Sei sulla pagina 1di 66

GENERAL CONCEPTS, ENZYMES AND

COENZYMES BIOENERGETICS AND


BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

I. BASIC CONCEPTS IN BIOCHEMISTRY Amino Acids with Acidic Side Chains


- proton donor at physiologic pH
A. Proteins - negatively charged at physiologic Ph
- abundant and functionally diverse molecules
- perform specific biological function due to their
unique three-dimensional structures
- composed of linear polymers of amino acids

Amino Acids
- there are only 20 amino acids in human proteins as
coded in the DNA
- composition: α- carbon atom, α-carboxyl group, α-
amino group and distinctive side chain (“R-group”)
- the side chains determine the role amino acids in
proteins Amino Acids with Basic Side Chains
- peptide bond: amide linkage between the α- - accept protons at physiologic pH
carboxyl group and α-amino group of two amino - positively charged at physiologic pH
acid residues

Amino Acids with Nonpolar Side Chains


- do not participate in hydrogen or ionic bonds
- do not lose or gain protons
- promotes hydrophobic interactions

Titration of Amino Acids


- amino acids contain weakly acidic α-carboxyl
groups and weakly basic α-amino groups as well as
ionizable R-groups
- act as buffers (i.e. resists change in pH following
addition of an acid or base)
- dissociation of the α-carboxyl group in alkaline pH
and dissociation of the α-amino group in acidic pH
- titration of alanine

Amino Acids with Uncharged Polar Side Chains

- at physiological pH, has a zero-net charge


- at alkaline pH, may donate proton (e.g. cysteine,
tyrosine)
- the hydroxyl, amide and carbonyl groups can
participate in hydrogen bond formation and be sites
of attachment of other compounds (i.e.
oligosaccharide in glycoproteins)
- sulfhydryl group of 2 cysteine molecules can - zwitterion: amino acid form with a zero zwitterion:
oxidize and form a dimer, cystine amino acid form with a zero-net charge

UST FMS MEDICAL BOARD REVIEW 2019 1 | BIOCHEMISTRY


GENERAL CONCEPTS, ENZYMES AND
COENZYMES BIOENERGETICS AND
BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

pH = pK1: equal amount of Forms I and II


- pH < pK1: predominantly Form I
- pH = pK2: equal amount of Forms II and III
- pH > pK2: predominantly Form III
- Isoelectric point (pI): predominantly Form II, net
charge is zero
- Buffering occurs within ± pH unit of the pKa

Primary Structure of Proteins


- consists of the sequence of amino acids along a
polypeptide chain
-Ser-Ala-Glu-Val-Leu-Arg-Gly-
Secondary Structure of Proteins
- formed by regular repeating pattern of hydrogen
bond formation between peptide bond carbonyl
oxygen and amide hydrogens
- atoms of the side chain are not involved B. Carbohydrates
- contain at least three carbon atoms, hydroxyl
α-helix groups and an aldehyde or ketone group
- Spiral configuration consisting of tightly packed, - empirical formula: (CH2O)n
coiled polypeptide - functions: main source of dietary energy (i.e.
- carbonyl of a peptide bond forms a hydrogen bond glucose), storage of energy (i.e. glycogen),
with an amide four amino acid residues further intracellular communication
along the chain
- the side chains extend outward Classification of Carbohydrates
- disrupted by proline, and amino acids with bulky or Monosaccharides
charged side chains -these are simple sugars containing one saccharide
group
β-sheet -can be further classified according to the number of
- formed by hydrogen bonds between two extended carbons and whether they have an aldehyde or
polypeptide chains or between two regions of a ketone group in their backbone structures
single folded chai
- the surface appears “pleated” Oligosaccharide
- the hydrogen bond are perpendicular to the -composed of two to ten saccharide groups
polypeptide backbone
- chains may run parallel or antiparallel Polysaccharide
-composed of ten or more saccharide groups that
Tertiary Structure of Proteins may be linear or branched
- refers to the unique three-dimensional
conformation of the protein determined by its amino Isomerism
acid sequence - isomers have the same chemical formula (e.g.
- protein folding produced by the interactions fructose and glucose)
between amino acid residues that may be located at - epimers are isomers that differ around one specific
a considerable distance from each other carbon atom (e.g. glucose is a C2- epimer of
- domains are the fundamental functional and mannose)
structural unit of polypeptides - enantiomers: the configuration of the asymmetric
- stabilize by electrostatic interactions, hydrogen carbon atom farthest from the aldehyde or ketone
bond, hydrophobic interactions, disulfide bonds group determines whether it is a D- (i.e. hydroxyl
group on the right) or L-sugar (i.e. hydroxyl group
Quaternary Structure of Proteins on the left)
- refers to the spatial arrangement of subunits in a - Anomers: the hydroxyl group on the anomeric
protein consisting of more than one polypeptide carbon may be in the α (i.e. below the plane of the
chain ring in the Hayworth projection or on the right in the
Fischer projection) or β (i.e. above the plane of the
Additional notes: pKa of side chains of amino acids ring or on the left in the Fischer projection)
configuration

UST FMS MEDICAL BOARD REVIEW 2019 2 | BIOCHEMISTRY


GENERAL CONCEPTS, ENZYMES AND
COENZYMES BIOENERGETICS AND
BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

- oxidized to provide energy, structural components


of triacylgycerols, cholesteryl esters, phospholipids
and glycosphingolipids
- in humans, fatty acids have an even number of
carbon atoms, are 16 to 20 carbon atoms in length,
and may be saturated (i.e. contains no double
bonds) or unsaturated (i.e. contains one or more
double bonds)
- glycosidic bonds form when the hydroxyl group on - essential fatty acids (cannot be synthesized by
the anomeric carbon reacts with an –OH (i.e.O- mammals): linoleic acid and linolenic acid
glycoside) or –NH group (i.e. N-glycoside) of - linoleic acid is the precursor of arachidonic acid
another compound

- Reducing properties: If the hydroxyl group on the


anomeric carbon of a cyclized sugar is not linked to
another compound the ring could open and act as a
reducing agent

Complex sugars
- includes the glycosaminoglycans, proteoglycans,
glycoprotein and glycolipids

Glycosaminoglycans (GAGs)
- aka mucopolysaccharides
- long, unbranched, negatively charged,
heteropolysaccharide chain composed of repeating
disaccharide unit [acidic sugar-amino sugar] n
- amino sugar is either D-glucosamine or D-
galactosamine, usually sulfated or acetylated
- acidic sugar is either D-glucuronic acid or
L-iduronic acid which contain carboxyl group
- the negative charge is due to the ionized carboxyl
and sulfate groups at physiologic pH and cause
GAGs to repel each other and bind water
- form most of the ground substance of extracellular
matrices and mucus secretions

Proteoglycans
- consist of a core protein with glycosaminoglycans
attached
- bottle brush appearance
- all GAGs form proteoglycans except hyaluronic acid

Glycoproteins
- proteins to which oligosaccharides are attached
- the carbohydrate moiety has no serial repeats, are
branched and may not be charged
- serves as enzymes, hormones, antibodies and
structural proteins

C. Lipids
- diverse group of molecules but are similar in that
they are water insoluble
- major source of energy, provide hydrophobic
partition, fat- soluble vitamins as coenzymes and
precursor of
prostaglandins and steroid hormones

Free or unesterified fatty acids


- empiric formula: CH3(CH2)nCOO-
- consists of a hydrophobic hydrocarbon chain with a
terminal carboxyl group that ionizes at physiologic
pH hence its amphipathic nature

UST FMS MEDICAL BOARD REVIEW 2019 3 | BIOCHEMISTRY


GENERAL CONCEPTS, ENZYMES AND
COENZYMES BIOENERGETICS AND
BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

Triacylglycerols Sphingophospholipids
- 3 molecules of fatty acid esterified to a molecule of
glycerol - sphingomyelin: the only significant
- esterification of fatty acid results in loss of negative sphingophospholipid in human as it is an important
charge constituent of the myelin sheath
- “neutral fat”- stored in adipose tissue as oily
droplet (i.e.body’s major fuel storage reserve) - backbone is the amino alcohol sphingosine
- the fatty acid in carbon 1 is typically saturated
- the fatty acid in carbon 2 is typically unsaturated
- the fatty acid in carbon 3 can be either

Phospholipids
- polar, ionic molecules composed of an alcohol that
is attached by a phosphodiester bridge to either
diacylglycerol or spingosine Glycosphingolipids
- amphipathic with a hydrophilic head and long,
hydrophobic tail - aka glycolipids
- predominant lipids of cell membrane, component
of lung surfactant and bile - differ from sphingophospholipid in that they do not
- classes depend on the backbone: glycerol (i.e. contain phosphate and the polar head function is
glycerophospholipids) and sphingosine (i.e. provided by a monosaccharide or oligosaccharide
glycosphingolipids)
attached directly to the ceramide
Glycerophospholipids
- classified as either neutral or acidic
- aka phosphoglycerides
glycosphingolipids
- Phosphatidic acid acid is the simplest
phosphoglyceride; contains fatty acids esterified to
- the cerebrosides are the simplest neutral
carbons 1 and 2 of the glycerol moiety and a
phosphate group at carbon 3 glycosphingolipids with the alcohol group attached
to monosaccharides

- rest of phosphoglycerides are formed from


phosphatidic acid and an alcohol [e.g.
phosphatidylserine, phosphatidylethanolamine
(cephalin), phosphatidylcholine (lecithin),
phosphatidylglycerol, phosphatidylinositol]

UST FMS MEDICAL BOARD REVIEW 2019 4 | BIOCHEMISTRY


GENERAL CONCEPTS, ENZYMES AND
COENZYMES BIOENERGETICS AND
BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

(e.g. galactocerebrosides) or oligosaccharides - shell: unesterified cholesterol, phospholipid,


(i.e. globosides) apolipoproteins

- acidic glycosphingolipids are negatively Chylomicrons


charged at physiologic pH due to the presence -assembled in intestinal mucosal cells and carry
dietary triacylglycerol, cholesterol, and fat-soluble
of N-acetylneuraminic acid (NANA) in
vitamins to the peripheral tissues
gangliosides or by sulfate groups in sulfatides
VLDL
- gangliosides are derivatives of ceramide
-produced in the liver and transport endogenous
oligosaccharides and contain one or more NANA triacylglycerol to the peripheral tissues

LDL
- derived from VLDL
- provide cholesterol to the peripheral tissues
- oxidized LDL are taken up by macrophages,
transformed into foam cell which participate in the
development of atherosclerotic plaque

HDL
- formed in the blood by the addition of lipid to apo
A-1, an apolipoprotein made by the liver and
intestine
- reservoir of apolipoproteins (apo C-II and apo E)
- sulfatides (or sulfoglycosphingolipids) are and take up cholesterol from peripheral tissues and
return it to the liver
cerebrosides that contained sulfated galactosyl
residues II. ENZYMES
A. General Features of Enzymes
1. Biological catalysts that accelerate
chemical reactions by lowering the
energy of activation of the reaction.

Cholesterol
- structural component of cell membrane, precursor
of hormones, bile and vitamins
- consists of four fused hydrocarbon rings (A-D) and
has an eight-carbon, branched hydrocarbon
attached to carbon 17 of the D ring, a hydroxyl
group at carbon 3 of ring A and a double bond
between carbon 5 and 6 in ring B
2. Conjugated proteins consisting of
protein portion (apoenzyme) and
nonprotein portion (cofactor, coenzyme,
prosthestic group); Apoenzyme + non
protein portion = Holoenzyme
3. Enzyme specificity resides in the active
site, where substrate binds. Models of
active site:
a. Rigid Template theory (Lock and
Key)- substrate fits snuggly into a
fixed active site
Plasma lipoproteins b. Induced Fit theory (Flexible)-
- spherical macromolecules of lipid and enzyme undergoes conformational
apolipoproteins change to accommodate the
- chylomicrons, VLDL, LDL and HDL differ in lipid substrate into its active site.
and protein composition, size, density and site of 4. Classification of enzymes based on type
origin of reaction catalyzed. Nomenclature
- function to transport lipids (e.g. cholesterol, based on an Enzyme commission
triacylglycerol) in the plasma and transport their lipid number (4-digit numbers) depicting
content to (and from) the tissues general class, sub class and sub-
- lipid core: triacylglycerol and cholesteryl esters subclass and serial number in the list
UST FMS MEDICAL BOARD REVIEW 2019 5 | BIOCHEMISTRY
GENERAL CONCEPTS, ENZYMES AND
COENZYMES BIOENERGETICS AND
BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

D. Enzyme Kinetics Principles based on studies


General Class Function of Michaelis and Menten

I. Oxido- Oxidation-reduction reactions e.g. 1. Study of reaction rates with


reductase dehydrogenase varying concentration of substrate
2. Km and Vmax are kinetic parameters
2.Transferases Transfers functional groups e.g. that characterize enzymes
transaminases  Km is a measure of affinity of
the E to S (affinity inversely
3. Hydrolases Split molecules in the presence of prpoprtional to Km value); is
water. e.g. peptidases equal to the [S] at half Vmax
 Vmax- velocity (vo) when the
4. Lyases Split molecules non hydrolytically active site is fully saturated
with S
e.g. aldolases

5.Isomerases Interconvert isomeric molecules 3. Michaelis Menten Equation:


e.g. epimerases

6. Ligases Join molecules creating bonds


using energy e.g. synthases

1. Enzymes are also classified based on


chemical nature of enzymes
a. Enzyme complex
b. Allosteric enzymes
c. Inducible/repressible enzymes vs
constitutive enzymes

B. Mechanisms of Catalysis explain how


enzymes are able to lower energy of
activation and reach the transitions state.
1. Proximity and orientation effects 4. Lineweaver Burke Equation and Plot
2. Desolvation Effects a. Equation is derived from Michaelis
3. Acid Base catalysis Menten equation
4. Covalent catalysis
5. Strain or bond distortion effects
6. Metal coordination effects

C. Factors Affecting Enzyme Activity


1. pH- extremes of pH denature enzymes b. double reciprocal plot of Michaelis
2. Temperature – high temperatures Menten
denature enzymes and low temperature
inactivate enzyme
3. Enzyme concentration- activity increases
with [E]
4. Substrate concentration- velocity
increases to maximum velocity (Vmax)
representing substrate saturation of
active site of Enzyme
5. Inhibitors
a. Irreversible- Inhibitors become
tightly bound to the enzyme
b. Reversible:
 Competitive- Inhibitor binds
to active site can differentiate different types of inhibition
 Noncompetitive – Inhibitor
binds to another site other
than active site
 Uncompetitive- Inhibitor
only binds to E-S complex

UST FMS MEDICAL BOARD REVIEW 2019 6 | BIOCHEMISTRY


GENERAL CONCEPTS, ENZYMES AND
COENZYMES BIOENERGETICS AND
BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

1. Small organic molecules required for the


activity of the enzymes, usually involved in
the transfer of chemical groups.
2. Maybe bound loosely near the active site
and act as second substrates or may be
tightly bound to the enzyme as a prosthetic
group
3. Many are derived from vitamins.

B. Cofactors
1. Small inorganic ions required for proper
structure or to aid in catalysis
2. Metal ions may serve as metal ions bridges
between enzyme and substrate
(metalloenzymes), electron sinks, proton
donors (acids).

Inhibition Km Vmax Coenzyme Vitamin Function


Competitive increased same component
NAD, NADP niacin Redox reactions
Non same decreased (transfer of electrons
competitive and H+)
Uncompetitive decreased decreased FAD, FMN riboflavin Redox
(transfer of electrons
and H+)
E. Regulation of Enzyme activity Coenzyme none Redox (transfer of
Q (Ubiqui- electrons in ETC)
1. Allosteric regulation. Positive and negative none)
effectors bind to allosteric sites and can Tetrahy- none Redox (hydroxylation)
increase and decrease activity, respectively. drobiop-
Allosteric enzymes give sigmoidal curves. terin
2. Covalent modification. Covalent attachment Thiamine Thiamine Transfers of acyl or
of chemical groups to enzymes render Pyrophos- active aldehyde,
enzyme active or inactive, e.g. phate decarboxylation of 
Phosphorylation and dephoshorylation (TPP) keto acids
3. Stimulation and inhibition of control Lipoic acid none Transfer of acyl group
proteins. Formation of Calcium-calmodulin Coenzyme Pantothenic Acyl group transfer
complex activates certain enzymes A acid
4. Proteolytic activation Pyridoxal Pyridoxine Transamination,
Proenzymes or zymogens get activated phosphate decarboxylation,
when peptide bonds in the molecule are isomerization of amino
cleaved. E.g. activation of blood coagulation acids
proteins Biotin biocytin carboxylation
Tetrahy- Folic acid One carbon group
drofolate transfer
F. Applications of Enzymes in Medicine
Cobamide Cobalamine Alkyl group transfer
coenzyme (vitamin
1. Clinical diagnosis and prognosis. B12)
Cell death release enzymes in the circulation
e.g. LDH and CPK isoenzymes in myocardial IV. BIOENERGETICS AND BIOLOGICAL
infarction OXIDATIONS
2. Enzymes as therapeutic agents
Enzymes as medications for treatment of A. Bioenergetics and Concept of Free Energy
certain medical problems. e.g. streptokinase 1. Bioenergetics is the study of energy changes
for blood clots that accompany chemical reactions. (energy
requiring vs energy liberating reactions)
3. Immobilized enzymes as reagents in desk
2. Bioenergetics is governed by Laws of
top clinical analyzers. e.g. glucose oxidase
Thermodynamics:
blood glucose kits
a. First Law- conservation of energy
4. Enzymes as indicators in laboratory
b. Second Law- law on entropy
procedures. e.g. ELISA
B. Free Energy Change ( G)
III. COENZYMES AND COFACTORS
1. Represents amount of energy available for
A. Coenzymes useful work

UST FMS MEDICAL BOARD REVIEW 2019 7 | BIOCHEMISTRY


GENERAL CONCEPTS, ENZYMES AND
COENZYMES BIOENERGETICS AND
BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

2. Calculated quantitative value determines 2. Oxidative phosphorylation is the


type of reaction phosphorylation of ADP to ATP coupled with
a) Exergonic-  G < 0; spontaneous oxidation of substrates via the Electron
reaction, Transport Chain
b) Endergonic –  G > than 0; Non
spontaneous; energy requiring. May be D. The Electron Transport Chain
coupled with exergonic reaction to 1. The ETC is composed of enzymes (oxidases
proceed and dehydrogenases) and coenzymes as
c) Equibrium  G=0 electron carriers within the inner membrane
of the mitochondria
3.  G0 is the standard free energy change at
2. The ETC is organized into complexes
standard conditions of T, pressure, at 1M
arranged in a chain like fashion with oxygen
reactants.
as final acceptor of electrons to form water:
4. Quantitative value of DG of reaction
a. Complex I- NADH CoQ dehydrogenase
determines whether compound is high
b. Complex II- Succinate CoQ reductase
energy or low energy.
c. Complex III- Cytochrome b-c1 oxidase
a. High energy compounds –  G0 of -7 to
d. Complex IV – Cytochrome aa3 oxidase
-15 kcal/mole
e. Complex V- ATPase (ATP synthetase)
b. Low energy counds-  G0 less than -7 3. Substrates donate electrons to the ETC via 2
kcal/mole points:
a. Complex I- NADH CoQ dehydrogenase
Compounds Examples DG0 producing a P/O ratio of 2.5/1
(kcal/mole) equivalent to 3 ATPs produced
Phosphoric acid ATP -7.3 b. Complex II- Succinate dehydrogenase
anhydride producing a P/O ratio of 1.5/1
Thiol esters AcetylCoA -7.7 equivalent to 2 ATPs produced
4. Three sites of oxidative phosphorylation in
the ETC where Go > -7.3 kcal/mole
a. Complex I- site 1
b. Complex III- site 2
c. Complex IV – site 3

Phosphoguanides Creatine -10.3


phosphate

Phosphoric-carboxylic - 1,3 BPG -10.1


acid anhydrides - acetyl -10.3
phosphate
- carbamoyl -12.3
phosphate

Enol phosphates Phosphoenol -14


pyruvate 5. Mechanism of oxidative phosphorylation
based on Chemiosmotic Coupling Hypothesis
Phosphate Esters - AMP -3.4 of Peter Mitchell. The oxidation of substrates
- glucose-6 - 3.3 in the ETC is accompanied by release of
phosphate protons in the mitochondrial matrix creating
- glycerol–3 - 3.2 a proton gradient between the matrix and
phosphate intermembranous space, with the
intermembranous space becoming acidic.
Movement of protons from the
C. Cellular Synthesis of ATP intermembranous space back to the matrix,
1. Substrate level phosphorylation via Fo and F1 complex of Complex V
Is direct cleavage of high energy bonds, produces ATP
coupled to synthesis of ATP by transfer of 6. Substances that affect the ETC
high energy group to ADP a. Site/Complex specific
e.g. pyruvate kinase reaction:  Complex I- barbiturates, rotenone,
piericidin A, guanethidine
phosphoenol pyruvate + ADP  pyruvate +
 Complex II- malonate, carboxin
ATP

UST FMS MEDICAL BOARD REVIEW 2019 8 | BIOCHEMISTRY


GENERAL CONCEPTS, ENZYMES AND
COENZYMES BIOENERGETICS AND
BIOLOGICAL OXIDATION
EARL LOUIS A. SEMPIO, MD

 Complex III- antimycin A, phenformin,


dimercaprol
 Complex IV – carbon monoxide,
cyanide, azide, H2S
 Complex IV – Oligomycin binds to the
Fo fraction of ATPase
b. Non site specific
 Inhibitor of ADP-ATP carrier-
bongkrekic acid, atractyloside
 Uncouplers – dissociates respiration
(electron flow) from phosphorylation
e.g. 2,4 dinitrophenol, gramicidin,
dicoumarol, valinomycin, thermogenin

UST FMS MEDICAL BOARD REVIEW 2019 9 | BIOCHEMISTRY


REVIEW TEST
EARL LOUIS A. SEMPIO, MD

CHOOSE THE BEST ANSWER:


_____8. Which of the following is formed in the
_____1. Which of the following is an essential amino blood by addition of lipid to apo A-1?
acid? A. LDL
A. Glycine B. Chylomicrons
B. Valine C. VLDL
C. Alanine D. HDL
D. Serine
_____9. Which of the following is TRUE regarding
_____2. A polypeptide chain with an amino acid enzyme activity?
sequence of Pro-Gly-Glu would have which of the A. activation energy is higher with the
following properties at physiologic pH? enzyme
A. No net charge B. change in free energy is lower without
B. Negative charge the enzyme
C. Positive charge C. free energy of the reactants is always
D. Non-polar lower than free energy of products
D. reaction is facilitated by lowering
_____3. Which of the following defines the
quaternary structure of a protein? _____10. Which of the following is TRUE regarding
A. Spatial arrangement of subunits the activity of Lyases?
consisting of more than one polypeptide A. Split molecules in the presence of water
chain B. They join molecules by creating bonds
B. Unique 3-dimensional conformation of C. They transfer functional groups
the protein determined by its amino acid D. Non-hydrolytic splitting
sequence
C. May run parallel or antiparallel _____11. Enzymes are affected by temperature,
D. Consists of the sequence of amino acids which among the following statements correctly
along a polypeptide chain states the effect of temperature on enzymes?
A. The higher the temperature the slower
_____4. Which component of the amino acid leucine the reaction
is deprotonated at pH 7.4? B. Reaction rates are similar in high and low
A. alpha-carbon temperatures
B. alpha-carboxyl group C. High temperatures denature enzymes
C. alpha-amino group D. Low temperatures activate enzymes
D. R-group
_____12. In enzyme kinetics, which of the following
_____5. Which among the following is TRUE is considered as the measure of affinity of E to S
regarding carbohydrates? A. Vmax
A. Contain at least 3 carbon atoms B. Km
B. Empirical formula (CH2OH) C. V0
C. Should contain both aldehyde and D. ½ Vmax
ketone group
D. Have reducing property if the hydroxyl _____13. In which type of enzyme activity inhibition
group on the non-anomeric carbon is would both Km and Vmax be decreased?
not linked to another compound A. Competitive
B. Non-Competitive
_____6. Which of the following is TRUE regarding C. Uncompetitive
Glycosaminoglycans? D. Partly Competive
A. The positive charge is due to ionized
carboxyl and sulfate groups at _____14. Which of the following is defined as small
physiologic Ph organic molecules required for the activity of
B. They are long unbranched charged enzymes?
heteropolysaccharide chains A. Cofactors
C. They repel each other and bind water B. Coenzymes
due to the net positive charge at C. Minerals
physiologic pH D. Apoenzyme
D. repeating disaccharide units of [basic
sugar-carboxyl sugar _____15. Which if the following characterizes the
reaction facilitated by tetrahydrofolate?
_____7. These are the simplest neutral A. transfer of acyl group
glycosphingolipids with the alcohol group attached B. alkyl group transfer
to monosaccharides? C. one carbon transfer
A. cerebrosides D. hydroxylation
B. ceramides
C. sphingomyelin
D. sulfoglycosphingolipids

