Vous êtes sur la page 1sur 17

C H A P T E R

4
Metabolic Pathways
and Cycles
Christina Werner, Torsten Doenst, Michael Schwarzer
Department of Cardiothoracic Surgery, Jena University Hospital,
Friedrich Schiller University of Jena, Jena, Germany

INTRODUCTION LIPIDS AND FATTY ACIDS

Metabolic pathways and cycles are reaction Lipids are the major source of energy (60–
chains where chemical products become the 90%) for cardiac contraction [1]. Almost the
substrate for the next step. All substrates are entire nutritional fat consists of one to three
chemically transformed in reactions that belong fatty acids esterified with glycerol. Lipase dis-
to either pathways (if the reactions are aligned sociates glycerol and prepares free fatty acids
in linear fashion) or metabolic cycles (if the moi- for cellular uptake. Cellular uptake of fatty ac-
eties of the reactions are preserved). The term ids is passive or mediated by fatty acid trans-
substrate oxidation is used for substrate deg- locase (FAT/CD36) and cardiac-specific fatty
radation ultimately leading to CO2 production. acid binding protein (H-FABP) [2]. Once inside
However, such an oxidative pathway may be the cell, fatty acids are esterified to coenzyme A
interrupted for several reasons, such as in the (CoA) in an ATP-requiring reaction that is cata-
case of glucose oxidation, where limited oxygen lyzed by fatty acid thiokinase. After this acti-
results in the production of lactate rather than vation step, 70–90% of the fatty acids undergo
water and CO2. Some but not all substrates can immediate oxidation. The rest are stored in the
also be converted to building blocks for biosyn- intracellular triglyceride pool [3].
thetic processes. This chapter will describe the In order to be oxidized, fatty acids must be
main metabolic pathways and cycles of cardiac transported into the mitochondria. Short-chain
substrate metabolism. It will also address their fatty acids and ketone bodies do not require a
interactions and their contributions to meet car- specific transport system but enter the mito-
diac energy requirements. chondria by diffusion. Since the majority of

The Scientist’s Guide to Cardiac Metabolism


http://dx.doi.org/10.1016/B978-0-12-802394-5.00004-2
39 Copyright © 2016 Elsevier Inc. All rights reserved.
40 4.  Metabolic Pathways and Cycles

fatty acids are long chained, a specific transport


mechanism, the carnitine-palmitoyl transferase
system, is required. This system consists of two
enzymes, carnitine-­palmitoyl transferase I and
II (CPT-I/II) and transfers the impermeable
acyl-CoA to the mitochondrial matrix by tem-
porarily converting it to acyl-carnitine. CPT-I
catalyzes the reaction from acyl-CoA to acyl-
carnitine at the outer mitochondrial membrane
and CPT-II reverts the reaction at the inner mi-
tochondrial membrane. Acyl-CoA inside the
mitochondrial matrix enters fatty acid oxidation
immediately. CPT-I activity is therefore seen as
a rate-­limiting step for fatty acid oxidation [4].
Fatty acid breakdown is driven by b-oxidation.
It mainly takes place inside the mitochondria,
but to a much smaller extent, may also occur in
peroxisomes.

FATTY ACID OXIDATION

The principle of b-oxidation is the stepwise


dissociation of two carbon atoms from fatty ac-
ids and the coenzyme A supported formation
of acetyl-CoA, NADH, and FADH2 [5]. The full FIGURE 4.1  Reaction steps of fatty acid oxidation
spiral of b-oxidation is shown in Fig. 4.1 and its (b-­oxidation).
energetic balance is given in Table 4.1.
First, acyl-CoA is oxidized by acyl-CoA
dehydrogenase and FAD+ and results in a,b-­ NAD+-dependent b-hydroxyacyl-CoA dehy-
unsaturated acyl-CoA and FADH2. This is the drogenase oxidizes b-hydroxyacyl-CoA to
first of four reactions in the b-oxidation chain. b-ketoacyl-CoA and NADH. This reaction is the
The a,b-­unsaturated acyl-CoA is hydrated by second oxidation step and has given b-oxidation
enoyltransferase to b-hydroxy-acyl-CoA. The pathway its name. The next step can be seen as

TABLE 4.1 Energetic Balance of Complete Fatty Acid Oxidation Using Stearyl-CoA as Example (18 C-Atoms Fatty
Acid). First Step: ß-Oxidation and Second Step: Oxidation of Acetyl-CoA in the Citric Acid Cycle

