You are on page 1of 21

Chapter 14

Carbohydrate Metabolism II:


Gluconeogenesis, Glycogen Synthesis and
Breakdown, and Alternative Pathways

Key Points proteins and by gene expression. Insulin and glucagon have
1. Gluconeogenesis, synthesis of new glucose from opposing effects on glycolysis and gluconeogenesis: insulin
noncarbohydrate precursors, provides glucose when dietary promotes glycolysis and inhibits gluconeogenesis, and the
intake is insufficient or absent (fasting). It also is essential in opposite is true for glucagon (Chapter 20). Cortisol, a
the regulation of acidbase balance, amino acid steroid hormone released from the adrenal cortex as a
metabolism, and synthesis of carbohydrate-derived physiological response to stress, promotes gluconeogenesis
structural components. by stimulating the enzyme synthesis required for the
2. Gluconeogenesis occurs in the liver and kidneys. pathway.
The precursors of gluconeogenesis are lactate, glycerol, and 7. Abnormalities in gluconeogenesis cause hypoglycemia with
amino acids, with propionate making a minor contribution. severe metabolic consequences. These abnormalities can
3. The gluconeogenesis pathway consumes ATP, which is result from a genetic deficiency of enzymes of
derived primarily from the oxidation of fatty acids. gluconeogenesis or of fatty acid oxidation (Chapter 16)
The pathway uses several enzymes of glycolysis with the pathways, ethanol abuse, or the plant-derived toxin
exception of enzymes of the irreversible steps, namely hypoglycin.
pyruvate kinase, 6-phosphofructokinase, and hexokinase. 8. Glucose is stored as glycogen when glucose levels are high
The irreversible reactions of glycolysis are bypassed by in virtually every cell of the body. Glycogen is especially
four alternate, unique reactions of gluconeogenesis. abundant in liver and skeletal muscle. Glycogen is
4. The four unique reactions of gluconeogenesis are degraded when the glucose supply is low. It is converted to
(a) pyruvate carboxylase, a biotin (a vitamin)-dependent glucose-6-phosphate (e.g., in muscle) or glucose,
enzyme, located in the mitochondrial matrix; depending on the presence of the tissue-specific glucose-6-
(b) phosphoenolpyruvate carboxykinase located in phosphatase. The liver and kidneys are able to convert
both mitochondrial matrix and cytosol; (c) fructose- glycogen to glucose. Quantitatively, the liver plays a major
1,6-bisphosphatase located in the cytosol; and (d) glucose- role in the maintenance of optimal blood glucose levels.
6-phosphatase located in the endoplasmic reticulum (ER). 9. Glycogenesis and glycogenolysis are reciprocally regulated
5. Glucose-6-phosphatase, which catalyzes the conversion by several allosteric modulators and hormones (insulin,
of glucose-6-phosphate to glucose in the final step of glucagon, epinephrine, and cortisol). The two key enzymes
gluconeogenesis, is also the final step in the conversion of that are regulated are glycogen synthase in glycogenesis
glycogen to glucose. The enzyme is present only in the ER and glycogen phosphorylase in glycogenolysis. Glucagon
of liver and kidney. and epinephrine activate a cell-membrane-mediated signal
6. Gluconeogenesis is regulated by the overall energy transduction process resulting in production of cAMP,
demands of the body, allosteric effectors, and hormones. which, in turn, activates protein kinase. Phosphorylation
Glycolysis and gluconeogenesis are reciprocally regulated activates glycogen phosphorylase and inactivates glycogen
by allosteric effectors so that both pathways do not occur synthase. Muscle is insensitive to glucagon’s action. Insulin
simultaneously. Acetyl-CoA inhibits pyruvate kinase and promotes glycogen synthesis by activating tyrosine kinase-
activates pyruvate carboxylase; fructose-2,6-bisphosphate mediated amplification systems. The effects of these
activates 6-phosphofructokinase and inhibits fructose-1,6- hormones are tissue specific.
bisphosphatase; and AMP and citrate inhibit 6- 10. Deficiencies of enzymes in glycogenolysis can result in
phosphofructokinase and activate fructose-1,6- glycogen storage disorders, hypoglycemia, or exercise
bisphosphatase. Hormones regulate gluconeogenesis by intolerance, depending on the tissue that is lacking the
means of phosphorylation and dephosphorylation of target enzyme.

N. V. Bhagavan and C.-E. Ha: Essentials of Medical Biochemistry, Second edition. DOI: http://dx.doi.org/10.1016/B978-0-12-416687-5.00014-2
© 2015 Elsevier Inc. All rights reserved. 205
206 Essentials of Medical Biochemistry

11. The terminal CH2OH group of the activated form of


testes, and embryonic tissues, all of which use glucose as
glucose (UDP-glucose) is oxidized to form UDP-glucuronic a major source of fuel. Gluconeogenesis has three addi-
acid (UDP-GA). UDP-GA is essential for the detoxification tional functions:
reactions in the liver for both endogenous metabolites
1. Control of acidbase balance. Production of lac-
(e.g., bilirubin, Chapter 27) and exogenous compounds
tate in excess of its clearance causes metabolic aci-
(e.g., drugs).
12. The monosaccharides fructose and galactose are converted
dosis, and resynthesis of glucose from lactate is a
to glucose by different pathways. Fructose enters the major route of lactate disposal. Since glycolysis is
pathway in the liver via fructokinase and in the muscle and almost totally anaerobic in erythrocytes, renal
kidney via hexokinase. Galactose is converted to glucose in medulla, and some other tissues, even under nor-
a circuitous route involving galactokinase and galactose-1- mal conditions, lactate is continually released.
phosphate uridyltransferase. Deficiencies of the enzymes Other tissues, particularly muscle during vigorous
involved in fructose and galactose metabolism can result in exercise, can produce large amounts of lactate,
serious clinical manifestations. which must be removed; otherwise, lactic acidosis
13. Amino sugars, constituents of glycoproteins and glycolipids, will result (Chapter 19).
are synthesized in pathways that originate from glucose-6-
The continuous conversion of lactate to glucose in
phosphate.
the liver, and of glucose to lactate by anaerobic gly-
14. Oxidation of glucose in the pentose phosphate pathway
(also called hexose monophosphate shunt) produces
colysis, particularly in muscle, forms a cyclical flow
NADPH and ribose-5-phosphate. The pathway consists of of carbon called the Cori cycle. Deamination of
two oxidative reactions followed by several nonoxidative amino acids prior to gluconeogenesis in the kidney
reactions. The intermediates of the pathway can be shuttled also provides a supply of NH3 to neutralize acids
back and forth with some of the intermediates of glycolysis. excreted in the urine (Chapter 37).
NADPH is required for several metabolic reactions such as 2. Maintenance of amino acid balance. Metabolic path-
fatty acid biosynthesis, steroid biosynthesis, hydroxylations, ways for the degradation of most amino acids and for
and antioxidant and oxidant generation reactions. The the synthesis of nonessential amino acids involve
pathway is active in tissue cells such as adipose tissue, some steps of the gluconeogenic pathway. Imbalances
liver, mammary glands, adrenal cortex, red blood cells, and
of most amino acids, whether due to diet or to an
leukocytes. The pentose phosphate pathway occurs in
altered metabolic state, are usually corrected in the
cytosol and begins with the first of two oxidation steps
initiated by glucose-6-phosphate dehydrogenase (G6PD).
liver by degradation of the excess amino acids or by
In red blood cells, G6PD deficiency can cause hemolysis, synthesis of the deficient amino acids through gluco-
and in leukocytes, deficiency disrupts the phagocytosis of neogenic intermediates.
bacteria. 3. Provision of biosynthetic precursors. In the absence of
adequate dietary carbohydrate intake, gluconeogenesis
supplies precursors for the synthesis of glycoproteins,
glycolipids, and structural carbohydrates.
In this chapter, carbohydrate metabolism is discussed in Gluconeogenesis from pyruvate is essentially the
terms of nondietary sources of glucose and nonglycolytic reverse of glycolysis, with the exception of three nonequi-
pathways. Gluconeogenesis and glycogen synthesis and librium reactions (Figure 14.1). These reactions are
breakdown make up the first category, while the pentose
phosphate pathway (also called hexose monophosphate hexokinase or glucokinase
Glucose1ATP42 !
shunt) and the glucuronic acid pathway make up the sec- (14.1)
ond. Metabolic pathways of fructose, galactose, and some glucose-6-phosphate22 1 ADP32 1 H1
other sugars belong in both categories.
6-phosphofructokinase
Fructose-6-phosphate22 1 ATP42 !
fructose-1;6-bisphosphate42 1 ADP32 1 H1
GLUCONEOGENESIS (14.2)
Metabolic Role
Phosphoenolpyruvate32 1 ADP32 1 H1
Gluconeogenesis (literally, “formation of new sugar”) is pyruvate kinase (14.3)
the metabolic process by which glucose is formed from ! pyruvate2 1 ATP42
noncarbohydrate sources, such as lactate, amino acids,
and glycerol. Gluconeogenesis provides glucose when In gluconeogenesis, these reactions are bypassed by
dietary intake is insufficient to supply the requirements of alternate steps also involving changes in free energy and
the brain and nervous system, erythrocytes, renal medulla, also physiologically irreversible reactions.
Carbohydrate Metabolism II Chapter | 14 207

Pyruvate Cytosol Intermembrane Mitochondrial


ATP + CO2 + H2O space matrix
Pyruvate carboxylase (in mitochondria)
ADP + Pi
Oxaloacetate Oxaloacetate
Oxaloacetate
GTP NADH+H + NADH+H +
Phosphoenolpyruvate carboxykinase 1
GDP + CO2 (in mitochondria and cytosol) 3
NAD+ NAD+
Phosphoenolpyruvate
2
H2O Malate Malate