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


REVIEW TEST
EARL LOUIS A. SEMPIO, MD

_____16. Free energy change is


A. The amount of energy unavailable for work
B. less than 0 in exergonic reactions
C. is more than 0 in energy releasing reactions
D. is 1 at equilibrium

_____17. Which of the following is defined as the


phosphorylation of ADP to ATP coupled with
oxidation of substrates via the ETC?
A. Substrate level phosphorylation
B. Oxidative phosphorylation
C. ATP phosphorylation
D. Cytosolic-Mitochondrial Phosphorylation

_____18. Carbon monoxide affect the electron


transport chain through which complex?
A. Complex I
B. Complex II
C. Complex III
D. Complex IV

_____19. Which of the following is described as a


substance affecting the electron transport chain by
dissociating electron flow from phosphorylation?
A. Inhibitor
B. Antimycin
C. Uncoupler
D. Ubiquinol

_____20. Which of the following is responsible for


producing ATP?
A. Movement of protons from the matrix to
the intermembranous space
B. Sequestration of protons outside the
mitochondrial matrix
C. Creation of proton gradient between the
matrix and intermembranous space
D. Making the intermembranous space
basic

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


CARBOHYDRATE METABOLISM
JOSE S. BLAS, MD

INTRODUCTION GLYCOLYSIS
Glucose, the major dietary sugar is the
1. Glycolysis is the metabolic pathway utilized by the
universal source of fuel for human cells. It is likewise
cells to oxidize glucose to provide energy as ATP
the form of sugar brought in the circulation to
and intermediates of other metabolic pathways.
ensure a continuous supply of fuel.
2. The glycolytic pathway is central to carbohydrate
I. DIGESTION, ABSORPTION AND TRANSPORT metabolism because sugars, whether obtained from
OF CARBOHYDRATES the diet or from the breakdown of other substrates
1. Carbohydrates are the largest source of dietary in the body, can eventually be chemically converted
calories. Major dietary forms are starch, lactose and to glucose.
sucrose
3. The ten reactions of glycolysis that produces
2. Main sites for digestion are the mouth and pyruvate from glucose can be subdivided into the
duodenum. preparatory (energy investment phase) and pay-off
phase.
3. Salivary amylase acts on masticated food to break
some  1, 4 glycosidic bonds of starch. 4. The energy investment phase utilizes 2ATPS to
phosphorylate glucose and fructose and form 2
4. Pancreatic amylase in the duodenum completes
triose (DHAP and glyceraldehyde 3 Phosphate. The
the digestion and produces a mixture of
payoff phase oxidizes 2 molecules of glyceraldehyde
monosaccharides, disaccharides and
3 phosphate and generates ATP via substrate level
oligosaccharides.
phosphorylation and NADH which is a potential
5. Disaccharides are hydrolyzed into source of ATP when oxidized in the ETC (aerobic
monosaccharides by specific disaccharidases in the glycolysis).
brush border of the small intestines e.g.
5. Anaerobic condition regenerates NAD when
 Sucrose- isomaltase
pyruvate is reduced by LDH to lactate e.g.
 Lactase-glucosylceramidases
erythrocytes and exercising skeletal muscles.
 Trehalase
6. Many enzymes and intermediates of glycolysis
6. Uptake of monosaccharides by intestinal mucosal
operate in gluconeogenesis.
cells is mediated by various transporters
 Na dependent active transport for 7. Key enzymes in glycolysis include
glucose and galactose hexokinase/glucokinase, phosphofructokinase-1
 Facilitative diffusion for fructose and (PFK-1,) and pyruvate kinase which catalyze
mannose irreversible reactions, while the rest catalyze
reversible reactions. Irreversible reactions have
7. Transport of absorbed monosaccharides from
counterparts in gluconeogenesis.
intestinal cells into the blood and circulate to the
liver and peripheral tissues by facilitative Hexokinase Glucokinase
transporters (GLUT I to V) in specific tissues.
Most tissues Liver
8. Dietary fibers, (e.g. cellulose) are not digestible
by human intestinal enzymes due to inherent lack of (hexokinase I, II, III)
(hexokinase IV)
 1,4 glucosidases
Inhibited by Glucose- Not inhibited by
6-phosphate glucose-6-phosphate

II. Low Km (high High Km (lower


Affinity) affinity)

Not induced by insulin Induced by insulin

8. The key regulatory enzyme PFK-1 catalyzes the


committed and rate limiting step is allosterically
inhibited by ATP, citrate and phosphoenol pyruvate.
It is activated by AMP (indicating a low energy
charge) and Fructose 2,6 bisphosphate.
9. Pyruvate Kinase (PK) is activated by fructose 1,6
bisphosphate, the product of the PFK-1
 Covalent Modification thru Phosphorylation
by a Camp - dependent protein kinase leads
to inactivation of the hepatic isozyme of PK.
Dephosphorylation of PK by a phosphatase
results in reactivation of the enzyme.
 Glucagon and insulin can affect the levels of
cAMP and covalent modification of PK

UST FMS MEDICAL BOARD REVIEW 2019 1 | BIOCHEMISTRY


CARBOHYDRATE METABOLISM
JOSE S. BLAS, MD

10. Two moles of pyruvate are formed from a


molecule of glucose because the energy investment
phase generates 2 triose phosphates as the 2 moles
of glyceraldehyde 3 phosphate and continues in the
payoff phase.
11. Stoichiometry of energy production produces a
gross yield of 2 ATPs from 2 substrate level
phosphorylations with 2 ATPs used in the investment
phase producing a net of 2 ATPs
12. NADH generated can be oxidized in the ETC and
generates additional ATPs the amount depending on
the Shuttle System used:
 Malate Aspartate shuttle: 3ATPs / NADH
 Alpha glycerophosphate Shuttle: 2 ATPs
/NADH
13. The pyruvate dehydrogenase reaction (PDH) in
the mitochondria oxidatively decarboxylates
pyruvate into acetylCoA is a preparatory reaction to
the TCA Cycle for complete oxidation of glucose into
CO2 and H2O. PDH is an enzyme complex composed
of 3 enzymes:
E1- Pyruvate decarboxylase bound to TPP
E2- Dihydrolipoyl transacetylase bound to
lipoic acid and CoASH
E3- Dihydrolipoyl dehydrogenase III. TRICARBOXYLIC ACID CYCLE
bound to FAD and NAD The TCA Cycle or Citric Acid Cycle (Krebs cycle) is
the final common pathway for the oxidation of the
 NADH enters the ETC and generates 3 ATPs/NADH major macro-- molecules, carbohydrates, fats and
oxidized proteins. Their metabolism results in the formation
 PDH is inhibited by acetylCoA, NADH, ATP and by of Acetyl CoA or other intermediates of the cycle.
covalent modification by phosphorylation by PDH 1. Occurring in the mitochondrial matrix the cycle
kinase starts with the entry of acetylCoA which
condenses with oxaloacetate to form citrate,
followed by series of reactions that removes 2
carbon dioxide molecules by dehydrogenases
and generates reduced coenzymes NADH and
FADH2, GTP (via substrate level
phosphorylation) and regeneration of
oxaloacetate to complete the cycle.
2. Intermediates of the cycle include isocitrate,
a keto glutarate, succinylCoA, fumarate and
malate. These intermediates plus oxaloacetate
are points of entry of amino acid catabolites for
gluconeogenesis and can also be used as
precursors of amino acids and other
Alternate Fates of Pyruvate compounds. AcetylCoA can be a source of
carbons for fatty acid synthesis via citrate.
3. The TCA cycle is controlled by the regulation of
several enzymes: citrate synthase, isocitrate
dehydrogenase, and α- Keto glutarate:
a. NADH inhibits the dehydrogenase
reactions.
b. SuccinylCoA inhibits KG
dehydrogenase and citrate synthase
c. Ciitrate blocks citrate synthase
d. ATP inhibits citrate synthase and ADP
relieves inhibition
e. Ca++ activates KG dehydrogenase
and isocitrate dehydrogenase
4. Energetics of the TCA Cycle
a. Each turn of the TCA cycle
produces three NADH, one FADH2, one

UST FMS MEDICAL BOARD REVIEW 2019 2 | BIOCHEMISTRY


CARBOHYDRATE METABOLISM
JOSE S. BLAS, MD

GTP (or ATP) and two CO2 released in 2 PEP to 2 pyruvate 2 ATP 2
oxidative decarboxylation reactions and 2 Pyruvate to 2 Acetyl 2 NADH 6
regeneration of oxaloacetic acid CoA
b. Each NADH oxidized in the ETC 2 Isocitrate to 2 a- KG 2 NADH 6
produces 3 ATPs and each FADH2 2 a-KG to 2 succinyl CoA 2 NADH 6
oxidized in the ETC produces 2 ATP 2 Succinyl CoA to 2 2 ATP (or 2
c. One GTP molecule is equivalent to one succinate 2 GTP)
ATP produced by substrate level 2 Succinate to 2 2 FADH2 4
phosphorylation fumarate
d. A total of 12 ATPs produced per 2 Malate to 2 2 NADH 6
acetylCoA entering the cycle with 2 CO2 oxaloacetate
produced number is either 4 or 6, 36-38
depending Shuttle
- Malate Aspartate= 6
-  Glycero PO4 = 4

IV. GLUCONEOGENESIS

Gluconeogenesis is the synthesis of


glucose from non-carbohydrate sources in order to
provide glucose to glucose dependent tissues. The
liver is the major organ for gluconeogenesis and
kidneys being a minor organ

1. Substrates for gluconeogenesis


a. Lactate produced from exercising muscles
and red blood cells is transported to the liver
and kidneys and converted to pyruvate and
eventually to glucose. The cycle is called
Lactic Acid Cycle or Cori Cycle.
b. Glucogenic amino acids derived from
hydrolysis of tissue proteins are the major
sources of glucose when catabolized to any of
5. Anaplerotic Reactions replenish intermediates of
these intermediates: pyruvate, -Keto
the TCA cycle which are siphoned off to serve
glutarate, succinylCoA, fumarate and
as biosynthetic precursors in other pathways
oxaloacetate.
such as amino acid synthesis and fatty acid
c. Glycerol released from the hydrolysis of
synthesis via citrate. Important anaplerotic
triacylglycerol in adipose is delivered to the
reactions:
liver. Glycerol is phosphorylated to Glycerol 3-
a. Pyruvate carboxylase reaction by
PO4, which is oxidized by Glycerol 3-PO4
pyruvate carboxylase to replenish
dehydrogenase to DHAP, an intermediate of
oxaloacetate (OXAA) is the major
glycolysis and gluconeogenesis
anaplerotic reaction.
b. Catabolism of glucogenic amino acids
2. Reactions in glycolysis and gluconeogenesis
generates intermediates of the TCA Cycle.
are the same except for those irreversible
c. Purine nucleotide cycle supplies fumarate.
reaction in glycolysis which have counterparts
unique to gluconeogenesis. These reactions
Stoichiometry of Coenzyme reduction and ATP
bypass the irreversible steps of glycolysis:
Formation in the Aerobic Oxidation of Glucose via
a. Pyruvate carboxylase converts pyruvate
Glycolysis, PDH Complex Reaction, the TCA Cycle
to OXAA in the mitochondria by passes
and Oxidative Phosphorylation the ETC
pyruvate kinase (PK)
b. Phosphoenolpyruvate (PEP)
Number Number
carboxykinase converts OXAA to PEP in
of ATP or of ATP
the cytosol, requiring GTP- by passes
Reactions reduced formed
PK.
coenzyme
c. Fructose 1,6 bisPhosphatase hydrolyzes
e directly
Fructose1,6 bisP to Fructose 6 P in the
formed
cytosol (by passes PFK-1)
Glucose to Glucose-6- -1 ATP -1 d. Glucose 6 phosphatase converts Glucose
PO4 6 P to Glucose in the cytosol (by passes
Fructose-6-PO4 to -1 ATP -1 Glucose 6 phosphatase)
Fructose 1,6-bisPO4
2 Glyceraldehyde 3-PO4 2 NADH 4 or 6 *
to 2 1,3 BPG
2 1,3- BPG to 2 3-BPG 2 ATP 2
UST FMS MEDICAL BOARD REVIEW 2019 3 | BIOCHEMISTRY
CARBOHYDRATE METABOLISM
JOSE S. BLAS, MD

Glycolysis Gluconeogenesis hours). Muscle glycogen provides a quick source of


Hexokinase/gluco- Glucose- 6 phosphatase energy during vigorous activity (strenuous exercise).
kinase
Phosphofructokinase- Fructose 1,6 bis- 1. GLYCOGENESIS
1 phosphatase Glycogen synthesis takes place in the cytosol of
Pyruvate kinase - Pyruvate carboxylase virtually all animal tissues but is essentially
- PEP carboxykinase prominent in the liver and skeletal muscle. Enzymes
involved are Glycogen phosphorylase and branching
Enzyme
3. Regulation of Gluconeogenesis
a. Regulation by compartmentation. First a. UDP Glucose (derived from reaction of
reaction (pyruvate carboxylase) takes place Glucose 6 P + UTP) in the presence of
in the mitochondria and the rest in the glycogen synthase adds or transfers glucosyl
cytosol to prevent a futile cycle units to the non-reducing end of a branched
b. Hormonal Regulation by Glucagon- lowers glycogen molecule or glycogen primer and
the level of fructose 2,6-bisphosphate, forms an 1,4 glycosidic bond
favoring gluconeogenesis over glycolysis. b. Branching enzyme, Amylo (14) to (16
Glucagon increases the transcription of the transglycosylase or glycogen (46)
gene for PEP-carboxykinase. Decreased transferase creates the branch points of
insulin levels favor mobilization of amino glycogen through the formation of (16)
acids from muscle protein bond. It catalyzes the transfer of a terminal
c. Substrate availability of gluconeogenic fragment of 6 or 7 glucose residues from
cursors particularly glucogenic amino acids. the non-reducing end of a glycogen branch
d. Allosteric Regulation of Gluconeogenesis having at least 11 residues to the C-6 OH
o AMP, Fructose 2,6 bisP inhibit group of a glucose residue at a more interior
gluconeogenesis position of the same or another glycogen
o ATP (high energy charge) activate chain, thus creating a new branch. The
gluconeogenesis process is repeated by glycogen synthase
o High insulin/glucagon ratio inhibit and branching enzyme.
gluconeogenesis
GLYCOGENOLYSIS
Glycogen degradation occurs in the liver and
skeletal muscles involving the enzymes, glycogen
phosphorylase, debranching enzyme and,
phosphoglucomutase.

a. Glycogen phosphorylase hydrolyzes (


14) glycosidic linkage between two
glucose residues at a non-reducing end of
glycogen with an attack to an Inorganic PO4
(Pi), removing the terminal glucose residue
as glucose-1-PO4. Pyridoxal PO4 is an
essential cofactor in the glycogen
phosphorylase reaction.
b. Glycogen phosphorylase acts repetitively on
the non-reducing ends of glycogen branches
until it reaches a point four glucose residues
away from an ( 16) branch point where
its action stops.
c. Debranching enzyme also called oligo (
16) to ( 14) glucan-transferase
transfers a block of 3 glucosyl units to a
nearby non-reducing end to which they are
attached in  1,4 linkage. The  1,6
glucosidase activity of the enzyme releases
V. GLYCOGEN METABOLISM a free glucose from the branched point.
d. Phosphoglucomutase converts the glucose-1
In vertebrates, glycogen is formed primarily in PO released from phosphorylase action into
the liver and skeletal muscle, representing glucose 6-PO4. In the liver, glucose 6- PO4
approximately 20% and 1-2% of the weight of liver is transported into the endoplasmic
and of the weight of muscle, respectively. Liver reticulum (ER) by glucose 6-PO4
glycogen serves as a reservoir of glucose for other translocase. There it is converted to glucose
tissues when dietary glucose is not available by glucose phosphatase (same enzyme used
(between meals or during a fast; for about 10-18 in gluconeogenesis. The glucose is

UST FMS MEDICAL BOARD REVIEW 2019 4 | BIOCHEMISTRY


CARBOHYDRATE METABOLISM
JOSE S. BLAS, MD

transported from the ER to the cytosol.


Glucose 6-phosphatase, the same enzyme in
gluconeogenesis is inherently absent in
skeletal muscles.

3. REGULATION OF GLYCOGEN METABOLISM


a. Hormonal Regulation via Covalent
Modification of the Enzymes. The hormones
that affect activity of the enzymes include
glucagon, epinephrine and insulin
(Phosphorylation Dephosphorylation)
 Glucagon and epinephrine binds specific
hepatocyte G protein coupled receptors
(GPCRs) and activates adenylyl cyclase
which in turn catalyzes the synthesis of the
second messenger, cAMP.
 Cyclic AMP-dependent protein kinase A
(PKA) is activated and leads to activation of
phosphorylase kinase and glycogen
phosphorylase b to phosphorylase a
 Phosphorylation of glycogen phosphorylase
activates glycogenolysis and
dephosphorylation leads to inactivation of
glycogenolysis.
 Cyclic AMP is decreased phosphodiesterase
into 5’AMP and inhibits the phosphorylation
cascade of glycogenolysis. Insulin activates
phosphodiesterase opposing the effects of - Cascade Mechanism of Glucagon and Epinephrine
glucagon and epinephrine favoring b. Allosteric Regulation
glycogenesis and inhibiting glycogenolysis.  Glycogenesis is stimulated when substrate
 Protein kinase phosphorylates glycogen availability and energy levels are high,
synthase a (active) to glycogen synthase b whereas glycogenolysis is increased when
Glycogen synthase b can b reconverted to glucose and energy levels are low.
the active form by dephosphorylation.  Glycogen synthase b in both liver and
muscle is activated by glucose 6-P
 Glycogen phosphorylase a is
 inhibited by glucose 6-P as well as by ATP, a
high energy signal in the cell.
 In liver, but not muscle, free glucose is also
an allosteric inhibitor of glycogen
phosphorylase a.
 In muscle glycogen phosphorylase is active
in the presence of the high AMP.
 AMP binds to glycogen phosphorylase b,
causing Its activation with phosphorylation.
AMP also activates PFK-1 of glycolysis
allowing glucose from glycogenolysis to be
oxidized in the muscle

c. Activation of glycogen degradation by Calcium.


 Ca2+ is released into the cytoplasm in
muscle in response to neural stimulation
and in liver in response to epinephrine
binding to  1- adrenergic receptors.
 Calcium released from the sarcoplasmic
reticulum during contraction binds to
calmodulin and binds to Calmodulin. The
Ca- Calmodulin complex (Ca-CaM) activates
muscle phosphorylase kinase which
phosphorylates glycogen phosphorylase and
promote muscle glycogen degradation
producing glucose 6-P which then can enter
muscle glycolysis (Note: Epinephrine at β-

UST FMS MEDICAL BOARD REVIEW 2019 5 | BIOCHEMISTRY


CARBOHYDRATE METABOLISM
JOSE S. BLAS, MD

adrenergic receptors signals through a rise A particular enzyme may be defective in a single
in cAMP, not Ca2+) tissue, such as liver (resulting in hypoglycemia) or
muscle (causing muscle weakness), or the defect
may be more generalized, affecting a variety of
tissue. Important glycogen storage diseases include:

 Von Gierke’s (GS I) – Glucose 6


phosphatase deficiency
 Pompe’s Disease (GS II) – lysosomal
glucosidase deficiency
 Cori or Forbe’s (GS III) – Debranching
enzyme deficiency
 Andersen’s (GS IV), branching enzyme
deficiency
 McArdle’s (GS V) – muscle phosphorylase
deficiency
 Her’s (GS VI)- liver phosphorylase deficiency

 Calcium activation of liver phosphorylase kinase. VI. HEXOSE MONOPHOSPHATE


Epinephrine released during stress binds to 1 SHUNT (Pentose Phosphate Pathway)
adrenergic GPCR in hepatocytes leading to activation 1. Functions of the HMP Shunt The pentose
of phospholipase C that hydrolyzes PIP 2 to phosphate pathway is an alternative route for
diacylglycerol (DAG) and IP3. IP3 causes movement the metabolism of glucose. It does not lead to
of Ca++ from the sarcoplasmic reticulum and forms formation of ATP but has two major functions:
a Ca++-Calmodulin complex. DAG and Ca++ a. the formation of NADPH for reductive
stimulates Protein kinase C. synthesis of fatty acids, hydroxylation
Ca-Calmodulin complex stimulates phosphorylase reactions of steroids, detoxification
kinase and calmodulin dependent protein kinase. reactions.
These three kinases phosphorylate glycogen b. the synthesis of ribose phosphate for
synthase at different sites and decrease its activity nucleotide and nucleic acid formation
while increasing activity of glycogen phosphorylase
2. The HMP Shunt operates in 2 phases:
thereby promoting glycogenolysis. a. Irreversible Oxidative phase - Glucose 6-
P is oxidized by G-6P dehydrogenase,
the rate limiting step of the reaction
resulting in formation of NADPH, CO2
and ribulose 5 phosphate.
b. Reversible Non oxidative phase
consisting of series of sugar - phosphate
interconversions resulting in
isomerization of ribulose-5 phosphate to
ribose-5 phosphate and xylulose 5-
phosphate. Series of transketolation and
transaldolation yield fructose 6-
phosphate and glyceraldehyde 3
phosphate that may enter glycolysis or
converted back to glucose-6 phosphate
and re-enter the oxidative phase of the
HMP Shunt.

3. The regulation of the HMP Shunt is dependent on


the metabolic demand of the cell for NADPH for
reductive reactions and ribose phosphates for
nucleotide synthesis:
4. Glycogen Storage Diseases
a. NADPH is an inhibitor of G6PD and
increased demand for NADP stimulates the
These are a group of genetic diseases that are
oxidative phase via G6PD stimulation. e.g.
caused by defects in enzymes required for glycogen exposure to oxidizing agents
degradation or, more rarely, glycogen synthesis. b. Increased demand for ribose 5
They result either in formation of glycogen that has phosphates stimulates the non-oxidative
an abnormal structure or in the accumulation of phase
excessive amounts of normal glycogen in specific
tissues as a result of impaired degradation.