Reducing equivalents Resulting ATP after oxidation of ATP output from


Metabolic step NADH FADH2 NADH FADH2 citric acid cycle

Stearyl-CoA → 9 Acetyl-CoA 8 8 21.6 12.8 –


9 Acetyl-CoA → 18 CO2 + 18 H2O 27 9 72.7 14.4 9
Sum 130.7

 
Glucose uptake and glycogen 41
the last and at the same time the first step of one GLUCOSE UTILIZATION
cycle of the b-oxidation spiral. Another coen-
zyme A is added to b-ketoacyl-CoA dissociat- The second important substrate for cardiac
ing one molecule of acetyl-CoA. The remaining energy metabolism is glucose. Glucose metabo-
acyl-CoA has two carbon atoms less than the lism involves the different pathways of glycolysis,
initial fatty acid. Fatty acids with an even num- glycogen synthesis, and the two alternate path-
ber of carbon atoms (practically all naturally ways pentose phosphate pathway (PPP) and hex-
occurring fatty acids) run through several pas- osamine biosynthesis pathway (HBP). Figure 4.2
sages of b-oxidation until all pairs of carbon at- gives an overview of the interactions of the PPP,
oms are converted to acetyl-CoA. Acetyl-CoA the HBP, and glycolysis.
directly enters the citric acid cycle (CAC). Glucose is first transported across the
Fatty acids with odd numbers of carbon atoms plasma membrane and then phosphorylated
only occur in some plants and have rather low by ­ hexokinase. These two steps are defined
importance for cardiac-energy production. How- as ­glucose uptake and this phosphorylation
ever, they can be catabolized to acetyl-CoA as de- step is irreversible since there is no glucose-­6-
scribed earlier. The last step leaves the activated phosphatase in the heart (e.g., as it is in the liv-
three-carbon fatty acid propionyl-CoA. Propio- er). If glucose is ultimately converted to water
nyl-CoA carboxylase catalyzes the ATP-depen- and CO2, the term “glucose oxidation” is used.
dent reaction of propionyl-CoA to 2-methylmal- Full oxidation of glucose represents 10–40% of
onyl-CoA. Activated carboxybiotin serves as the cardiac energy production.
carbon donor for this carboxylation. 2-methyl-
malonyl-CoA is converted into succinyl-CoA by
an isomerase. Finally, succinyl-CoA is able to en-
ter the CAC. This anaplerotic reaction serves as GLUCOSE UPTAKE
replenishment of the cycle i­ntermediates rather AND GLYCOGEN
than for energy generation. For more detailed in-
formation on anaplerotic reactions the reader is Glucose transport in myocytes is driven by
referred to the following text. the translocation of monosaccharide transport-
b-Oxidation is mainly controlled by the ers (GLUT-4 and GLUT-1) to the sarcolemma.
presence of activated fatty acids in the cardio- Insulin-mediated GLUT-4 is the major glucose
myocyte. The mechanisms of regulation will transporter in cardiac and skeletal muscle [7]. In
be discussed in Chapter 5. In times of fasting contrast, GLUT-1 shows a much lower glucose
with low substrate supply and glucose restric- uptake rate and rather mediates general glucose
tion, the heart is able to use ketone bodies as uptake in most tissues. Additionally, intracellu-
substrate for energy generation. In the liver, lar glycogen stores are another potential source
acetyl-CoA is transformed to the ketone bod- of glucose. Cardiac glycogen stores are small
ies acetoacetic acid and b-hydroxybutyric acid. compared to other tissues such as liver or skel-
They can enter the cardiomyocyte and mito- etal muscle. There is a rapid turnover of glucose
chondria directly through monocarboxylate to glycogen for storage and of glycogen to glu-
transporters or diffusion and are regenerated cose as substrate in glycolysis (see Chapter 5).
to acetyl-CoA by b-ketoacyl-CoA transferase. Cardiomyocytes therefore present with quite
They contribute to substrate competition by stable glycogen concentration. However, high
the same mechanisms as fatty acids through in- extracellular glucose concentrations increase the
hibition of pyruvate dehydrogenase activity by glycogen pool [8]. In contrast, elevated amounts
acetyl-CoA [6]. of AMP, inorganic phosphate, and a fall in ATP

