Sie sind auf Seite 1von 15

This article appeared in a journal published by Elsevier.

The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elseviers archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/authorsrights

Author's personal copy


Clinical Biochemistry 46 (2013) 13391352

Contents lists available at ScienceDirect

Clinical Biochemistry
journal homepage: www.elsevier.com/locate/clinbiochem

Review

A quick look at biochemistry: Carbohydrate metabolism


Monireh Dashty
Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands

a r t i c l e

i n f o

Article history:
Received 17 October 2012
Received in revised form 18 April 2013
Accepted 20 April 2013
Available online 14 May 2013
Keywords:
Carbohydrate
Glycolysis
Pyruvate decarboxylation
Oxidative pathway
Pentose phosphate pathway
Glycogenesis
Glycogenolysis
Gluconeogenesis

a b s t r a c t
In mammals, there are different metabolic pathways in cells that break down fuel molecules to transfer their
energy into high energy compounds such as adenosine-5-triphosphate (ATP), guanosine-5-triphosphate
(GTP), reduced nicotinamide adenine dinucleotide (NADH2), reduced avin adenine dinucleotide (FADH2)
and reduced nicotinamide adenine dinucleotide phosphate (NADPH2). This process is called cellular respiration. In carbohydrate metabolism, the breakdown starts from digestion of food in the gastrointestinal tract
and is followed by absorption of carbohydrate components by the enterocytes in the form of monosaccharides. Monosaccharides are transferred to cells for aerobic and anaerobic respiration via glycolysis, citric
acid cycle and pentose phosphate pathway to be used in the starvation state. In the normal state, the skeletal
muscle and liver cells store monosaccharides in the form of glycogen. In the obesity state, the extra glucose is
converted to triglycerides via lipogenesis and is stored in the lipid droplets of adipocytes. In the lipotoxicity
state, the lipid droplets of other tissues such as the liver, skeletal muscle and pancreatic beta cells also
accumulate triacylglycerol. This event is the axis of the pathogenesis of metabolic dysregulation in insulin
resistance, metabolic syndrome and type 2 diabetes. In this paper a summary of the metabolism of carbohydrates is presented in a way that researchers can follow the biochemical processes easily.
2013 The Canadian Society of Clinical Chemists. Published by Elsevier Inc. All rights reserved.

Contents
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Descriptions of gures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Carbohydrate digestion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Fructose metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Galactose metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Glucose oxidation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Non-oxidative pathway (glycolysis) . . . . . . . . . . . . . . . . . . . . . . . .
Pyruvate decarboxylation (oxidative decarboxylation reaction) . . . . . . . . . . .
Oxidative pathway (Krebs cycle and OXPHOS reaction (electron transport chain)) . . .
Glycolysis (EmbdenMeyerhofParnas pathway) . . . . . . . . . . . . . . . . . . . .
Pyruvate dehydrogenase (PDH) or oxidative decarboxylation reaction . . . . . . . . . .
Regulation of PDH complex . . . . . . . . . . . . . . . . . . . . . . . . . . .
Krebs cycle (citric acid or tricarboxylic acid (TCA) cycle) . . . . . . . . . . . . . . . .
Malate-aspartate shuttle . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Energy balance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
TCA regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Glycogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Glycogenolysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Phosphorylase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Pentose phosphate pathway (PPP), phosphogluconate pathway or hexose monophosphate

. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
. . .
shunt

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.

1340
1342
1342
1342
1343
1343
1343
1343
1344
1345
1345
1346
1346
1346
1347
1347
1347
1349
1349
1349

Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands. Fax: + 31 50
3632522.
E-mail address: M.Dashti.Rahmatabadi@med.umcg.nl.
0009-9120/$ see front matter 2013 The Canadian Society of Clinical Chemists. Published by Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.clinbiochem.2013.04.027

Author's personal copy


1340

Gluconeogenesis . . . . . . . . .
Cori cycle . . . . . . . . . .
Glucose-alanine cycle . . . .
Amino acids . . . . . . . . .
Glycerol . . . . . . . . . . .
Role of renal gluconeogenesis .
Regulation of gluconeogenesis
Conclusion . . . . . . . . . . . .
Acknowledgment . . . . . . . . .
References . . . . . . . . . . . .

M. Dashty / Clinical Biochemistry 46 (2013) 13391352

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

Introduction
The study of biochemistry is always one of the most complicated
topics among medical and biology students in spite of being one of the

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.

1350
1350
1350
1351
1351
1351
1351
1351
1352
1352

main subjects during their study. In addition, there seems to be a shortage


of papers on pure biochemistry for biology students to be used as reference in their papers. In this paper, the main topics and denitions of carbohydrate pathways are summarized and the related gures are shown in

Graphic abstract gure. Presentation of the association between 6 biochemical pathways. The biochemical reactions happen in the cytoplasm or mitochondria;
therefore, a close interaction between these two parts always exists. These interactions are mostly, regulated by transporters that are in the mitochondrial membrane
(red ovals). The energetic pathways in the cell metabolize energetic molecules such as glucose and lipids. These pathways are glycolysis, gluconeogenesis, lipolysis,
lipogenesis and the electron transport chain. Pyruvate (product of glycolysis) can be used in amino acid reactions as well. The energetic products of the oxidative reaction of glucose in the mitochondria (NADH2 and FADH2) are delivered to the inner mitochondrial intermembrane (IMM) for complete conversion of their potential
energy to chemical energy in the form of ATP. ATP is the energetic parcel in cells that is used in different biochemical reactions. In this gure, the main energetic biochemical reactions and their association to each other are illustrated. These reactions are 1. Glycerol phosphate shuttle, 2. NADH2 shuttle into IMM, 3. Transhydrogenase
cycle (blue), 4. Citrate/malate cycle (pink), 5. Krebs cycle (red) and 6. Malate-aspartate shuttle (green). The correlative function of the lipid and carbohydrate biochemical
pathways together with the electron transport chain of the mitochondria for maintenance of glucose is also shown. Abbreviations are explained in gure 2.

Author's personal copy


M. Dashty / Clinical Biochemistry 46 (2013) 13391352

1341

Fig. 1. Formulas of biochemical molecules. Abbreviations are explained in gure 2.

a way to facilitate the study of this topic and its link to other pathways.
The subjects that are investigated here are listed below:
1. Carbohydrate digestion (in the intestine)
2. Fructose metabolism (in the liver)

Fig. 2. Abbreviations.

3. Galactose metabolism (in the liver)


4. Glucose oxidation via glycolysis (in the cytoplasm), oxidative
decarboxylation reaction (in the mitochondria), citric acid cycle

Author's personal copy


1342

M. Dashty / Clinical Biochemistry 46 (2013) 13391352

Fig. 3. Summary of biochemical reactions in carbohydrate metabolism.

5.
6.
7.
8.

(in the mitochondria) and the electron transport chain (ETC) (in
the inner mitochondrial membrane (IMM))
Glycogenesis (in the liver and skeletal muscle)
Glycogenolysis (in the liver, skeletal muscle and kidney)
Pentose phosphate pathway (in the liver, adipose tissue, adrenal cortex, testis, milk glands, phagocyte cells and red blood cells (RBCs))
Gluconeogenesis (in the liver, kidney, brain, testes and erythrocytes) [1].