UST FMS MEDICAL BOARD REVIEW 2019 6 | BIOCHEMISTRY


CARBOHYDRATE METABOLISM
JOSE S. BLAS, MD

VI. Uronic Acid Pathway cells of peripheral nerves, liver, kidney,


placenta, RBCs, and cells of the ovaries and
1. In the liver, the Uronic acid pathway, like the
seminal vesicles. Sperm cells use fructose for
HMP Shunt is an alternative oxidative pathway
energy.
for glucose that does not lead to the formation
f. The pathway from sorbitol to fructose in the
of ATP.
liver provides a mechanism by which any
2. The initial reactions in the Uronic Acid is similar
available sorbitol is converted into a
to glycogenesis when glucose 6-phosphate is
substrate that can enter glycolysis or
somerized to glucose-1 phosphate. Glucose-1
gluconeogenesis.
phosphate is activated by UTP to form
g. The accumulation of sorbitol in cells due to
UDPGlucose. Oxidation of UDP Glucose forms
increase glucose and NADPH results in
UDPGlucuronic acid, the active form of
strong osmotic effects and, therefore, cell
glucuronic acid used in various reactions.
swelling as a result of water retention. Some
3. UDP Glucuronic acid is the source of glucuronate
of the pathologic alterations associated with
for reactions involving its incorporation into
diabetes can be attributed, in part, to this
proteoglycans or for reaction with substrates
phenomenon, including cataract formation,
such as steroid hormones, bilirubin, and a
peripheral neuropathy, and microvascular
number of drugs that are excreted in urine or
problems leading to nephropathy and
bile as glucuronide conjugates. UDPGlucose acid
retinopathy.
may be isomerized to UDP Galactose which is
used in the synthesis of lactose. 2. Galactose Metabolism
4. Glucuronate is reduced to L-gulonate, the direct a. The major dietary source of galactose is
precursor of ascorbate in those animals capable lactose (galactosyl β-1,4-glucose) obtained
of synthesizing this vitamin, in an NADPH- from milk and milk products. Some lactose
dependent reaction. In humans and other can also be obtained by lysosomal
primates, as well as guinea pigs, bats, and some degradation of complex carbohydrates, such
birds and fishes, -ascorbic acid cannot be as glycoproteins and glycolipids, which are
synthesized because of the absence of L- important membrane components. Like
gulonolactone oxidase. fructose, the transport of galactose into cells
5. L-gulonate continues with the pathway is not insulin dependent.
ultimately forming xylulose 5 phosphate to link b. Phosphorylation of galactose by
the pathway to the non-oxidative phase of the galactokinase to galactose -1- phosphate is
HMP shunt. the initial reaction followed by formation of
UDP galactose thru an exchange reaction
VII. Metabolism of other Monosaccharides with UDP Glucose catalyzed by galactose 1-
1. Fructose Metabolism phosphate uridyltransferase (GALT).
a. Fructose, is derived from the disaccharide Deficiency of this enzyme causes
sucrose which when cleaved in the intestine, galactosemia.
releases equimolar amounts of fructose and c. UDP Galactose can be epimerized to UDP
glucose. Fructose transport into cells is not Glucose and find its way in Glycogenesis.
insulin dependent (unlike that of glucose into UDP Galactose may be used as sugar donor
certain tissues), and, in contrast to glucose, in lactose synthesis.
fructose does not promote the secretion of d. In lactose intolerance, galactose cannot be
insulin. produced from lactose due to deficiency of
b. Fructokinase primarily phosphorylates lactase which leads to osmotic diarrhea and
fructose to fructose 1- PO4 because of its low flatulence, production of organic acids due
Km (high affinity for fructose) and high to lactose fermentation by intestinal
Vmax. Deficiency of fructokinase causes bacteria.
Essential fructosuria. Cleavage by aldolase b 3. Mannose Metabolism
forms DHAP and glyceraldehyde. Deficiency a. Mannose is initially phosphorylated to
aldolase b leads to Hereditary Fructose Mannose-6 P and then isomerized to
Intolerance. Fructose 6-P, which then enters the EMP.
c. DHAP enters glycolysis or gluconeogenesis b. GDP Mannose is used in substances
and glyceraldehyde upon phosphorylation to- requiring mannose.
glyceraldehyde 3-phosphate likewise enters
the glycolytic and gluconeogenetic pathway REFERENCES:
d. The rate of fructose metabolism is more  Greenstein, Ben, Greenstein Adam, Medical
Biochemistry at a Glance. University Press, Cambridge
rapid than that of glucose because the
1996
trioses formed from fructose 1-PO4 bypass
 Liebermann, Michael et al. Marks’ Basic Medical
PFK-1, the major rate-limiting step in Biochemistry, A Clinical Approach, 4th edition
glycolysis. Lippincott Williams and Wilkins, 2013
e. Fructose can be synthesized from other  Rodwell Victor, Bender David et al. Harper’s
monosaccharide (glucose) via the sorbitol Illustrated Biochemistry 31sr edition, International
pathway. Aldose reductase, which reduces edition. Lange.
glucose, producing sorbitol, is found in many  Wilcox. R. Bruce. High Yield Biochemistry. Lippincott
tissues, including the lens, retina, Schwann Williams and Wilkins.

UST FMS MEDICAL BOARD REVIEW 2019 7 | BIOCHEMISTRY


REVIEW TEST
JOSE S. BLAS, MD

CHOOSE THE BEST ANSWER:


_____ 8. The total number of ATPs produced per
_____ 1. Which of the following statements is
turn of the TCA Cycle per mole of glucose is:
correct of carbohydrate digestion?
A. 15
A. Salivary amylase acts on  1,4 and 1,6
B. 12
glycosidic bonds of starch
C. 30
B. Specific disaccharidases in the brush
D. 24
border of the intestines produce
monosaccharides as final products
_____ 9. The complete oxidation of a mole of
C. Cellulose are degraded into dextrins and
glucose-6- phosphate into carbon dioxide and water,
oligosaccharides upon digestion
using the Malate Aspartate Shuttle is:
D. The acidic pH of the stomach helps in
A. 36
digestion by denaturing dietary
B. 37
saccharides
C. 38
D. 39
_____ 2. The absorption and transport of glucose
across the lumen of the gastrointestinal tract is:
_____ 10. Which of the following reactions in the
A. Through facilitative diffusion
TCA Cycle is coupled with substrate level
B. Co-transported with sodium, coupled
phosphorylation?
with Na-K ATPase pump
A. malate dehydrogenase
C. Dependent on insulin
B. succinylCoA synthase (thiokinase)
D. Enhanced by phosphorylation of glucose
C. - keto glutarate dehydrogenase
to glucose -6 phosphate
D. succinate dehydrogenase
_____ 3. Hexokinase differs from glucokinase in
which of the following points? _____ 11. Which of the following is the major
A. Hexokinase is an insulin inducible anaplerotic reaction?
enzyme A. Fumarase
B. Glucokinase is inhibited by its product, B. Pyruvate carboxylase
glucose-6-phosphate C. Glutamate dehydrogenase
C. Hexokinase has a higher Km for glucose D. Pyruvate kinase
D. Glucokinase phosphorylates glucose
when glucose concentration is high _____ 12. Which of the following cannot be used as
substrate for glucose synthesis?
_____ 4. The rate limiting and committed step in A. Lactate
glycolysis is: B. Acetate
A. In the energy investment phase of C. Glycerol
glycolysis D. Pyruvate
B. catalyzed by phosphofructokinase-2
C. activated by ATP and a high energy _____ 13. All of the following enzymes are unique to
charge gluconeogenesis, EXCEPT?
D. coupled with substrate level A. Pyruvate carboxylase
phosphorylation B. PEP carboxykinase
C. Fructose 2,6 bisphosphatase
_____ 5. In the presence of limiting amounts of D. Glucose-6 phosphatase
oxygen, pyruvate is metabolized into:
A. Oxaloacetic acid _____ 14. Which of the following conditions will
B. acetylCoA favor gluconeogenesis over glycolysis?
C. Lactic acid A. High energy charge
D. Alanine B. Fructose 2,6 bisphosphate
C. Hyperglycemia
_____ 6. This glycolytic intermediate in the D. High insulin/ glucagon ratio
erythrocyte enhances the oxygen delivery to the
peripheral tissues: _____ 15. Which is the difference between liver
A. Phosphoenolpyruvate glycogen and muscle glycogen?
B. 1,3 bisphosphogylcerate A. Muscle glycogen concentration is higher
C. 2,3 bisphosphoglycerate per dry weight of tissue
D. dihydrohyacetone phosphate (DHAP) B. Liver glycogen maintains blood glucose
level during fasting
_____ 7. Which of the following is true of the C. Muscle glycogen is structurally different
pyruvate dehydrogenase reaction? from liver glycogen
A. A mitochondrial enzyme complex D. Liver glycogen synthesis involves
B. Active when covalently modified by addition of glucosyl units to nonreducing
phosphorylation end of glycogen primer
C. Starts the TCA Cycle
D. Catalyzes a reversible reaction

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


REVIEW TEST
JOSE S. BLAS, MD

_____ 16. Which of the following cellular conditions _____ 22. Which of the following is true of fructose
or scenarios is compatible with increased metabolism?
glycogenolysis and decreased glycogenesis? A. Enters glycolysis as dihydroxyacetone 3
A. Increased 3’5’ cAMP dependent protein phosphate
kinase activity B. Slower rate of metabolism compared to
B. Dephosphorylation of glycogen synthase glucose
enzyme C. Initially phosphorylated to fructose 6
C. Decreased glucagon/insulin ratio phosphate
D. Increased glucose -6-phosphate D. Intestinal absorption via Na-symport
system.
_____ 17. Epinephrine can promote liver
glycogenolysis upon binding to  adrenergic _____ 23. Galactose enters glucose metabolism as:
receptors and cause: A. Glucose- 6 phosphate
A. Increased cyclic AMP dependent protein B. Uridine Diphosphate Glucose (UDPG)
kinase activity C. Galactose -1- phosphate
B. Inositol triphosphate (IP3)- mediated D. Fructose 1,6 bisphosphate
calcium - Calmodulin stimulation of
phosphorylase kinase _____ 24. Mannose enters the mainstream of
C. Increase 5’ AMP concentration glycolysis thru:
D. Phosphorylation of glycogen synthase A. glucose-6 phosphate
enzyme B. fructose-6-phosphate
C. fructose-1-phosphate
_____ 18. Which of the following Glycogen Storage D. glucose-1-phosphate
diseases is characterized by fasting hypoglycemia,
acidosis, lipemia, hyperuricemia but with normal
liver glycogen structure?
A. Von Gierke’s disease
B. Andersen’s disease
C. Pompe’s disease
D. Mc Ardle’s Disease

_____ 19. The Hexose Monophosphate Shunt is


important in nucleotide metabolism because it
provides:
A. Ribose 5 phosphate
B. NADPH
C. C and N atoms in purine/pyrimidine ring
de novo synthesis
D. Intermediates for glycolysis

_____ 20. Intake of oxidant drugs by Glucose-6-


Phosphate dehydrogenase deficient patents leads to
hemolytic anemia. Decreased levels of which of the
following is a consequence of the enzyme deficiency
and the cause of hemolysis?
A. Glucose – 6-phosphate
B. NADP
C. Reduced glutathione
D. Ribose-5 phosphate

_____ 21. The important function of the Uronic Acid


Pathway in man:
A. Synthesis of ascorbic acid
B. Alternate pathway of glycolysis
C. Source of sugar acids for proteoglycan
D. Provision of sugar alcohols for synthesis
of monosaccharides

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


LIPID METABOLISM
JUDELYN T. UY, MD

GENERAL CHARACTERISTICS OF FATTY ACIDS


1. ‘Free’’ or esterified to glycerol
2. Usually have an even number of Carbon atoms,
(16 to 20), saturated or unsaturated (containing
double bonds).
3. Described by the number of carbons and the
positions of the double bonds (e.g., arachidonic
acid, which has 20 carbons and 4 double bonds,
is 20:4,Δ5,8,11,14).

Figure 2. The Citrate Cleavage Pathway.

C. Fatty Acid Synthesis


1. Glucose is the major precursor via acetyl CoA.
FATTY ACID SYNTHESIS
2. Synthesis involves a sequential addition of 2
A. Glucose is the Major Precursor for Fatty
carbon units, as Acetyl CoA, to the growing
Acid Synthesis (Figure 1)
chain.
1. Dietary carbohydrates in excess of that needed
for energy production and glycerol synthesis
3. Fatty acid synthase (FAS) is a multi-enzyme
is converted to fatty acids in the liver during
complex located in the cytosol composed of
the fed state.
two large identical subunits with 7 catalytic
activities.
2. Glucose provides the carbon (via acetyl-CoA)
for fatty acid synthesis and the reducing
4. This enzyme contains a phosphopantetheine
equivalents in the form of NADPH (pentose
residue, derived from the vitamin pantothenic
phosphate pathway) required for the process.
acid (B5), and a cysteine residue; both contain
sulfhydryl groups that can form thioesters with
acyl groups.

5. The growing fatty acyl chain moves during the


synthesis of two carbon units from one to the
other of these sulfhydryl residues as it is
elongated.

6. The initial step is the formation of Malonyl CoA


– rate limiting step catalyzed by Acetyl CoA
carboxylase (ACC; Figure 3 and 4)

Figure 1. Lipogenesis, the synthesis of fatty acids


(FA) and triacylglycerols (TAG) from glucose, occurs
Fig 3. Formation of Malonyl CoA by Acetyl CoA
mainly in the liver. (BRS, Biochemistry, Molecular
Carboxylase
Biology and Genetics 5th Edition)

B. The Citrate Cleavage Pathway provides


Acetyl CoA and NADPH for Lipogenesis in
the cytosol (Figure 2)
1. Citrate from the TCA is transported across the
mitochondrial inner membrane via the
tricarboxylate or citrate transporter. It is then
cleaved in the cytosol by ATP-citrate lyase
(citrate cleaving enzyme) to form acetyl CoA
and oxaloacetate.
Acetyl CoA carboxylase
2. The oxaloacetate formed is then reduced to is regulated by
Malate by the cytosolic form of NAD Malate phosphorylation and
dehydrogenase. Malate then undergoes allosteric controls
oxidative decarboxylation to pyruvate,
catalyzed by NADP Malate dehydrogenase
Figure 4. Regulation of Acetyl CoA Carboxylase
(malic enzyme).
(ACC)
UST FMS MEDICAL BOARD REVIEW 2019 1 | BIOCHEMISTRY
LIPID METABOLISM
JUDELYN T. UY, MD

7. Addition of two-carbon units (Figure 5) SYNTHESIS OF TRIACYLGLYCEROL (TAG)


a. Initially, a priming molecule of acetyl CoA A. In intestinal epithelial cells, TAG synthesis occurs
reacts with the phosphopantetheinyl by a different pathway than in other tissues
residue, and then the acetyl group is becoming a component of chylomicrons.
transferred to the cysteinyl residue. Ultimately, the fatty acyl groups are stored in
b. The acetyl group on the FAS complex adipose TAGs.
condenses with the malonyl group; the CO2
that was added to the malonyl group by B. In liver and adipose tissue, glycerol 3-PO4
- provides the glycerol moiety that reacts with 2
ketoacyl group, now containing four fatty acyl CoA molecules to form phosphatidic
carbons, is produced. acid. The phosphate group is cleaved to form a
diacylglycerol, which reacts with another fatty
acyl CoA to form a TAG.

1. The liver can use glycerol to produce glycerol


3-phosphate by a reaction that requires ATP
and is catalyzed by glycerol kinase.
2. Adipose tissue, which lacks glycerol kinase,
cannot generate glycerol 3-phosphate from
glycerol.
3. Both liver and adipose tissue can convert
Figure 5. Synthesis of Palmitic Acid glucose, through glycolysis, to DHAP, which is
reduced by NADH to glycerol 3-PO4 (Figure 7).
8. Reduction of the β-ketoacyl group TAG is stored in adipose tissue.
a. The β-ketogroup is reduced by NADPH to a 4. In the liver, TAG is incorporated into very low-
β-hydroxyl group. density lipoprotein (VLDL), entering blood.
b. Then dehydration occurs, producing a double Ultimately, fatty acyl groups are stored in
bond between carbons 2 and 3. adipose TAGs.
c. Finally, the double bond is reduced by 5. Fatty acids released from chylomicrons and
NADPH, and a four-carbon acyl group is VLDL by lipoprotein lipase are taken up by
formed. adipose cells and converted to TAGs, but
d. NADPH is generated from the pentose glycerol is not used because adipose tissue
phosphate pathway and malic enzyme. lacks glycerol kinase.
6. Glucose, transported into adipose cells as
9. Elongation of the growing fatty acyl chain influenced by insulin, is converted to DHAP
(Figure 6) and reduced by NADH to form glycerol 3-PO4,
a. The acyl group is transferred to the cysteinyl which produces the glycerol moiety of the
sulfhydryl group, and malonylCoA reacts TAG. The TAGs are stored in large fat globules
with the phosphopantetheinyl group. in adipose cells.
b. Condensation of the acyl and malonyl
groups releases CO2, followed by 3 reactions
reducing the β-keto group. The chain grows
by two carbons.
c. This sequence of reactions repeats until the
growing chain is 16 carbons in length.
d. Palmitate, a 16-carbon saturated fatty acid,
is the final product released by hydrolysis
from the fatty acid synthase complex.

Figure 7. Glycerol 3-PO4 from liver and adipose

7. In the fasted state in adipose tissue and liver,


glycerol 3-PO4 is derived from
glyceroneogenesis. The key enzyme in this
process is phosphoenolpyruvate (PEP)
carboxykinase, which is induced in the liver
and adipose tissue in the fasted state. PEP
carboxykinase converts oxaloacetate from TCA
to phosphoenolpyruvate (PEP). PEP is
converted to DHAP and then to glycerol-3-PO4
which is used for TAG synthesis. (Figure 8)
Figure 6. Elongation of the growing fatty acyl chain

UST FMS MEDICAL BOARD REVIEW 2019 2 | BIOCHEMISTRY


LIPID METABOLISM
JUDELYN T. UY, MD

4. Thiolysis - in the final step, 3-ketoacyl-CoA is


split at the 2,3 position by 3-ketoacyl-CoA-
thiolase, forming acetyl-CoA and a new acyl-
CoA which is two carbons shorter than the
original acyl-CoA molecule.

Each round of β-oxidation produces one mole of


NADH, one mole of FADH2 and one mole of acetyl-
CoA. The acetyl-CoA, the end product of each round
of β-oxidation, then enters the TCA cycle, where it is
further oxidized to CO2 with the concomitant
generation of three moles of NADH, one mole of
FADH2 and one mole of ATP. The NADH and FADH 2
Figure 8. Glyceroneogenesis in the liver and adipose generated during the lipid oxidation and acetyl-CoA
oxidation in the TCA cycle then can enter the
FATTY ACID OXIDATION respiratory pathway for the production of ATP. The
A. Initial step involves activation of the fatty acid in oxidation of fatty acids yields significantly more
the cytoplasm prior to oxidation in the energy per carbon atom than does the oxidation of
mitochondria. carbohydrates.

Fatty acid + ATP + CoA ----> Acyl-CoA + PPi


+AMP

B. Cytosolic fatty acyl CoA reacts with carnitine in


the outer mitochondrial membrane, forming fatty
acyl carnitine via carnitine acyl transferase I (CAT
I), also called carnitine palmitoyl transferase I
(CPT I). Fatty acyl carnitine passes to the inner
membrane, where it reacts with carnitine acyl
transferase II (CAT II) to reform fatty acyl CoA,
which enters the mitochondrial matrix. (Figure 9)

Figure 9. Transport of long chain Fatty Acyl CoAs in


mitochondria Figure 10. Beta (β)-Oxidation of even chain fatty
acids
C. Beta (β)-Oxidation of even chain fatty acids
is a four-step spiral (Figure 10)
KETONE BODY SYNTHESIS AND UTILIZATION
1. Dehydrogenation- the first step is the removal A. Occurs in liver mitochondria when fatty acids are
of two hydrogen atoms (dehydrogenation) in high concentration in the blood (during fasting,
catalyzed by acyl-CoA dehydrogenase and starvation, or as a result of a high-fat diet).
requiring FAD. This results in the formation of
Δ2-trans-enoyl-CoA and FADH2. B. Ketone bodies: (Figure 11)
a. Acetoacetate  Acetone
2. Hydration - water is then added to saturate b. β-hydroxybutyrate
the double bonds (hydration) and form 3-
hydroxyacyl-CoA, catalyzed by Δ2-enoyl-CoA C. STEPS:
hydratase. 1. Two molecules of acetyl CoA condense to
produce acetoacetyl CoA. This reaction is
3. Dehydrogenation - the 3-hydroxy derivative catalyzed by thiolase.
undergoes further dehydrogenation on the 3- 2. Acetoacetyl CoA and acetyl CoA form
carbon catalyzed by L-β-3-hydroxyacyl-CoA hydroxymethylglutaryl CoA (HMG-CoA) in a
dehydrogenase to form the corresponding 3- reaction catalyzed by HMG-CoA synthase.
ketoacyl-CoA compound. The coenzyme 3. HMG-CoA is cleaved by HMG-CoA lyase to
utilized in this reaction step is NAD+ which is form acetyl CoA and acetoacetate.
reduced to NADH + H+. 4. Acetoacetate can be reduced by an NAD-
requiring dehydrogenase (3-hydroxybutyrate
dehydrogenase) to 3-hydroxybutyrate (also

UST FMS MEDICAL BOARD REVIEW 2019 3 | BIOCHEMISTRY


LIPID METABOLISM
JUDELYN T. UY, MD

known as b-hydroxybutyrate). This is a


reversible reaction.

5. Acetoacetate is also spontaneously


decarboxylated in a nonenzymatic reaction,
forming acetone (the source of the odor on
the breath of ketotic diabetic patients).

Figure 12. Ketone Body Utilization

LIPID METABOLISM IN FASTING STATE


A. Mobilization of stored TAGs in the adipose is
initiated by hormone-sensitive lipase. This
enzyme is activated when it is phosphorylated by
cAMP-dependent protein kinase A. Conversely,
insulin inhibits the activity of this enzyme by
inducing dephosphorylation. (Figure 13)

B. Increase in epinephrine and glucagon elevates


cAMP and activates protein kinase A.
1. Adipose – activation of hormone sensitive
lipase and perilipin by phosphorylation
2. Liver – decrease in fatty acid synthesis due to
Figure 11. Ketone Body Synthesis inhibition of ACC via cAMP-dependent
phosphorylation
D. The liver lacks the enzyme needed to metabolize 3. Glycolysis – inhibited which decreases supply
ketone bodies (succinyl CoA-acetoacetate- CoA of acetyl CoA
transferase, so it cannot use the ketone bodies 4. Ketogenesis - stimulated due to increased FA
it produces. Therefore, acetoacetate and 3- oxidation and increased enzymes for
hydroxybutyrate are released into the blood by ketogenesis
the liver.

E. During starvation, ketone bodies released from


the liver in the blood increase to a level that
permits entry into brain cells, muscle and kidney
where they are oxidized.

F. Acetoacetate can enter cells directly, or it can be


produced from the oxidation of 3-
hydroxybutyrate by 3-hydroxybutyrate
dehydrogenase. NADH is produced by this
reaction and can generate adenosine
triphosphate (ATP).

G. Acetoacetate is activated by reacting with


Succinyl CoA to form acetoacetylCoA and
succinate. The enzyme is succinyl CoA-
acetoacetate-CoA transferase. Finally,
acetoacetyl CoA is cleaved by thiolase to form
two acetyl CoA’s, which enter the TCA cycle and Figure 13. Hormone-induced fatty acid mobilization
are oxidized to molecules of CO2. (Figure 12) in adipocytes

UST FMS MEDICAL BOARD REVIEW 2019 4 | BIOCHEMISTRY


LIPID METABOLISM
JUDELYN T. UY, MD

LIPID METABOLISM IN FED STATE


A. After a meal that contains lipids, carbohydrates F. The ring structure of cholesterol cannot be
and proteins, the dietary lipid is deposited as TAG degraded in the body. The bile salts in the feces
in adipose tissue. The dietary carbohydrates and are the major form in which the steroid nucleus is
proteins in excess of that required for energy or excreted.
for protein synthesis are converted into fatty
acids and deposited in adipose tissue as TAGs.

B. Insulin, the major anabolic hormone is required


for both fatty acid synthesis and for the formation
of TAG in adipose tissue. This hormone acts at
two levels:
1. Long term regulation
a. Promotes fatty acid synthesis through
increase in the levels of key enzymes in the
liver:
 Fatty acid synthase
 NADP-malate dehydrogenase (malic
enzyme)
 Acetyl CoA carboxylase
b. Stimulates synthesis of oxidative enzymes
in HMP to generate NADPH for fatty acid
synthesis.
c. Promotes glucose uptake via GLUT 4
transporter in adipose to provide glycerol-
3-PO4 for TAG synthesis

2. Short term regulation


a. Promotes activation of phosphoprotein Figure 14 Synthesis of Mevalonate
phosphatase in the liver, which activates
ACC through dephosphorylation.
b. Dephosphorylation of key enzymes in the
adipose inactivates the hormone sensitive
lipase and perilipin

CHOLESTEROL AND BILE SALT METABOLISM


A. Cholesterol is synthesized from cytosolic acetyl
coenzyme A (CoA) derived from Glucose. (Figure
14) Cytosolic acetyl CoA forms acetoacetyl CoA,
which condenses with another acetyl CoA to form
HMG-CoA.

B. Cytosolic HMG-CoA, a key intermediate in


cholesterol biosynthesis, is reduced in the
endoplasmic reticulum to mevalonic acid by the
regulatory enzyme HMG-CoA reductase.

C. HMG-CoA reductase is inhibited by cholesterol,


and phosphorylation by (AMP)-activated protein
kinase. In the liver, HMG-CoA reductase is also
inhibited by bile salts and is induced when blood
insulin levels are elevated. (Figure 16)

D. Mevalonic acid is phosphorylated and


decarboxylated to form the five-carbon (C-5)
isoprenoid, isopentenyl pyrophosphate. Two
isopentenyl pyrophosphate units condense, Figure 15a. Synthesis of Squalene
forming a C-10 compound, geranyl
pyrophosphate, which reacts with another C-5
unit to form a C-15 compound, farnesyl
pyrophosphate.