2-Phosphoglycerate
Malate shuttle

3-Phosphoglycerate Oxaloacetate Oxaloacetate


ATP
ADP NAD+

1,3-Bisphosphoglycerate Glutamate Glutamate


7 4 5
NADH + H + α-Keto- α-Keto-
NAD+ glutarate glutarate
Pi
NH4+ +
Aspartate
6
Aspartate NADH+H +
Glyceraldehyde-3-phosphate Dihydroxyacetone phosphate

Aspartate shuttle
FIGURE 14.2 Shuttle pathways for transporting oxaloacetate from
Fructose-1,6-bisphosphate mitochondria into the cytosol. The shuttles are named for the molecule
H2O that actually moves across the mitochondrial membrane. 1 and 3 5 malate
Fructose-1,6-bisphosphatase
Pi dehydrogenase; 2 5 malate translocase; 4 and 7 5 aspartate aminotrans-
ferase; 5 5 glutamate dehydrogenase; 6 5 aspartate translocase.
Fructose-6-phosphate

Like many CO2-fixing enzymes, pyruvate carboxylase


Glucose-6-phosphate contains biotin bound through the ε-NH2 of a lysyl resi-
H2O Glucose-6-phosphatase (endoplasmic due (Chapter 16).
Pi reticulum)
The second reaction is the conversion of oxaloacetate
Glucose to phosphoenolpyruvate:
FIGURE 14.1 Pathway of gluconeogenesis from pyruvate to glucose.
Only enzymes required for gluconeogenesis are indicated; others are from Oxaloacetate22 1 GTP42 ðor ITP42 Þ
glycolysis. The overall reaction for the synthesis of one molecule of glucose phosphoenolpyruvate carboxykinase ðPEPCKÞ
from two molecules of pyruvate is 2 Pyruvate 1 4ATP42 1 2GTP42 1 !
2NADH 1 2H1 1 6H2O-Glucose 1 2NAD1 1 4ADP32 1 2GDP32 1
6Pi22 1 4H1. phosphoenolpyruvate32 1 CO2 1 GDP32 ðor IDP32 Þ
In this reaction, inosine triphosphate (ITP) can substi-
Conversion of pyruvate to phosphoenolpyruvate involves tute for guanosine triphosphate (GTP), and the CO2 lost is
two enzymes and the transport of substrates and reactants the one gained in the carboxylase reaction. The net result
into and out of the mitochondrion. In glycolysis, conversion of these reactions is
of phosphoenolpyruvate to pyruvate results in the formation Pyruvate 1 ATP 1 GTP ðor ITPÞ-
of one high-energy phosphate bond. In gluconeogenesis, two phosphoenolpyruvate1ADP1Pi 1 GDP ðor IDPÞ
high-energy phosphate bonds are consumed (ATP-ADP 1
Pi; GTP-GDP 1 Pi) in reversing the reaction. Pyruvate carboxylase is a mitochondrial enzyme in
Gluconeogenesis begins when pyruvate, generated in the animal cells. In humans (and guinea pigs), PEPCK occurs
cytosol, is transported into the mitochondrion—through the in both mitochondria and cytosol.
action of a specific carrier—and converted to oxaloacetate: In humans, oxaloacetate must be transported out of the
mitochondrion to supply the cytosolic PEPCK. Because there
42 acetyl-CoA; Mg
21
is no mitochondrial carrier for oxaloacetate and its diffusion
Pyruvate2 1 HCO2
3 1 ATP !
pyruvate carboxylase across the mitochondrial membrane is slow, it is transported
1
oxaloacetate22 1 ADP32 1 P22
i 1H as malate or aspartate (Figure 14.2). The malate shuttle
208 Essentials of Medical Biochemistry

carries oxaloacetate and reducing equivalents, whereas the prolonged starvation, however, this proportion decreases,
aspartate shuttle, which does not require a preliminary reduc- while that synthesized in the kidney increases to nearly
tion step, depends on the availability of glutamate and half of the total, possibly in response to a need for NH3 to
α-ketoglutarate in excess of tricarboxylic acid (TCA) cycle neutralize the metabolic acids eliminated in the urine in
requirements. The proportion of oxaloacetate carried by each increased amounts (Chapter 20).
shuttle probably depends on the redox state of the cytosol. If Gluconeogenesis is a costly metabolic process.
most of the pyruvate is derived from lactate, the NADH/ Conversion of two molecules of pyruvate to one of
NAD1 ratio in the cytosol is elevated. In this situation, there glucose consumes six high-energy phosphate bonds
is no need to transport reducing equivalents out of the mito- (4ATP 1 2GTP-4ADP 1 2GDP 1 6Pi) and results in the
chondria, and the aspartate shuttle predominates. However, oxidation of two NADH molecules (Figure 14.1). In contrast,
if alanine is the principal source of pyruvate, no cytosolic glycolytic metabolism of one molecule of glucose to two of
reduction occurs, and the glyceraldehyde-phosphate dehy- pyruvate produces two high-energy phosphate bonds
drogenase reaction (which requires NADH) requires trans- (2ADP 1 2Pi-2ATP) and reduces two molecules of NAD1.
port of reducing equivalents via the malate shuttle. In For gluconeogenesis to operate, the precursor supply and the
species in which oxaloacetate is converted in mitochondria energy state of the tissue must be greatly increased. Using
to phosphoenolpyruvate (which is readily transported to the some gluconeogenic precursors to provide energy (via glyco-
cytosol, perhaps via its own carrier system), no transport of lysis and the TCA cycle) and to convert the remainder of the
oxaloacetate or reducing equivalents is required. precursors to glucose would be inefficient, even under aero-
Phosphoenolpyruvate is converted to fructose-1,6- bic conditions. Usually, the catabolic signals (catecholamines,
bisphosphate by reversal of glycolysis in the cytosol via cortisol, and increase in glucagon/insulin ratio) that increase
reactions that are at near-equilibrium and whose direction the supply of gluconeogenic precursors also favor lipolysis,
is dictated by substrate concentration. Conversion of fruc- which provides fatty acids to supply the necessary ATP.
tose-l,6-bisphosphate to fructose-6-phosphate is a nonequi- When amino acid carbons are the principal gluconeo-
librium step, catalyzed by fructose-l,6-bisphosphatase: genic precursors, the metabolic and physiological debts
are particularly large compared to those incurred when
Mg21
Fructose-1;6-bisphosphate42 1 H2 O! lactate or glycerol is used. Amino acids are derived
fructose-6-phosphate22 1 P22
i
through breakdown of muscle protein, which is accompa-
nied by a loss of electrolytes and tissue water.
Fructose-6-phosphate is then converted to glucose-6-
phosphate by reversal of another near-equilibrium reaction of
glycolysis. In the last reaction in gluconeogenesis, glucose-6- Gluconeogenic Precursors
phosphate is converted to glucose by glucose-6-phosphatase: Gluconeogenic precursors include lactate, alanine and several
Glucose-6-phosphate22 1 H2 O-glucose1P22
i
other amino acids, glycerol, and propionate (Chapter 16).
Lactate, the end product of anaerobic glucose metabo-
Glucose-6-phosphatase is part of a multicomponent lism, is produced by most tissues of the body, particularly
integral membrane protein system that consists of six dif- skin, muscle, erythrocytes, brain, and intestinal mucosa.
ferent proteins located in the endoplasmic reticulum of In a normal adult, under basal conditions, these tissues
liver, kidney, and intestine, but not of muscle or adipose produce 1300 mM of lactate per day, and the normal
tissue. Deficiency of any one of these five proteins leads serum lactate concentration is less than 1.2 mM/L. During
to glycogen storage disease (discussed later). vigorous exercise, the production of lactate can increase
Thus, gluconeogenesis requires the participation of several-fold. Lactate is normally removed from the circu-
enzymes of the cytosol, mitochondrion, and smooth endo- lation by the liver and kidneys. Because of its great
plasmic reticulum, as well as of several transport systems, capacity to use lactate, liver plays an important role in the
and it may involve more than one tissue. The complete glu- pathogenesis of lactic acidosis, which may be thought of
coneogenic pathway, culminating in the release of glucose as an imbalance between the relative rates of production
into the circulation, is present only in liver and kidney. and utilization of lactate (Chapter 37).
Most tissues contain only some of the necessary enzymes. Alanine, derived from muscle protein and also synthe-
These “partial pathways” are probably used in glycerogen- sized in the small intestine, is quantitatively the most impor-
esis and in replenishing tricarboxylic acid (TCA) intermedi- tant amino acid substrate for gluconeogenesis. It is converted
ates. Muscle can also convert lactate to glycogen, but this to pyruvate by alanine aminotransferase (Chapter 15):
probably takes place only in one type of muscle fiber and
B6 PO4
only when glycogen stores are severely depleted and lactate Alanine 1 α-ketoglutarate22 ! pyruvate2
concentrations are high, such as after heavy exercise. 1 glutamate2
Under normal conditions, the liver provides 80% or
more of the glucose produced in the body. During where B6-PO4 5 pyridoxal phosphate.
Carbohydrate Metabolism II Chapter | 14 209

Cysteine FIGURE 14.3 Points of entry of amino acids into


Alanine the gluconeogenesis pathway.
Glycine
Hydroxyproline
Serine
Glucose Tryptophan
Aspartate
Pyruvate
Oxaloacetate
Oxaloacetate Acetyl-CoA

Malate
Citrate

Tyrosine
phenylalanine Fumarate α-Ketoglutarate

Arginine
Glutamate
Glutamine
Succinyl-CoA
Histidine
Proline

Isoleucine
Methionine
Valine
Threonine

An amino acid is classified as ketogenic, glucogenic, Glycerol 3-phosphate is oxidized to dihydroxyacetone


or glucogenic/ketogenic depending on whether feeding phosphate by glycerol-3-phosphate dehydrogenase:
it to a starved animal increases plasma concentrations of
ketone bodies (Chapter 16) or of glucose. Leucine and CH2OH CH2OH
lysine are purely ketogenic because they are catabolized CHOH + NAD+ C O + NADH + H+
to acetyl-CoA, which cannot be used to synthesize glu-
2− 2−
cose (Chapter 15). Isoleucine, phenylalanine, tyrosine, CH2OPO3 CH2OPO3
and tryptophan are both glucogenic and ketogenic, and
the remaining amino acids (including alanine) are gluco- Glycerol 3-phosphate Dihydroxyacetone phosphate
genic. Entry points of the amino acids into the gluconeo-
genesis pathway are shown in Figure 14.3. Dihydroxyacetone phosphate is the entry point of
Glycerol is more reduced than the other gluconeo- glycerol into gluconeogenesis. Glycerol cannot be metab-
genic precursors, and it results primarily from triacyl- olized in adipose tissue, which lacks glycerol kinase, and
glycerol hydrolysis in adipose tissue. In liver and the glycerol 3-phosphate required for triacylglycerol syn-
kidney, glycerol is converted to glycerol 3-phosphate by thesis in this tissue is derived from glucose (Chapter 20).
glycerol kinase: During fasting in a resting adult, about 210 mM of glyc-
erol per day is released, most of which is converted to
CH2OH CH2OH glucose in the liver. During stress or exercise, glycerol
CHOH + ATP4− CHOH + ADP3− + H+ release is markedly increased (Chapter 20).