Descriptions of gures
In this paper, the biochemical gures are shown in a new style in
order to facilitate the study and follow up of the pathways. Substrates
and products are presented in brown, enzymes in blue, number of carbons in each molecule in red, utilized materials at the left side and
yielded materials at the right side. S is the stimulators of the enzymes
and I is the inhibitors of the enzymes. Red arrows represent the inhibitory effects of agents. For interpretation of the references to color in the
gure legends, the reader is referred to the web version of this article.
Graphic abstract gure presents the association between pathways.
The general information of the pathways including formulas, abbreviations and a summary of the biochemical reactions in carbohydrate
metabolism are shown in gures 1, 2, 3 and table 1.

sucrase for degradation of sucrose to its constituent components


(glucose and fructose) [6] and maltase for degradation of maltose
to two glucose molecules. The 5C-carbohydrates such as xylulose,
arabinose, ribose and ribulose easily diffuse into the intestinal absorptive cells and do not need degradation.
Absorption of the 6C-carbohydrates from intestinal epithelium
happens in two ways: passive and active transport systems. In the
passive diffusion form, phosphorylation of carbohydrate (e.g., glucose
or galactose) in the intestinal cells leads to their facilitated transfer
to the circulation. Glucose-6-phosphate (G6P) and galactose-6phosphate (Gal6P) are then dephosphorylated and enter the liver.
In the active diffusion form, carbohydrates utilizing a mobile carrier
protein coupled with the sodium/potassium (Na +/K +) pump and
against the gradient together with Na + ion enter enterocytes [7].
Therefore, the Na +/K + pump using ATP as its source of energy exchanges 3 Na + ions with 3 K + ions [7].
Galactose and fructose are converted to glucose in the liver. Glucose
is used in different metabolic pathways for (i) stability of blood sugar in
the hypoglycemic state, (ii) energy supplier of the peripheral tissues,
(iii) energy storage in the liver and skeletal muscle in the form of glycogen to be used in exercise [8,9], (iv) energy storage in the adipose
tissue following conversion to triglycerides (TG, TAG, triacylglycerol
or triacylglyceride) [10] in the case of excess glucose and (v) stability
of body temperature [11,12].

Carbohydrate digestion
Fructose metabolism
Dietary carbohydrates of greatest importance are composed of
hexoses such as sucrose (saccharose or table sugar), lactose (milk
sugar), galactose (derived from fermented products) and maltose
(derived from hydrolysis of starch) and also pentoses such as xylose
and arabinose (from fruits) [2]. Food digestion starts in the mouth
through secretion of salivary alpha-amylase (or ptyalin) that hydrolyses alpha-1,4 (-1,4) linkage of starch (or amylum) and converts it
to maltose. The next enzyme is pancreatic-amylase (or amylopsin) in
the small intestine that digests 60% of starches. Intestinal epithelial
cell enzymes degrade 6-carbon (6C-) carbohydrates [35]. These enzymes are lactase for degradation of lactose to glucose and galactose,

Glucose is the main source of energy in cells; however, with high


consumption of sucrose (composed of glucose and fructose) cells
can use fructose as well. In the muscles, adipose tissue and kidney,
which contain hexokinase (HK), fructose gets phosphorylated to become fructose-6-phosphate (F6P) to be directly used in the glycolysis
pathway. However, in the liver, which contains glucokinase (GK),
fructose must rst be converted to glucose for consumption in the
glycolysis pathway. Therefore, fructose rst gets phosphorylated and
is then divided to two 3-carbon glycolysis intermediates: glyceraldehyde
and dihydroxyacetone phosphate (DHAP). These intermediates can easily

Author's personal copy


M. Dashty / Clinical Biochemistry 46 (2013) 13391352

1343

Table 1
A summary of carbohydrate biochemical pathways. Numbering in glycolysis, PDH reaction and the Krebs cycle is based on breakdown of one glucose molecule. VR: Vital reaction,
C: Catabolic or exergonic reaction, A: Anabolic or endergonic reaction, Cyt: Cytoplasm, Mit: Mitochondria, 4C-P: Phosphorylated form of 4C-carbohydrate (erythrose-4P), 5C-P: Phosphorylated form of 5C-carbohydrate (xylulose-5P, riboluse-5P and ribose-5P), 7C-P: Phosphorylated form of 7C-carbohydrate (sedoheptulose-7P).
Pathway
Glycolysis (aerobic)
Glycolysis (anaerobic)
PDH reaction (aerobic)
Krebs cycle (aerobic)
Electron transport chain
Glycogenesis
Glycogenolysis
Gluconeogenesis
Fructose metabolism
Galactose metabolism
Pentose phosphate

VR
C
C
C
C
C
A
C
A
A
A
A

Place
Cyt
Cyt
Mit
Mit
Mit
Cyt
Cyt
Both
Cyt
Cyt
Cyt

Substrate
1 Glucose
1 Glucose
2 Pyruvate
2 Acetyl-CoA, 2 OAA
1 NADH2 & 1 FADH2
Glucose
Glycogen
2 Pyruvate (or 2 lactate)
Fructose
Galactose
G6P

Product

Utilized

2 Pyruvate
2 Lactic acid
2 Acetyl-CoA
2 Citrate
3 ATP & 2 ATP
Glycogen
G6P, glucose, F6P
G6P, glucose, F6P
G6P, glycogen
G6P, lactose
F6P, GA3P, 4C-P, 5C-P, 7C-P

be converted to each other. Thereafter, glyceraldehyde is phosphorylated


to become glyceraldehyde-3-phosphate (GA3P) and is again joined
to DHAD to form the double-phosphorylated product, fructose1,6-bisphosphate (F1,6BP). Dephosphorylation of F1,6BP by phosphatase produces F6P. F6P is either used as the substrate of the glycolysis
pathway or is converted to G6P using isomerase. G6P is used in
two ways. It either loses its phosphate with glucose-6-phosphatase
(G6Pase) activity and is converted to glucose to enter the circulation
or via utilizing mutase is converted to glucose-1-phosphate (G1P)
(substrate of glycogen synthesis pathway). G6Pase exists only in the
liver and kidney cells and not in the muscle cells (Figs. 4 and 5).

Galactose metabolism
Galactose enters the body following consumption of milk. Milk
sugar or lactose is composed of galactose and glucose. Lactose is
converted to its constituents with lactase activity in the brush border
of the small intestine. Galactose enters the blood stream following absorption by enterocytes and enters the liver through the portal vein to
be metabolized and converted to glucose for consumption as energy.
Glucose and galactose are the sugars whose active forms are transferred
by the uridine diphosphate (UDP) coenzymes in the form of uridine
diphosphate-glucose (UDPG) and UDP-galactose (UDPGal). Galactose
metabolism starts from phosphorylation and conversion of galactose
to galactose-1-phosphate (Gal1P). Thereafter, galactose-1-phosphate
uridylyltransferase (GALT) and its UDPG coenzyme, exchange the
galactose of Gal1P with glucose to form G1P and as a result UDPGal is