E. Squalene is formed from two C-15 units and then


oxidized and cyclized, forming lanosterol which is
converted to cholesterol in a series of steps.
(Figure 15a and 15b)
UST FMS MEDICAL BOARD REVIEW 2019 5 | BIOCHEMISTRY
LIPID METABOLISM
JUDELYN T. UY, MD

B. Chylomicrons are synthesized in intestinal


epithelial cells. Their triacylglycerols are derived
from dietary lipid, and their major apoprotein is
apo B-48.
1. Chylomicrons travel through the lymph into
the blood. Apo C-II, the activator of
lipoprotein lipase, and apo E are transferred to
nascent chylomicrons from HDL, and mature
chylomicrons are formed.

2. In peripheral tissues, particularly adipose and


muscle, the triacylglycerols are digested by
lipoprotein lipase. As the chylomicron loses
triacylglycerol, a chylomicron remnant is
formed. The chylomicron remnants interact
with receptors on liver cells and are taken up
by endocytosis. The contents are degraded by
lysosomal enzymes, and the products (amino
acids, fatty acids, glycerol, cholesterol, and
Figure 15b. Synthesis of Cholesterol phosphate) are released into the cytosol and
reused.

C. VLDL is synthesized in the liver, particularly after


a high-carbohydrate meal. It is formed from
TAGs that are packaged with cholesterol,
apoproteins (particularly apo B-100), and
phospholipids, and it is released into the blood.
1. In peripheral tissues, particularly adipose and
muscle, VLDL triacylglycerols are digested by
lipoprotein lipase, and VLDL is converted to
IDL.

2. IDL returns to the liver, is taken up by


endocytosis, and is degraded by lysosomal
enzymes. IDL can also be further degraded,
forming LDL.

Figure 16. Regulation of HMG CoA reductase activity D. LDL reacts with receptors on various cells, is
taken up by endocytosis, and is digested by
G. Bile acids are synthesized in the liver from lysosomal enzymes.
cholesterol through the rate-limiting step 1. Cholesterol, released from cholesterol esters
catalyzed by 7α-hydroxylase forming 7α-hydroxyl by a lysosomal esterase, can be used for the
cholesterol. synthesis of cell membranes or for the
synthesis of bile salts in the liver or steroid
H. The primary bile acids, chenodeoxycholic acid hormones in endocrine tissue.
and cholic acid are acted upon by bacteria and
converted to the secondary bile acids in the 2. Cholesterol inhibits HMG-CoA reductase and,
intestines. The secondary bile acids are thus, decreases the rate of cholesterol
deoxycholate (from cholate) and lithocholate synthesis by the cell.
(from chenodeoxycholate). Both primary and
secondary bile acids are reabsorbed by the 3. Cholesterol inhibits synthesis of LDL receptors
intestines and delivered back to the liver via the (downregulation) and, thus, reduces the
portal circulation. amount of cholesterol taken up by cells.

I. Bile salts are formed by conjugation of the 4. Cholesterol activates acyl: cholesterol
carboxyl groups of bile acids via an amide bond acyltransferase (ACAT), which converts
to either glycine or taurine prior to secretion into cholesterol to cholesterol esters for storage in
the bile canaliculi. cells.

LIPOPROTEIN METABOLISM (FIGURE 17) E. HDL is synthesized by the liver and released into
A. Four major groups of lipoproteins the blood as small, disk-shaped particles. The
1. CHYLOMICRONS major protein of HDL is apo A.
2. VLDL or (pre-β-lipoprotein) 1. ApoC-II, which is transferred by HDL to
3. LDL or (β-lipoprotein) chylomicrons and VLDL, serves as an activator
4. HDL or (α-lipoprotein) of lipoprotein lipase.

UST FMS MEDICAL BOARD REVIEW 2019 6 | BIOCHEMISTRY


LIPID METABOLISM
JUDELYN T. UY, MD

2. Apo E is also transferred and serves as a B. Degradation of phosphoglycerides (Figure


recognition factor for cell surface receptors. 18)
Apo C-II and apo E are transferred back to
HDL after digestion of triacylglycerols of
chylomicrons and VLDL.

3. Cholesterol, obtained by HDL from cell


membranes or from other lipoproteins, is
converted to cholesterol esters within the HDL
particle by the lecithin:cholesterol
acyltransferase (LCAT) reaction, which is
activated by apo A-I.

4. As cholesterol esters accumulate in the core of


the lipoprotein, HDL particles become
spheroidal. Figure 18. Degradation of Phosphoglycerides

C. Sphingolipidoses (Table 1)
5. HDL transfers cholesterol esters to other
lipoproteins in exchange for various lipids via
Cholesterol ester transfer protein (CETP). Table 1. Sphingolipidoses
VLDL and other lipoproteins carry the Disease Enzyme Major Symptoms
cholesterol esters back to the liver. Deficiency
GM1 GM1 Mental retardation, liver
6. HDL particles and other lipoproteins are taken Gangliosid β- enlargement, skeletal
up by the liver by endocytosis and hydrolyzed osis Galactosidas involvement, death by
by lysosomal enzymes. e age 2
Tay-Sachs Hexosamini- Mental retardation,
7. Cholesterol, released from cholesterol esters, Disease dase A blindness death by age
can be packaged by the liver in VLDL and 3
released into the blood or converted to bile Fabry’s α- Skin rash, kidney
salts and secreted into the bile. Disease Galactosidas failure, pain in lower
e extremities
Sandhoff’s Hexosamini Similar to Tay-Sach’s
Disease da-ses A Disease but more
and B rapidly progressing
Gaucher’s Glucocerebr Liver and Spleen
Disease osidase enlarge-ment, long
bone erosion, infantile
mental retardation
Niemann- Sphingomye Liver and Spleen
Pick li-nase enlarge-ment, mental
Disease retardation
Farber’s Ceramidase Painful and
Lipogranul progressively deformed
o-matosis joints, skin nodules,
early death
Krabbe’s Galactocere- Loss of myelin, mental
Disease brosidase retardation, death by
age 2
Sulfatide Arylsulfatas Mental retardation,
Lipidosis eA death in first decade
Figure 17. Transport of Cholesterol between Tissues
IMPORTANT ASPECTS OF EICOSANOID
IMPORTANT ASPECTS OF METABOLISM
PHOSPHOGLYCERIDE AND SPHINGOLIPID
METABOLISM A. Prostaglandins, Prostacyclins, and
A. Respiratory distress syndrome (RDS) of the Thromboxanes (Figure 19)
newborn occurs in premature infants due to a 1. Arachidonic acid, derived from membrane
deficiency of surfactant in the lungs, which leads phospholipids, is the major precursor for
to a decrease in lung compliance. Dipalmitoyl synthesis of the Prostaglandins.
phosphatidylcholine (DPPC, also called lecithin),
is the primary phospholipid in surfactant, which 2. The polyunsaturated fatty acid is cleaved from
lowers surface tension at the alveolar air–fluid the membrane phospholipid by phospholipase
interface. Surfactant is normally produced at A2, which is inhibited by the steroidal anti-
gestational week 30. inflammatory agents (steroids).

UST FMS MEDICAL BOARD REVIEW 2019 7 | BIOCHEMISTRY


LIPID METABOLISM
JUDELYN T. UY, MD

3. Oxygen is added, and a five-carbon ring is


formed by the enzyme cyclooxygenase, which
produces the initial prostaglandin. The initial
prostaglandin is converted to other classes of
prostaglandins and to the thromboxanes.
Aspirin, acetaminophen, and other NSAIDS
inhibit this isozyme of cyclo-oxygenase.

4. The prostaglandins are identified as PG and


the thromboxanes as TX. Prostaglandin PGI2 is
also known as prostacyclin. The subscript 2 in
each molecule refers to the number of -C=C-
present.

5. Certain prostacyclins (PGI2), produced by


vascular endothelial cells, inhibit platelet
aggregation, whereas certain thromboxanes Figure 20. Synthesis of the clinically relevant
(TXA2) promote platelet aggregation. Aspirin leukotrienes from arachidonic acid through the
has been shown to be cardioprotective in Lipoxygenase Pathway.
myocardial infarction. Although PGI2 is also
inhibited, the cardioprotective effect is REFERENCE:
mediated by inhibiting TXA2. SWANSON, et al. Basic Review Series Biochemistry,
Molecular Biology and Genetics 5th edition.
B. Leukotrienes (Figure 20)
1. Arachidonic acid, derived from membrane Lierberman, and Ricer. BRS Biochemistry, Molecular
phospholipids, is the major precursor for Biology and Genetics, 6th ed copyright 2014
synthesis of the leukotrienes.
Ferrier. Lippincott’s Illustrated Review Biochemistry
2. In the first step, oxygen is added by 6th ed copyright 2014
lipoxygenases, and a family of linear
molecules, hydroperoxyeicosatetraenoic acids Murray et al. Harper’s Illustrated Biochemistry, 29th
(HPETEs), is formed. A series of compounds, ed copyright 2012
comprising the family of leukotrienes, is
produced from these HPETEs. The Nelson and Cox. Lehninger Principles of Biochemistry
leukotrienes are involved in allergic reactions. 5th edition copyright 2008

3. The leukotrienes are identified as LT. The Nelson and Cox. Lehninger Principles of Biochemistry
leukotrienes, LTC4, LTD4, LTE4 and LTF4 are 6th edition
known as the peptidoleukotrienes because of
the presence of amino acids. The Lieberman and Mark’s Basic Medical Biochemistry, A
peptidoleukotrienes, LTC4, LTD4 and LTE4 are Clinical Approach, 3rd ed copyright 2009
components of slow-reacting substance of
anaphylaxis The subscript 4 in each molecule
refers to the number of -C=C- present.

Figure 19. Synthesis of the clinically relevant


prostaglandins and thromboxanes from arachidonic
acid through the Cyclooxygenase Pathway.

UST FMS MEDICAL BOARD REVIEW 2019 8 | BIOCHEMISTRY


REVIEW TEST
JUDELYN T. UY, MD

CHOOSE THE BEST ANSWER: 7. _____ The first dehydrogenation step in


betaoxidation involves:
1. _____ CH3(CH2)4CH=CHCH2CH=CH(CH2)7COOH A. Addition of water to trans Δ2 enoyl CoA
What can be deduced from the structural formula of B. Condensation of malonyl CoA with an acetyl
this fatty acid? CoA primer
A. Conjugated double bonds exist either in cis C. Formation of a double bond between the α
or trans configuration. and β carbons of acyl CoA
B. Delta system of nomenclature reads the D. Thiolytic cleavage β-ketoacyl CoA to yield
fatty acid as C18:2 Δ (9,12) acetyl CoA.
C. Hydrocarbon chain is entirely linear in
arrangement 8. _____ How many moles of acetyl CoA will be
D. Omega (Ω) carbon is also the carboxylic produced from the complete degradation of a 16-
carbon carbon fatty acid such as palmitic acid?
A. 7
2. _____ Which statement is true regarding Citrate B. 8
Cleavage Pathway in relation to lipogenesis? C. 9
A. Acetyl CoA generated from the pathway D. 10
enters the krebs cycle in the mitochondria
B. Citrate is transported and cleaved in the 9. _____ How many moles of ATP are generated by
cytosol to provide acetyl CoA for the complete oxidation of 1 mole of Palmitate?
biosynthesis of fats A. 35
C. It provides the reducing equivalents, NADH B. 96
+ H+ required for lipogenesis C. 129
D. Malic enzyme utilizes NADPH from the D. 131
hexose monophosphate shunt
10. _____ "Ketone Bodies" are produced:
3. _____ What is the activity of acetyl-CoA A. In the liver by the condensation of acetyl-CoA
carboxylase? and acetoacetyl-CoA
A. Controlled by the presence of citrate B. In the liver from α-ketoglutarate by excessive
B. Inhibited by carnitine stimulation of the TCA cycle
C. Increased by epinephrine and glucagon C. In the muscle as a result of the incomplete
D. Unregulated at all times oxidation of fatty acids
D. Only under abnormal conditions such as
4. _____ Which of the following statements correctly starvation and diabetes
describes lipogenesis?
A. Primary source of carbons for fatty acid 11. _____ Which metabolic process correctly
synthesis is glycerol describes Lipid metabolism when the cell is in the
B. Fatty acids are synthesized from acetyl CoA “Fed state”?
in the mitochondria A. Accelerated rate of fatty acid degradation
C. Fatty acid synthesis and esterification occurs B. Increased fatty acid and triacylglycerol
primarily in muscle cells synthesis in the liver and adipose
D. Fatty acyl chain on the fatty acid synthase C. Inhibition of fatty acid synthesis due
complex is elongated two carbons at a time inactivation of the rate limiting enzyme
D. Rapid utilization of ketone bodies by the
5. _____ Glyceroneogenesis is an important source extra-hepatic tissues
of glycerol-3-PO4 in the liver and adipose during
fasting. Which key enzyme is involved? 12. _____ Insulin affects Lipid metabolism by
A. Glycerol-3-phosphatase activation of this metabolic process:
B. Glycerol-3-phosphate dehydrogenase A. Fatty Acid mobilization from the adipocytes to
C. Glycerol kinase the brain and muscle cells
D. Phosphoenolpyruvate carboxykinase B. Glyceroneogenesis in the hepatocytes
C. Lipogenesis in the liver and adipose tissues
6. _____ Prior to betaoxidation, what does activation D. Triaclyglycerol hydrolysis in the adipose by
of fatty acids to fatty acyl-CoAs require? the Hormone Sensitive Lipase
A. Breakdown of long chain fatty acids into
short and medium chains 13. _____ HDL is considered as good cholesterol
B. Condensation with an acyl carrier protein because it:
(ACP) A. Inhibits formation of LDL from VLDL and IDL
C. Hydrolysis of ATP to AMP + PPi B. Promotes degradation of cholesterol into bile
D. Transport of free fatty acids into the inner acids
mitochondrial membrane C. Transports cholesterol from peripheral tissue
back to the liver
D. Transports cholesterol from the liver to the
peripheral tissues

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


REVIEW TEST
JUDELYN T. UY, MD

14. _____ The cholesterol lowering effect of the 21. _____ An 11-year-old Ashkenazi Jewish girl
Statins directly involves: presents with an enlarged liver and spleen, low
A. Decreased formation of foam cells by the white and red blood cell counts, bone pain, and
macrophages bruising. She is diagnosed with Gaucher disease.
B. Enhanced uptake of LDL-cholesterol by Which of the following compounds is accumulating
peripheral cells in her lysosomes?
C. Increased production of HDL in the liver A. Ceramide
D. Inhibits activity of HMGCoA reductase B. Galactocerebroside
C. Glucocerebroside
15. _____ Which lipoproteins are the major carriers D. Sphingosine
of triacylglycerols?
A. Chylomicrons and VLDL 22. _____ Aspirin is a nonsteroidal anti-inflammatory
B. HDL and LDL drug that inhibits cyclooxygenase, an enzyme
C. IDL and LDL required for the conversion of this compound from
D. VLDL and LDL arachidonic acid
A. HPETEs
16. _____ Apoprotein C-II is important in the B. Leukotrienes
metabolism of chylomicrons and VLDL because: C. Peptidoleukotrienes
A. Activates hepatic lipase D. Thromboxanes
B. Acts as ligand for LDL-receptor-mediated
endocytosis 23. _____ A 40-year-old woman has rheumatoid
C. Helps clear the lipoproteins into remnants by arthritis and was prescribed prednisone. What is the
lipoprotein lipase mechanism of action of this drug?
D. Promotes esterification of cholesterol into A. Inhibits conversion of arachidonic acid to
esters by LCAT epoxides
B. Inhibits phospholipase A2
17. _____ A 30-year-old man is diagnosed with an C. Promote activation of prostacyclins
acute middle cerebral artery stroke secondary to D. Promote leukotriene formation from HPETEs
atherosclerosis. Genetic studies show that he has
familial hypercholesterolemia, type II. Which defect
is seen in this type of hypercholesterolemia?
A. Defect in Apoprotein E2 synthesis
B. Deficiency of LDL receptor
C. Lipoprotein lipase deficiency
D. Overproduction of VLDL associated with
glucose intolerance

18. _____ A 25-year-old woman is diagnosed with


LCAT deficiency. LCAT is involved in which of the
following processes?
A. Converting cholesterol to cholesterol esters
B. Decreased uptake of cholesterol by
hepatocytes
C. Endocytosis of HDL particles into
hepatocytes
D. Transfer of cholesterol esters from HDL to
other lipoproteins

19. _____ What is the rate limiting step in the


biosynthesis of bile acids from cholesterol?
A. 7-α hydroxylation of the steroid nucleus
B. C-17 side chain cleavage to propionyl CoA
C. Conjugation with taurine and glycine
D. Esterification of C-3 OH group with a fatty
acid

20. _____ An infant born prematurely at 28 weeks is


diagnosed with respiratory distress syndrome due to
a deficiency of surfactant. Which type of
phospholipid is deficient in the surfactant of the
infant’s lung?
A. Dipalmitoyl phosphatidylcholine
B. Dipalmitoyl phosphatidylethanolamine
C. Dipalmitoyl phosphatidylglycerol
D. Dipalmitoyl phosphatidylserine

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

I. STRUCTURE, CLASSIFICATION AND


FUNCTIONS OF AMINO ACIDS E. The synthesis of new proteins
- requires amino acids. The primary source of
A. Structure amino acids is dietary protein. Breakdown of
tissue proteins also provides amino acids.

F. Amino acids provide nitrogen-containing


substrates for the biosynthesis of:
1. Non-essential amino acids
2. Purines and pyrimidines
3. Porphyrins
4. Neurotransmitters and hormones

G. The carbon skeletons of the surplus amino


acids not needed for synthetic pathways
serve as fuel. They may be:
1. Oxidized in the tricarboxylic acid (TCA)
B. Nomenclature cycle to produce energy.
2. Used as substrates for gluconeogenesis.
3. Used as substrates for fatty acid
synthesis.

II. DIGESTION OF PROTEINS

C. Classification

Essential Amino Acids: PVT TIM HALL


(Phenylalanine, Valine, Threonine, A. Proteolytic enzymes (proteases) break down
Tryptophan, Isoleucine, Methionine,
dietary proteins into their constituent amino
Histidine, Arginine*, Leucine, Lysine)
acids in the stomach and the intestine. Many
D. Architecture: Levels of Structure of these digestive proteases are synthesized as
larger, inactive forms known as zymogens.

B. In the stomach, pepsin begins the digestion of


proteins by hydrolyzing them to smaller
polypeptides. The contents of the stomach pass
into the small intestine, where enzymes
produced by the exocrine pancreas act. The
pancreatic proteases (trypsin, chymotrypsin,
elastase, and the carboxypeptidases) cleave
the polypeptides into oligopeptides and amino
acids.
UST FMS MEDICAL BOARD REVIEW 2019 1 | BIOCHEMISTRY
AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

C. Further cleavage of the oligopeptides to amino D. Transamination reactions are readily reversible
acids is accomplished by intestinal enzymes that and can be used in the synthesis or the
include aminopeptidases located on the brush degradation of amino acids.
border and other peptidases located within the
cells. Ultimately, the amino acids produced by E. Most amino acids participate in transamination
protein digestion are absorbed through the reactions. Lysine is an exception; it is not
intestinal epithelial cells and enter the transaminated.
blood.
F. Pyridoxal phosphate (PLP) serves as the
D. A large number of overlapping transport systems cofactor for transamination reactions. PLP is
exist for amino acids in cells: facilitative derived from vitamin B6.
transporters, sodium-linked tranporters,
IV. REMOVAL OF AMINO ACID AS AMMONIA
which allow the active transport of amino
acids into cells. - In comparison with carbohydrate and lipid
metabolism, the metabolism of amino acids
E. Defects in amino acid transport can lead to is complex. The body is concerned not only
disease. with the fate of the carbon atoms of the
amino acids but also with the fate of the
III. ADDITION AND REMOVAL OF AMINO nitrogen. During their metabolism, amino
ACID NITROGEN acids travel in the blood from one tissue to
another. Ultimately, most of the nitrogen is
A. Transamination involves the transfer of an converted to urea in the liver
amino group from one amino acid (which
is converted to its corresponding a-ketoacid) A. A number of amino acids undergo reactions in
to an a-ketoacid (which is converted to its which their nitrogen is released as ammonia
-amino acid). Thus, the (NH3) or ammonium ion (NH4+).
nitrogen from one amino acid appears in
B. Glutamate dehydrogenase catalyzes the
another amino acid.
oxidative deamination of glutamate. Ammonium
ion is released, and -ketoglutarate is formed.
The glutamate dehydrogenase reaction, which is
readily reversible, requires NAD or NADP.

B. The enzymes that catalyze transamination


reactions are known as transaminases or
aminotransferases.
Glutamate
C. C. Glutamate and -ketoglutarate are Dehydrogenase
often involved in transamination reactions,
-ketoacid
pairs. C. Histidine is deaminated by histidase to form NH 4
+
and urocanate.

D. Serine and threonine are deaminated by serine


dehydratase, which requires PLP. Serine is
converted to pyruvate, and threonine is
-ketobutyrate; NH4+ is released.

E. The amide groups of glutamine and asparagine


are released as ammonium ions by hydrolysis.
Glutaminase converts glutamine to glutamate
and NH4+. Asparaginase converts asparagine to
aspartate and NH4+.

F. The purine nucleotide cycle serves to release


NH4+ from amino acids, particularly in muscle.

UST FMS MEDICAL BOARD REVIEW 2019 2 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

1. Glutamate collects nitrogen from other amino VI. FATE OF AMINO ACID NITROGEN
acids and transfers it to aspartate by a
transamination reaction.

2. Aspartate reacts with inosine monophosphate


(IMP) to form adenosine monophosphate (AMP)
and generate fumarate.

3. NH4+ is released from AMP, and IMP is re-


formed.

V. ROLE OF GLUTAMATE

A. Deamination, the first step in metabolizing


surplus amino acids, yields an -keto acid
and an ammonium ion (NH4).

A. Glutamate provides nitrogen for synthesis B. Transdeamination effects deamination


through the sequential actions of the
of many amino acids.
enzymes transaminase
(aminotransferase) and glutamate
1. NH4+ provides the nitrogen for amino acid
dehydrogenase.
-
ketoglutarate to form glutamate in the C. The appearance of aspartate
glutamate dehydrogenase reaction. aminotransferase (AST) or alanine
aminotransferase (ALT) in the blood is an
2. Glutamate transfers nitrogen by indication of tissue damage, especially
-ketoacids cardiac muscle (AST) and liver (AST and
-amino ALT).
acids.
VII. UREA CYCLE AND DETOXIFICATION OF
B. Glutamate plays a key role in removing NH4+
nitrogen from amino acids.
A. NH4+ is toxic to the human body,
particularly the central nervous system
-Ketoglutarate collects nitrogen from
(CNS).
other amino acids by means of
transamination reactions, forming B. Conversion of NH4+ to urea occurs in the
glutamate. liver via the urea cycle. Urea is excreted in
the urine.
2. The nitrogen of glutamate is released as
NH4+ via the glutamate dehydrogenase C. The concentration of ammonia and
reaction. ammonium ions in the blood is normally
very low. Ammonia is in equilibrium with
3. NH4+ and aspartate provide nitrogen for ammonium ion (NH3 + H+  NH4+), with
urea synthesis via the urea cycle. a pKa of 9.3. NH3 is freely diffusible across
membranes, but at physiologic pH, the
Aspartate obtains its nitrogen from
concentration of ammonia is 1/100 the
glutamate by transamination of concentration of the NH4+ ion (Henderson-
oxaloacetate. Hasselbach equation).

D. Ammonia travels to the liver from other


tissues, mainly in the form of alanine and
glutamine. It is released from amino acids in
the liver by a series of transamination and
deamination reactions.

