.6-bisphosphatc 279, 281
regulation 92, 93 Fructose-2,6-bisphosphatc 96, 279
specificity, stereo 64 Fructosc-6-bisphosphatasc 274
V (maximal velocity) 66, 67 Fructose-6-phosphaie 266, 274, 279
unit of activity (U J 69 Fumarase 227, 243
Enzymopathies 103 Fumarate 97, 227. 433
Epinephrine (adrenaline) 1.5, 197, 201. 295
Ethanol 100, 302, 563-566 G
Ethanolaminc 309, 329 Galactocc re broside 313
Ethanolaminc mcthyllransfcrasc 452 Gallstones disease (cholclilhias) 379, 400
Euchromatin 165 Gangliosidc(s) 191, 313, 367, 369
Eumelanin 437 Gastrin 16, 398,492
Exergonic 218 Gaucher’s disease 369
Exogenous cholesterol 381 Gel electrophoresis 182, 184
Exon(s) 135, 170 Gel filtration chromatography 51
Exopeptidases 397, 400 Gene cloning 179, 184
Gene rearrangement 165, 166
F Gene regulation 159, 166, 170
FAD (Flavin adenine dinucleotidc) 60, 75, Genetic code 141 — 144, 156
555 Gierke’s disease 299
Familial lipoprotein lipase deficiency, dis Glycogen 93, 254
lipoproteinemia type I 319 synthesis 289, 290
Farber’s disease 369 degradation 293, 289
Fasting state, (Poslabsorptivc state) 259, hormonal regulation 353, 509
374 Glucagon 280, 294, 500
Fat (See also TAG) 313-319 action 343
Fat malabsorption 319 biosynthesis 500
Fat-soluble vitamin(s) 73, 308. 313 Glucoamylase complex 255
Fat tv acid (s) 308-312, 315 Glucoccrcbroside 333
essential 311, 319,327 Glucocorticoids 361, 504. 508
structure 325 Glucokinase 259, 500, 511
synthesis 323, 328, 500 Gluconeogenesis 78. 271-282
index 577
S I
S-adenosyl homocysteine (SAH) 446 TAG (triacylglycerols) 63, 84, 311, 31.3
S-adenosyl me ih ion inc (SAM) 435, digestion 315, 319
446-449 hormonal regulation 323
Salicylic acid LOO. 562,563 mobilization 519
Salvage pathway 467.^ 468, 472, 473 synthesis of 317, 330
Scatole 560 transport by lipoproteins 323
Scavenger re cepto r (S R) 384-387 TATA-box 130
Secondary bile sails, 379. 380, 392 Taurochenocholic acid 379
dcoxycholic 379 Taurocholic acid 380, 391
l ilac hoi i c 379 Tay-Sachs disease 369
Secondary proleinopathy 52, 53 TCA cycle (See also Krebs cicle) 78, 223,
Serine decarboxylase 452 227, 243
ScroLonin 15, 451, 453 Tctrahydrobiopterin (H+-hio pterin) 435
Sickle cell anemia 44. 45, 171. 178 Telrahydrofolate (THF, H^-folatc) 442
Somatostatin (SS) 485. 489, 491, 492 Thermogcnin 245. 252. 503
Southern blot 185, 186 Thiamine pyrophosphate (TPP) 74, 221.
Specific catabolic pathway(s) 218, 21.9 225
Specificity of transformations 64 Thromboxane (TXA) 354—360
Sphingolipids 191,308, 31.2, 362 Thymidylate synthase 471, 478
Sphingomyelin 191. 309, 312, 362 Thyroglobulin 501., 502
Sphingosine 77, 191, 312, 362, 365 Thyroid hormones (T1, T4) 169, 170, 194,
Squalene 373, 374 210, 493.501
SSB (single strand binding proteins) 119. receptors 169, 170, 487
120 peroxidase 524
Starch 53, 218, 253-256 Thyroid-stimulating hormone (TSH), or
Stearic acid 364 Thyrotropin 485, 493
Steatorrhea 319, 322 Thyrotropin releasing hormone (TRH)
Stercobilin 465, 466, 481 485, 491
Steroid hormones 169, 176, 282, 370, 382. Thyroxine (Tj 495, 501
503 Tophi gouty arthritis 475
Slop codon 141, 149, 172, 173 Topoisomerase 119. 153, 156
Substrate cycle 278-281 Transatdolasc 284
Substrate level phosphorylation 226, 265, Transaminase 79, 228, 403-405
522 Transcription 141, 153, 168
Subunit 33, 60, 94, 117, 145 Factors 26, 130, 131
Succinate dehydrogenase 226, 236 Transferases 79, 82
Succinyl CoA 220, 225, 226, 459 Transketolase 74, 79. 284
Succinyl CoA: accloacctale CoA Translation 113, 115, 133, 141-153
transferase 346 Translocation 147, 149, 173
Sucrase isomahasc complex 255 Transmembrane proton gradient 235. 245,
Sucrose 253, 255, 300 252
Sulfo lipid 366 Transmethylation reaction 445-447
Sulfonamide 99, 445. 458 Transport
Superoxide anion (O2 ) 245, 246 Passive 194
Superoxide dismutase (SOD2) 248 Active 194
Surfactant 312, 363, 364, 369 Triacylglycerol (TAG) 63, 311-319
Sympon 194-196, 501 Triiodothyronine (Ta) 501
index 583
Textbook
y'ic6uoc iiwiihc
EA3OBMH KY PC EHOXKMHH
JJIH CTWHTOB MEAMHHHCKHXCnEHMAflbHOCTEiL
BKJIIOqAfOlUHH CHTyAUMOHHLIE 3.OAHU
Fiori pcjiaxiiucfl A.M. EjYXOBa, A.E. TybapeBoW
http://www.geotar.ru/ISBN/978-5-9704-5650-7.html
r wwwjrouuru
L www.mcdknigitMnis.rv