2+

Yielded

Mg
Mg2+
Lipoic acid, 2 CoA
4 H2O, Fe2+, Mg2+, Mn2+, Fe/S
Oxygen
2 ATP, Mg2+

2 ATP, 2 NADH2
2 ATP
2 NADH2
6 NADH2, 4 CO2, 2 GTP, 2 FADH2, 2 CoA
H2O

2 HCO3, 4 NADH2, 4 ATP, 2 GTP, 2 H2O


2ATP
1 ATP, UDPG
1 H2O, TPP

2 NADPH2, 2 CO2, 6 Pi
Pi
2 NADPH2, 1 CO2, 1 H+

released. UDPGal can be converted to UDPG with UDP-galactose-4epimerase activity (Figs. 4 and 5).
Glucose oxidation
There are two non-oxidative and oxidative pathways that oxidize
glucose to prepare the energy source of cells. Oxidative decarboxylation
reaction is the linker reaction between these two pathways.
Non-oxidative pathway (glycolysis)
This is an anaerobic respiration or pyruvic acid (pyruvate) fermentation that happens in the cytoplasm in the absence of oxygen.
In glycolysis, partial oxidation of glucose produces pyruvic acid
(Fig. 6). In anaerobic states (heavy exercise or cells without
mitochondria), pyruvate is converted to lactate through a bilateral
reaction catalyzed by lactate dehydrogenase (LDH) and consumption of one molecule NADH2 (Fig. 3/12). Lactate is transferred to
the liver for gluconeogenesis and glucose production to be used
again in the glycolysis pathway (Cori cycle) (Fig. 7).
Pyruvate decarboxylation (oxidative decarboxylation reaction)
This reaction is the linker between glycolysis and the Krebs cycle
and is catalyzed by pyruvate dehydrogenase complex (PDC) (Fig. 8).
In this process, two pyruvic acids of the glycolysis pathway are consumed to produce two molecules of acetyl coenzyme A (acetyl-CoA)

Fig. 4. Presentation of the association between carbohydrate metabolisms. Galactose, fructose, mannose and glucose monosaccharides have a close association with each other and also
with the polysaccharide pathways including glycogenesis and glycogenolysis. In this gure, the roles of the main metabolic hormones on these pathways are also shown.
Abbreviations are explained in gure 2.

Author's personal copy


1344

M. Dashty / Clinical Biochemistry 46 (2013) 13391352

Fig. 5. Fructose and galactose metabolism. In overconsumption of sucrose and lactose,


cells can also use fructose and galactose as their source of energy. These products
can be converted to G6P and F6P (the intermediates of the glycolysis pathway) and
G1P (the substrate of glycogenesis). The gure and abbreviations are explained in
the text and in gure 2.

and 2 NADH2. NADH2 is consumed in the oxidative phosphorylation


(OXPHOS) reaction. Acetyl-CoA is either used in the Krebs cycle
(Fig. 9) and OXPHOS reaction in mitochondria for full oxidization of
glucose or is converted to citrate. Citrate is exported to the cytosol for
biosynthesis of fatty acid (FA) and isoprenoid.
PDC is composed of multiple copies of 3 separate enzymes, namely
pyruvate dehydrogenase (PDH), dihydrolipoamide S-acetyltransferase
(DLAT) and dihydrolipoamide dehydrogenase (DLD) and 5 coenzymes,
namely coenzyme A (CoA or CoASH), NAD+, FAD+, lipoic acid and
thiamine pyrophosphate (TPP). TPP, lipoic acid and FAD+ are tightly
bound to the enzymes of the complex and CoA and NAD + are the
carriers of the products of PDC.
Oxidative pathway (Krebs cycle and OXPHOS reaction (electron transport
chain))
It is an aerobic respiration that happens in the mitochondria in
the presence of oxygen. In the Krebs cycle, two pyruvates of glycolysis
are degraded to release their energy in the form of 2 GTP and the reduced forms of high energy molecules (6 NADH2 and 2 FADH2).
NADH2 and FADH2 are consumed in the respiratory chain process

Fig. 6. Glycolysis. The non-oxidative glycolysis pathway is a cytoplasmic anaerobic


pathway that happens in the absence of oxygen. It degrades the glucose molecule to two
3C-pyruvate molecules and yields 2 ATP and one energetic molecule, NADH2. The gure
and abbreviations are explained in the text and in gure 2.

of the mitochondria (OXPHOS reaction). These reduced coenzymes


(NADH2 and FADH2) release their hydrogen (proton), which at the
end of the process combines with the oxygen atom to produce
water. The electron of the hydrogen has a high transfer potential energy that gradually is transferred to oxygen through a series of enzymes (complexes I to IV). In each step a small amount of its energy is
released and the level of its energy fall down to a lower energy state. Enzymes of the electron transport system use the energy released from
the oxidation of NADH2 and FADH2 to pump protons across the IMM
to the intermembrane space (IMS). This generates a proton gradient
across the IMM. Reverse movement of protons to the mitochondrial
matrix across the IMM releases energy, which is used for creation of
ATP (Graphic abstract gure). Therefore, the reducing potentials of
the NADH2 and FADH2 are converted to chemical energy in the form
of ATP. The energy, which is released from the OXPHOS reaction of
NADH2 is equal to 3 ATP and the energy of FADH2 is equal to 2 ATP.
As a result, the nal products of complete glucose oxidation are two
waste products (CO2 and H2O) and energy (Fig. 3/1). Succinate is also
oxidized by the electron transport chain, but feeds into the pathway
at a different point. In conclusion, during glucose oxidation most of
the ATP, which is produced via aerobic cellular respiration, is made by
OXPHOS reaction through a series of complex electron transfer processes. Aerobic metabolism is 19 times more efcient than anaerobic
metabolism.

Author's personal copy


M. Dashty / Clinical Biochemistry 46 (2013) 13391352

1345

Fig. 7. Cori cycle and glucose-alanine cycle. These are the cycles that link glucose production in the liver to energy production in other tissues. In the Cori cycle, bilateral association
between glycolysis in the skeletal muscle cells with gluconeogenesis in the hepatocytes is shown. Waste product of the skeletal muscles (lactate) is used in the hepatocytes to produce glucose for consumption as energy in the skeletal muscle. In the glucose-alanine cycle, the nitrogen product of non-hepatic tissues, produced from transamination reaction and
amino acid production, is transferred to the hepatocytes to be used for either gluconeogenesis and glucose production or excretion as urea. Abbreviations are explained in gure 2.

Glycolysis (EmbdenMeyerhofParnas pathway)


Degradation of glucose for releasing its energy for the anabolic pathways starts from glycolysis and continues to the Krebs or tricarboxylic
acid (TCA) cycle in the mitochondria. Glycolysis is a cytoplasmic
non-oxidative reaction for glucose degradation that is composed of 9 processes. A non-specic HK enzyme by using ATP phosphorylates glucose
following entrance to the cell and converts it to G6P. In the liver both

HK as well as GK (the specic kinase for glucose substrate) exist. Therefore, in high concentration of glucose (>100 mg/100 ml) GK is also active.
These enzymes are stimulated by insulin, adenosine monophosphate
(AMP) and adenosine diphosphate (ADP) and inhibited by long chain
fatty acids (LCFAs) and their own products, G6P and ATP. G6P is converted
to one 6C-F1,6BP molecule. Aldolase divides F1,6BP to two 3C-molecules,
namely DHAP or glyceraldehyde 3 phosphate (GA3P). These two molecules can be converted to each other using triosephosphate isomerase
(TPI). The high energy products of glycolysis pathway are ATP,
1,3-di(or bis)phosphoglyceric acid (1,3D(B)PGA) and phosphoenolpyruvate (PEP) that produce 8 kcal, 10 kcal and 14 kcal energy respectively. The nal product of the glycolysis pathway is two pyruvate molecules
that are used in the PDH reaction (Fig. 3/2). The allosteric enzymes of
glycolysis are phosphofructokinase (PFK), pyruvate kinase (PK) and
either GK or HK depending on the tissue.
One glucose produces 8 ATP during glycolysis. Glycolysis happens in two phases: preparatory and pay-off phases. In the preparatory phase, HK and PFK consume 2 ATP molecules. In the pay-off
phase glyceraldehyde-3-phosphate dehydrogenase (GA3PDH),
phosphoglycerate kinase (PGK) and PK produce 10 ATP via production of 2 NADH2 (equal to 6 ATP) and 4 ATP (Fig. 6).
In anaerobic states, each pyruvate produces lactate and 2 ATP by
utilizing one NADH2. Therefore, it produces 6 ATP less than the aerobic
state. Pyruvic acid is used in each of the following pathways and its nal
fate is regulated by acetyl-CoA (Fig. 10).
1.
2.
3.
4.