UST FMS MEDICAL BOARD REVIEW 2019 3 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

E. Ammonia is also produced by bacteria in the gut 2. Arginine stimulates the synthesis of N-
and travels to the liver via the hepatic portal vein. acetylglutamate from acetyl coenzyme A
The agent lactulose is used to treat this condition
(CoA) and glutamate.
and is thought to work to reduce ammonia levels by
either increasing bacterial assimilation of ammonia 3. Although the liver normally has a great
or reducing deamination of nitrogenous compounds. capacity for urea synthesis, the enzymes
of the urea cycle are induced if a high-
F. In peripheral tissues, detoxification of NH4+, protein diet is consumed for 4 days or
which is ultimately converted to urea in the liver, more.
occurs by different mechanisms. 4. The key relationship between the urea
1. In most tissues, the enzyme glutamine
cycle and the tricarboxylic acid (TCA)
synthetase incorporates NH4+ into
glutamate to form glutamine, cycle is that one of the urea nitrogen
which is carried by the circulation to molecules is supplied to the urea cycle
the liver. There the enzyme as aspartic acid, which is formed from
glutaminase hydrolyzes glutamine the TCA cycle intermediate oxaloacetic
back to NH4+ and glutamate. acid.
2. In skeletal muscle, sequential action of
the enzymes glutamate H. Hyperammonemia
dehydrogenase and glutamate-
pyruvate aminotransferase can 1. This condition may be caused by
lead to the incorporation of NH4+ insufficient removal of NH4+,
into alanine. The alanine is carried resulting from disorders that involve
to the liver, where one of the enzymes in the urea
transdeamination results in the cycle.
conversion of the alanine back to 2. Blood ammonia concentrations above the
pyruvate and NH4+. normal range (30-60 uM) may cause
coma due to ammonia intoxication.
G. Regulation of the Urea Cycle 3. Ammonia intoxication can lead to mental
retardation, seizure, coma, and
1. N-Acetylglutamate is an activator of death.
CPS I, the first enzyme of the urea 4. Enzyme defects:
cycle. a. When the activity of the enzyme
carbamoyl phosphate
synthetase or ornithine-
UST FMS MEDICAL BOARD REVIEW 2019 4 | BIOCHEMISTRY
AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

carbamoyl transferase is low,


ammonia concentrations in the
blood and urine rise, and
ammonia intoxication occurs.
b. When any of the enzymes
argininosuccinate synthetase,
argininosuccinase, or arginase is
defective, blood levels of the
metabolite immediately preceding
the defect increase. Ammonia
levels may also rise.
5. Treatment consists of restriction of
dietary protein, intake of mixtures of
keto acids that correspond to essential
amino acids and feeding benzoate and
phenylacetate to provide an alternate
pathway for ammonia excretion.

VIII. DEGRADATION OF AMINO ACIDS

UST FMS MEDICAL BOARD REVIEW 2019 5 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

The amino acids can be grouped into families based which is an enzyme that requires
on the point where their carbon skeletons, the PLP.
structural portions that remain after deamination,
enter the TCA cycle. c. Glycine, in a reversal of the reaction
used for its synthesis, reacts with
A. The amino acid carbon skeletons
methylene FH4 to form serine.
undergo a series of reactions whose
products may be glucogenic, - Glycine also reacts with FH4 and NAD+
ketogenic, or both.
to produce CO2 and NH4+.
Leu, Lys - Glycine can be converted to glyoxylate,
Degraded to acetyl-CoA.
Ketogenic which can be oxidized to CO2 and
Glucose cannot be made from
these. H2O, or converted to oxalate.
Glucogenic
Phe, Ile, Tyr, Trp, Thr d. Cysteine forms pyruvate. Its sulfur,
and
Goes both ways. which was derived from methionine,
Ketogenic
is converted to sulfuric acid
Everything else (H2SO4), which is excreted by the
Degraded to pyruvate or a
Glucogenic kidneys.
member of the TCA cycle
Glucose can be made from these.
e. Alanine can be transaminated to
pyruvate.
B. Degradation of amino acids
(Glucogenic) Amino acids that produce
pyruvate or intermediates of the TCA
cycle. These amino acids are considered
glucogenic because they can produce
glucose in the liver. The fumarate group
of amino acids produces cytoplasmic
fumarate. Potential mechanisms
whereby the cytoplasmic fumarate can
be oxidized.

1. Amino acids that are converted to pyruvate

- Glutamate can be deaminated by


glutamate dehydrogenase or
-
ketoglutarate.

- Glutamine is converted by glutaminase


to glutamate with the release of its
amide nitrogen as NH4+.

- Proline is oxidized so that its ring


opens, forming glutamate
semialdehyde, which is reduced to
glutamate.

a. The amino acids that are synthesized - Arginine is cleaved by arginase in the
from intermediates of glycolysis liver to form urea and ornithine.
(serine, glycine, cysteine, and Ornithine is transaminated to
alanine) are degraded to form glutamate semialdehyde, which is
pyruvate. oxidized to glutamate.

b. Serine is converted to 2- - Histidine is converted to


phosphoglycerate, an intermediate formiminoglutamate (FIGLU). The
of glycolysis, or directly to pyruvate formimino group is transferred to
and NH4+ by serine dehydratase, - FH4, and the remaining five carbons
form glutamate.

UST FMS MEDICAL BOARD REVIEW 2019 6 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

c. Amino acids that form succinyl CoA group from the FH4 pool via vitamin
B12.
- Four amino acids (threonine,
methionine, valine, and - Homocysteine can also react with
isoleucine) are converted to serine to form cystathionine. The
propionyl CoA. cleavage of cystathionine produces
cysteine, NH4+, and a-ketobutyrate,
- Propionyl CoA is carboxylated in a which is converted to propionyl CoA.
biotin-requiring reaction to
formmethylmalonyl CoA. (3) Valine and (4) isoleucine, two of the
three branched-chain amino acids,
- Methylmalonyl CoA is rearranged to form succinyl CoA:
form succinyl CoA in a reaction
that requires vitamin B12. - Degradation of all three branched-
chain amino acids begins with a
transamination, followed by an
oxidative decarboxylation catalyzed
by the branched- -ketoacid
dehydrogenase complex. This
enzyme, like pyruvate
-ketoglutarate
dehydrogenase, requires thiamine
pyrophosphate, lipoic acid, CoA,
flavin adenine dinucleotide (FAD),
and NAD+.

- Valine is eventually converted to


succinyl CoA via propionyl CoA and
methylmalonyl CoA.

- Isoleucine also forms succinyl CoA after two of its


carbons are released as acetyl CoA.

(1) Threonine is converted by a


dehydratase to NH4+ -
ketobutyrate, which is oxidatively
decarboxylated to propionyl CoA. In
a different set of reactions,
threonine is converted to glycine
and acetyl CoA.

(2) Methionine provides methyl groups for


the synthesis of various compounds;
d. Amino acids that form fumarate
its sulfur is incorporated into
cysteine; and the remaining carbons (1) Three amino acids (phenylalanine,
form succinyl CoA. tyrosine, and aspartate) are
converted to fumarate
- Methionine and ATP form S-
adenosylmethionine (SAM), which (2) Phenylalanine is converted to
donates a methyl group and forms tyrosine by phenylalanine
homocysteine. hydroxylase in a reaction requiring
tetrahydrobiopterin and O2.
- Homocysteine is reconverted to
methionine by accepting a methyl-
UST FMS MEDICAL BOARD REVIEW 2019 7 | BIOCHEMISTRY
AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

(3) Tyrosine, which is obtained from the c. Leucine is degraded to form both
diet or by hydroxylation of acetyl CoA and acetoacetate.
phenylalanine, is converted to
D. Generalities of Amino Acid Catabolism
homogentisic acid. The aromatic
ring is opened and cleaved, forming - If a vitamin or cofactor is involved in amino
fumarate and acetoacetate. acid metabolism, it’s most likely pyridoxal
phosphate (B6), unless it involves serine,
(4) Aspartate is converted to fumarate and then its B6 and folic acid.
through reactions of the urea cycle - Nitrogen is dumped into the urea cycle by
and the purine nucleotide cycle. transamination to make Asp or Glu or by
Aspartate reacts with IMP to form deamination to make ammonia.
AMP and fumarate in the purine
IX. SYNTHESIS OF NON-ESSENTIAL AMINO
nucleotide cycle.
ACIDS
e. Amino acids that form oxaloacetate
Synthesis of the amino acids: Eleven of the
twenty common amino acids can be synthesized
(1) Aspartate is transaminated to form
in the body. The other nine are considered
oxaloacetate. “essential” and must be obtained from the
diet. Almost all of the amino acids that can be
(2) Asparagine loses its amide nitrogen as
synthesized by humans are amino acids used for
NH4+, forming aspartate in a reaction the synthesis of additional nitrogen-containing
catalyzed by asparaginase. compounds. Examples include glycine, which is
used for porphyrin and purine synthesis;
C. Degradation of amino acids (Ketogenic) glutamate, which is required for
neurotransmitter and purine synthesis; and
Amino acids that produce acetyl CoA or aspartate, which is required for both purine and
ketone bodies. These amino acids are pyrimidine biosynthesis.
considered ketogenic. Nine of the eleven “nonessential” amino acids
can be produced from glucose plus, of course, a
source of nitrogen, such as another amino acid
or ammonia. The other two nonessential amino
acids, tyrosine and cysteine, require an
essential amino acid for their synthesis
(phenylalanine for tyrosine, and methionine for
cysteine). The carbons for cysteine synthesis
come from glucose; the methionine only
donates the sulfur.

1. Messenger RNA contains codons for 20 amino


acids. Eleven of these amino acids can
be synthesized in the body. The carbon
skeletons of 10 of these amino acids can
be derived from glucose. These 10 are
serine, glycine, cysteine, alanine,
glutamic acid, glutamine, aspartic
acid, asparagine, proline, and
arginine. The essential amino acids
derived from diet are histidine,
isoleucine, leucine, lysine, methionine,
1. Acetyl CoA family (also called the phenylalanine, threonine, tryptophan,
ketogenic amino acid family) and valine. Note that tyrosine is derived
[isoleucine, leucine, lysine, from phenylalanine.
phenylalanine, tryptophan, and tyrosine]
2. Amino acids derived from intermediates of
glycolysis
a. Acetyl CoA is the starting point for
ketogenesis but cannot be used for
net gluconeogenesis. Leucine and
lysine are the only ketogenic
amino acids. The other four amino
acids that form acetyl CoA are both
ketogenic and glucogenic.
b. Phenylalanine and tyrosine form
acetoacetate.

UST FMS MEDICAL BOARD REVIEW 2019 8 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

a. Intermediates of glycolysis serve as


precursors for serine, glycine,
cysteine, and alanine.
b. Serine can be synthesized from the
glycolytic intermediate 3-
phosphoglycerate, which is oxidized,
transaminated by glutamate, and
dephosphorylated.
c. Glycine and cysteine can be derived
from serine.
(1) Glycine can be produced from serine
by a reaction in which a methylene
group is transferred to
tetrahydrofolate (FH4).
(2) Cysteine derives its carbon and
nitrogen from serine. The essential
amino acid methionine supplies the
sulfur.
d. Alanine can be derived by
transamination of pyruvate.

3. Amino acids derived from TCA cycle


intermediates

UST FMS MEDICAL BOARD REVIEW 2019 9 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

Glu  Glu-semialdehyde 
Arg
ornithine  Arg
Pro Glu  Glu-semialdehyde  Pro
Phe  Tyr (phenylalanine
Tyr
hydroxylase)
Met  homoCys + Ser 
Cys
cystathionine  Cys

The regulation of individual amino acid


biosynthesis can be quite complex, but the
overriding feature is that the pathways are
feedback regulated such that as the
concentration of free amino acid
increases, a key biosynthetic enzyme is
allosterically or transcriptionally inhibited.
Amino acid levels, however, are always
maintained at a level such that the aminoacyl-
a. Aspartate can be derived from
tRNA synthetases can remain active, and protein
oxaloacetate by transamination.
synthesis can continue.
b. Asparagine is produced from
X. CLINICAL RELEVANCE: INHERITED
aspartate by amidation.
(INBORN) ERRORS OF AMINO ACID
METABOLISM
c. Glutamate -
ketoglutarate by the addition of NH4+
A. Phenylketonuria (PKU)
via the glutamate dehydrogenase
1. Phenylalanine accumulates in the
reaction or by transamination.
blood (hyperphenylalaninemia).
Glutamine, proline, and arginine
a. Phenylalanine builds up to toxic
can be derived from glutamate.
concentrations in body fluids,
resulting in CNS damage with
(1) Glutamine is produced by amidation
mental retardation.
of glutamate.
b. Elevated phenylalanine inhibits
(2) Proline and arginine can be derived
melanin synthesis, leading to
from glutamate semialdehyde,
hypopigmentation.
which is formed by reduction of
2. This condition results from a deficiency
glutamate.
of phenylalanine hydroxylase or
(3) Proline can be produced by
dihydropterine reductase
cyclization of glutamate
3. An alternative pathway for phenylalanine
semialdehyde.
breakdown produces
(4) Arginine, via three reactions of the
phenylketones (phenylpyruvic,
urea cycle, can be derived from
phenyllactic, and phenylacetic
ornithine, which is produced by
acids), which spill into the urine.
transamination of glutamate
4. In affected individuals, tyrosine is an
semialdehyde.
essential dietary amino acid.
5. Treatment consists of restricting dietary
4. Tyrosine, the 11th nonessential amino acid,
protein (phenylaknine).
is synthesized by hydroxylation of the
essential amino acid phenylalanine in a
B. Albinism
reaction that requires
1. Tyrosinase, the first enzyme on the
tetrahydrobiopterin.
pathway to melanin, is absent.
2. Albinos have little or no melanin (skin
Summary of Synthesis of Non-Essential Amino
pigment). They sunburn easily,
Acids
and are:
Amino Acid Synthetic Route
a. Particularly susceptible to skin
Ala from pyruvate by transamination
carcinoma.
-ketoglutarate by
Glu b. Photophobic because of lack of
transamination
pigment in the iris of the eye.
Asp from oxaloacetate by transamination
Gln Glu + NH4 + ATP  Gln C. Homocystinuria
Asp + Gln + ATP  Asn + 1. In this disorder, homocysteine, which
Asn
AMP + PPi + Glu accumulates in blood and body
Glucose  hydroxypyruvate  Ser; fluids, appears in the urine.
Ser Glucose  phosphohydroxypyruvate 2. Homocystinuria may result from several
 Ser defects
Ser + THfolate  Gly + Ch2- a. Cystathionine synthase deficiency
Gly
THfolate b. Reduced affinity of cystathionine
UST FMS MEDICAL BOARD REVIEW 2019 10 | BIOCHEMISTRY
AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

synthase for its coenzyme, 2. Creatine travels from the liver to other
pyridoxal phosphate (PLP) tissues, where it is converted to creatine
[This form may respond to phosphate. Adenosine triphosphate (ATP)
megadoses of pyridoxine
phosphorylates creatine to form creatine
(vitamin B6).]
5
c. N -Methyl tetrahydrofolate phosphate in a reaction catalyzed by
homocysteine creatine kinase (CK).
methyltransferase deficiency
d. Vitamin B12 coenzyme a. Muscle and brain contain large amounts of
(methylcobalamin) deficiency creatine phosphate.
[This form may respond to
vitamin B12 supplements] b. Creatine phosphate provides a small
3. Pathologic changes reservoir of high-energy phosphate that
a. Dislocation of the optic lens readily regenerates ATP from adenosine
b. Mental retardation diphosphate (ADP). It plays a particularly
c. Osteoporosis and other skeletal
important role during the early stages of
abnormalities
d. Atherosclerosis and thromboembolism exercise in muscle, where the largest
4. Patients who are unresponsive to vitamin quantities of creatine phosphate are found.
therapy may be treated with
synthetic diet low in methionine, c. Creatine also transports high-energy
and by administering betaine (N, phosphate from mitochondria to actomyosin
N, N-trimethylglycine) as an fibers.
alternative methyl group donor.
3. Creatine phosphate spontaneously cyclizes,
D. Maple-syrup urine disease forming creatinine, which is excreted by
1. In this disorder, the branched chain keto the kidney.
acids derived from isoleucine,
leucine and valine appear in the B. Glutathione
urine, giving it a maply syrup-like
odor. 1. Structure
2. This condition results from a deficiency in
the branched-chain 2-keto acid - GSH is a tripeptide synthesized from
decarboxylase. glutamate, cysteine, and glycine. It contains
3. The elevated keto acids cause severe
an unusual linkage between the glutamate
brain damage, with death in the first
year of life. side-chain carboxylate group and the
4. Treatment. A few cases respond to nitrogen of cysteine.
megadoses of thiamine (vitamin B1).
Otherwise, synthetic diets low in
branched-chain amino acids are
given.

E. Histidinemia
1. This disorder is characterized by elevated
histidine in the blood plasma and
excessive histidine metabolites in the
urine.
2. The enzyme histidine- -deaminase,
the first enzyme in histidine
catabolism, is deficient.
3. Mental retardation and speech defects
may occur but are rare.
4. Treatment is not usually indicated.

XI. SPECIAL PRODUCTS DERIVED FROM


AMINO ACIDS

A. Creatine
2. Function
1. Creatine is produced from glycine, arginine,
a. Involved in the transport of amino acids
and S-adenosylmethionine (SAM). Glycine
across the cell membranes (the c-
combines with arginine to form ornithine -
glutamyl cycle)
and guanidinoacetate, which is methylated
by SAM to form creatine.

UST FMS MEDICAL BOARD REVIEW 2019 11 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

b. Aids in the rearrangement of protein D. Products formed by amino acid


disulfide bonds under oxidizing decarboxylations
conditions and detoxification reactions
1. Amines are produced by decarboxylation of
(1) The sulfhydryl groups of GSH are amino acids in reactions that use pyridoxal
used to reduce oxidized proteins, phosphate (PLP) as a cofactor.
resulting in the oxidation of two
molecules of GSH to form GSSG 2. -Aminobutyric acid (GABA), an inhibitory
(two glutathione molecules linked by neurotransmitter, is produced by
a disulfide bond). decarboxylation of glutamate

(2) GSSG is reduced back to two


molecules of GSH through the
action of glutathione reductase, an
NADPH-requiring enzyme.

C. Nitric Oxide (NO)


1. Synthesis

a. Liberated in the conversion of L-arginine


3. Histamine is produced by decarboxylation of
to citrulline
histidine.
b. The enzyme nitric oxide synthase (NOS)
a. Histamine causes vasodilation and
is a complex enzyme requiring NADPH,
bronchoconstriction.
flavin adenine dinucleotide (FAD), flavin
mononucleotide (FMN), and b. In the stomach, it stimulates the
tetrahydrobiopterin (BH4). secretion of hydrochloric acid (HCl).
c. NOS is found in three major isoforms. 4. The initial step in ceramide formation
involves the condensation of palmitoyl
(1) Neuronal NOS (nNOS or NOS-1).
coenzyme A (CoA) with serine, which
(2) Macrophage or inducible NOS (iNOS undergoes a simultaneous decarboxylation.
or NOS-2). Ceramide forms the sphingolipids (e.g.,
sphingomyelin, cerebrosides, and
(3) Endothelial NOS (eNOS or NOS-3). gangliosides).

2. Function 5. The production of serotonin from tryptophan


and of dopamine from tyrosine involves
a. iNOS is important in macrophages for
decarboxylations of amino acids.
creating NO for the generation of free
radicals, which are bactericidal. E. Products derived from tryptophan

b. NO stimulates the influx of Ca2+ into 1. Serotonin, melatonin, and the


vascular endothelial cells, with the nicotinamide moiety of NAD and NADP are
activation of cyclic guanosine formed from tryptophan
monophosphate (cGMP) resulting in
relaxation of vascular smooth muscle
(NO is also known as endothelium-
derived relaxation factor [EDRF]).

UST FMS MEDICAL BOARD REVIEW 2019 12 | BIOCHEMISTRY


AMINO ACID METABOLISM
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

H. S-Adenosylmethionine (SAM)

- synthesized from methionine and ATP

- supplies methyl groups

2. Tryptophan is hydroxylated in a BH4-requiring


reaction similar to the hydroxylation of
phenylalanine. The product, 5-
hydroxytryptophan, is decarboxylated to
form serotonin.

3. Serotonin undergoes acetylation by acetyl


CoA and methylation by SAM to form
melatonin in the pineal gland.

4. Tryptophan can be converted to the


nicotinamide moiety of NAD and NADP
although the major precursor of
nicotinamide is the vitamin niacin (nicotinic
acid). Thus, to a limited extent, tryptophan
can spare the dietary requirement for niacin.

F. Products derived from phenylalanine and


tyrosine

1. Phenylalanine can be hydroxylated to form


tyrosine in a reaction that requires BH4.
Tyrosine can be hydroxylated to form dopa
(3,4-dihydroxyphenylalanine)

2. Thyroid hormones

3. Melanins, which are pigments in skin and


hair, are formed by polymerization of
oxidation products (quinones) of dopa.

4. The catecholamines (dopamine,


norepinephrine, and epinephrine) are
derived from tyrosine in a series of reactions

G. Tetrahydrofolate

- Serine, glycine, and formaldehyde produce


N5,N10-methylene-FH4
UST FMS MEDICAL BOARD REVIEW 2019 13 | BIOCHEMISTRY
REVIEW TEST
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

CHOOSE THE BEST ANSWER: _____6. Which of the following statements


concerning amino acids is correct?
_____1. Which one of the following statements A. Alanine is ketogenic
concerning the peptide shown below is correct? B. Amino acids that are catabolized to acetyl
V–C–E–S–D–R–C coenzyme A are glucogenic
A. The peptide contains asparagine C. Branched-chain amino acids are
B. The peptide contains a side chain with a catabolized primarily in the liver
secondary amino group D. Cysteine is essential for individuals
C. The peptide would move to the cathode consuming a diet severely limited in
during electrophoresis at pH5 methionine
D. The peptide contains a side chain that
can form hydrogen bond with another _____7. This vitamin cofactor is required for
amino acid transamination reactions?
A. cobamide
_____2. The principal function of amino acids in B. nicotinic acid
intermediary metabolism is to: C. tetrahydrofolic acid
A. synthesize tissue and blood proteins D. pyridoxal phosphate
B. furnish energy to the cell
C. provide intermediates for synthesis of _____8. The molecule of urea is synthesized in
specialized products which compartment of the cell?
D. provide nitrogen for urea synthesis A. mitochondria
B. peroxisomes
_____3. Which of the following statements is TRUE C. cytosol
regarding trypsin, a major enzyme in the pancreatic D. microsomes
phase of protein digestion?
A. an exopeptidase _____9. Which of the following metabolites of the
B. is activated by H+ ions released from the urea cycle links it to the citric acid cycle?
stomach A. arginine
C. is activated by an enzyme (enterokinase) B. fumarate
secreted from the brush border of the C. aspartic acid
small intestine D. malate
D. activation of its zymogen form involves
addition of more amino acid residues to _____10. A 3-month-old child presents with
the polypeptide vomiting and convulsions. Notable findings include
hepatomegaly and hyperammonemia. A deficiency in
_____4. Presence of these enzymes explains why which of the following enzymes would most likely
cause an elevation of blood ammonia levels?
practically only free amino acids are found in the
A. CPS I
portal blood after a meal: B. Glutaminase
A. endopeptidases (trypsin, chymotrypsin, etc) C. Argininosuccinate lyase
B. cytoplasmic dipeptidases and tripeptidases D. Asparagine synthetase
C. carboxypeptidases B
D. aminopeptidases _____11. Which of the following amino acids can be
synthesized in the Krebs-Henseleit Cycle?
_____5. Means of transepithelial transport of amino A. fumarate
acids from the lumen to the intestinal cell involving B. arginine
an ion pump that creates a gradient of ions between C. aspartate
the lumen and the intestinal cell. D. alanine
A. facilitated diffusion
B. active transport _____12. N-acetlyglutamate (NAG) is a positive
C. simple diffusion allosteric effector of which enzyme of the urea
D. Na+-dependent co-transport cycle?
A. ornithine transcarbamoylase
B. carbamoyl phosphate synthetase I
C. arginase
D. arginosuccinase

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


REVIEW TEST
NOEL MARTIN S. BAUTISTA, MD, MBAH
MARY ANNE D. CHIONG, MD, MSC, FPPS

_____13. When the carbon skeletons of amino acids _____19. A 1-week old infant, who was born at
are metabolized to any glycolytic and TCA cycle home in a rural area, has undetected classic
intermediates, the amino acid can be used for any of phenylketonuria. Which statement about this baby
the following EXCEPT: and/or her treatment is correct?
A. synthesis of some non-essential amino acids A. A diet devoid of phenylalanine should be
B. complete oxidation to provide energy to the cell initiated immediately
C. synthesis of ketone bodies B. Dietary treatment will be recommended
D. maintenance of glucose through gluconeogenesis to be discontinued in adulthood
C. Supplementation with vitamin B6 is required
_____14. Which of the following amino acids when D. Tyrosine is an essential amino acid
catabolized may be converted to ketone bodies and
fatty acids? _____20. A 56-year-old man with long-standing,
A. aspartate poorly controlled diabetes visits his primary
care physician for a follow-up after a recent
B. histidine
hospitalization. The patient experienced an episode
C. valine of acute renal failure while in the hospital, and his
D. leucine creatinine level rose to 3.4 (normal, 0.7 to
1.5). Creatinine, a marker of kidney function, is
_____15. All of the following amino acids when produced from which of the following precursors?
catabolized enter through the Citric Acid Cycle, A. Glycine, arginine, and SAM
EXCEPT: B. Glutamine, aspartic acid, and CO2
A. histidine C. Glutamine, cysteine, and glycine
D. Serine and palmityl CoA
B. methionine
C. asparagine
D. alanine

_____16. A new test is developed that can non-


radioactively ‘‘label’’ compounds in the human body.
As a physician with a background in the new field of
metabolomics, you assess a 21-year-old with classic
PKU. The patient is fed phenylalanine with a label in
the phenyl ring, and a 24-hour urine sample is
collected. Which of the following compounds would
you expect to contain the greatest amount of label
in this urine sample?
A. Tyrosine
B. Tryptophan
C. Epinephrine
D. Phenylketone

_____17. If an individual has a vitamin B6


deficiency, which of the following amino acids could
still be synthesized and be considered non-essential?
A. Tyrosine
B. Serine
C. Cysteine
D. Aspartate

_____18. In Hartnup’s Disease, pellagra-like


manifestations are seen because of relative
deficiency of nicotinic acid coenzyme derived from:
A. tryptophan
B. phenylalanine
C. tyrosine
D. threonine

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


METABOLIC INTEGRATION
ASSOC. PROF. IMELDA A. DAKIS M.D.