CH2OH CH2OPO3
2− Propionate is not a quantitatively significant gluconeo-
genic precursor in humans, but it is a major source of glucose
Glycerol Glycerol 3-phosphate in ruminants. It is derived from the catabolism of isoleucine,
210 Essentials of Medical Biochemistry

valine, methionine, and threonine; from β-oxidation of odd- activators of 6-phosphofructo-1-kinase (PFK-1), the com-
chain fatty acids; and from the degradation of the side chain peting glycolytic enzyme (Figure 14.4). This inhibition
of cholesterol. Propionate enters gluconeogenesis via the prevents the simultaneous activation of the two enzymes
TCA cycle after conversion to succinyl-CoA (Chapter 16). and helps prevent a futile ATP cycle.

Regulation of Gluconeogenesis Conversion of Glucose-6-Phosphate to


Gluconeogenesis is regulated by the production and Glucose
release of precursors and by the activation and inactiva- Glucose-6-phosphatase is regulated only by the concentra-
tion of key enzymes. Glucagon and glucocorticoids stimu- tion of glucose-6-phosphate, which is increased by gluco-
late gluconeogenesis, whereas insulin suppresses it. See corticoids, thyroxine, and glucagon.
Chapter 20 for a discussion of the overall metabolic con-
trol and physiological implications of the several seem- ATP ADP
Glucose Glucose-6-phosphate Gluconeogenesis
ingly competing pathways.
Pi
Carboxylation of Pyruvate to Oxaloacetate
The pyruvate carboxylase reaction is activated by Mg21
and, through mass action, by an increase in either the
[ATP]/[ADP] or the [pyruvate]/[oxaloacetate] ratio. It is
virtually inactive in the absence of acetyl-CoA, an alloste- For extrahepatic Glycogen Glycolysis Pentose
ric activator. The enzyme is allosterically inhibited by tissue utilization synthesis phosphate
pathway
glutamate, since oxaloacetate formed in excess would
flood the TCA cycle and result in a buildup of α-ketoglu-
tarate and glutamate. Abnormalities of Gluconeogenesis
Glucose is the predominant fuel for cells that depend
Conversion of Oxaloacetate to largely on anaerobic metabolism, cells that lack mito-
Phosphoenolpyruvate chondria, and tissues such as brain that normally cannot
use other metabolic fuels. An adult brain represents only
Short-term regulation of this reaction is accomplished by
2% of total body weight, but it oxidizes about 100 g of
changes in the relative proportions of substrates and pro-
glucose per day, accounting for 25% of the basal metabo-
ducts. Increased concentrations of oxaloacetate and GTP
lism. Brain cannot utilize fatty acids because they are
(or ITP) increase the rate, and accumulation of phospho-
bound to serum albumin and so do not cross the blood
enolpyruvate and GDP (or IDP) decreases it. The cyto-
brain barrier. Ketone bodies are alternative fuels, but their
solic PEPCK is also under long-term regulation by
concentration in blood is negligible, except in prolonged
hormones. Its synthesis is increased by corticosteroids.
fasting or diabetic ketoacidosis (Chapters 16 and 20).
Starvation and diabetes mellitus increase the synthesis of
Brain and liver glycogen stores are small relative to the
cytosolic PEPCK, whereas refeeding and insulin have the
needs of the brain, and gluconeogenesis is essential for
opposite effect.
survival. Decreased gluconeogenesis leads to hypoglyce-
mia and may cause irreversible damage to the brain.
Conversion of Fructose-1,6-Bisphosphate to Blood glucose levels below 40 mg/dL (2.2 mM/L) in
Fructose-6-Phosphate adults or below 30 mg/dL (1.7 mM/L) in neonates repre-
This reaction is catalyzed by fructose-1,6-bisphosphatase sent severe hypoglycemia. The low levels may result
(FBPase-l). It is inhibited by AMP, inorganic phosphate, from increased glucose utilization, decreased glucose pro-
and fructose-2,6-bisphosphate, all of which are allosteric duction, or both. Increased glucose utilization can occur

FIGURE 14.4 Regulation of liver 6-phosphofructokinase and fructose-1,6-bisphosphatase. These multimodulated enzymes catalyze nonequilibrium
reactions, the former in glycolysis and the latter in gluconeogenesis. Note the dual action of fructose-2,6-bisphosphate (F-2,6-BP), which activates
phosphofructokinase (PFK-1) and inactivates fructose-1,6-bisphosphatase. The activity of F-2,6-BP is under hormonal and substrate regulation.
" 5 positive effectors; ~ 5 negative effectors.
Carbohydrate Metabolism II Chapter | 14 211

in hyperinsulinemia secondary to an insulinoma. In a dia- H2


C NH2
betic pregnancy, the mother’s hyperglycemia leads to OH
H2
fetal hyperglycemia and causes fetal and neonatal hyper- H2C C C C C C
insulinemia. Insufficiency of glucocorticoids, glucagon, or H H
Hypoglycin A
growth hormone and severe liver disease can produce O
hypoglycemia by depressing gluconeogenesis (Chapters 20 α-Ketoglutarate
and 30). Ethanol consumption can cause hypoglycemia,
Transamination
since a major fraction of ethanol is oxidized in the liver by
cytosolic alcohol dehydrogenase, and the NADH and Glutamate
acetaldehyde generated inhibit gluconeogenesis:
H2
CH3 CH2 OH1NAD1 -CH3 CHO1NADH1H1 C O
OH
Excessive NADH decreases the cytosolic [NAD1]/ H2C C C
H2
C C C
[NADH] ratio, increasing lactate dehydrogenase activity H
and thereby increasing conversion of pyruvate to lactate. O
The decrease in pyruvate concentration inhibits pyruvate Methylenecyclopropyl α-ketopropionic acid
carboxylase. Acetaldehyde inhibits oxidative phosphory-
lation, increasing the [ADP]/[ATP] ratio, promoting gly- NAD+
colysis, and inhibiting gluconeogenesis. Hypoglycemia CoASH
and lactic acidosis are common findings in chronic
Oxidative decarboxylation
alcoholics.
Pyruvate carboxylase deficiency can cause intermittent CO2
hypoglycemia, ketosis, severe psychomotor retardation, NADH + H+
and lactic acidosis. The deficiency of either cytosolic or
mitochondrial isoenzyme forms of phosphoenolpyruvate H2 O
carboxykinase (PEPCK) is characterized by a failure of C
gluconeogenesis. Both these disorders are rare, and the H2
H2 C C C C C S CoA
mitochondrial PEPCK deficiency is inherited as an auto- H
somal recessive trait. The major clinical manifestations Methylenecyclopropyl α-ketoacetyl-CoA
include hypoglycemia, lactic acidosis, hypotonia, hepato- FIGURE 14.5 Hypoglycin A metabolism in the liver. Hypoglycin A is
megaly, and failure to thrive. The treatment is supportive converted to a toxic metabolite, methylenecyclopropyl α-ketopropionyl-
and is based on symptoms. Fructose-1,6-bisphosphatase CoA, which inhibits several acyl-CoA dehydrogenases and thereby inhi-
bits mitochondrial fatty acid oxidation. Inhibition of fatty acid oxidation
deficiency, inherited as an autosomal recessive trait,
causes decreased rates of gluconeogenesis due to short supply of ATP,
severely impairs gluconeogenesis, causing hypoglycemia, which induces hypoglycemia.
ketosis, and lactic acidosis. Glucose-6-phosphatase defi-
ciency, also an autosomal recessive trait, causes a similar
condition but with excessive deposition of glycogen in Overactivity of gluconeogenesis due to increased
liver and kidney (discussed later). secretion of catecholamines, cortisol, or growth hormone,
Hypoglycin A (2-methylenecyclopropylalanine), the or an increase in the glucagon/insulin ratio (Chapter 20)
principal toxin of the unripe akee fruit indigenous to west- leads to hyperglycemia and causes many metabolic
ern Africa that also grows in Central America and the problems.
Caribbean, produces severe hypoglycemia when ingested. Metformin, a commonly used drug to treat type 2 diabe-
Hypoglycin A causes hypoglycemia by inhibiting gluco- tes, inhibits hepatic gluconeogenesis, leading to reduction in
neogenesis. In the liver, hypoglycin A forms nonmetabo- plasma glucose levels. The mechanism of action of metfor-
lizable esters with CoA (Figure 14.5) and carnitine, min in suppressing hepatic gluconeogenesis is not
depleting the CoA and carnitine pools, and thereby inhi- completely understood. Several mechanisms of action may
biting fatty acid oxidation (Chapter 16). Since the princi- be contributory to metformin’s hypoglycemic effect. They
pal source of ATP for gluconeogenesis is mitochondrial include (1) decreased ATP production required for gluco-
oxidation of long-chain fatty acids, gluconeogenesis is neogenesis by inhibiting complex I of OXPHOS,
stopped. Intravenous administration of glucose relieves (2) decreased glucagon-stimulated cAMP production, and
the hypoglycemia. Inherited fatty acid oxidation defects (3) activation of the AMP-activated protein kinase (AMPK)
that lead to a deficit in ATP production can cause hypo- system. AMPK orchestrates cellular energy homeostasis
glycemia (Chapter 16). (Chapter 20).
212 Essentials of Medical Biochemistry