Acetyl-CoA synthesis inside the mitochondria during the PDH reaction


Lactate synthesis inside the cytoplasm following LDH activity
Alanine (Ala) synthesis during the transamination reaction
Acetaldehyde synthesis after decarboxylation in bacteria, mice and
moles (Fig. 3/3)
5. Ethanol synthesis via using alcohol dehydrogenase and NADH2
(Fig. 3/4)
6. Oxaloacetic acid (OAA) synthesis after carboxylation by pyruvate
carboxylase (PC). OAA can be consumed in either the Krebs cycle
or the gluconeogenesis (Figs. 3/5 and 3/7).

Fig. 8. Pyruvate dehydrogenase (PDH) reaction. This mitochondrial oxidative decarboxylation


reaction is the linker between glycolysis and the Krebs cycle and produces acetyl-CoA.
Acetyl-CoA can be used in different pathways including the Krebs cycle. In this pathway,
one molecule of NADH2 is produced. NADH2 enters the OXPHOS process and releases 3
ATP. The gure and abbreviations are explained in the text and in gure 2.

Pyruvate dehydrogenase (PDH) or oxidative decarboxylation


reaction
This mitochondrial reaction is the process that happens following
glycolysis and preceding the Krebs cycle. PDH reaction utilizes

Author's personal copy


1346

M. Dashty / Clinical Biochemistry 46 (2013) 13391352

PDC loses its phosphate using PDH phosphatase and becomes active.
Insulin stimulates this reaction and consequently the Krebs cycle.
Mg 2 + and Ca 2 + are the stimulators of PDH phosphatase (Fig. 8).
The level of acetyl-CoA (product of PDH reaction) is the regulator
of PDK as well as PC (enzyme of gluconeogenesis) and the PDH itself
(enzyme of PDH reaction). Acetyl-CoA is also the substrate of FA synthesis and the Krebs cycle. Therefore, in the high level of acetyl-CoA
(or in excess of intracellular energy), gluconeogenesis (through OAA
production) and FA synthesis pathways are activated.
Translocation of the acetyl-CoA from the mitochondria to the cytoplasm happens via conversion to citrate. First, acetyl-CoA is converted
to citrate using citrate synthesis, and then citrate translocates to the cytoplasm and is degraded back to acetyl-CoA using citrate lyase. In the cytoplasm, acetyl-CoA carboxylase (ACC) converts acetyl-CoA to malonylCoA and proceeds toward the FA synthesis (Graphic abstract gure).
Malonyl-CoA is the inhibitor of carnitine palmitoyltransferase I (CPT-I)
and consequently FAO. This is the way that increased rate of glucose oxidation results in inhibition of FAO. The reason for high regulation of the
PDC is that there is no pathway in the body to convert acetyl-CoA to glucose. Adenosine monophosphate-activated protein kinase (AMPK) that
is the energy sensor of cells has the opposite effect on the function of
ACC. Low energy in cells stimulates both glucose degradation and the
function of AMPK. AMPK consequently inhibits ACC and decreases the
concentration of malonyl-CoA. As a result, FAO and production of
acetyl-CoA is increased.
Carbohydrates, lipids and amino acids are the substrates which
produce acetyl-CoA. Acetyl-CoA is used in different biochemical
pathways including (i) the Krebs cycle to produce H2O, CO2 and energy,
(ii) biosynthesis of cholesterol, squalene, bile acids, vitamin D3 and
steroidal hormones such as estrogen, progesterone, testosterone and
adrenal hormones, (iii) synthesis of FAs, (iv) synthesis of ketone bodies
such as acetone, beta-hydroxybutyric acid (BHB) and acetoacetic acid,
in the liver in starvation and the diabetic states and (v) synthesis of
acetylcholine that is the carrier of nerve impulses (Fig. 10).
Krebs cycle (citric acid or tricarboxylic acid (TCA) cycle)

Fig. 9. Tricarboxylic acid (TCA) cycle or Krebs cycle. The mitochondrial oxidative Krebs cycle is
a process, whereby the product of the PDH complex (acetyl-CoA) joins to OAA. In this process glucose is degraded to produce energetic molecules such as GTP, NADH2 and FADH2.
These reduced products release their energy in the IMM and via the OXPHOS reaction.
The gure and abbreviations are explained in the text and in gure 2.

coenzyme A (CoA or CoASH), thiamine pyrophosphate (TPP) and PDC


to oxidize cytoplasmic pyruvate, which is transferred to the mitochondria by a carrier protein, to acetyl-CoA and CO2. In this reaction, one
molecule NAD + is reduced to NADH2 (Fig. 3/6). In the absence of TPP,
pyruvate is concentrated in the cytoplasm and converted to lactate
(Fig. 10).
Regulation of PDH complex
The fate of pyruvate in the mitochondria depends on the level of
intracellular energy. In the absence of energy, pyruvate is used in the
Krebs cycle to produce ATP, while in the excess of energy (resulted
from high concentration of acetyl-CoA) pyruvate can be used in the
gluconeogenesis pathway and the excess acetyl-CoA can be consumed
in the lipogenesis pathway. Therefore, acetyl-CoA and PDC are the regulators of glycolysis, gluconeogenesis, fatty acid oxidation (FAO that
produce acetyl-CoA) and the Krebs cycle (Fig. 10) [13].
PDH kinase (PDK) phosphorylates and inhibits the PDC (Fig. 3/8).
The products of the PDC (acetyl-CoA and NADH2) and ATP are the activators of PDK and the substrates of the PDC (pyruvate, NAD + and
CoASH), ADP, Mg 2 + and Ca 2 + are the inhibitors of PDK. In high concentrations of ADP (or in shortage of energy), the phosphorylated