I. PRINCIPAL FUELS OF METABOLISM b. Recommended Nutrient Intake (RNI)


is the level of intake of a specific nutrient
The body’s major fuel depots are glycogen in liver and which is adequate for the maintenance of
muscle, triacylglycerols (TAGs) in adipose tissue, and health and well-being of nearly all healthy
protein in skeletal muscle. Ketone bodies form a persons in the population.
secondary source particularly in the starved state. It defines the quantity of essential
nutrients needed to meet the minimal
A. Carbohydrate requirement to maintain health and
1. Glucose is stored as glycogen in liver and provide reasonable levels of reserves plus
muscle. an added amount to allow for incomplete
a. Liver glycogen maintains blood glucose digestion.
concentrations between meals. There is c. Adequate Intake (AI) is the daily
approximately 200 grams of liver glycogen nutrient intake level that is based on
during the post-absorptive state and this observed or experimentally determined
amount may go down to as low as 80 approximation of the average nutrient
grams after an overnight fast. intake by a group (or groups) of
b. Muscle glycogen furnishes energy for apparently healthy people that is assumed
muscle contraction during exercise. The to sustain a defined nutritional state. It is
glycogen content of muscle is about 150 used when there is insufficient data to
grams. establish the EAR.
d. Tolerable Upper Intake Level or
B. Fat Upper Limit (UL) is the highest average
1. TAGs in adipose tissue serve as the chief fuel daily nutrient intake level likely to pose no
store in the body. adverse health effects to almost all
2. About 15 kg. of TAGs accounts for 85% of individuals in the general population.
total stored calories.
2. Minimum Daily Requirement (MDR) refers
C. Protein to the least amount of a nutrient needed by the
1. Unlike glycogen and fat, protein is not body to prevent manifestations of deficiency.
primarily a fuel reserve.
2. Only about 6 kg. can be oxidized before the 3. Essential Nutrients are substances which the
body functions become compromised. body cannot synthesize in sufficient amounts
to meet the demands of the body and
D. Ketone Bodies therefore, must be included in the diet.

1. -hydroxybutyrate, acetoacetate, and acetone 4. Food Guides are tools to interpret and apply
are synthesized from acetyl CoA in the liver sound nutrient standards for use in food
and are normally found at 3 mg/dl in the planning of individuals and families.
blood; they are used by the heart, muscle, and
brain tissues especially during prolonged 5. Basal Metabolic Rate is the energy required
fasting or starvation. by an individual in the awake state, during
2. One gram supplies 4 kcal upon oxidation. physical, digestive, and emotional rest to
sustain the metabolic activities of the body and
E. Alcohol to maintain circulatory, respiratory,
1. While it is not a dietary essential, each gram of gastrointestinal, and renal processes.
ethanol yields 7kcal.
2. Ethanol is catabolized in the liver, by two NAD+- 6. Energy Requirement is the level of energy
linked enzymes: alcohol dehydrogenase and intake that will balance energy expenditure
acetaldehyde dehydrogenase. when the individual has a body size and
composition, and a level of physical activity,
II. NUTRITIONAL ASPECTS OF DIETARY FUELS consistent with long-term good health.

A. Definitions B. Carbohydrates
1.Philippine Dietary Reference Intake (PDRI) 1. Each gram yields 4 kcal of energy.
is the collective term comprising reference 2. 55% to 70% of dietary energy should come
values for energy and nutrient levels of from carbohydrates, 70% of which should
intakes. It has four components: come from complex carbohydrate, and not
more than 10% should come from simple
a. Estimated Average Requirement sugars.
(EAR) is the daily nutrient intake level 3. A daily intake of 20-25 grams of dietary fiber is
that meets the median or average recommended.
requirement of healthy individuals in a
particular life stage and sex group,
corrected for incomplete utilization or
dietary nutrient bioavailability.
UST FMS MEDICAL BOARD REVIEW 2019 1 | BIOCHEMISTRY
METABOLIC INTEGRATION
ASSOC. PROF. IMELDA A. DAKIS M.D.

C. Lipids C. Acetyl CoA


1. A gram of fat provides 9 kcal of energy. 1. Acetyl CoA is produced by oxidative
2. The recommended intake is 20%-25% of total decarboxylation of pyruvate or by the -
daily dietary energy, with 10% each of oxidation of fatty acids and oxidation of
monounsaturated, polyunsaturated, and ketogenic amino acids.
saturated fatty acids. 2. Acetyl CoA may be completely oxidized to CO2
3. Daily cholesterol consumption should be and H2O, converted to HMGCoA
limited to 300mg or less. (hydroxymethylglutaryl CoA), or used in the
4. Linoleic acid, the only absolute essential fatty synthesis of fatty acids.
acid should be taken at 1%-2% of total daily
calories. V. THE INFLUENCE OF HORMONES ON FUEL
METABOLISM
D. Proteins
1. Protein provides 4 kcal of energy per gram. A. Insulin
2. Recommended daily intake for Filipinos is 1. The pancreatic -cells sense glucose levels and
1.14grams/kilogram of desirable body weight secrete insulin in response to elevation of
(DBW), or 10%-15% of the Total Caloric blood glucose concentration.
Allowance (TCA). 2. Insulin signals the fed state. It promotes
3. Threonine, tryptophan, lysine, leucine, uptake of fuel substrates into skeletal muscle
isoleucine, methionine, valine, phenylalanine and adipose cells, storage of glycogen and
and histidine are indispensable amino acids. TAGs, and biosynthesis of proteins and nucleic
acids.

III. CENTRAL THEMES OF METABOLIC B. Glucagon


PATHWAYS 1. The -cells of the pancreas sense the blood
glucose level and release glucagon in
A. Acetyl CoA is a common intermediate of all response to low levels.
metabolic pathways. 2. The foremost target of glucagon is the liver;
B. Oxidation of dietary fuel leads to the capture of its principal effect is to increase cyclic AMP
energy in the form of ATP and NADH/ FADH2. (cAMP) levels in liver cells. The resulting
C. NADH and FADH2 transfer their electrons to O2 cascades promote glycogenolysis.
via the ETC. The energy released is used to 3. Glucagon raises cAMP levels in adipose tissue,
synthesize ATP. in which TAG mobilization is favored, yielding
D. ATP is the biochemical currency of energy. glycerol and fatty acids.
E. Biosynthesis involves the reduction of simple
carbon compounds to complex polymers. C. Epinephrine
F. Most biosynthetic processes use NADPH as the 1. It is released from the adrenal medulla in
electron donor. response to low blood glucose. It then
G. Biosynthetic and degradative pathways are interacts with second-messenger systems in
distinct and coordinately regulated. many tissues.
2. Epinephrine also holds back the secretion of
IV. KEY JUNCTIONS IN INTEGRATED insulin and stimulates glucagon secretion.
METABOLISM
VI. METABOLIC PROFILES OF MAJOR TISSUES
A. Glucose 6-Phosphate AND ORGANS
1. When glucose is transported into the cell, it is
rapidly phosphorylated to glucose 6-phosphate. A. Brain
Glucose 6-phosphate may be catabolized into 1. The primary fuel for the brain is glucose. Only
pyruvate, stored as glycogen, or converted to under prolonged starvation does the brain use
ribose 5-phosphate by the HMP Shunt. ketone bodies as fuel source, to spare proteins.
2. Glucose 6-phosphate is also formed when 2. The average brain consumes 120 grams of
glycogen is mobilized in liver and muscle, or it glucose in a day or 60% of the utilization of
can be synthesized from pyruvate and glucose by the body in the resting state.
glucogenic amino acids by the gluconeogenic 3. Glucose gains entry into the brain via GLUT3
route. whose Km for glucose is 1.6mM. This transporter
is saturated under normal blood glucose-
B. Pyruvate 4. conditions of 5mM, thus the brain has a steady
1. The primary source of pyruvate is glycolysis. share of glucose.
Other important origins are alanine and lactate. 5. Fatty acids cannot cross the blood-brain barrier
2. On the other hand, pyruvate may be converted as they are bound to albumin in plasma. Ketone-
to lactate, alanine, oxaloacetic acid, and acetyl bodies are the transportable equivalents of fatty
CoA. acids.

UST FMS MEDICAL BOARD REVIEW 2019 2 | BIOCHEMISTRY


METABOLIC INTEGRATION
ASSOC. PROF. IMELDA A. DAKIS M.D.

B. Skeletal Muscle 2. Blood glucose levels rise, stimulating insulin


1. The muscles can use glucose, fatty acids, and release from the pancreatic -cells creating a
ketone bodies for fuel. high insulin-to-glucagon ratio.
2. Muscle has stores of glycogen (1200 kcal) or 3. Because of an increased rate of glucose uptake
75% of all the glycogen stored in the body. by the tissues, glucose becomes the major fuel
3. During vigorous muscle activity, the muscles for oxidation in the liver, muscle, and adipose
account for 90% of the total O2 consumption of tissue.
the body (against the 50% consumption at rest). 4. Activation of the following pathways occurs:
4. The rate of glycolysis exceeds that of the TCA a. glycogen synthesis in liver and muscle
Cycle so that much of the pyruvate is reduced to b. glycolysis in liver
lactate. Lactate is readily brought to the liver for c. HMP Shunt
conversion to glucose. d. TAG synthesis and storage in adipose
5. Protein in muscle is not an efficient fuel store e. protein synthesis
and its degradation proves deleterious. 5. Glycogenolysis, gluconeogenesis, Cori Cycle, -
6. Skeletal muscle contains 10–30 mM oxidation of fatty acid and lipolysis are inhibited.
phosphocreatine which rapidly regenerates ATP
from ADP using phosphocreatine kinase. B. Early Fasting
1. This is represented by an overnight fast, which
C. Liver begins 4 hours after the evening meal and lasts
1. The liver is the center of metabolism, providing for 16 to 24 hours.
fuel to the brain, muscle, and other peripheral 2. Fuel stops coming in from the digestive tract,
tissues. blood glucose level falls and glucagon release
2. It stores glucose as glycogen. It carries out from the pancreas is stimulated.
glycogenolysis and gluconeogenesis to release 3. Glucose uptake and utilization by the cells is
glucose into the blood, being the primary organ suppressed, except in the brain.
that maintains blood glucose level. 4. Glucagon stimulates the cAMP-dependent
3. The liver processes lipoproteins and ketone body protein kinase A, activating glycogenolysis in the
formation from fatty acids. liver to maintain normal blood glucose
4. It is the site of disposal of nitrogen derived from concentration.
amino acid degradation, by means of urea 5. Gluconeogenesis is activated: lactate, pyruvate,
formation. and alanine are diverted to glucose production
5. Keto acids from amino acid degradation are through the Cori Cycle and Glucose-Alanine
preferred to glucose for its own fuel. Cycle.
6. -oxidation of fatty acids is increased to fuel
muscle in order to spare glucose for brain
activity; consequent increase in ketogenesis
D. Adipose Tissue occurs.
1. Adipose tissue stores TAGs and releases fatty 7. Hydrolysis of TAG stores in adipose tissue is
acids as needed. stimulated by hormone-sensitive lipase.
2. Fatty acids are synthesized in the liver, while in 8. Hydrolysis of muscle protein is stimulated to
adipose tissue fatty acids become activated to harvest energy from amino acid carbons.
acyl-CoA which, in turn, esterifies to glycerol to 9. Glycogenesis, glycolysis, lipogenesis, and amino
form TAG. acid degradation are inactivated.
3. Adipose tissue is dependent on glucose for the
supply of glycerol 3-phosphate in TAG C. Starvation/Prolonged Fasting
production. 1. After 2 to 3 days without food, the body enters
4. TAG stores in adipose tissue are hydrolyzed by the starvation state.
hormone-sensitive lipases releasing free fatty 2. Persistence of a low insulin-glucagon ratio
acids and glycerol. exaggerates metabolic events of early fasting.
3. Even during starvation, blood glucose levels
E. Heart have to be maintained at 40mg/dl to offer fuel to
1. Heart muscle, in contrast to skeletal muscle, the brain. Glycogen stores have been depleted
functions only under aerobic conditions. at this stage and the liver can no longer keep up
2. There are virtually no glycogen reserves in heart with gluconeogenesis because of lack of
muscle. substrates (glycerol, lactate, pyruvate, etc.)
3. The fuel of choice is fatty acids while the least 4. Most of the stored energy is in the form of fat
preferred fuel is glucose. Ketone bodies and which cannot be converted to glucose (except
lactate are used under stress. the glycerol from TAGs); accelerated lipolysis
and -oxidation of fatty acids in the liver
VII. METABOLIC ADAPTATIONS IN produce high levels of acetyl CoA and increased
NUTRITIONAL STATES NADH/NAD+ ratio – events that retard the TCA
Cycle.
A. Well-fed State
1. For about four hours after a meal, there is an
abundant supply of metabolic fuels.

UST FMS MEDICAL BOARD REVIEW 2019 3 | BIOCHEMISTRY


METABOLIC INTEGRATION
ASSOC. PROF. IMELDA A. DAKIS M.D.

5. Increased acetyl CoA but lack of oxaloacetic acid  The well-fed state of tissues stays the same
results in exaggerated production of ketone except for a reduction in TAGs.
bodies whose levels go up steadily about 3 days  The switch to fasting state occurs sooner
into starvation. after meals.
6. The brain switches to ketone bodies, sparing
proteins from being used for gluconeogenesis. 3. Another way of losing weight is to have an
7. The heart and skeletal muscle rely on ketone extremely low carbohydrate-high protein-moderate-fat
bodies for energy. diet.
8. During this state, fatty acids are fuelling every  The fasting state is little changed from that
tissue except the red blood cells, which still of other diets.
depend on glucose.  The liver remains gluconeogenic as well as
9. Starvation can be fatal because of eventual ketogenic in the fed state.
protein loss as well as ketoacidosis.  Little rise in blood glucose occurs and insulin
exhibits a less than normal elevation in response
D. Early Re-fed State to meals.
1. This state sets in soon after fuels are again  The excess amino acids are converted to liver
absorbed in the gut after an absence of food. glycogen, blood glucose, and ketone bodies.
2. Fat is metabolized normally while normal glucose  Fatty acids delivered to the liver are converted to
metabolism is gradually reestablished. ketone bodies.
3. The liver remains in gluconeogenic mode for a  Glucose and ketone bodies are produced in both
few hours after feeding, but this the fed and fasted states.
gluconeogenesis does not provide blood glucose  The need for a supply of energy in the form
but rather glucose 6-phosphate for glycogenesis ketone bodies in peripheral tissues largely balances
(“indirect glycogenesis”). Also, gluconeogenesis the production of ketone bodies by the liver.
from amino acids coming in from the gut assists
in relaunching glycogen reserves. G. Alcohol Ingestion
4. After maintenance of a well-fed state for a few 1. Ethanol is primarily catabolized in the liver, by
hours, the metabolic inter-connections of the two NAD+-linked enzymes: alcohol
well-fed state become sustained. dehydrogenase, and acetaldehyde
Gluconeogenesis decelerates, glycolysis dehydrogenase. The two-step process produces
predominates as a means of glucose utilization acetaldehyde and acetate, respectively, with
and liver glycogen stores are maintained by concomitant generation of NADH.
“direct” synthesis from glucose. 2. The major portion of acetate is released into the
blood and delivered to the extrahepatic tissues.
E. Obesity Some of this acetate is changed to acetyl CoA in
1. It is characterized by an excessive accumulation the liver. The extra acetyl CoA is stored as long-
of body fat, with an actual body weight of over chain fatty acids in the liver that may later
20% beyond the desirable body weight. become deposited in the hepatocytes (fatty
2. It is similar to a “prolonged well-fed state”, with liver). Little acetyl CoA enters the TCA Cycle
a too short fasting phase during which stored fat because of a surplus in NADH.
is incompletely used up. 3. Aerobic glycolysis is likewise inactivated because
3. Insulin-glucagon ratio is high. of the high NADH/NAD+ ratio. Pyruvate then
4. Glycolysis and glycogenesis are increased; becomes transformed to lactate and may lead to
glycogenolysis and gluconeogenesis are acidosis.
suppressed. 4. Gluconeogenesis is also inhibited because an
5. Acetyl CoA is produced in increased amounts elevated NADH/NAD+ ratio depresses TCA cycle
due to increased availability of pyruvate from enzymes thus restricting the supply of
glycolysis; excess acetyl CoA is diverted towards oxaloacetate and PEP for gluconeogenesis.
fatty acid and TAG synthesis. 5. Glucose 6-phosphate is shifted to the HMP shunt
6. Lactate from muscle and red blood cell is also because of accumulation of glycolytic
converted to acetyl CoA and into fat through intermediates brought about by a high
pyruvate; lipolysis is inhibited. NADH/NAD+ ratio. Flooding of the HMP shunt
7. Other contributors to exaggerated fat synthesis with glucose 6-phosphate increases the
are dietary fat (chylomicrons) and endogenous production of NADPH for fatty acid synthesis as
TAGs (VLDL) as well as ketogenic amino acids. well as of ribose 5-phosphate for nucleotide
synthesis.
F. Dieting 6. Chronic and excessive ethanol ingestion
1. Consuming less food with the same macronutrient associated with malnutrition is a stimulus for
composition has little effect upon the starve-feed glycogenolysis in the liver.
cycle.
 The roles of the tissues remain the same in
the well-fed state.
 There less glycogen and TAGs stored.

2. Consuming less fat to cut calories is another way to


lose weight.

UST FMS MEDICAL BOARD REVIEW 2019 4 | BIOCHEMISTRY


METABOLIC INTEGRATION
ASSOC. PROF. IMELDA A. DAKIS M.D.

VIII. METABOLIC ADAPTATIONS IN amounts. This is truly a form of “starvation in


PHYSIOLOGICAL STATES the midst of plenty.”
3. With failure of insulin to promote glucose
A. Aerobic Exercise consumption, the sugar accumulates in the
1. As in the starved state, liver and adipose blood and starves the cells of nutrients,
tissue adapt to supply fuel for muscle producing metabolic responses similar to those
contraction. of fasting.
2. With a low insulin-glucagon ratio, lipolysis is a. Glycolysis and glycogenesis are inhibited
stimulated, the liberated fatty acids fueling the in all major tissues.
muscle. b. Liver cells attempt to generate more
3. Glycogenolysis in liver and muscle, and glucose through gluconeogenesis. This
ketogenesis in liver furnish the muscle with- increases further the blood glucose level
more glucose and ketone bodies. Cori Cycle in a well-fed state.
supplies additional glucose to muscle. c. TAG depots are mobilized. Fatty acid
oxidation is enhanced, with concomitant
B. Anaerobic Exercise increase in production of acetyl CoA and
1. Anaerobic exercise involves little inter-organ ketone bodies, which can eventually lead
interaction because the blood vessels within to ketoacidosis.
the muscles are compressed during peak d. Increase in blood fatty acid levels leads
contraction. This causes the muscle cells to to an increased intake of fatty acids by
become separated from the rest of the body. the liver. The excess is exported as VLDL
2. Muscle relies for the most part on its own and this leads to hypertriglyceridemia.
glycogen reserve and phosphocreatine for
energy. Phosphocreatine provides high-energy 4. Type I (insulin-dependent) diabetes mellitus is
phosphate bonds for synthesis of ATP until characterized by a defect in -cell production of
glycogenolysis and glycolysis are stimulated. insulin in the presence of high blood glucose
concentration. Since insulin-glucagon ratio
C. Pregnancy cannot increase, the liver is always
1. The fetus has high demands for nutrients. It gluconeogenic and ketogenic. Ketoacidosis
mainly uses glucose but may also derive energy develops from accumulation of ketone bodies
from amino acids, lactate, and ketone bodies. and H+ ions.
2. Fatty acids are not a source of fuel for the fetus
because these compounds cannot cross the 5. Type II (insulin-independent) diabetes mellitus is
placental barrier. caused by -cell failure and insulin resistance is
3. Placental lactogen activates lipolysis in adipose characteristic. The amount of insulin produced is
tissue; placental estradiol and progesterone inadequate to control glucose production by the
induce insulin resistance. liver or promote glucose uptake by muscle.
4. Pregnant women enter the starved state more Ketoacidosis rarely develops because enough
rapidly. insulin is available to prevent uncontrolled fatty
acid release from adipose tissue.
D. Lactation
1. In late pregnancy, progesterone and prolactin B. Liver Disease
induce lipoprotein lipase in the mammary gland 1. Liver damage, especially in the advanced-
to promote the development of milk-secreting stages, is coupled with major metabolic
tissues. derangements, particularly involving the amino
2. During this period, the major energy source of acids.
breast tissue is glucose. 2. In liver cirrhosis, for example, the hepatocytes
3. Amino acids are taken up for protein synthesis. are not capable of converting ammonia to urea-
Chylomicrons and VLDL furnish fatty acids for and glutamine rapidly enough, and the blood
TAG production. ammonia rises.

IX. METABOLIC ADAPTATIONS IN C. Cancer


PATHOLOGICAL STATES 1. Tumor cells function independently of the
starve-feed cycle. They exhibit an incessant
A. Diabetes Mellitus demand for glucose as a source of energy and
1. This complex metabolic derangement is amino acids for protein synthesis.
characterized by a grossly abnormal usage of- 2. Glucose is the preferred fuel of tumor tissues.
fuel. In untreated diabetes, the level of insulin is 3. Cancer cells rarely adapt in the fasting state to
inappropriately low while that of glucagon is too use fatty acids and ketone bodies.
high relative to the needs of the body. This 4. They are unresponsive to hormonal changes
deficiency in insulin impairs the entry of glucose that influence metabolic processes in normal
into the cells. tissues.
2. As in starvation, glucose utilization in diabetes is
poor, but glucose is actually present in excessive

UST FMS MEDICAL BOARD REVIEW 2019 5 | BIOCHEMISTRY


METABOLIC INTEGRATION
ASSOC. PROF. IMELDA A. DAKIS M.D.

X. REGULATION OF APPETITE, ENERGY


CONSUMPTION, AND BODY WEIGHT

A. Neuronal Control of Appetite


 The arcuate nucleus contains two types of
cells that influence the intake of food and the
utilization of energy:
a. Neuropeptide Y (NPY) and Agouti-related
peptide (AgRP) are potent stimulators of
food intake and inhibitors of energy
expenditure.
b. Pro-opiomelanocortin (POMC) and
Cocaine-Amphetamine-Regulated
Transcript (CART).