GLYCOGEN METABOLISM primarily in response to the availability of glucose and


gluconeogenic precursors. Muscle glycogen stores vary
Animals have developed a method of storing glucose less in response to dietary signals, but they depend on the
that reduces the need to catabolize protein as a source of rate of muscular contraction and of oxidative metabolism
gluconeogenic precursors between meals. The principal (tissue respiration).
storage form of glucose in mammals is glycogen. Glycogenesis (glycogen synthesis and storage) and
Storage of glucose as a polymer reduces the intracellular glycogenolysis (glycogen breakdown) are separate meta-
osmotic load, thereby decreasing the amount of water of bolic pathways that have only one enzyme in common,
hydration and increasing the energy density of the stored namely, phosphoglucomutase. Glycogen synthesis and
glucose. breakdown are often reciprocally regulated, so that stim-
In muscle, glycogen is stored in the cytosol and endo- ulation of one inhibits the other. The control mechan-
plasmic reticulum as granules, called β-particles, each of isms are more closely interrelated than those for
which is an individual glycogen molecule. In the liver, glycolysis and gluconeogenesis, perhaps because the
the β-particles aggregate, forming larger, rosette-shaped glycogen pathways ensure the availability of only one
α-particles that can be seen with the electron microscope. substrate, whereas intermediates of several other meta-
The average molecular weight of glycogen is several mil- bolic pathways are produced and metabolized in glycol-
lion (10,00050,000 glucose residues per molecule). The ysis and gluconeogenesis.
storage granules also contain the enzymes needed for gly-
cogen synthesis and degradation, and for regulation of
these two pathways. Glycogen Synthesis
Glycogen is present in virtually every cell in the body,
but it is especially abundant in liver and skeletal muscle. Glycogenesis begins with the phosphorylation of glucose
The amount stored in tissues varies greatly in response to by glucokinase in liver and by hexokinase in muscle and
metabolic and physiological demands, but in a resting other tissues (Chapter 12):
individual after a meal, liver usually contains roughly
Mg21
4%7% of its wet weight as glycogen and muscle about Glucose1ATP42 !glucose-6-phosphate22
1%. Since the body contains 10 times more muscle than 1 ADP32 1 H1
hepatic tissue, the total amount of glycogen stored in
muscle is greater than that in liver. The second step in glycogenesis is conversion of
Muscle needs a rapidly available supply of glucose to glucose-6-phosphate to glucose-1-phosphate by phospho-
provide fuel for anaerobic glycolysis when, during bursts glucomutase in a reaction similar to that catalyzed by
of muscle contraction, blood may provide inadequate sup- phosphoglyceromutase.
plies of oxygen and fuel. The liver, under most condi- In the third step, glucose-1-phosphate is converted to
tions, oxidizes fatty acids for this purpose; however, liver uridine diphosphate (UDP)-glucose, the immediate pre-
is responsible for maintaining blood glucose levels during cursor of glycogen synthesis, by reaction with uridine
short fasts, and it integrates the supply of available fuels triphosphate (UTP). This reaction is catalyzed by glucose-
with the metabolic requirements of other tissues in differ- 1-phosphate uridylyltransferase (or UDP-glucose pyro-
ent physiological states. Liver glycogen content changes phosphorylase) (Figure 14.6).

HN
CH 2 OH

O O N
O O
H2
UTP4– + Glucose-1-phosphate2– OH C
O + PPi4–
β α
O P O P O
OH

OH
O– O–
OH OH
UDP-glucose
FIGURE 14.6 Formation of UDP-glucose for glycogenesis.
Carbohydrate Metabolism II Chapter | 14 213

Glycogenin Tyrosyl group

Mg2+
Autocatalysis UDP-glucose
UDP
Glucosyl unit

Autocatalysis UDP-glucose
UDP

UDP-glucose
Glycogen synthase
UDP

Branching enzyme

Glycogen synthase
and branching enzyme UDP-glucose
UDP
Mature glycogen (60,000 glucosyl units)
(β-particle)
FIGURE 14.7 Schematic representation of glycogen biosynthesis. — , Glucosyl units in (1-6) linkage.

The reaction is freely reversible, but it is rendered The branching of the glycogen is accomplished by
practically irreversible by hydrolysis of the product pyro- transferring a minimum of six α(1-4) glucan units from
phosphate (PPi) through the action of pyrophosphatase: the elongated external chain into the same or a neighbor-
ing chain by α(l-6) linkage. This reaction of α(l-4) to
PPi 1 H2 O-2Pi
α(1-6) transglucosylation creates new nonreducing ends
Thus, two high-energy phosphate bonds are hydrolyzed and is catalyzed by a branching enzyme. A mature glyco-
in the formation of UDP-glucose. Nucleoside diphosphate gen particle is spherical, containing one molecule of gly-
sugars are commonly used as intermediates in carbohy- cogenin and up to 60,000 glucosyl units (β-particles). In
drate condensation reactions. The high-energy bond the liver, 2040 β-particles are aggregated into rosettes,
between the sugar and the nucleoside diphosphate provides known as α-particles.
the energy needed to link the new sugar to the nonreducing As discussed previously, the glycogen synthesis cata-
component of another sugar or polysaccharide. lyzed by glycogen synthase consists of a glycosyl moiety
The next step in the biogenesis of glycogen is addition from UDP-glucose being transferred to a nonreducing end
of a glucosyl residue at the C-1 position of UDP-glucose of a growing glycan chain with the release of UDP. In a
to a tyrosyl group located at position 194 of the enzyme very rare instance, presumably as a catalytic error, a
glycogenin, which is a Mg21-dependent autocatalytic β-phosphate residue of the substrate UDP-glucose (see
reaction. Glycogenin (M.W. 37,000) extends the glucan Figure 14.6) is added to a glucose residue of a growing
chain, again by autocatalysis, by six to seven glucosyl glycan chain by the glycogen synthase. In this reaction,
units in α(1-4) glycosidic linkages using UDP-glucose the product is UMP. However, a phosphatase enzyme
as substrate (Figure 14.7). The glucan primer of glyco- known as laforin subsequently removes the inappropri-
genin is elongated by glycogen synthase using UDP- ately incorporated phosphate group. The deficiency of
glucose. Initially, the primer glycogenin and glycogen laforin leads to serious clinical consequences [13].
synthase are firmly bound in a 1:1 complex. As the glu-
can chain grows, glycogen synthase dissociates from gly-
cogenin. Deficiency of glycogenin leads to lack of
Glycogen Breakdown
glycogen synthases in muscle tissues and is associated Glycogenolysis is catalyzed by two enzymes unique to
with clinical manifestations including cardiac malfunc- the pathway: glycogen phosphorylase and debranching
tion (see Clinical Case Study 14.1). enzyme. The former normally regulates the rate of
214 Essentials of Medical Biochemistry

Inaccessible to
phosphorylase
α(1→ 6) bonds; all other
linkages are α(1→ 4) Glycogen
phosphorylase

14Pi 14 G-1-P Inaccessible to


phosphorylase

A portion of glycogen Debranching enzyme


(α1,4 → α1,4 glucan
transferase) activity

H2O
Debranching enzyme
(amylo-1,6 glucosidase)
16 G-1-P activity
Glucose
Glycogen
phosphorylase 16Pi

10 Pi
Glucose Glycogen
Debranching
phosphorylase
enzyme 10 G-1-P
H2O
Debranching
enzyme

FIGURE 14.8 Catabolism of a small glycogen molecule by glycogen phosphorylase and debranching enzyme. P i 5 inorganic phosphate;
G-1-P 5 glucose-1-phosphate.

glucose release from glycogen. The progressive degrada- enter the glycolytic pathway, but if glucose-6-phosphatase
tion of glycogen is illustrated in Figure 14.8. Glycogen is present, free glucose can be formed.
phosphorylase catalyzes the release of glucose-1-
phosphate from the terminal residue of a nonreducing end
of a glycogen branch by means of phosphorolysis. A mol- Regulation of Glycogen Metabolism
ecule of inorganic phosphate attacks the C1 side of an
Metabolism of glycogen in muscle and liver is regulated
α(1-4) glycosidic bond, leaving a hydroxyl group on C4
primarily through reciprocal control of glycogen synthase
that remains in the glycogen polymer:
and glycogen phosphorylase. The activities of these
Phosphorylase enzymes vary according to the metabolic needs of the tis-
Glycogen ðglucosyl residues nÞ 1 Pi !
sue (as in muscle) or of other tissues that use glucose as a
glycogen ðn 2 1 glucosyl residuesÞ 1 glucose-1-phosphate
fuel (as in liver). Proximal control is exerted on synthase
The energy stored in the α(1-4) glycosidic bond dur- and phosphorylase by phosphorylation/dephosphorylation
ing the condensation reaction in glycogen synthesis is suf- and by allosteric effectors such as glucose, glucose-6-
ficient to permit the formation of a glucosephosphate phosphate, and several nucleotides (ATP, ADP, AMP,
bond without using ATP. and UDP). Glucagon and epinephrine activate a cell
Glucose-1-phosphate is next converted by phosphoglu- membranemediated signal transduction cascade result-
comutase to glucose-6-phosphate. The latter may then ing in production of cAMP, which, in turn, activates
Carbohydrate Metabolism II Chapter | 14 215