The Krebs cycle is an aerobic biodegradation process that starts from


catabolism of acetyl-CoA to produce the reduced coenzymes (NADH2
and FADH2) and CO2. OAA (derived from pyruvate carboxylation) is
the rst substrate of the Krebs cycle. OAA joins to acetyl-CoA to form
citric acid (CA). Therefore, the source of OAA is sugar. In diabetic
patients, who have less sugar and pyruvate in their cells, the level of
OAA and consequently the activity of the Krebs cycle is low.
In the higher energy level, the rate of citrate synthesis increases
and the rate of ux via the Krebs cycle decreases. This excess citrate
is then transferred to the cytosol and degraded to acetyl-CoA. Thereafter, acetyl-CoA can be used in the FA and cholesterol synthesis.
Citrate also has a positive regulatory effect on the enzymes of lipogenesis (e.g., ACC), and a negative regulatory effect on PFK1 (the enzyme of glycolysis) (Fig. 6). Citrate is also required for ketogenesis in
the non-hepatic tissues.
Alpha-ketoglutarate (KG) converts to succinyl-CoA via several
processes of the alpha-ketoglutarate dehydrogenase (KGDH) complex (Fig. 9). Succinyl-CoA is a high energetic molecule and produces
the only GTP molecule of this process under the inuence of succinate
thiokinase. The energetic molecules produced in this oxidative decarboxylation cycle are NADH2 and FADH2. These products release their
energy during the OXPHOS reaction and consequently their energy is
reserved as high energy phosphate in ATP.
Malate-aspartate shuttle
The malate-aspartate shuttle is a process, which transfers electrons of
the reducing equivalents of the glycolysis pathway (NADH2) from
cytoplasm to the mitochondria in the form of malate. In this process,

Author's personal copy


M. Dashty / Clinical Biochemistry 46 (2013) 13391352

1347

Fig. 10. Presentation of the linking role of pyruvate, G6P and acetyl-CoA in carbohydrate and lipid pathways. In this gure, the interrelation between glycogenesis, glycogenolysis, glycolysis, gluconeogenesis, the Krebs cycle, the urea cycle and hexose monophosphate (HMP) shunt (blue rectangle) is illustrated. In HMP shunt, G6P (product of glycogenolysis) is used in the pentose
phosphate pathway to produce Xu5P. Xu5P is also formed from F6P and GA3P by transketolase activity. Xu5P activates PP2A, which is the inducer of PFK2 and ChREBP. ChREBP is a lipogenic
transcription factor that which can induce triglyceride synthesis. Here also the linking role of pyruvate (product of glycolysis) and acetyl-CoA (product of PDH reaction) in carbohydrate and
lipid pathways are highlighted. As is shown, acetyl-CoA regulates the fate of pyruvate, gluconeogenesis and lipogenesis and is the substrate for acetylcholine, ketone bodies, biliary acids,
cholesterol, steroids and triglyceride synthesis. PDC is the main regulatory enzyme of acetyl-CoA. This complex is regulated by the PDH phosphatase and PDK enzymes. Ca2+ and Mg2+
are the activators of PDH phosphatase and the inhibitors of PDK. ATP, NADH2 and acetyl-CoA are the stimulators of PDK. The gure and abbreviations are explained in the text and gure 2.

inter-conversion of the malate and OAA in the cytoplasmic membrane


and OAA and aspartate in the mitochondrial membrane lead to transfer
of NADH2 from the cytosol to the mitochondria (Graphic abstract gure).

produces 29 to 30 ATP molecules and this maximum amount is never


yielded.
TCA regulation

Energy balance
Theoretically, 38 ATP molecules are made from one molecule of
glucose during cellular respiration. 8 ATP are directly formed from the
anaerobic glycolysis via degradation of one glucose molecule to two
pyruvate molecules. In the pyruvate decarboxylation reaction, each
3C-pyruvate produces one NADH2 (equal to 3 ATP). Therefore, each
6C-glucose in this reaction produces 6 ATP. In the Krebs cycle, the
total amount of energy that is made from each 3C-pyruvate is 12 ATP.
This amount is produced at the level of isocitrate dehydrogenase
(NADH2 = 3 ATP), -ketoglutarate dehydrogenase (NADH2 = 3
ATP), malate dehydrogenase (MDH) (NADH2 = 3 ATP), succinate dehydrogenase (FADH2 = 2 ATP) and succinate thiokinase (GTP). Since
each 6C-glucose molecule produces two 3C-pyruvate molecules, the
total amount of energy, which is made from one glucose is 24 ATP.
The 8 ATP of glycolysis, 6 ATP of the PDH complex and 24 ATP of the
Krebs cycle together produce 38 ATP.
Since the energy of each ATP is equal to 8 kcal, therefore the total
amount of the released energy is 304 kcal. This amount of obtained
energy in the chemical form is equal to 44% of the total amount of
energy which is measured by a calorimeter. Therefore, 56% of the
energy of glucose is released as heat to stabilize the body temperature. This wasting energy is through proton loss via the leaky mitochondrial membranes that ow back the protons from the IMS to the
matrix. Moreover, moving pyruvate and ADP into the mitochondrial
matrix requires energy. Therefore, in reality each glucose molecule

TCA is regulated through different ways including (i) allosteric


regulation by Ca 2+, ATP and ADP, (ii) the level of acetyl-CoA (the
entry substrate of the cycle), (iii) substrate availability (e.g., reduced
OAA regulates citrate synthase), (iv) NAD +/NADH ratio (e.g., the
TCA cycle needs NAD + as cofactor) and (v) product inhibition (e.g.,
citrate inhibits citrate synthase as well as NADH2 and succinyl-CoA
inhibit KGDH).
Glycogenesis
Glycogenesis is the process of glycogen synthesis. Glycogen is a
polymer of glucose residues that is linked by -1,4 and -1,6 glycosidic
bonds. Therefore, it is the glucose storage molecule in the hepatocytes
and skeletal muscle cells. The total amount of glycogen storage among
these two tissues depends on the mass of the hepatocytes and skeletal
muscle cells. Glycogen amount per mass unit of the liver is higher
than the skeletal muscle; however, since in body the total mass of the
skeletal muscle is more than that of the liver, thus 2/3 of the total
amount of glycogen is stored in the muscle cells. Liver glycogen is for
the stability of the blood sugar to supply enough energy for other tissues
(mainly the brain that consumes 75% of the blood glucose). However, glycogen in the skeletal muscle is used just as their sources of energy because
muscle cells don't have G6Pase. In glycogenesis, glucose is joined to a core
of glycogen via -1,4 linkage. G1P converts to the active form of glucose (UDPG) using Glucosyl-1-phosphate uridyl-transferase (UGP2
or UDPG pyrophosphorylase) and one uridine triphosphate (UTP)

Author's personal copy


1348

M. Dashty / Clinical Biochemistry 46 (2013) 13391352

Fig. 11. Glycogenesis and glycogenolysis. Regulation of blood glucose is in close association with the biochemical regulation of glycogen pathways. G6P and F6P (products of glycolysis) are the
linkers of glycolysis and glycogen pathways. The gure and abbreviations are explained in the text and in gure 2.

[14]. Consequently, one group of pyrophosphate is made. Active glucose joins to the core of glycogen via -1/4 linkage using glycogen
synthase to elongate the glycogen molecule. By this way, C1 of

glucose joins to the C4 of glycogen and UDP molecule is released.