B. Hormonal Control of Appetite

1. Leptin is the protein product of the Ob gene.


Secreted by the adipose cells, it informs the
brain of the body fat store level. It acts on the
hypothalamus to directly stimulate the oxidation
of fatty acids and inhibit accumulation of lipids
in non-adipose tissue.
2. Insulin stimulates the uptake of glucose by
muscle and adipose cells and storage of glucose
as glycogen in liver and muscle. It stimulates
POMC/CART neurons that will inhibit food intake
and stimulate energy expenditure.
3. Ghrelin, secreted by an empty stomach and a
short-term regulator of appetite, is a gastric
peptide containing 28 amino acid residues. It is
the only known appetite-stimulating hormone.
4. PYY3-36 is secreted in the gastrointestinal tract
proportionally with caloric intake. As a short-
term regulator of appetite, it inhibits further
food intake.

-------------------------------------------------------------------

REFERENCES
1. Devlin TM, Textbook of Biochemistry with Clinical
Correlations, John Wiley Sons, Inc.
2. Murray RK, Granner DK, Rodwell VW, Harper’s
Illustrated Biochemistry
McGraw-Hill Companies, Inc.
3. Marks DB, Marks AD, Smith CM, Basic Medical
Biochemistry A Clinical Approach
Williams & Wilkins Company
4. Gilbert HF, Basic Concepts in Biochemistry McGraw-
Hill Companies
5. FNRI-DOST, PDRI 2015 Philippine Dietary
Reference Intakes Summary of
Recommendations

UST FMS MEDICAL BOARD REVIEW 2019 6 | BIOCHEMISTRY


REVIEW TEST
ASSOC. PROF. IMELDA A. DAKIS M.D.

CHOOSE THE BEST ANSWER:


_____ 8. Which metabolic characteristic is typical of
_____ 1. Which metabolic enzyme does not promote fuel utilization by the heart muscle cells?
fuel storage? A. fuel substrate metabolism regardless of
A. acetyl CoA carboxylase availability of oxygen
B. branching enzyme B. storage of significant amounts of fuel
C. glucose 6-phosphatase reserve
D. glycogen synthase C. preference for fatty acids for primary fuel
D. mobilization of triacylglerol stores by
_____ 2. Which metabolic intermediate interlocks hormone-sensitive lipase
glycolysis, fatty acid degradation, and oxidation of
ketogenic amino acids? _____ 9. A young man slept from 11:00 p.m. to
A. acetyl CoA 6:00 a.m. the following day. It is most likely that
B. citrate during the hours that he was asleep:
C. glucose 6-phosphate A. glucose generation from lactate, pyruvate
D. oxaloacetic acid and alanine was significantly rapid
B. glycogen degradation in liver was the
_____ 3. The reason for protein not being an main source of fuel of the tissues
efficient metabolic fuel is that: C. triacylglycerol stores in adipose tissue
A. a gram of protein provides only 2 kcal of were increased to a great extent
energy D. uptake and utilization of glucose in most
B. it is the chief source of fuel for muscle cells were activated
tissues only
C. its only function is to build up and repair _____ 10. The alcohol intake of an individual should
tissues be counted in the daily planning of meals because:
D. only limited amounts of it can be oxidized A. alcohol is metabolized slowly in the liver
for energy B. ethanol is directly converted to glucose
and fatty acids during metabolism
_____ 4. Glycogen that is stored in skeletal muscle C. each gram of ethanol yields 7kcal of
is chiefly intended for: energy
A. allowing the recycling of metabolic D. starch and alcohol provide equal amounts
intermediates during muscle activity of calories per gram
B. maintaining normal blood glucose
concentration _____ 11. A healthy and non-diabetic 25-year old
C. providing immediate fuel supply during office secretary goes into a very low carbohydrate,
muscle activity moderate fat, high protein diet. Which metabolic
D. saving phosphocreatine for future ATP effect is expected while she is on this dietary
synthesis regimen?
A. a less than normal insulin response
_____ 5. The chief role of glycogen stored in the B. a long well-fed state
liver is to: C. a pronounced elevation of blood glucose
A. abort hyperglycemia after a meal D. ketogenesis that leads to ketoacidosis
B. furnish energy to the skeletal muscles
during physical activity _____ 12. A 29-year old ex-ramp model whose
C. keep blood glucose levels between meals desirable body weight is 56 kg, presently weighs 70
within normal range kg. Becoming less attractive than before, she
D. sustain adequate energy reserve for at becomes depressed and binges on fast-food meals
least three days and takes in-between meals about every two hours.
In her current nutritional state:
_____ 6. Which is correct regarding lipid as a major A. glucagon is the predominant hormonal signal
metabolic fuel? B. glycolysis and glycogenesis are inactive
A. Around 85% of the total body fuel C. stored fat is incompletely used up during
reserves is triacylglycerol. the too short fasting phase
B. Each gram of fat yields less energy than a D. there is lack of acetyl CoA for energy
gram of glucose does. generation
C. A huge portion of dietary fat is
transformed to glucose in the liver. _____ 13. A 57-year old company executive, a
D. Its storage form is mobilized to keep the diabetic, is non-compliant with his medications and
energy needs of the brain in sleep. prescribed dietary program for over a year now.
Recent laboratory tests show elevated FBS and
_____ 7. A low insulin-glucagon ratio promotes: HbA1c values. Which metabolic event is expected to
A. glucose uptake of adipose and muscle supply the fuel needs of his tissues?
B. production of glucose from lactate, A. inhibition of ketone body synthesis
glycerol, and alanine B. inactivation of gluconeogenesis
C. storage of fat as triacylglycerols in adipose C. mobilization of TAG depot
D. synthesis of liver and muscle glycogen D. promotion of glycolysis and glycogenesis
UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY
REVIEW TEST
ASSOC. PROF. IMELDA A. DAKIS M.D.

For nos. 20-21. A 25-year old call center agent,


For nos. 14-15. A 52-year old traffic enforcer is whose desirable body weight is 60kg, consumes, on
found stuporous, with face flushed, breath strongly the average, 300 grams of carbohydrate each day.
smelling of alcohol. His blood pressure is 170/90 mm
Hg., cardiac rate 115/min, respiration shallow, _____ 20. How many calories from carbohydrate
respiratory rate 15/min; plasma glucose 2.5 mmol/L does he actually obtain daily?
(normal 3.3-8.4 mmol/L); blood lactate 2.9 mmol/L A. 2700
(normal 0.7-2.0 mmol/L); plasma alcohol B. 2100
concentration 98 mmol/L (normal 17.4 mmol/L). C. 1800
D. 1200
_____ 14. The blood glucose level of the patient is a
result of: _____ 21. Approximately, in what amount (in
A. activation of triacylglycerol synthesis into grams) should he take in protein from his diet
VLDL according to the recommended intake for a normal
B. hyperactivity of the Krebs cycle in the Filipino adult?
peripheral tissues A. 100
C. inhibition of gluconeogenesis in the liver B. 70
D. stimulation of beta-oxidation inside the C. 50
mitochondria D. 30

_____ 15. The blood lactate concentration is _____ 22. Which feature is correct about leptin?
explained by: A. enhances food intake
A. a high NADH-NAD+ ratio B. inhibits fatty acid oxidation
B. an excess of intermediates from muscle C. promotes lipid storage in non-adipose
glycogenolysis tissues
C. failure of fatty acid biosynthesis in D. secreted by the adipose cells
adipose
D. lack of substrate for the Cori cycle _____ 23. Ghrelin is a peptide that stimulates the
intake of food by:
_____ 16. Because of the increased availability of A. accelerating energy consumption by the
glycolytic enzymes and glucose transporters in body
cancer tissue, tumor cells exhibit an incessant B. activating the NPY/AgRP neurons in the
demand for: arcuate nucleus of the hypothalamus
A. acetyl CoA C. blocking the action of appetite-reducing
B. fatty acids hormones in the brain
C. glucose D. causing the release of melanocyte-
D. oxaloacetic acid stimulating hormone

_____ 17. Which amino acid must be supplied in the


diet?
A. alanine
B. aspartic acid
C. glutamic acid
D. threonine

_____ 18. Which recommendation is correct for


dietary intake of lipids?
A. Linolenic acid and arachidonic acid should
always be present in the diet.
B. Saturated fats must comprise 50% of the
total fat intake.
C. The daily consumption of cholesterol
should be less than 300mg/day.
D. Trans-fatty acids may replace saturated
fat in the daily meals.

_____ 19. An individual who is watchful of his blood


cholesterol levels should avoid:
A. avocado
B. cheese from skim milk
C. egg white
D. fish roe

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

I. CHEMISTRY OF NUCLEIC ACIDS E. Structure of Nucleic Acids


1. DNA Structure (Watson and Crick model)
A. Nucleic acids, DNA and RNA are polymers of a. Polymer of deoxyribonucleotides
nucleotides, the basic building blocks of nucleic acids linked by 3’-5’ phosphodiester
bonds; sugar- phosphate as
B. Nucleotides contain three units: backbone; Bases found include
1. Sugar (ribose or deoxyribose) A, G, C and T
2. Base
a. purines: adenine (A), guanine (G), b. Double stranded right handed
hypoxanthine (H) helix, with antiparallel complementary
b. pyrimidines: cytosine (C), thymine strands. One chain runs in a 5’ to 3’
(T), uracil (U) direction while the other in a 3’ to 5’
3. Phosphate group direction. Adenine (A) always base
pairs thru hydrogen bonding with
C. Nucleoside consists of a sugar with a base in thymine (T) and guanine (G) with
glycosidic linkage to C1 and a nucleotide is a cytosine(C); there are 10 bases per
nucleoside with one or more phosphate groups in an turn of the helix and makes a
ester linkage to C5. complete turn every 3.4 nm; bases
are perpendicular to the helix;
Purine nucleoside - root word + osine,
e.g. Adenine + osine = adenosine; c. Base pairing is reflected by
Pyrimidine nucleoside – root word + idine Chargaff’s rule that states:
e.g. cytosine + idine = cytidine Purines (A +G) = pyrimidines (C+T)
and purine/ pyrimidine = 1 (A/T = 1;
G/C = 1)

d. DNA helix is stabilized by hydrogen


bonding between bases on complementary
strands as well as stacking and hydrophobic,
Van der Waals forces between bases, and
ion-ion interactions, AT base pairs have two
H bonds and GC have three H bonds.

D. Function of Nucleic Acids/ Nucleotides:


1. Nucleic acids are repository molecules for
genetic information.
2. Nucleotides are substrates for DNA synthesis
(replication) and RNA synthesis
(transcription)
3. Nucleotides are carriers of high energy
groups e.g. ATP, UDP glucose, CDP choline
acetylCoA, acylCoA S- adenosylmethionine.
4. Nucleotides are components of
coenzymes: NAD, NADP, FAD, CoASH.
5. Nucleotides as regulatory molecules: e. Eukaryotic DNA is stabilized and bound to
cyclic AMP, cyclic GMP. basic proteins called histones. Their basic
charge enables the histones to bind to the
phosphate backbone of DNA. DNA wraps
around a core of eight histone molecules,
(two molecules each of H4, H3, H2B and
UST FMS MEDICAL BOARD REVIEW 2019 1 | BIOCHEMISTRY
NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

H2A) producing a nucleosome. The translated into proteins; considered an adapter-


polynucleosome consists of several molecule that binds to mRNA through its
nucleosome joined by a linker histone H1. anticodon arm.
The arrangement produces an appearance
of beads on a string. The chromatin formed III. rRNA in eukaryotes constitutes 65% of
from the DNA-protein complex is condensed ribosomes; contains a large (60S) and smaller
into chromosomes. subunit (40S) to form an 80S molecule with other
protein molecules; prokaryotes have 50S (larger)
and 30S (smaller), collectively called the 70S
rRNA.

E. Denaturation of DNA:
I. Unwinding of double stranded DNA when
subjected to high temperatures, pH extremes
and certain chemicals.

II. Produces a hyper chromic effect, which is


increased in ultraviolet (UV)absorption at 260nm II. NUCLEOTIDE METABOLISM
and a decrease in viscosity.
A. Purine Nucleotide Synthesis
III. Polynucleotides denature at certain 1. De Novo Pathway of purine synthesis involves
temperatures called melting temperature (Tm). the assembly of all the C and N atoms from
GC rich regions form more stable double helices various precursor
than AT rich regions, thus GC rich DNA has molecules: glycine (C4, C5, N7), glutamine (N3,
higher Tm than AT rich DNA. N9), aspartate (N1), Carbon dioxide (C6), and
formyltetrahydrofolate (C2, C8)
IV. When denatured polynucleotides are cooled
or when denaturing agents are removed,
complementary single stranded regions
reassociate in a process called annealing.
Annealing is the basis for hybridization
(DNA/RNA) in the use of probes in recombinant
DNA technology, e.g. Southern Blot

2. RNA Structure
a. Polymer of ribonucleotides linked by 3’-5’
phosphodiester bonds; single stranded but may
form internal double stranded regions
sometimes called hairpin loops.
2. Synthesis of 5’ phosphoribosyl-1-pyrophosphate
b. Bases found are A, G, C and uracil (U) instead of (PRPP) occurs at the beginning of the process
T catalyzed by ribose phosphate
pyrophosphokinase or PRPP synthase from ATP
c. Three classes of RNA: and ribose-5-phosphate. IMP, AMP and GMP
I. mRNA: carries genetic information (codon) to inhibit this step.
be translated into proteins;
unique base sequences and a 7-methyl 3. The committed step involves the conversion of
guanosine cap at the 5’ end and a polyadenylic PRPP to 5’ phosphoribosyl-1- amine. PRPP
acid tail at 3’ end. activates the enzyme glutamine PRPP
amidotransferase and the products of the
II. tRNA: smallest RNA which assumes pathway inhibit the enzyme. The end products
secondary structure similar to a cloverleaf are:
appearance; contains atypical bases; carries the
specific amino acid to the ribosomes to be
UST FMS MEDICAL BOARD REVIEW 2019 2 | BIOCHEMISTRY
NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

a. IMP- formed on the amino group of


phosphoribosylamine by a 9-reaction
sequence
b. GMP, formed by the addition of an amino
group to C2 of IMP
c. AMP, formed by the substitution of
an amino group for the oxygen at C6.

4. Regulation of Purine Synthesis


a. Regulation occurs at the final branches
of the de novo pathway to provide a
steady supply of purine nucleotides
- GMP and AMP both inhibit the first step 3. De novo synthesis starts with the synthesis of
in their own synthesis from IMP carbamoyl phosphate (CAP) using carbon dioxide,
- GTP is a substrate in AMP synthesis and glutamine and ATP by the action of cytosolic enzyme
ATP is a substrate in GMP synthesis. carbamoyl phosphate synthase II. (CPS II)
Reciprocal substrate effect balances the
supply of adenine and guanine 4. Synthesis of dihydroorotic acid, a pyrimidine,
ribonucleotides involves a 2-step process.
a. The committed step is the addition
b. Interconversion among purine aspartate to CAP, catalyzed by
nucleotides ensures control of the transcarbamoylase, to form carbamoyl
levels of adenine and guanine aspartate.
nucleotides b. Ring closure with a loss of water, catalyzed
- IMP is the starting point for synthesis of by dihydroorotase produces dihydroorotic
AMP and GMP acid.
- AMP deaminase converts AMP back to
IMP 5. Dihydroorotate forms UMP
- GMP reductase converts GMP back to a. Addition of ribose-phosphate
IMP moiety from PRPP by orotate
phosphoribosyl transferase
5. Salvage pathway involves the synthesis of produces orotidylate (OMP)
purine nucleotides from preformed bases b. Decarboxylation of OMP produces
through two enzymes: uridylate (UMP)
a. Hypoxanthine-guanine c. From UMP the other pyrimidine
phosphoribosyltransferase (HGPRT). IMP nucleotides (CTP and TMP) are
and GMP are competitive inhibitors of synthesized
HGPRT
Mg++ 6. Regulation of pyrimidine synthesis occurs at
Guanine + PRPP  GMP + PPi several levels:
a. UTP inhibits carbamoyl phosphate
Mg++ synthase II while ATP and PRPP
Hypoxanthine + PRPP  IMP +PPi activate this enzyme
b. UMP and CMP inhibit OMP
b. Adenine phosphoribosyl transferase. AMP decarboxylase
inhibits this enzyme c. CTP inhibits CTP synthase

Mg++ 7. Salvage of pyrimidines is accomplished by


Adenine + PRPP  AMP + PP the enzyme pyrimidine phosphoribosyl which can
use orotic acid, uracil or thymine but not cytosine,
A. Pyrimidine Nucleotide Synthesis with PRPP as source of the ribose phosphate
1. Synthesis of pyrimidines differs from that
of purines in that the pyrimidine ring is Orotic acid + PRPP  OMP + PPi
synthesized first and is then attached to Uracil + PRPP UMP +PPi
ribose phosphate. Thymine +PRPP  TMP + PPi

2. The origin of the atoms in the pyrimidine


ring are carbon dioxide and glutamine
via carbamoyl phosphate (C2, N3) and
aspartate (C4, C5, C6, N1)

UST FMS MEDICAL BOARD REVIEW 2019 3 | BIOCHEMISTRY


NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

e. Reaction is inhibited by anticancer


agents:
- 5 fluorouracil (5FU) inhibits
thymidylate synthase
- aminopterin and methotrexate
inhibit dihydrofolate reductase

D. Nucleotide Degradation
1. Purine degradation in man involves uric acid
formation and its urinary excretion; other species
excrete different forms e.g. allantoin, ammonia.
a. Sequential actions of nucleases and
nucleotidases lead to hydrolysis of nucleic
acids to nucleosides.
b. Adenosine deaminase converts adenosine
and deoxyadenosine to inosine or
deoxyinosine
c. Purine nucleoside phosphorylase splits
inosine and guanosine to ribose- 1-phoshate
and the free bases, hypoxanthine and
guanine.
d. Guanine is deaminated to xanthine.
e. Hypoxanthine and xanthine are oxidized to
uric acid by xanthine oxidase.
f. Xanthine oxidase is inhibited by allopurinol

C. Deoxyribonucleotide Synthesis
1. Formation of deoxyribonucleotides, required for
DNA synthesis, involves reduction of the ribose
moiety of ribonucleoside diphosphates
a. Ribonucleotide reductase converts ADP,
GDP, CDP, and UDP to dADP, dGDP, dCDP,
and dUDP, respectively.
b. Thioredoxin with sulfhydryl groups acts as
reducing agent.
c. Thioredoxin reductase converts oxidized
thioredoxin back to reduced form, using
NADPH + H.
d. Reduction requires presence of
nucleoside triphosphate as allosteric
activator, with dATP as an allosteric
inhibitor.

2. Thymidylate synthase catalyzes the formation of


deoxythymidylate (dTMP) from dUMP,
2. Pyrimidine degradation produces amino
a. Transfer of a one-carbon unit from
acids, CO2 and NH4+
N5, N10- methylene etrahydrofolate
a. Excess nucleotides are degraded to
(THF) to C5 of the uracil ring
the free bases uracil or thymine
b. Reduction of methylene group to
b. Reaction sequence consisting of
methyl group and oxidation of THF
reduction, ring opening and
to dihydrofolate (DHF)
deamination- decarboxylation converts
c. Reduction of dihydrofolate back to
uracil to CO2, NH4+ and -alanine.
THF by dihydrofolate reductase
Thymine is converted to CO2, NH4+
(DHFR), requiring NADPH as cofactor
and -amino isobutyrate, an indicator
d. Remethylation of FH4 at the expense
or DNA turnover.
of serine, by enzyme serine
hydroxymethyl transferase
UST FMS MEDICAL BOARD REVIEW 2019 4 | BIOCHEMISTRY
NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

E. Clinical Disorders of Purine and Pyrimidine III. GENE EXPRESSION (INFORMATION


Metabolism METABOLISM)
1. Hereditary Orotic Aciduria: deficiency of
orotate phosphoribosyl transferase and/or A. Genetic information found in DNA is
OMP decarboxylase leads to increased ultimately expressed into proteins,
urinary excretion of orotic acid and following the Central Dogma of Molecular
decreased production of pyridine nucleotides Biology. The flow of genetic information
needed for both RNA and DNA synthesis, proceeds from DNA replication,transcription
characterized by retarded growth, severe into RNA and translation into proteins.
anemia (megaloblastic).

2. Purine nucleoside phosphorylase deficiency DNA  RNA  Proteins:


leads to increased levels of purine Replication Transcription Translation
nucleosides with decreased uric acid
formation and impaired T cell function B. The three major processes (replication,
transcription, translation) consist of
3. Severe combined immunodeficiency (SCID) subprocesses which includes initiation,
caused by adenosine deaminase deficiency. elongation and termination
It leads to impaired T and B cell dysfunction
with death due to overwhelming infection. C. The genetic code describes the relationship
SCID has been treated using gene therapy between the polynucleotide alphabet of
the four bases (A, G, T, C) in DNA and the 20
4. Lesch-Nyhan Syndrome. Partial or absence amino acids in proteins. The base
of HGPRT, salvage enzyme leads to sequences in one strand of parental DNA
excessive purine synthesis, hyperuricemia, dictate the amino acid sequences of
severe neurologic problems like spasticity, proteins. Characteristics and properties of
mental retardation, self-mutilation. the genetic code:
Unsalvaged hypoxanthine and guanine leads 1. Universal- same in all organisms
to decreased IMP and GMP and de novo 2. Contiguous- the codons do not overlap
pathway becomes stimulated due to and are not separated by spacers
increased levels of PRPP. Unsalvaged 3. Specific or unambiguous – the three
purines are then degraded to uric acid. nucleotide sense codon specifies only
one amino acid
5. Gout, a form of arthritis associated with 4. Degenerate- there is more than one
hyperuricemia could be primary due to codon for the same amino acids
defects in enzymes of purine metabolism or
secondary to other clinical disorders D. DNA synthesis (Replication) which takes
place during the S phase of the cell cycle is
a. Uric acid which is not very soluble in body the production of two double stranded DNA
fluids are deposited in joints and soft tissues molecules that are identical in every way to
causing the inflammation characteristic of the parent DNA. Eukaryotic replication is
gouty arthritis. Crystals can also be semiconservative. i.e. each daughter DNA
deposited in the kidneys leading to renal contains one strand of parental DNA and
damage (gouty nephropathy) one newly synthesized daughter strand.
Replication is unidirectional.
b. Primary gout involves overproduction Stages:
of purine nucleotides via de novo 1. Pre-priming stage involves a ssDNA
pathway which could be due to: binding proteins and DNA unwinding
- mutations in PRPP synthase with proteins or helicases that unwind the
loss of feedback inhibition by DNA duplex. Topoisomerases (DNA
purine nucleotides gyrase) relieve the strain imposed by
- partial HGPRTase which causes unwinding.
accumulation of PRPP that 2. Initiation involves the use of primase
activates PRPP amidotransferase Which makes RNA primers that are
- abnormal structure of PRPP complementary to the DNA template
Amidotransferase strand. This process moves in the 5’-to
3’ direction on the newly synthesized
c. Secondary Gout may be due to excessive primer.
turnover of purines due to cancer 3. Elongation involves the addition of
chemotherapy, radiation therapy, leukemia. nucleotides (dNTPs) at the 3’ end of
the primer by DNA polymerase (DNAP)
d. Gout is frequently treated with allopurinol, a III in prokaryotes ( and  in
structural analog of that inhibits xanthine eukaryotes). DNAP III possesses
oxidase Xanthine oxidase can convert editing function due to its 3’-5
allopurinol to alloxanthine. exonuclease activity, i.e. detects and
removes mismatched base pairs and

UST FMS MEDICAL BOARD REVIEW 2019 5 | BIOCHEMISTRY


NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

replaces correct bases. DNAP  in


eukaryotes carries out repair. PPi is
released during polymerization.
4. Replication is continuous in the leading
strand and discontinuous in the lagging
strand. DNAP  in eukayotes copies
the lagging strand and DNAP copies
the leading strand RNA primers are
removed with nick translation by
DNAP I. DNAP  carries out
mitochondrial replication in
eukaryotes. Segments of newly
synthesized DNA, in the lagging
strand are called Okazaki fragments
5. Termination involves the joining of the
ends of the synthesized Okazaki
fragments by DNA ligase. (sealing of
nicks)
6. In eukaryotes, DNA is associated with
nucleoprotein histones.
7. Replication is inhibited by the Ara-A and
Ara-C and by Actinomycin D which
intercalates between GC sequences,
blocking the elongation.process;
Ofloxacin inhibits gyrase.

E. Transcription is the process that leads to


synthesis of RNA, with a sequence that is
complementary to that of the DNA template.