protein kinase followed by phosphorylation of target pro- pathway (discussed later), it provides biosynthetic precur-
teins. Phosphorylation activates glycogen phosphorylase, sors and interconverts some less common sugars to ones
whereas it inactivates glycogen synthase. that can be metabolized. The first steps are identical to
those of glycogen synthesis, i.e., formation of glucose-6-
phosphate, its isomerization to glucose-1-phosphate, and
Glycogen Storage Diseases activation of glucose-1-phosphate to form UDP-glucose.
The group of glycogen storage diseases is characterized UDP-glucose is then oxidized to UDP-glucuronic acid by
by the accumulation of normal or abnormal glycogen due NAD1 and UDP-glucose dehydrogenase (Figure 14.10).
to a deficiency of one of the enzymes of glycogen metab- UDP-glucuronic acid is utilized in biosynthetic reac-
olism. Although all are rare (overall incidence of tions that involve condensation of glucuronic acid with a
B1:25,000 births), they have contributed greatly to the variety of molecules to form an ether (glycoside), an
understanding of glycogen metabolism. They are summa- ester, or an amide, depending on the nature of the accep-
rized in Table 14.1 and Figure 14.9. tor molecule. As in condensation reactions that use nucle-
Infants with type I disease (subtypes Ia, Ib, Ic, and Id) otide sugars as substrates, the high-energy bond between
develop hypoglycemia even after feeding because of their UDP and glucuronic acid provides the energy to form the
inability to convert glucose-6-phosphate to glucose. new bond in the product. UDP is hydrolyzed to UMP (uri-
Lactate is produced at a high rate in extrahepatic tissues dine monophosphate) and inorganic phosphate, further
and is transported to the liver for gluconeogenesis. ensuring the irreversibility of the coupling reaction.
Many characteristics of type I disease are attributed to Because glucuronic acid is highly polar, its conjugation
the attendant hypoglycemia, and patients have been trea- with less polar compounds such as steroids, bilirubin, and
ted with frequent daytime feedings and continuous noctur- some drugs can reduce their activity and make them more
nal intragastric feeding with a high-glucose formula. This water-soluble, thus facilitating renal excretion. Glucuronic
regimen produces substantial improvement in growth, acid is a component of the structural polysaccharides called
reduction in hepatomegaly, and normalization of other glycosaminoglycans (hyaluronic acid and other connective
biochemical parameters. Feeding uncooked cornstarch tissue polysaccharides; see Chapter 10). Glucuronic acid is
every 6 hours resulted in normoglycemia, resumption of usually not a component of glycoproteins or glycolipids.
normal growth, and reduction in substrate cycling and
liver size. The success of this simple nutritional therapy is
thought to depend on slow hydrolysis of uncooked starch
Fructose and Sorbitol Metabolism
in the small intestine by pancreatic amylase, with continu- Fructose is a ketohexose found in honey and a wide vari-
ous release and absorption of glucose. (Cornstarch that is ety of fruits and vegetables. Combined with glucose in an
cooked or altered in other ways is ineffective, presumably α(1-2)β linkage, it forms sucrose. It makes up one-sixth
because of too rapid hydrolysis.) The therapy is ineffec- to one-third of the total carbohydrate intake of most indi-
tive in patients with low pancreatic amylase activity due, viduals in industrialized nations.
for example, to prematurity. Sorbitol, a sugar alcohol, is a minor dietary constitu-
Defects in the glucose-6-phosphatase system are asso- ent. It can be synthesized in the body from glucose by
ciated with severe chronic neutropenia due to altered glu- NADPH-dependent aldose reductase (Figure 14.11). It is
cose metabolism in neutrophils. clinically important because of its relationship to cataract
Myophosphorylase deficiency (also known as formation in diabetic patients. Fructose and sorbitol are
McArdle’s disease) is a common form of glycogen storage catabolized by a common pathway (Figure 14.11).
disease. It exhibits pain, cramps, and fatigue during exer- Fructose transport and metabolism are insulin-indepen-
cise. Sucrose ingestion before exercise in these subjects, the dent; only a few tissues (e.g., liver, kidney, intestinal
sucrose being rapidly hydrolyzed to glucose and fructose in mucosa, and adipose tissue, but not brain) can metabolize
the gastrointestinal tract, alleviates symptoms of exercise it. Fructose metabolism, which is much less tightly regu-
intolerance by providing glucose for muscle contraction. lated, is more rapid than glucose metabolism. The renal
threshold for fructose is very low, and fructose is more
readily excreted in urine than glucose. Despite these dif-
ALTERNATIVE PATHWAYS OF GLUCOSE
ferences, the metabolic fates of glucose and fructose are
METABOLISM AND HEXOSE closely related because most fructose is ultimately con-
INTERCONVERSIONS verted to glucose. Fructose metabolism (Figure 14.11)
starts with phosphorylation and formation of fructose-1-
Glucuronic Acid Pathway phosphate, and this reaction is the rate-limiting step.
The glucuronic acid pathway is a quantitatively minor The Km of hexokinase for fructose is several orders of
route of glucose metabolism. Like the pentose phosphate magnitude higher than that for glucose, and at the
TABLE 14.1 Glycogen Storage Diseases

Type Eponym Deficiency Glycogen Structure Comments


Ia von Gierke’s Glucose-6-phosphatase Normal Hypoglycemia; lack of glycogenolysis
disease in liver, intestine, and by epinephrine or glucagon; ketosis,
(hepatorenal kidney hyperlipidemia, hyperuricemia;
glycogenosis) hepatomegaly; autosomal recessive.
Galactose and fructose not converted
to glucose.
Ib ... Glucose-6-phosphate Normal Clinically identical to type Ia.
transporter in
hepatocyte microsomal
membrane
Ic ... Phosphate transporter in Normal Clinically identical to type Ia.
hepatocyte microsomal
membrane
Id ... Glucose transporter Normal Clinically identical to type Ia.
GLUT7
II Pompe’s disease Lysosomal α-1,4- Normal How this deficiency leads to glycogen
(generalized glucosidase (acid storage is not well understood. In some
glycogenosis) maltase) cases, the heart is the main organ
involved; in others, the nervous system is
severely affected; autosomal recessive.
III Forbes’s disease, Amylo-1,6-glucosidase Abnormal; very long inner Hypoglycemia; diminished hyperglycemic
Cori’s disease (debranching enzyme) and outer unbranched response to epinephrine or glucagon;
(limit dextrinosis) chain normal hyperglycemic response to fructose
or galactose; autosomal recessive. Six
subtypes have been defined based on
relative effects on liver and muscle, and on
properties of the enzyme.
IV Andersen’s (1,4-1,6)- Abnormal; outer chains Rare, or difficult to recognize; cirrhosis
disease (branching transglucosylase missing or very short; and storage of abnormal glycogen;
deficiency, (branching enzyme) increased number of diminished hyperglycemic response to
amylopectinosis) branched points epinephrine; abnormal liver function;
autosomal recessive.
V McArdle’s disease Muscle glycogen Normal High muscle glycogen content (2.5%
phosphorylase 4.1% versus 0.2%0.9% normal); fall in
(myophosphorylase) blood lactate and pyruvate after exercise
(normal is sharp rise) with no postexercise
drop in pH; normal hyperglycemic
response to epinephrine (thus normal
hepatic enzyme); myoglobinuria after
strenuous exercise; autosomal recessive.
VI Hers’ disease Liver glycogen Normal Not as serious as glucose-6-phosphatase
phosphorylase deficiency; liver cannot make glucose
(hepatophosphorylase) from glycogen but can make it from
pyruvate; mild hypoglycemia and ketosis;
hepatomegaly due to glycogen
accumulation; probably more than one
disease; must be distinguished from defects
in the glycogen phosphorylase-activating
system.
VII Tarui’s disease Muscle Normal Shows properties similar to type V;
phosphofructokinase autosomal recessive; it is not completely
clear why this defect results in increased
glycogen storage.
VIII ... Reduced activation of Normal Hepatomegaly; increased hepatic glycogen
phosphorylase in stores; probably X-linked, but there may
hepatocytes and be more than one type, with some
leukocytes autosomally inherited; must be
distinguished from glycogen
phosphorylase deficiency.
Carbohydrate Metabolism II Chapter | 14 217

Inactive hepatophosphorylase
cAMP-
Branched glycogen dependent
(1,4 and 1,6 glucosyl units) Pi Active protein
Inactive
phosphorylase kinase
phosphorylase
IV V kinase kinase
Oligosaccharides & VIII
VI
Lysosomal Unbranched glycogen Active hepatophosphorylase
1, 4-glucosidase (1,4 glucosyl units)
II
Glucose Limit dextrin

UDP
III

Glycogen synthase
Debranching enzyme
(amylo-1,6-glucosidase)

UDP-glucose
Glucose from the action
UDP-pyrophosphorylase of the debranching enzyme
PPi
UTP
Glucose-1-phosphate
Lactate Phosphofructo-
kinase-1 Phosphoglucomutase
Fructose-1,6- VIII Fructose-6-phosphate Glucose-6-phosphate
bisphosphate
Glucose-6-phosphate
translocase
lb

Glucose-6-phosphate
Phosphate
Endoplasmic
transporter
reticulum
lc

H2O Pi Pi
Glucose transporter, GLUT 7
Glucose-6-phosphatase ld
la Glucose

FIGURE 14.9 Locations of the glycogen storage disease enzyme defects in the overall scheme of glycogenesis and glycogenolysis. The numbers
correspond to the disease types listed in Table 14.1.

concentrations of glucose found in most tissues, fructose 3-phosphate and glyceraldehyde. Glyceraldehyde is phos-
phosphorylation by hexokinase is competitively inhibited. phorylated by triosekinase, and glyceraldehyde 3-phosphate
In tissues that contain fructokinase, such as liver, the rate and dihydroxyacetone 3-phosphate can either enter the gly-
of fructose phosphorylation depends primarily on fructose colytic pathway or be combined to form fructose-1,6-
concentration, and an increase in fructose concentration bisphosphate by the action of fructose-1,6-bisphosphate
depletes intracellular ATP. Essential fructosuria is aldolase. Thus, fructose metabolism bypasses phosphofruc-
caused by the absence of hepatic fructokinase activity. tokinase, the major regulatory site of glycolysis.
The condition is asymptomatic but, as with pentosuria, Most dietary fructose is converted to glucose by way
may be misdiagnosed as diabetes mellitus. of gluconeogenesis, through condensation of the triose
The next step in fructose metabolism is catalyzed by phosphates to fructose-1,6-bisphosphate. However,
aldolase B (fructose-1-phosphate aldolase), which cleaves administration of large doses of fructose (i.e., by intrave-
fructose-1-phosphate to the trioses dihydroxyacetone nous feeding) may lead to hypoglycemia and lactic
218 Essentials of Medical Biochemistry

Glucose-6-P CH2OH CH2OH


2NAD+
O UDPG- O
+ H2O
pyrophosphorylase
2NADH + 2H+
OH OH
HO O P UTP PPi HO O P P U
UDPG-
HO HO dehydrogenase
Glucose-1-P UDP-glucose
COOH
O
Substrate
Glucuronyl-
OH transferase
HO O P P U
UDP
HO

UDP-glucuronic acid Glucuronides


FIGURE 14.10 Synthesis of UDP-glucuronic acid. P 5 phosphate.