When the number of the joined glucose increases till 1012 glucose
molecules in each branch, amylo--(1,4 1,6)-transglycosylase

Fig. 12. Inuence of glucagon on carbohydrate pathways. In the fasted state, glucagon stimulates both liver glycogenolysis via activation of glycogen phosphorylase and gluconeogenesis via
stimulation of FBPase2 in order to elevate the blood sugar. Glucagon also inhibits glycogenesis via phosphorylation of glycogen synthase. Glucagon inuences beta receptors and uses
adenylate cyclase to stimulate conversion of ATP to cAMP. PKA is a protein kinase, which is cAMP-dependent and is activated through this receptor-mediated mechanism. The cAMP/
PKA complex phosphorylates (and activates) phosphorylase kinase (a form), which phosphorylates (and activates) phosphorylase (a form). Both phosphoprotein kinase and phosphorylase are dephosphorylated (and inhibited-b form) by phosphoprotein phosphatase. The inhibitor of phosphoprotein phosphatase is PPI1. PPI1 dephosphorylates (and inhibits) phosphorylase kinase (b form), phosphorylase (b form) and itself (b form). Phosphorylase kinase, phosphorylase and PPI1 are all activated (a-form) via phosphorylation by using ATP. PPI1
is phosphorylated (and activated) by cAMP/PKA and dephosphorylated (and inhibited-b form) by phosphoprotein phosphatase. Phosphorylase kinase is also activated by calcium
even without phosphorylation. This allows acetylcholine to increase glycogenolysis via neuromuscular stimulation and without receptor stimulation. In the normal state and excess
glucose, glycogenesis is activated and glycogenolysis is inhibited to reserve the glucose in the body. The gure and abbreviations are explained in the text and in gure 2.

Author's personal copy


M. Dashty / Clinical Biochemistry 46 (2013) 13391352

1349

Fig. 13. Pentose phosphate pathway (PPP). PPP is a cytoplasmic pathway and is composed of
two oxidative and non-oxidative (highlighted) stages. This glucose oxidation pathway consumes G6P as substrate to produce F6P, Xu5P and NADPH2. NADPH2 is used in the lipid biochemical pathways. The gure and abbreviations are explained in the text and in gure 2.

(branching enzyme) picks 6- to 7-glucose molecules up from the branch


and via -1/6 linkage makes a new branch in the glycogen molecule
(Fig. 11). Insulin is the activator of glycogen synthase.
Glycogenolysis
Glycogenolysis is the process of glycogen degradation. Glycogenolysis happens in the liver and kidney to produce glucose for balancing the
blood sugar; however, it produces G6P in muscle cells to be used as the
energy supplier of myocytes.
Glycogen phosphorylase using inorganic phosphate group (Pi)
hydrolyzes the -1,4 glycosidic linkages of glycogen and produces G1P
(or glucose in the heart). G1P is converted to G6P and thereafter to
glucose using G6Pase in the liver and kidney. In the skeletal muscle due
to the absence of G6Pase, G6P converts to F6P to be used in glycolysis.
Phosphorylase continues degradation of the -1,4 linkages until
only four glucose molecules remain on the branch. Then three
glucose molecules are transferred to another glucose-branch via
glucan transferase (amylo--(1,4 1,6) transferase) and the last
glucose molecule (-1,6 linkage) is degraded with glucosidase
(amylo--1,6-glucosidase) activity (Fig. 11). During exercise, adrenaline in the skeletal muscle and during hypoglycemia, glucagon in the
liver is the stimulator of glycogenolysis. This happens by activating the
glycogen phosphorylase. Therefore, glycogenolysis results in nutrition
of the skeletal muscle cells and maintenance of blood glucose. Adrenaline and glucagon affect beta receptor activity and use adenylate cyclase
to convert ATP to cyclic AMP (cAMP) (Fig. 12).
Phosphorylase
Glycogen phosphorylase is a lytic enzyme in glycogenolysis. Phosphorylase joins a phosphate group to the glucose residue of glycogen
and catalyzes the phosphorylation reaction and consequently the cleavage of glucose from glycogen in the form of G1P (phosphorolysis

Fig. 14. Gluconeogenesis. Gluconeogenesis is a mitochondrialcytoplasmic pathway


that happens mainly in the liver and kidney. The substrates of this pathway are pyruvate
(product of glycolysis), lactate (produced from pyruvate), alanine, glutamic acid and glycerol
(product of triglyceride breakdown). Glycerol is used in gluconeogenesis via glycerol
phosphate shuttle. Shifting of oxaloacetate from the mitochondria to cytoplasm happens
via transition to malate. PEP is the substrate of glycolysis and G6P is the substrate of
glycogen synthesis. The gure and abbreviations are explained in the text and in gure 2.

reaction). Phosphorylase is in either the active (R) form or the inactive


(T) form. Liver phosphorylase is in dimeric form and skeletal muscle
phosphorylase is in tetrameric form (Fig. 3/9). Each monomer of this
enzyme is composed of the C-terminal domain, N-terminal domain, active site and pyridoxal phosphate (PLP, derived from vitamin B6) cofactor, which binds near the active site to facilitate the reaction. The active
site contains a serine amino acid that can be affected by the phosphorylase kinase and phosphorylase phosphatase. Phosphorylation of serine
by phosphorylase kinase and using ATP produces an activated form,
whereby the monomers are attached to each other. This happens after
change in the shape of phosphorylase and production of an ester linkage between OH of serine and the phosphoric acid. In addition, serine
can join to PLP (B6-al-PO4) and form an active phosphorylase. Phosphorylase phosphatase inactivates phosphorylase via losing its phosphate group.
Pentose phosphate pathway (PPP), phosphogluconate pathway or
hexose monophosphate shunt
In the liver, adipose tissue, adrenal cortex, testis, milk glands,
phagocyte cells and RBCs another glucose oxidation pathway exists
that is called pentose phosphate pathway (PPP). G6P is the substrate

Author's personal copy


1350

M. Dashty / Clinical Biochemistry 46 (2013) 13391352

Fig. 15. Association between glycolysis, gluconeogenesis and pentose phosphate pathways and the role of Xu5P (product of PPP) in PP2A activation and FA synthesis. Xu5P-activated PP2A
by using ATP and H2O and following activation of phosphoprotein phosphatase and hydroxylation of the PFK2/FBPase2 complex phosphorylates F6P to form F2,6BP. F2,6BP activates PFK1 (regulatory enzyme of glycolysis) and inhibits FBPase1 (regulatory enzyme of gluconeogenesis). These effects lead to increase in the level of F1,6 BP and glycolysis.
Xu5P-activated PP2A also activates ChREBP and gene expression of lipogenic enzymes, including ACL, ACC, and FAS. F1,6BP, PFK1 and FBPase1 are regulated by F2,6BP and the ratio of
ATP/ADP. In high level of F2,6BP and low level of this ratio, glycolysis will proceed maximally. The gure and abbreviations are explained in the text and gure 2.

of PPP to produce ribose-5-phosphate (R5P), riboluse-5-phosphate


(Ru5P) and reduced coenzyme NADPH2. R5P is either used for nucleotide synthesis of nucleic acids or is recycled in the PPP for more production of NADPH2. The reduced equivalent (NADPH2) is consumed
for the reductive biosynthesis reactions such as synthesis of FA and
cortical steroidal hormones of the adrenal gland. In RBCs, NADPH2
is consumed for glutathione reduction. Oxidized glutathione (GSSG)
converts to the reduced form of glutathione (GSH) using glutathione
reductase and NADPH2 as coenzyme. In the absence of the GSH, the
phospholipids of RBCs that contain unsaturated FAs become oxidized
and lyse the RBC membrane (Fig. 3/10).
Therefore, NADPH2 has three functions in RBC: (i) supporting the RBC
membrane, (ii) supporting the ferrous (Fe2+) group of hemoglobin and
(iii) reduction of enzymes which have cysteine (Cys) in their active site
(Cys has SH). In the phagocytic cells (e.g., neutrophils and macrophages),
NADPH2 is used to produce superoxide radicals from oxygen and also reactive oxygen species (ROS) to kill microorganisms. Therefore, during
phagocytosis the phagocytic cells increase their oxygen utilization dramatically in order to attack microorganisms (it is called oxygen burst). Reductive and oxidative enzymes utilize the NADP+/NADPH and NAD+/
NADH cofactor pairs respectively. 50% of glucose oxidation in adipose tissue and 30% of glucose oxidation in liver happens through PPP. Thereby,
adipocytes, hepatocytes and also mammalian gland cells can produce
fats due to the existence of NADPH2 in their tissues. NADPH2 can be produced both in PPP and gluconeogenesis (Figs. 3/11 and 13).