1. RNA polymerase (RNAP) is the main


enzyme that synthesizes RNA from DNA.
In eukaryotes several types of RNAP exist:
a. RNAP I: synthesizes rRNA
b. RNAP II: synthesizes mRNA
c. RNAP III:synthesizes tRNA and 5S Rrna
d. mitochondrial RNAP: transcribes RNA
from mitochondrial genes

2. Transcription Cycle occurs as follows:

a. Binding: RNAP binds to specific promoter


sequences on the DNA, which orients the
RNAP on the sense strand in apposition to
begin transcription. Both prokaryotic and
eukaryotic promoters have consensus
sequences “upstream” of the start site.
 Promoter regions in eukaryotes include a -25
TATA box (Hogness box) and a -75 CAAT
region.
 Promoter regions in prokaryotes include a -10
TATAAT region (Pribnow box) and a -75
TTGACA region. A short stretch of DNA duplex
unwinds to form a transcription bubble.
 Unlike DNA replication, no RNA primers are
needed for transcription, and only one DNA
strand is transcribed.
 RNAP holoenzyme is composed of ’
subunits. Polymerase activity resides in the
’; the  subunit recognizes the promoter
region

UST FMS MEDICAL BOARD REVIEW 2019 6 | BIOCHEMISTRY


NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

F. Translation (Protein Synthesis) involves the


polymerization of amino acids in a precise
sequence directed by the sequence of bases
in mRNA. Steps consist of the following:

1. Activation of amino acids. An amino acid is


activated by binding to the 3’ end of a tRNA
catalyzed by aminoacyltRNA synthase at
the expense of ATP producing an
aminoacyltRNA + AMP + PPi. The first
amino acid to be translated is methionine
(fmet in prokaryotes) as would be indicated
b. Initiation involves the formation of the first by the first codon AUG, in the mRNA.
phosphodiester bond. ATP or GTP forms a
base pair with the template base on the 2. Formation of the Initiation complex.
antisense strand at the origin, and then the a. The 40S initiation complex is formed
base of the next nucleoside triphosphate when the 40S rRNA binds to initiation
pairs with the next template base and factors (eIFs), GTP and met- tRNA. The
forms a phosphodiester bond with the ATP or binding enables the mRNA to bind to the
GTP, eliminating PPi 40S subunit.
c. Elongation proceeds along the DNA sense b. As certain initiation factors are released
strand, with the RNA growing in the 5’-to-3’ and GTP is hydrolyzed, the 40S initiation
direction. NTPs are added with release of PPi complex joins the 60S subunit.
The DNA duplex reforms behind the c. The met- tRNA occupies the P site of
enzyme, and the 5’ end of the RNA is the ribosome with the AUG codon and
released as a single strand. with an empty A site, the resulting
d. Termination. In prokaryotes, this occurs at complex is called 80S initiation complex.
the site of a stem loop (hairpin loop followed In prokaryotes a purine rich region in
by a string of Uracils). The presence of a the mRNA called Shine Dalgarno
rho protein makes this process more sequence binds to the 16S subunit of
efficient. Termination signals in eukaryotes the 30S rRNA to stabilize the binding of
are poorly understood. the mRNA to the 30S subunit.
The 7-methyl guanosine cap in
3. RNA processing or post transcriptional eukaryotes is its counterpart
processing involves modification of RNA in
eukaryotes. Unprocessed eukaryotic mRNA 3. Elongation phase is a three-step cycle that
is also called heterogenous nuclear RNA repeats each time an amino acid is added.
(hnRNA) a. The incoming AA-tRNA binds to the
aminoacyl (A) site of the large 80S
a. 7 methyl guanosine cap at the 5’ end of subunit, requiring several elongation
mRNA provides protection against nuclease factors (EFs) and the hydrolysis of GTP.
digestion and help in alignment of the mRNA b. Peptidyl transferase catalyzes the transfer
during translation. of the amino acid or the peptide from the
b. Poly (A) tail in the 3’ end of mRNA P site to the AA-tRNA on the A site, with
c. RNA splicing removes introns or intervening The formation of the peptide bond. The
segments in mRNA and joining of exons. “uncharged” tRNA dissociates from the
Small nuclear RNA helps in splicing complex.
d. Addition of ACC at 3’ end and modification c. The new peptidyl t-RNA moves to the P
of bases in tRNA. site (i.e., the ribosome moves three
nucleotides over on the mRNA), requiring
4. Certain drugs that inhibit transcription EF-2 and GTP hydrolysis. The ribosome
include rifampicin and streptolydigin which moves along the mRNA in the 5’-to-3’
binds and inhibits RNAP. Actinomycin D direction and the peptide chain grows
binds to DNA template and inhibits from the N terminus to the C terminus.
elongation process. Coumermycin, Nalidixic d. After the ribosome has “moved” out of
acid and Novobiocin are antibiotics that the way, another ribosome can begin
inhibit topoisomerases translation at the initiation codon. An
mRNA with several attached ribosomes
are carrying out translation is known as a
polyribosome or polysome.

4. Termination. The process occurs when the


ribosome encounters a nonsense
(termination) codon, i.e. UAA, UAG, UGA,
which signals termination and release of
the polypeptide.
UST FMS MEDICAL BOARD REVIEW 2019 7 | BIOCHEMISTRY
NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

a. A protein releasing factor (RF) G. Regulation of Gene Expression. Regulation


together with GTP binds to the A site, can take place in all phases of gene
instead of a AA-tRNA) expression, from replication through
b. Peptidyl transferase hydrolyzes the translation but major control takes place
peptidyl-tRNA, with the release of during the transcription process.
the completed polypeptide. GTP is Regulation in prokaryotes differ that
hydrolyzed to GDP and Pi occurs. that of eukaryotes.
c. The ribosomes, dissociate into subunits
1. Prokaryotic gene regulation.
5. Wobble. The codon in mRNA (3’ base) and a. An operon refers to the prokaryotic
the anticodon in tRNA (5’ base), can genetic unit in which several genes are
“wobble” at the nucleotide –nucleotide clustered and transcribed into a
pairing site. In the tRNA anticodon, the polycistronic mRNA.
wobble base is often inosine, which can b. Regulatory genes code for proteins that
pair with U, C, or A in the mRNA. In in turn control the expression of genes
mRNA, G in the wobble position can pair by binding to control elements at sites
with U or C; U in the wobble position can on the DNA near the structural gene.
pair with A or G. Because of wobble fewer Regulatory proteins control the degree
than 61 tRNAs are needed to translate of access that the enzyme RNA
the 61 sense codons of the genetic code. polymerase has to its binding site on
the gene. Two types of regulatory
6. Post-translational modifications. The first protein have been found
amino acid translated, methionine - negatively acting which represses the
(f-met in prokaryotes) are removed. The operon by binding to the operator
amino acid sequence, e.g. signal - positively acting which enhance the
recognition sequence, and conformational affinity of RNAP for its binding sites
shape of a protein will determine its fate, on the gene.
whether it is to be targeted to a particular
site or to be a substrate for modifying c. A good example of an operon is the
enzymes. They will also determine its half lactose operon in E. coli. Expression of
life. Proteins can also be co-translationally the operon is regulated by an inducer
modified. Modifications include: (lactose or allo lactose) and by a
a. crosslinking by disulfide bonds repressor protein, expressed by the
b. excision of a polypeptide chain. i gene. The i gene, situated just
e.g. proinsulin to insulin before the controlling element for the
c. glycosylation cluster of genes coding for three
enzymes is important for lactose
7. Various antibiotics inhibit translation in utilization by the cell. The i gene is
prokaryotes. Tetracyclines inhibit binding continually expressed into a repressor
of tRNA to the 30S ribosomal subunit. protein which binds to a specific DNA
Streptomycin affects all phases of sequence between the promoter and
translation, Erythromycin inhibits the operator gene. This binding blocks the
function of 50S rRNA, Chloraphenicol binding of RNA to the promoter, and
inhibits peptidyl transferase and the operon is said to be repressed,
Puromycin mimics an aminoacylt RNA especially so if glucose is abundant in
causing premature termination. the cell.
d. Derepression of the operon could be
achieved by an inducer, lactose or
allolactose in the case of lactose
operon. Lactose (and
isopropylthiogalactose)
binds to the repressor protein causing it
to change in conformation which
drastically lowers its affinity to the DNA
sequence to which it usually binds,
paving way to the binding of RNAP to
the promoter.
e. The lac operon is repressed if there is
plenty of glucose even despite
abundance of lactose. This is called
catabolite repression. The cell will only
turn to lactose as a substrate when
glucose concentrations fall. When
glucose concentration is high, the
second messenger cyclic AMP are low.
When glucose concentrations are low,

UST FMS MEDICAL BOARD REVIEW 2019 8 | BIOCHEMISTRY


NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

cAMP rises and binds to a protein called


catabolite activator protein (CAP.). CAP
undergoes a conformational change as a
result of the binding reaction. This
enables CAP to bind to the promoter,
and this in turn facilitates binding of
RNA polymerase to the promoter
f. Another regulation using the operon
model is the regulation thru repression.
A co- repressor needs to bind to the
repressor in order to inhibit the RNAP
from binding to the promoter. This
model is exemplified by the tryptophan
(trp) operon with tryptophan as a co-
repressor for expression of enzymes for
trp synthesis.

2. Eukaryotic gene regulation. Gene control IV. MUTATIONS AND REPAIR


has three main components namely,
signals, levels of regulation, and mechanisms A. Mutations are heritable, unrepaired
of transcriptional control. alteration in nucleotide sequence in DNA.
a. Signals include hormones, protein factors Mutations can be spontaneous or induced
and environmental conditions such as and may or may not be phenotypically
heat shock. expressed. Gene alterations are an
b. Control in the levels of regulation important factor in biological evolution.
involve, nuclear RNA synthesis, On the other hand, when mutation rates
differential processing of primary are too high, they can threaten the
transcripts, and alteration in mRNA survival of individual organisms or entire
stability in the cytoplasm. species. This is why cells possess repair
i. Control of nuclear RNA synthesis is mechanisms that correct most of the DNA
effected mainly at the initiation stage alterations caused by mutations.
Initiation is activated by transcription B. Various mutagenic agents can cause gene
factors (activators) which may alterations. This could arise as a result of
interact with genomic promoters, in physical or chemical damage, from errors
order to guide the RNA polymerase II during DNA replication and recombination,
to the correct expression of an mRNA and post replication errors. Viruses can also
species lead to mutations and cancer.
ii. Differential nuclear processing of C. Mutations may cause cellular death and if
transcripts involves differential unrepaired may affect surviving cells
choice of polyA sites on the primary undergoing organogenesis and cause
transcripts which will determine physical malformations in the organism.
tissue specificity of gene expression. Affected germ cells may lead to sterility or
iii. Cytoplasmic gene control involves genetic disorders while growth regulation
The rate of protein synthesis which in somatic cells may be lost and lead to
could be affected by the rate of cancer. Mutagenic agents causing cancer
transport of mRNA into the can also be classified as carcinogen.
cytoplasm, half-life of the mRNA, D. Tumors can arise when mutations occur
frequency of mRNA translation, and in proto- oncogenes and tumor suppressor
post translational control. genes. Proto-oncogenes code for proteins
involved in normal cell growth and
differentiation but are converted to
oncogenes when mutated. e.g. ras, abl,
src, myc, jun. Tumor suppressor genes or
anti –oncogenes regulate cell growth but
upon mutation their suppressor function is
lost. Tumor suppressor genes are involved
in regulation of cell cycle, apoptosis and
DNA repair.e, g. p53, Rb.
E. Types of mutations include base substitution and
frameshift mutation.
1. Base substitution
a. Transition- a change from a purine
base to another purine base or a
pyrimidine to another pyrimidine. e.g.
adenine to guanine; cytosine to
thymine.
UST FMS MEDICAL BOARD REVIEW 2019 9 | BIOCHEMISTRY
NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

b. Transversion – a change from purine to I. DNA repair mechanisms are efficient and aim
pyrimidine or a pyrimidine to a to maintain cellular function of both germ
purine. e.g. adenine to thymine; . cells to preserve species and of somatic cells
guanine to cytosine to prevent cancer formation. Repair
2. Frameshift mutation; a base deletion or mechanisms include:
base insertion alters the reading frame 1. Excision repair- nuclease removes a
of the codon in the mRNA. complete segment of DNA on both sides
of the error site. The segment is
F. Base substitution and frameshift mutations replaced by DNA polymerase using
lead to: the opposite strand as template. DNA
1. Missense mutation. A different protein ligase closes the gaps
may be translated with a different 2. Photoreactivation- a photolyase binds at
protein and function. e.g. sickle cell the site of the defect (thymine dimer)
anemia and upon illumination, cleaves dimer
2. Nonsense mutation. Fomation of a stop to yield two single bases.
codon prematurely terminates protein 3. Recombinational repair- the region
synthesis containing the defect is omitted during
3. Silent mutation. No change in amino acid replication. The resultant gap is closed
being translated due to degeneracy of by shifting the corresponding segment
the genetic code, and no change in from the correctly replicated strand. The
protein structure and function. new gap formed is then filled by
polymerases and ligases. Finally, the
G. Spontaneous mutations may be caused by original defect is corrected by excision.
tautomeric base mispairs, simple
misalignment of repeated bases, J. Protective mechanisms against mutations
palindromic misalignment and could be inherently present such as the
insertional mutagenesis (jumping genes or protective structural function of cellular and
transposons) among others. nuclear membranes. Protective enzymes
such as superoxide dismutase, glutathione
H. Induced mutations are frequently caused peroxidase, catalase protects the cell from
by various agents such as ionizing the damaging effects of reactive oxygen
radiation and chemical agents. species and radicals. Compounds with
protective groups such as glutathione,
1. Ionizing radiation results in the acetylCoA, some amino acids and antioxidant
production of free radicals and can vitamins and minerals prevent DNA
damage DNA., with thymine the most damage.
radiosensitive and adenine the least
radiosensitive. Short wavelength VI. GENE TECHNOLOGY
ultraviolet light also has a mutagenic
effect on the skin cells, particularly A. Techniques for isolating and amplifying
formation of thymine dimers. Such genes and studying and manipulating DNA
dimers results in errors when DNA is sequences involve the use of restriction
read during replication and enzymes, cloning, polymerase chain
transcription reaction, gel electrophoresis, Southern
blotting and gene sequencing.
2. Chemical agents can be directly
mutagenic or indirectly mutagenic 1. Restriction Analysis involves the use of
when metabolized by the body to a restriction endonucleases. This cuts
mutagen (promutagenic). DNA into reproducible pieces of
a. Intercalating agents such as manageable size. These enzymes, derived
polycyclic aromatic hydrocarbons from bacteria, cleave DNA at specific
insert between successive GC base palindromic restriction sites of 4 to 8
pairs and distorts the helix. This base pairs. e.g. EcoRI, SmaI, HaeIII
interferes with unwinding during
replication. 2. Gel Electrophoresis is used to separate
b. Alkylating agents carry reactive DNA fragments on the basis of size.
groups than can form covalent bonds a. Agarose Gel Electrophoresis is used to
with DNA bases, e.g. separate larger fragments,
methylnitrosoamines, benzo(A) b. Polyachrylamide Gel Electrophoresis
pyrenes, aflatoxin, ethylene separates smaller fragments.; used
dibromide for sequencing DNA and preferred-
c. Deaminating and oxidizing agents technique for protein separation
such as nitrous acid and (SDS-PAGE).
hydroxylamine converts cytosine to
uracil and adenine to inosine. 3. Southern Blotting is used to detect DNA
fragments that contain specific base

UST FMS MEDICAL BOARD REVIEW 2019 10 | BIOCHEMISTRY


NUCLEOTIDE METABOLISM AND MOLECULAR BIOLOGY
JOSE S. BLAS, MD

sequences. After denaturing the DNA in -number of cycles in the program. Each cycle
the gel, fragments are transferred to is programmed to denature the target DNA.
nitrocellulose and the latter is hybridized by high temperature of 90 degrees, followed
with a DNA or RNA probe. The by lowering to 60 degrees to allow the forward
hybridized DNA is detected by auto- and reverse primers to anneal at the 3’ ends
radiography if using a radioactive probe. of the target DNA to be amplified. The
temperature is raised to 72 degrees to allow
4. Restriction Fragment Length extension of primers by polymerization using
Polymorphism (RFLP) uses restriction dNTPs by using heat stable enzyme usually
enzymes to cut DNA from different Taq polymerase.
individuals having different DNA
sequences, followed by probe
hybridization. This results in DNA
fragments of different lengths due to
polymorphisms that exist among
individuals. The technique may use PCR
to amplify DNA prior to digestion by
RE, and gel electrophoresis to separate
DNA fragments and Southern blotting.

5. Cloning is a process of producing


multiple copies of identical DNA. This
could be by use of recombinant DNA in
bacteria (in vivo) or by use of PCR (in
vitro). Cloning in vivo involves production
of DNA inside genetically engineered
cultured bacteria such as E. coli by
recombinant vectors such as plasmids,
thru transformation. This is followed by
selection of recombinant clones inside
the bacteria. The recombinant DNA B. Application of Recombinant DNA
can be isolated from the bacterial colones 1. Production of vaccines and other useful
and can be propagated to create gene proteins, e.g. hormones, growth factors
libraries 2. Genetic counseling
3. Gene therapy
In Vivo Cloning: 4. Production of transgenic animals
5. Production of genetically modified
organisms (GMOs)
5. DNA fingerprinting (forensic applications)

REFERENCES:

Greenstein, Ben, Greenstein Adam, Medical Biochemistry


at a Glance.University Press, Cambridge 1996 pp 38-
39,42-45,52-55 ,90-93,
Koolman, Jan, ; Rohm, Klaus. Color Atlas of
Biochemistry.Thieme- Stuttgart, New York 1996. pp
174- 177; 218-241
Liebermann, Michael et al. Marks’ Basic Medical
Biochemistry, A Clinical Approach, 4th edition
Lippincott Williams and Wilkins, 2013
Rodwell Victor, Bender David et al. Harper’s Illustrated
Biochemistry 31sr edition, International edition.
6. DNA sequencing involves two methods.
Lange.
a. The Sanger-dideoxy method (chain
Swanson, Todd, Kim Sandra,et al BRS Review Series in
termination or enzymatic) Sanger which
Biochemistry, Molecular Biology and Genetics. Fifth
is used frequently and is the technique
Edition, Lippincott pp 203- 210; 251-276; 319-331
in automated sequencing machines.
Wilcox. R. Bruce. High Yield Biochemistry. Lippincott
b. Maxam-Gilbert method (chemical
Williams and Wilkins. pp 54-63; 77-99
method)

7. Polymerase Chain Reaction (PCR) is an


automated in vitro method of cloning
DNA fragments. Introduced by Karl Mullis.
It is used to amplify DNA exponentially
within a short period of time, based on the

UST FMS MEDICAL BOARD REVIEW 2019 11 | BIOCHEMISTRY


REVIEW TEST
JOSE S. BLAS, MD

CHOOSE THE BEST ANSWER: _____ 9. Synthesis de novo of pyrimidine


nucleotides differs from that of purine nucleotides in
_____ 1. The basic building block or unit of nucleic that:
acid: A. PRPP is added after synthesis of the
A. nitrogenous base pyrimidine ring
B. nucleotide B. the C and N atoms are built around the
C. sugar phosphate backbone PRPP molecule
D. nucleoside C. there are more energy requiring steps
D. PRPP is the source of the sugar and
_____ 2. Which of the following is NOT a function of phosphate
nucleotides?
A. Substrate for RNA and DNA synthesis _____ 10. The final end product of purine
B. carriers of high energy groups catabolism in man is:
C. Components of coenzymes A. ammonia
D. Major energy source of metabolic fuel B. allantoin
C. urea
_____ 3. Which of the following is considered a D. uric acid
nucleoside?
A. adenosine _____ 11. In treatment of gout, allopurinol lowers
B. cytosine the blood level of uric acid by inhibiting:
C. guanine A. Xanthine oxidase
D. thymine B. HGPRTase
C. PRPP synthetase
_____ 4. Which of the following best describes the D. uricase
Watson Crick Model of DNA?
A. right-handed double helix with both _____ 12. The anticancer agents, aminopterin and
strands running in parallel direction methotrexate can inhibit DNA synthesis and cellular
B. hydrogen base pairing between purine proliferation by inhibiting:
and pyrimidine A. PRPP amidotransferase
C. adenine + thymine = cytosine + guanine B. dihydrofolate reductase
D. 11 base pairs per helical turn with 2.6nm C. thymidylate synthase
distance D. cabamoyl phosphate synthetase II

_____ 5. Which of the following is true of the RNA _____ 13. The final end product of gene expression:
molecule? A. DNA
A. double stranded nucleic acid in a left B. RNA
handed twist or coil C. Proteins
B. mRNA is derived from DNA thru reverse D. glycoproteins
transcription
C. tRNA is the smallest RNA _____ 14. All of the following is a correct statement
D. eukaryotic rRNA is called 70S with 50S about DNA replication EXCEPT:
and 30S subunits A. Both strands of paternal DNA are copied,
and daughter DNAs are synthesized in
_____ 6. In de novo nucleotide synthesis, which of the 5’  3’ direction
the following are sources of atoms common to both B. Replication is continuous in the leading
purine and pyrimidine rings? strands while discontinuous in the lagging
A. formyl tetrahydrofolate strand
B. glycine C. Replication in eukaryotes is semi
C. aspartate conservative
D. glutamate D. Replication is an error free process

_____ 7. The sugar and phosphate moieties in both _____ 15. A type of point mutation wherein an
purine and pyrimidine nucleotide synthesis comes adenine is replaced by a thymine:
from: A. transition
A. phosphoribosyl pyrophosphate (PRPP) B. transversion
B. carbamoyl phosphate C. transformation
C. glucose 6 phosphate D. transfection
D. adenosine triphosphate
_____ 16. An error free type of DNA repair:
_____ 8. The committed step in purine nucleotide A. Excision repair
synthesis de novo is the synthesis of: B. Recombination repair
A. carbamoyl phosphate C. Photoreactivation repair
B. phosphoribosyl pyrophosphate (PRPP) D. Deletion repair
C. phosphoribosyl amine
D. carbamoyl aspartate

UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY


REVIEW TEST
JOSE S. BLAS, MD

_____ 17. Which of the following statements is


correct of gene transcription? _____ 24. The standard method used in separating
A. Both parental DNA stands are and identification of large DNA fragments in the
transcribed into daughter RNAs in the 5’ laboratory:
 3’ direction A. polyachrylamide gel electrophoresis
B. Initiation of transcription is signaled by B. SDS- PAGE
release of rho factor C. agarose gel electrophoresis
C. RNA polymerase recognizes promoters’ D. paper chromatography
regions to start RNA synthesis
D. RNA primers are required for elongation _____ 25. This automated technique can amplify or
of RNA clone DNA exponentially in a short period of time:
A. Polymerase Chain reaction (PCR)
_____ 18. Which of the following is NOT a post B. Maxam-Gilbert method
transcriptional processing of mRNA? C. Sanger technique
A. polyadenylation D. Southern Blot technique
B. ligation of exons
C. addition of 7 methyl guanosine cap
D. addition of histones

_____ 19. During protein synthesis (translation),


which of the following is a correct statement?
A. Amino acyl-tRNA binds to the smaller
subunit of the ribosome while the mRNA
binds to the larger subunit
B. Incoming amino acyl-tRNA binds to the P
site of the large ribosomal subunit
B. Peptidyl transferase forms a peptide
bond between the amino acid in the A
and P sites
D. Termination of translation occurs when
the codon AUG is encountered in the mRNA

_____ 20. Degeneracy of the genetic code means


that:
A. the genetic code used by all prokaryotes
and eukaryotes is one and the same
B. an amino acid may have more than one
triplet codon
C. a triplet codon can be coded by more
than one amino acid
D. more than one codon can be read by
one tRNA

_____ 21. All of the following have been found to


inhibit translation process of gene expression,
EXCEPT:
A. Chloramphenicol
B. Tetracyline
C. Erythromycin
D. Ofloxacin

_____ 22. All of the following molecular mechanisms


can promote proto-oncogene activation and
carcinogenesis, EXCEPT?
A. point mutation
B. genetic rearrangement
C. activation of p53 gene
D. promoter and enhancer insertion

_____ 23. Enzymes that are used in recombinant


DNA technology, that recognizes and cut palindromic
sequences:
A. Restriction endonucleases
B. DNA exonucleases
C. Taq polymerase
D. T4 DNA ligase
UST FMS MEDICAL BOARD REVIEW 2019 | BIOCHEMISTRY

Potrebbero piacerti anche