Glucose
NADP+
Aldose
reductase
NADH + H+
Sorbitol
NAD+
Sorbitol
dehydrogenase
NADH + H+
Fructose From sucrose and other
ATP dietary sources
Fructokinase
ADP

Fructose-1-phosphate
Aldolase B

Dihydroxyacetone 3-phosphate + Glyceraldehyde


ATP
Triosekinase
ADP
Glyceraldehyde 3-phosphate
Fructose-1,6-
bisphosphate aldolase

Fructose-1,6-bisphosphate

Glycolysis Gluconeogenesis

Pyruvate Glucose
FIGURE 14.11 Metabolic pathway for sorbitol and fructose. Sorbitol dehydrogenase is sometimes known as iditol dehydrogenase. Aldolase B is
also called fructose-1-phosphate aldolase, in contrast to fructose-1,6-bisphosphate aldolase.

acidosis because of saturation of aldolase B, causing with this condition exhibit hypoglycemia, metabolic aci-
accumulation of fructose-1-phosphate, and depletion of dosis, vomiting, convulsions, coma, and signs of liver
intracellular ATP and inorganic phosphate. This situation failure following ingestion of fructose or sucrose.
may be likened to hereditary fructose intolerance, The fructose-induced hypoglycemia arises from accumu-
which is caused by inadequate amounts of aldolase B lation of fructose-1-phosphate, reduction in the
activity. Although normally asymptomatic, individuals [ATP]/[ADP] ratio, and depletion of inorganic phosphate.
Carbohydrate Metabolism II Chapter | 14 219

Fructose-1-phosphate depresses gluconeogenesis and pro- glucose-1-phosphate, and high concentrations of UDP-
motes glycolysis, while inorganic phosphate depletion glucose. Deficiency of galactokinase or transferase can
inhibits ATP synthesis (Chapter 13). Glycogenolysis is cause galactosemia.
inhibited by fructose-1-phosphate at the level of phos- Galactose is isomerized to glucose by UDP-galactose-
phorylase. If sucrose, fructose, and sorbitol are eliminated 4-epimerase in what may be the rate-limiting step in galac-
from the diet, complete recovery occurs. High levels of tose metabolism. The reaction, which is freely reversible,
fructose also increase purine turnover owing to enhanced converts UDP-glucose to UDP-galactose for the synthesis
ATP utilization, which leads to increased production of of lactose, glycoproteins, and glycolipids. The epimerase
purine degradation products: inosine, hypoxanthine, xan- requires NAD1 and is inhibited by NADH. It may also be
thine, and uric acid (Chapter 25). regulated by the concentrations of UDP-glucose and other
uridine nucleotides. Because of this epimerase, preformed
galactose is not normally required in the diet. However,
Galactose Metabolism infants with deficiency of epimerase in hepatic and extra-
Most galactose ingested by humans is in the form of lac- hepatic tissues require small quantities of dietary galactose
tose, the principal sugar in human and bovine milk. Milk for normal development and growth. An isolated defi-
sugar other than lactose is found in the sea lion and mar- ciency of epimerase in erythrocytes, which is clinically
supials, whose first pouch milk contains a trisaccharide of benign, has been described.
galactose. Lactose is hydrolyzed to galactose and glucose Genetically determined deficiencies of galactokinase
by lactase, located on the microvillar membrane of the and galactose-1-phosphate uridylyltransferase cause clini-
small intestine (Chapter 11). Following absorption, galac- cally significant galactosemia. Galactokinase deficiency is
tose is transported to the liver, where it is converted to a rare, autosomal recessive trait in which high concentra-
glucose (Figure 14.12). The enzymes required are found tions of galactose are found in the blood, particularly after
in many tissues, but the liver is quantitatively the most a meal that includes lactose-rich foods, such as milk and
important site for this epimerization. nonfermented milk products. Patients frequently develop
Galactose is a poor substrate for hexokinase; it is phos- cataracts before one year of age because of the accumula-
phorylated by galactokinase. Galactose-1-phosphate is tion of galactitol in the lens. Galactose diffuses freely into
converted to UDP-galactose by galactose-1-phosphate the lens, where it is reduced by aldose reductase, the
uridylyltransferase. This enzyme may be regulated by enzyme that converts glucose to sorbitol. Galactitol may
substrate availability, since the normal hepatic concentra- cause lens opacity by the same mechanisms as for sorbitol.
tion of galactose-1-phosphate is close to the Km for this Galactose in the urine is detected by nonspecific tests for
enzyme. The transferase is inhibited by UDP, UTP, reducing substances, as are fructose and glucose.

Galactose FIGURE 14.12 Metabolic pathway of galactose. *Inherited


defects that lead to galactosemia.
ATP
Mg2+ Galactokinase*
ADP
Galactose-1-phosphate
UDP-glucose
Galactose-1-phosphate
uridylyltransferase*
Glucose UDP

Glucose-1-phosphate UDP-galactose Lactose


Lactose synthase
Phospho- Uridine
gluco- diphosphogalactose-
mutase 4-epimerase (NAD+)

Glucose-6-phosphate UDP-glucose
H2O Glucose-
Glycogen synthesis
6-phos-
Pi phatase Glycogen
Glucose
Glycogen breakdown

Glucose
220 Essentials of Medical Biochemistry

Deficiency of the transferase causes a more severe form Glucose


of galactosemia. Symptoms include cataracts, vomiting, diar- ATP
rhea, jaundice, hepatosplenomegaly, failure to thrive, and Glycogenolysis
mental retardation. If galactosuria is severe, nephrotoxicity ADP
with albuminuria and aminoaciduria may occur. The more Gluconeogenesis
Glucose-6-phosphate
severe clinical course may be due to accumulation of
galactose-1-phosphate in cells. Measurement of transferase NADP+
activity in red blood cells is the definitive test for this disorder. NADPH + H+
In about 70% of transferase alleles in the white population,
the DNA in cells of transferase-deficient patients possesses an CO2
A-to-G transition that leads to the Q186R mutation.
Both forms of galactosemia are treated by rigorous Ribulose 5-phosphate
exclusion of galactose from the diet. In pregnancies where
the family history suggests that the infant may be affected, FIGURE 14.13 Summary of the pentose phosphate pathway. This dia-
the best outcomes have been reported when the mother gram is intended to show the two major parts of the pathway: oxidation
was maintained on a galactose-free diet during gestation. and decarboxylation of glucose-6-phosphate to ribulose-5-phosphate; and
resynthesis of the former from the latter. The stoichiometry of the path-
Condensation of glucose with UDP-galactose, catalyzed
way is ignored.
by lactose synthase (Figure 14.12), forms lactose, the only
disaccharide made in large quantities by mammals. This
enzyme is a complex of galactosyltransferase, a membrane- glucose-6-phosphate, it is a cycle, or shunt. For every six
bound enzyme that participates in glycoprotein synthesis molecules of glucose-6-phosphate that enter the path-
(Chapter 10), and α-lactalbumin, a soluble protein secreted way, five molecules of glucose-6-phosphate 1 6CO2 are
by the lactating mammary gland. Binding of α-lactalbumin produced. Thus, there is a net loss of one carbon from
to galactosyltransferase changes the Km of the transferase each glucose-6-phosphate that enters the cycle.
for glucose from 12 mol/L to 1023 mol/L. Synthesis of
α-lactalbumin by the mammary gland is initiated late in The pathway performs a variety of functions.
pregnancy or at parturition by the sudden decrease in proges- Production of ribose 5-phosphate for nucleotide synthesis
terone levels that occurs at that time. Prolactin promotes the by de novo and salvage pathways (Chapter 25), generation
rate of synthesis of galactosyltransferase and α-lactalbumin. of NADPH for biosynthetic reactions in the cytosol, and
interconversion of pentoses and hexoses are more valu-
able processes. Thus, the pathway provides a means for
Metabolism of Amino Sugars generating glucose from ribose and other pentoses that
can be converted to ribose 5-phosphate.
In amino sugars, one hydroxyl group, usually at C2, is
The pentose phosphate pathway is active in a wide
replaced by an amino group. Amino sugars are important
variety of cell types, particularly those that have a high
constituents of many complex polysaccharides, including
rate of nucleotide synthesis or that utilize NADPH in
glycoproteins and glycolipids, and of glycosaminoglycans
large amounts. Nucleotide synthesis is greatest in rapidly
(Chapter 10). De novo synthesis of amino sugars starts from
dividing tissues, such as bone marrow, skin, and gastric
glucose-6-phosphate, but salvage pathways can also operate.
mucosa. NADPH is needed in the biosynthesis of fatty
acids (liver, adipose tissue, lactating mammary gland),
Pentose Phosphate Pathway cholesterol (liver, adrenal cortex, skin, intestine, gonads),
steroid hormones (adrenal cortex, gonads), and catechola-
The series of cytoplasmic reactions known as the pentose mines (nervous system, adrenal medulla), and in other
phosphate pathway are also called the hexose monopho- reactions that involve tetrahydrobiopterin. NADPH is also
sphate (HMP) shunt (or cycle) or the phosphogluconate needed for maintenance of a reducing atmosphere in cells
pathway. The qualitative interconversions that take place are exposed to high concentrations of oxygen radicals includ-
summarized in Figure 14.13, in which stoichiometry is ing erythrocytes, lens and cornea of the eye, and phago-
ignored. cytic cells, which generate peroxide and superoxide
The pentose phosphate pathway can be thought of as anions during the process of killing bacteria.
two separate pathways:
1. The oxidative conversion of glucose-6-phosphate to
ribulose 5-phosphate and CO2; and Oxidative Phase
2. Resynthesis of glucose-6-phosphate from ribulose 5- The first reaction in the oxidative phase is the oxidation
phosphate. Because the pathway begins and ends with of glucose-6-phosphate to 6-phosphoglucono-δ-lactone
Carbohydrate Metabolism II Chapter | 14 221