(PEPCK-M), which is the most important allosteric enzyme in the kidney


(part of the transhydrogenase cycle), (ii) transamination to aspartate
and (iii) reduction to malate [15]. In reaction (iii), oxaloacetate is
converted to malate using MDH and NADH2. This process happens in
the mitochondria and is the opposite of the Krebs cycle. Malate passes
through the mitochondrial membrane and is reconverted to oxaloacetate in the cytoplasm using NADPH2 as coenzyme. The NADH2 of this
process is used in the GA3PDH reaction of glycolysis. Therefore, coupling
of these two oxidation-reduction reactions is necessary for the functionality of gluconeogenesis. Conversion of OAA to malate predominates
when pyruvate is the substrate of gluconeogenesis. Gluconeogenesis is
active in the shortage of glucose supply, for instance during a long
time starvation or diabetic states where the body uses fats and amino
acids instead of carbohydrates.
The regulators of this pathway are adrenocorticotropic hormone
(ACTH), ATP, AMP, ADP and insulin. ATP by stimulation of phosphatase and ACTH by stimulation of cortisol secretion enhances
gluconeogenesis. In contrast, AMP, ADP and insulin by inhibition of
phosphatase regulate gluconeogenesis negatively. Thyroxin, which increases the basal metabolic rate and the catabolism of fats and proteins,
stimulates gluconeogenesis. In this state, the excess amount of amino
acids, FAs and glycerol convert to carbohydrate derivatives via gluconeogenesis (Figs. 3/7 and 14).

Gluconeogenesis

Cori cycle is a hepatic gluconeogenesis that consumes lactate as its


substrate. During the Cori cycle, the non-hepatocyte cells including skeletal muscle cells and erythrocytes consume glucose to produce lactate via
glycolysis. Lactate is then transferred to the hepatocytes for gluconeogenesis. Since during hepatic gluconeogenesis 6 ATP are consumed and during the Cori cycle in the non-hepatic tissues 2ATP are produced, therefore
the nal result is consumption of 4 ATP in each cycle. Thus, the Cori cycle
cannot be a permanent process for energy production (Figs. 3/12 and 7).

Gluconeogenesis is the process in which non-carbohydrate molecules


(pyruvate, lactate, glycerol, alanine and glutamine) are converted to glucose in the liver, kidneys, brain, testes and erythrocytes. Gluconeogenesis
is the reverse process of glycolysis and happens mostly in the cytoplasm.
It starts from conversion of pyruvate to oxalate in the mitochondria
using PC and biotin as its coenzyme. Oxalate is required to be
used in the cytoplasm; therefore, it has to be transferred across the
mitochondrial membrane. The mitochondrial membrane is not diffusible to oxalate. Therefore, OAA is transported to the cytoplasm in
one of the following ways (i) conversion to PEP through function
of the mitochondrial isoform of phosphoenolpyruvate carboxykinase

Cori cycle

Glucose-alanine cycle
Alanine is another source of gluconeogenesis in the liver. Alanine
is produced from the transamination process in non-hepatic tissues.

Author's personal copy


M. Dashty / Clinical Biochemistry 46 (2013) 13391352

1351

is produced in the skeletal muscles during the prolonged fasting state,


when muscles use amino acids as their source of energy. In this state,
kidney gluconeogenesis removes the waste nitrogen that is produced by
the catabolism of amino acids [1618].

Regulation of gluconeogenesis

Fig. 16. Presentation of gluconeogenesis transcription factors. PGC1 is the key regulator of
gluconeogenesis. Glucagon/PKA phosphorylates CREB transcription factor and recruits
CBP coactivator to induce PGC1 coactivator. PGC1 coactivates and forms complexes
with other gluconeogenesis transcription factors including FOXO1, GR, HNF4 (as well
as PPAR) to induce the gluconeogenesis promoters. PEPCK and G6Pase are the gluconeogenesis genes (the same name of the related enzyme). AF2 is the PEPCK promoter and IRU
is the G6Pase promoter. Post-translational modication of PGC1 by reversible acetylation
and phosphorylation of PGC1 regulates the function of PGC1. Insulin/Akt inhibits
PGC1 via induction of PGC1 phosphorylation and degradation of the hepatic PGC1.
SIRT1 induces PGC1 activity via deacetylation of PGC1. SIRT1 stimulates gluconeogenesis through coactivation of FOXO1 and HNF4. GCN5 acetylates and inhibits PGC1 and
gluconeogenic activity. GCN5 is a member of the N-acetyltransferase superfamily. This gure
is mainly the idea of Sugden et al. The gure and abbreviations are explained in the text
and gure 2.

During transamination, one -amino acid donates the amino group


and generates one -keto acid. During the glucose-alanine cycle, the
non-hepatic tissues send the amino part of the catabolized amino
acids to the liver to be excreted as urea; therefore, nitrogen is eliminated from muscles while replenishing its energy supply (Fig. 7).
Amino acids
Except for leucine and lysine, the carbon skeleton of all other
amino acids can be degraded to the intermediates of the TCA cycle
(e.g., oxaloacetate and pyruvate) and are used in gluconeogenesis. By
this way, during starvation amino acids are used for the maintenance
of glucose in circulation.
Glycerol
FAs are highly energetic molecules; however, because their
carbons cannot be used for glucose synthesis, body cannot use FAs directly as a source of energy. The glycerol backbone of lipids is linked
to glycolysis via the glycerol-phosphate shuttle.
In the glycerol-phosphate shuttle, glycerol is phosphorylated
to glycerol-3-phosphate (G3P) and then dehydrogenated to DHAP
by Glycerol-3-phosphate dehydrogenase (G3PDH) in the cytoplasm.
G3PDH is the same enzyme which is used for transporting electrons
from the cytosolic NADH2 (produced by glycolysis) into the mitochondrial oxidized carriers (FAD+) to be used in the OXPHOS reaction. Therefore,
continuous conversion of DHAP and G3P (products of glycolysis) to each
other leads to transfer of electron from the cytosolic reduced NADH2 to
the mitochondrial oxidized FAD+ (Graphic abstract gure).
Role of renal gluconeogenesis
Gluconeogenesis happens in the liver as well as in the kidney. Therefore, in liver failure, the kidney is involved in the maintenance of blood
glucose. The substrate of kidney gluconeogenesis is glutamine. Glutamine

Glycolysis and gluconeogenesis are regulated simultaneously and


through the same parameters, however, in the opposite direction to
each other. The main regulatory enzymes of glycolysis and gluconeogenesis are PFK1 and F1,6BPase respectively. Both of these enzymes are regulated by the bifunctional PFK2/F2,6BPase2 enzyme. Insulin and glucagon
regulate glycolysis and gluconeogenesis via inuence on the component
of this enzyme. Glucagon (and also catecholamine) reduces the level of
F2,6BP and insulin increases the activity of PFK2 and consequently increases the function of F2,6BP. F2,6BP has an inhibitory effect on F1,6BP
and a stimulatory effect on PFK1. Therefore, glucagon stimulates gluconeogenesis and insulin stimulates glycolysis (Fig. 15). Insulin also activates phosphodiesterase (PDE), which hydrolyses cAMP to AMP [13,19].
Another point for gluconeogenesis regulation is at the level of
pyruvate to PEP bypass. PK (the glycolysis enzyme, which converts PEP
to pyruvate) is negatively regulated through phosphorylation by glucagon and epinephrine. This modies the pathway to the gluconeogenesis
direction. ATP and alanine are the other inhibitors of PK. This means
that in the high level of energy and availability of the gluconeogenesis
substrate the gluconeogenesis pathway is active.
The other regulatory enzymes of gluconeogenesis are G6Pase, PC and
PEPCK (rst control point). Cortisol is the activator of these enzymes and
ADP (or low energy level in the cell) is the inhibitor of them. During
prolonged starvation, glucagon is secreted. Glucagon increases the
cAMP level and stimulates PEPCK and gluconeogenesis and produces glucose for organs like the brain, which are dependent on glucose.
cAMP (activated by glucagon and epinephrine) phosphorylates the
transcription factor cAMP response element-binding protein (CREB) of a
target gene and results in the recruitment of the transcriptional P300/
CBP (CREB-binding protein) coactivator. The CREBCREP300/CBP complex can alone or by recruitment of other coactivator molecules stimulate
the gene expression of the gluconeogenesis enzymes. These coactivator
molecules are peroxisome proliferator-activated receptor-gamma
coactivator-1 alpha (PGC1), glucocorticoid receptor (GR or GCR), hepatic nuclear factor-4 alpha (HNF4) and forkhead transcription factor
(FOXO).
Insulin has the opposite effect on gluconeogenesis. It activates PDE
and hydrolyzes cAMP to AMP and switches off the gluconeogenesis transcription factors. Insulin/phosphoinositide 3-kinase (PI3K) phosphorylates CBP, which is adjacent to CREB-binding domain (CREB-BD). Insulin
also excludes FOXO from the nucleus to the cytoplasm via FOXO phosphorylation and inhibits its stimulatory effect on the gluconeogenesis
gene expression (Fig. 16) [19,20,21].

Conclusion
Energy homeostasis is one of the main tasks of the body. Regulation
and retour of energetic molecules like glucose and FAs is a complex process in the body in which all cells are involved. Adipose tissue, skeletal
muscle and liver are the main metabolic organs in the body. The normal
function of these metabolic organs is represented as normoglycemic
and normolipidemic states in the circulation. Understanding the
biochemistry of carbohydrates and lipids is one of the main steps towards comprehending the pathophysiology of metabolic diseases like
metabolic syndrome, diabetes and dyslipidemia. This review summarizes and illustrates a general view on the biochemistry of carbohydrates. The goal of this attempt is to facilitate the understanding of
complex biochemical pathways for researchers.

Author's personal copy


1352

M. Dashty / Clinical Biochemistry 46 (2013) 13391352

Acknowledgment
I would like to extend my appreciation to Dr Nessa Dashty
Rahmatabady for her useful comments in this paper.
References
[1] Tymoczko B, Stryer T. Biochemistry Sixth Edition, 2007, by W.H. Freeman and Company.
[2] Rosensweig NS, Herman RH. Control of jejunal sucrase and maltase activity by
dietary sucrose or fructose in man. A model for the study of enzyme regulation
in man. J Clin Invest 1968;47:225362.
[3] Hauri HP, Sander B, Naim H. Induction of lactase biosynthesis in the human intestinal epithelial cell line Caco-2. Eur J Biochem 1994;219:53946.
[4] Taketa K, Watanabe A, Shimamura J, Takesue A, Ueda M. Undifferentiated patterns of
key glycolytic and gluconeogenic enzymes in epithelial cell lines derived from rat
liver. Acta Med Okayama 1975;29:1519.
[5] Yeh KY, Yeh M, Holt PR. Intestinal lactase expression and epithelial cell transit in
hormone-treated suckling rats. Am J Physiol 1991;260:G37984.
[6] Mac DO, Chediack JG, Caviedes-Vidal E. Isolation of epithelial cells, villi and crypts
from small intestine of pigeons (Columba livia). Biocell 2008;32:21927.
[7] Stevens BR, Kaunitz JD, Wright EM. Intestinal transport of amino acids and sugars:
advances using membrane vesicles. Annu Rev Physiol 1984;46:41733.
[8] Fairchild TJ, Armstrong AA, Rao A, Liu H, Lawrence S, Fournier PA. Glycogen synthesis
in muscle bers during active recovery from intense exercise. Med Sci Sports Exerc
2003;35:595602.
[9] Hessman Y, Thoren L. Glycogen storage in rat liver and skeletal muscle in thermal
trauma. I. Effect of exogenous insulin. Acta Chir Scand 1975;141:38592.

[10] Marin P, Hogh-Kristiansen I, Jansson S, Krotkiewski M, Holm G, Bjorntorp P.


Uptake of glucose carbon in muscle glycogen and adipose tissue triglycerides in
vivo in humans. Am J Physiol 1992;263:E47380.
[11] Gale EA, Bennett T, Green JH, MacDonald IA. Hypoglycaemia, hypothermia and
shivering in man. Clin Sci (Lond) 1981;61:4639.
[12] Green JH, MacDonald IA. The inuence of intravenous glucose on body temperature.
Q J Exp Physiol 1981;66:46573.
[13] Kabashima T, Kawaguchi T, Wadzinski BE, Uyeda K. Xylulose 5-phosphate
mediates glucose-induced lipogenesis by xylulose 5-phosphate-activated protein
phosphatase in rat liver. Proc Natl Acad Sci USA 2003;100:510712.
[14] Dillard WL, Graf L, Schweiger HG. Regulation of UDPG pyrophosphorylase in
Acetabularia mediterranea. Eur J Cell Biol 1983;29:1459.
[15] Wynn JP, Hamid AA, Li Y, Ratledge C. Biochemical events leading to the diversion
of carbon into storage lipids in the oleaginous fungi Mucor circinelloides and
Mortierella alpina. Microbiology 2001;147:285764.
[16] DeFronzo RA, Davidson JA, Del PS. The role of the kidneys in glucose
homeostasis: a new path towards normalizing glycaemia. Diabetes Obes Metab
2012;14:514.
[17] Goodman AD, Fuisz RE, Cahill Jr GF. Renal gluconeogenesis in acidosis, alkalosis,
and potassium deciency: its possible role in regulation of renal ammonia
production. J Clin Invest 1966;45:6129.
[18] Mather A, Pollock C. Glucose handling by the kidney. Kidney Int Suppl 2011:
S16.
[19] Veech RL. A humble hexose monophosphate pathway metabolite regulates
short- and long-term control of lipogenesis. Proc Natl Acad Sci USA 2003;100:
557880.
[20] Sugden MC, Caton PW, Holness MJ. PPAR control: it's SIRTainly as easy as PGC.
J Endocrinol 2010;204:93104.
[21] Wallace JC, Barritt GJ. Gluconeogenesis. Encyclopedia of life sciences 2002;
Macmillan Publishers Ltd; 18.

Das könnte Ihnen auch gefallen