and reduction of NADP1 to NADPH, catalyzed by a number of sugars rather than acts as a unidirectional ana-
glucose-6-phosphate dehydrogenase (G6PD): bolic or catabolic route for carbohydrates.
2− 2−
CH2OPO3 CH2OPO3
Pentose Phosphate Pathway in Red Blood Cells
H O H H
NADP+ NADPH + H+ H In the erythrocyte, glycolysis, the pentose phosphate path-
H
OH H O way, and the metabolism of 2,3-bisphosphoglycerate (see
OH H
HO OH HO Chapters 12 and 26) are the predominant pathways of carbo-
hydrate metabolism. Glycolysis supplies ATP for membrane
H OH H OH
ion pumps and NADH for reoxidation of methemoglobin.
Glucose-6-phosphate 6-phosphoglucono- The pentose phosphate pathway supplies NADPH to reduce
δ-lactone glutathione for protection against oxidant injury. Glutathione
(γ-glutamylcysteinylglycine; GSH) is synthesized by γ-
In the second step, 6-phosphoglucono-δ-lactone is hydro- glutamylcysteine synthase and GSH synthase (Figure 14.14).
lyzed to 6-phosphogluconate by 6-phosphogluconolactonase: In the steady state, 99.8% of the glutathione is in the reduced
COO−
form (GSH), and only 0.2% is in the oxidized form (GSSG),
2−
because of the NADPH-dependent reduction of oxidized glu-
CH2OPO3 HCOH tathione, catalyzed by glutathione reductase:
O
H2O HOCH γ-Glutamylcysteinylglycine
OH H O
S NADPH + H+ NADP
+

HO HCOH
S
H OH HCOH γ-Glutamylcysteinylglycine
2− (GSSG)
H2COPO3
SH
6-phosphoglucono-δ-lactone 6-phosphogluconate
2γ−Glutamylcysteinylglycine
In the third step, 6-phosphogluconate is oxidatively dec- (GSH)
arboxylated to ribulose-5-phosphate in the presence of
NADP1, catalyzed by 6-phosphogluconate dehydrogenase: Glutathione reductase also catalyzes reduction of
mixed disulfides of glutathione and proteins (Pr):
NADP+ NADPH + H+
+ +
Pr 2 S 2 SG 1 NADPH 1 H1 -Pr 2 SH
COO− COO− CO2 1 GSH1NADP1
+
HCOH HCOH CH2OH
Thus, the active sulfhydryl groups of hexokinase, glycer-
HOCH Mn2+
C O C O aldehydephosphate dehydrogenase, glutathione reductase,
HCOH HCOH HCOH and hemoglobin (SH group at the β-93 position) are main-
HCOH HCOH HCOH
tained in the reduced form. Glutathione reductase is an FAD
H2CPO3
2− 2− 2−
enzyme composed of two identical subunits encoded by a
H2COPO3 H2COPO3
single locus on the short arm of human chromosome 8.
GSH is used in the inactivation of potentially damaging
6-phosphogluconate 3-Keto-6-phosphogluconate D-ribulose-5-phosphate
organic peroxides (e.g., a peroxidized unsaturated fatty acid)
and of hydrogen peroxide. Hydrogen peroxide is formed
Nonoxidative Phase
through the action of superoxide dismutase (Chapter 13):
In the nonoxidative phase, ribulose-6-phosphate is converted
to glucose-6-phosphate. Stoichiometrically, this process 2O2 1
2 1 2H -H2 O2 1 O2
requires the rearrangement of six molecules of ketopentose
phosphate to five molecules of aldohexose phosphate. One Peroxide inactivation is catalyzed by glutathione per-
of the steps is catalyzed by transketolase, which requires thi- oxidase, a selenium-containing enzyme, in the following
amine pyrophosphate and Mg21 as cofactors. reactions:
The versatility of this phase of the pathway allows for
2GSH 1 ROOH-GSSG1H2 O 1 ROH
interconversion of a number of sugars and glycolytic inter-
mediates, in part because of the ready reversibility of the and
reactions and regulation of the enzymes by substrate avail-
ability. Thus, the pathway modulates the concentrations of 2GSH1H2 O2 -GSSG 1 2H2 O
222 Essentials of Medical Biochemistry

ATP ADP + Pi FIGURE 14.14 Metabolism of glutathione.


1 (1) γ-Glutamylcysteine synthase; (2) GSH synthase;
Glutamate + Cysteine γ-Glutamylcysteine (3) glutathione peroxidase (Se-containing enzyme);
Mg2+ (4) glutathione reductase (an FAD enzyme).
ATP Glycine
K+ 2
ADP + Pi
NADP+ γ-Glutamylcysteinylglycine (GSH)
ROOH or H2O2
4 3
ROH or H2O
NADPH + H+ Oxidized glutathione (GSSG)

From pentose phosphate pathway

The concentrations of substrates for glutathione reduc- circulating red cells. The urine may turn dark, even black,
tase and peroxidase determine the rate at which the pen- from the high concentration of hemoglobin, and a high
tose phosphate pathway operates in erythrocytes. urine flow must be maintained to prevent damage to the
Genetically determined deficiencies of γ-glutamyl- renal tubules by the high protein load. Because G6PD
cysteine synthase and glutathione synthase can cause activity is highest in reticulocytes and decreases as the
hemolytic anemia. Abnormalities of glutathione metabo- cell ages, the older erythrocytes (more than 70 days old)
lism can also result from nutritional deficiencies of ribo- are destroyed. For this reason, measurement of red cell
flavin or selenium. Glutathione reductase is an FAD G6PD activity following a hemolytic crisis can lead to a
enzyme that requires riboflavin for activity, and ribofla- spuriously high value, even within the normal range. If
vin deficiency can cause hemolytic anemia. An inherited the patient survives the initial crisis, with or without
lack of glutathione reductase apoenzyme has been transfusions, recovery usually occurs as the reticulocyte
described. Selenium deficiency diminishes activity of count increases.
glutathione peroxidase and can lead to peroxidative dam- The characteristics of the disorder were elucidated
age. This can be partially ameliorated by vitamin E, an during investigations into the hemolytic crises observed
antioxidant (Chapters 35 and 36). in some patients following administration of
8-aminoquinoline derivatives, such as primaquine and
pamaquine, used for prophylaxis and treatment of
Glucose-6-Phosphate Dehydrogenase malaria. The relatively high frequency of such crises in
Deficiency some geographic areas is due to the extensive overlap in
G6PD deficiency is the most common inherited enzyme the distributions of endemic malaria and G6PD defi-
deficiency known to cause human disease, occurring in ciency. The geographic distribution of G6PD variants,
about 100 million people (see Clinical Case Study 14.2). like that of sickle cell trait (Chapter 26), suggests that
Most clinical manifestations are related to hemolysis, heterozygosity for G6PD deficiency may confer some
which results from impaired ability to produce cytosolic protection against falciparum malaria. A number of
NADPH. Over 150 variants of the G6PD structural gene other drugs that cause oxidative stress also cause hemo-
are known, many of which show either abnormal kinetics lytic crises.
or instability of the enzyme. The erythrocytes are most Oxidation of glutathione by these drugs beyond the
severely affected because of their long half-lives and capacity of the cell to generate NADPH for GSSG reduc-
inability to carry out protein synthesis. Since persons with tion causes the acute crisis. Since these drugs do not cause
G6PD deficiency can usually make an adequate supply of hemolysis in vitro, additional steps must take place
NADPH under normal conditions, the defect may not in vivo. Following administration of a drug known to pro-
become apparent until the patient takes a drug, such as mote hemolysis, Heinz bodies are seen in erythrocytes in
primaquine, that greatly increases the demand for the peripheral blood. These also occur in some thalasse-
NADPH. The severity of the reaction depends, in part, on mias and consist of oxidized and denatured forms of
the particular inherited mutation. hemoglobin known as hemichromes (Chapter 27). Heinz
In most persons with G6PD deficiency, hemolysis is bodies impair movement of the cells through the splenic
observed as an acute phenomenon only after severe oxida- pulp and probably are excised there, presumably together
tive stress, leading to loss of perhaps 30%50% of the with the adjacent piece of plasma membrane, leaving a
Carbohydrate Metabolism II Chapter | 14 223

red cell that is more susceptible to destruction by the administration. Hemolysis may cease even with continued
reticuloendothelial system. Infection and diabetic ketoaci- administration of the drug because the reticulocytes,
dosis can also cause hemolysis in persons with G6PD which increase in proportion following hemolysis, have
deficiency, possibly through depletion of NADPH. adequate G6PD activity. In persons of Mediterranean and
Favism is characterized by acute hemolysis following Middle Eastern ancestry, the most common abnormal
ingestion of fava beans (Vicia fava) by people with G6PD allele is G6PD M (G6PD Mediterranean; class II) associ-
deficiency. The fava bean is a vegetable staple of the ated with severe hemolysis following administration of an
Mediterranean region, an area in which G6PD deficiency appropriate drug. The average incidence of this allele is
is endemic. Infants are especially susceptible to favism. approximately 5%10%, but a subpopulation of Kurdish
The disorder is frequently fatal unless a large amount of Jews is reported to have an incidence of 50%. Enzyme
blood is transfused rapidly. activity in erythrocytes from these patients is often less
Studies of the genetics of human G6PD variants have than 1% of normal, and transfusion is usually required
contributed to the understanding of G6PD deficiency and of following a hemolytic crisis. The difference in the clinical
more general aspects of human genetics. G6PD deficiency is severity of the diseases associated with G6PD A2 is a
inherited as an X-linked trait, as are hemophilia (Chapter 34) normal Km for glucose-6-phosphate (5070 μmol/L) and
and color blindness (Chapter 36). If the X-chromosome car- for NADP1 (2.94 μmol/L), but it exhibits an abnormal
rying an abnormal G6PD allele is designated X*, then the pH activity curve. The deficiency is due to an accelerated
three possible genotypes containing X are rate of inactivation of G6PD A2 protein. Bone marrow
cells and reticulocytes have normal amounts of G6PD
1. X*Y-hemizygous male, with full phenotypic expres-
activity, while activity in older red cells is very low.
sion of the abnormal allele;
However, the residual enzyme seems to be more resistant
2. XX*-heterozygous female, with a clinically normal
to inhibition by NADPH. In contrast, G6PD M molecules
phenotype in spite of the abnormal allele expressed in
have an intrinsically lower catalytic activity, and both
about half her cells; and
reticulocytes and older red cells have decreased amounts
3. X*X*-homozygous female, with full phenotypic
of G6PD activity. Consequently, when one of the drugs is
expression of the abnormal allele. Sons of affected
given, more cells are susceptible, and hemolysis is greater
males are usually normal (because they receive their
than with G6PD A2. The low Km of G6PD M for G6P
X-chromosome from their mothers), and daughters of
and NADP1 may account for the near-normal survival of
affected males are usually heterozygotes (because they
erythrocytes in the absence of oxidative stress.
receive one X-chromosome from their father). The rar-
A number of other enzymopathic disorders (e.g., pyru-
est genotype is that of the homozygous female, since
vate kinase, Chapter 12; and pyrimidine-50 -nucleotidase,
it requires that both parents have at least one abnormal
Chapter 25), abnormal hemoglobins (Chapter 26), and
X-chromosome.
abnormalities of the erythrocyte cytoskeleton (Chapter 10)
Females heterozygous for a G6PD variant are pheno- may cause hemolytic anemia. Because many enzymes in the
typic mosaics. They have two erythrocyte populations: red cell are identical to those in other tissues, defects in these
one containing normal G6PD, the other containing the enzymes may have pleiotropic effects. Thus, in addition to
variant. In fact, in heterozygotes, every tissue has some hemolytic anemia, triose phosphate isomerase deficiency
cells expressing the normal, and some the abnormal, causes severe neuromuscular disease, and phosphofructoki-
G6PD gene. Random X-chromosome inactivation early in nase deficiency causes a muscle glycogen storage disease.
embryonic development causes only one of the two X- Mutations that result in decreased enzyme stability are usu-
chromosomes to be active (Lyon’s hypothesis). ally most strongly expressed in erythrocytes because of their
Severity of the disorder in homozygotes and hemizy- inability to synthesize proteins.
gotes depends on a number of factors. G6PD variants are
classified into five groups (class I being the most severe) Phagocytosis and the Pentose Phosphate
depending on the presence or absence of chronic anemia
and on the amount of enzyme activity present in the ery-
Pathway
throcytes. The most common normal activity (class IV) The pentose phosphate pathway is crucial to the survival
allele is G6PD B. Another common electrophoretic vari- of erythrocytes because of its ability to provide NADPH
ant with normal activity is G6PD A. Among north for reduction of toxic, spontaneously produced oxidants.
American blacks, the gene for the absence of G6PD A In phagocytic cells, the pentose phosphate pathway gener-
(G6PD A2; class III) has an incidence of about 11%. ates oxidizing agents using molecular oxygen and
Hemizygous males have only 5%15% of normal eryth- NADPH oxidase that participate in the killing of bacteria
rocyte G6PD activity and exhibit a mild hemolytic ane- and abnormal cells engulfed by phagocytes (see
mia following an oxidative insult, such as primaquine Chapter 13 for a discussion on this topic).
224 Essentials of Medical Biochemistry

CLINICAL CASE STUDY 14.1 Deficiency of Glycogenin-1 Leading to Glycogen Depletion in


Skeletal Muscle Fibers and Cardiac Myocytes
This clinical case was abstracted from: A.-R. Moslemi, C. tissues. Its synthesis requires three enzymes: autocata-
Lindberg, J. Nilsson, H. Tajsharghi, B. Andersson, A. Oldfors, lytic glucosylation of glycogenin, which provides a prim-
Glycogenin-1 deficiency and inactivated priming of glycogen ing oligosaccharide chain; glycogen synthase, which
synthesis, N. Engl. J. Med. 362 (2010) 12031210. extends the oligosaccharide chain; and branching
enzyme, which is responsible for the synthesis of highly
Synopsis branched polymers.
A 27-year-old man, after exercise, experienced dizziness and 2. Glycogenin-1, the predominant isoform of skeletal mus-
cardiac symptoms and was brought to emergency medical ser- cle, is also present in cardiac muscle, along with the
vices at the hospital by ambulance. In the ambulance, the glycogenin-2 isoform. The deficiency of glycogenin-1
subject required cardiac defibrillation to establish normal leads to lack of glycogen energy stores and is accompa-
sinus rhythm. After several laboratory tests, echocardiography, nied by metabolic abnormalities as discussed in this case.
and coronary angiography, the subject’s cardiac problems It should be emphasized that, despite the fact that cardiac
were corrected by cardioverter-defibrillator implantation, and muscle is capable of the oxidation of fatty acids as an
pharmacotherapy with a β1-adrenergic-receptor blocker and energy source, glucose is an important energy source to
an angiotensin-converting enzyme inhibitor. meet its metabolic demands.
In order to definitively establish the biochemical basis of 3. Glycogen synthase deficiency also leads to metabolic
the subject’s abnormalities, biopsies of deltoid muscle and abnormalities similar to glycogenin-1 deficiency (see ref-
endomyocardium were obtained. The histochemical and elec- erence 1).
tron microscopic studies revealed depleted glycogen levels in 4. Deficiency of glycogenin-1 or glycogen synthase affects
both tissues, and mitochondrial proliferation and hypertrophy plasma glucose clearance. This observation has been
of cardiomyocytes. Relevant molecular biochemical studies attributed to a compensatory mitochondrial proliferation,
revealed a missense mutation (Thr83Met) in the glycogenin-1 which augments skeletal muscle glucose oxidation.
gene, and Western blotting showed the accumulation of Integration of this topic with Chapters 8, 14, and 19.
unglycosylated defective glycogenin-1 protein.
Reference
[1] G. Kollberg, M. Tulinius, T. Gilljam, I. Ostman-Smith, G.
Teaching Points
Forsander, P. Jotorp, et al., Cardiomyopathy and exercise intoler-
1. Glycogen, a branched polymer of glucose with α(1-4) ance in muscle glycogen storage disease, N. Engl. J. Med. 357
and α(1-6) linkages, serves as an energy store for many (2007) 15071514.

CLINICAL CASE STUDY 14.2 Glucose-6-Phosphate Dehydrogenase (G6PD) Deficiency


This clinical case was abstracted from: A. Puig, A.S. Dighe, Teaching Points
Case 20-2013: A 29-year-old man with anemia and jaundice, 1. Ten to fifteen percent of the subject’s black African decent
N. Engl. J. Med. 368 (2013) 25022509. ancestry have G6PD deficiency designated as A2. The
mutation causes the production of an unstable G6PD
Synopsis whose levels also decrease as the lifespan of the red blood
A 29-year-old man from North Africa was admitted to hospital cells (about 128 days) declines. Thus, in an acute episode
with key complaints and clinical features of fatigue, jaundice, with a preponderance of young red blood cells entering
and dark urine. His pertinent laboratory findings included nor- the circulation, the G6PD level may be normal. In that
mocytic and normochromic anemia, elevated serum unconju- case, the G6PD level should be determined after about
gated bilirubin, and decreased haptoglobin levels. Other 46 weeks.
laboratory investigations such as liver function tests and 2. Any oxidative stress or illness renders red blood cells sus-
hemoglobin electrophoresis to detect abnormal variants were ceptible to a hemolysis episode in G6PD deficiency.
all negative. Based on history, laboratory findings led to inves- 3. The key laboratory indications for the in vivo hemolysis in
tigation of the G6PD level in the subject’s red blood cells. this subject were normocytic normochromic anemia, ele-
The G6PD level was significantly decreased to 1.9 U per gram vated serum level of unconjugated bilirubin, decreased
hemoglobin (reference interval 8.8 to 13.4). The subject’s haptoglobin level due to its consumption with hemoglo-
diagnosis of intravascular hemolysis was attributed to G6PD bin, and elevated lactate dehydrogenase level (see also
deficiency. Chapter 27).
Resolution of the case: The subject’s clinical findings 4. The subject’s black urine was caused by filtered hemoglo-
resolved without any intervention after a few days. bin and its oxidized products.
Carbohydrate Metabolism II Chapter | 14 225

REQUIRED READING [3] V.S. Tagliabracci, J. Turnbull, W. Wang, J. Girard, X. Zhao, A.V.
Skurat, et al., Laforin is a glycogen phosphatase, deficiency of
[1] C.A. Worby, J.E. Dixon, Glycogen synthase: an old enzyme with a which leads to elevated phosphorylation of glycogen in vivo, Proc.
new trick, Cell Metab. 13 (2011) 233234. Natl Acad. Sci. USA 104 (2007) 1926219266.
[2] V.S. Tagliabracci, C. Heiss, C. Karthik, C.J. Contreras, J. Glushka,
M. Ishihara, et al., Phosphate incorporation during glycogen synthe-
sis and lafora disease, Cell Metab. 13 (2011) 274282.

You might also like