 
42

 
4.  Metabolic Pathways and Cycles

FIGURE 4.2  Interactions of the pentose phosphate pathway, the hexosamine biosynthesis pathway, and glycolysis.
Glycolysis 43
activate glycogenolysis and result in enhanced droxyacetone phosphate and glyceraldehyde-
substrate supply for glycolysis [9]. 3-phosphate, which are in a dynamic equilibri-
um driven by triosephosphate isomerase. Only
glyceraldehyde-3-phosphate underlies further
GLYCOLYSIS reaction and is oxidized and phosphorylated by
the NAD+-­specific glyceraldehyde-­3-phosphate
Glycolytic enzymes are located in the sarco- dehydrogenase (GAPDH) to 1,3-bisphospho-
plasm and are associated with the sarcoplasmic glycerate and NADH. The following reaction of
reticulum [10,11]. They convert glucose-6-phos- 1,3-­ bisphosphoglycerat to 3-phosphoglycerate,
phate and nicotinamide adenine dinucleotides catalyzed by phosphoglycerate kinase, transfers
(NAD+) to pyruvate and NADH by producing inorganic phosphate to ADP and results in the
two molecules of ATP. Table 4.2 shows the key first substrate chain phosphorylation generating
chemical reactions of glycolysis and their ener- ATP. The enzyme mutase transforms 3-phos-
getic efficiency [12]. phoglycerate to 2-­ phosphoglycerate, which
A scheme of the full glycolysis pathway is dehydrated by enolase under formation of
is shown in Fig. 4.3. First, sarcoplasmic, free energy-rich phosphoenolpyruvate and water.
glucose is activated to glucose-6-phosphate Phosphoenolpyruvate is dephosphorylated to
by hexokinase and transformed to fructose-6-­ pyruvate by pyruvate kinase. Within this reac-
phosphate by phosphohexoisomerase. Hexo- tion step, inorganic phosphate is transferred to
kinase has a high affinity to glucose but is also ADP for a second time during glycolysis and re-
able to utilize fructose or galactose as substrates. sults in the production of the second molecule
The first step of glycolysis is catalyzed by phos- ATP. This reaction is again irreversible due to
phofructokinase (PFK) and converts fructose- the high energetic gradient between phospho-
6-phosphate to fructose-1,6-­ bisphosphate. The enolpyruvate and pyruvate.
two initial phosphorylations by hexokinase and Glycolytic breakdown of glucose ends with
PFK require ATP and are therefore irrevers- the formation of pyruvate. There are four differ-
ible. PFK is one of the key regulators in glycoly- ent reactions pyruvate is involved in afterwards:
sis. It is activated by fructose-2,6-bisphosphate decarboxylation to acetyl-CoA, reduction to lac-
and AMP and inhibited by increased forma- tate, carboxylation to oxaloacetate or malate or
tion of citrate and ATP during oxidative sub- transamination with glutamate to alanine. While
strate phosphorylation in the CAC. In the next acetyl-CoA and lactate are further metabolized
step, the enzyme aldolase divides the hexose within the process of ATP generation, the for-
fructose-1,6-bisphosphate in two trioses, dihy- mation of oxaloacetate, malate, or alanine have

TABLE 4.2 Catalytic Efficiency and ATP Production of Glycolysis


Enzyme Metabolic step ATP output

Hexokinase Glucose + ATP → Glucose-6-phosphate + ADP −1


PFK Fructose-6-phosphate + ATP →
Fructose-1,6-bisphosphate + ADP −1
Phosphoglycerate kinase 1,3-Bisphosphoglycerate + ADP →
3-Phosphoglycerate + ATP 2
Pyruvate kinase Phosphoenolpyruvate + ADP → Pyruvate + ATP 2
Sum 2

 
44 4.  Metabolic Pathways and Cycles

FIGURE 4.3  Reaction steps of glycolysis.

anaplerotic function (see later in the chapter) and ation and oxidation of pyruvate to acetyl-CoA
are favored in case of a lack of CAC intermedi- is supported by the pyruvate dehydrogenase
ates. Under aerobic conditions, pyruvate is trans- complex (PDH), a highly regulated multien-
ported to the inner mitochondrial membrane zyme complex with coenzyme A and thiamine
by the mitochondrial pyruvate carrier MCP1/2 pyrophosphate as coenzymes. PDH serves as
[13,14]. The following irreversible decarboxyl- a key regulator and catalyzes the irreversible

 
Pentose phosphate pathway 45
step in carbohydrate decarboxylation or glu- Besides glycolysis and glycogen synthesis,
cose oxidation. The regulation of this enzyme there are two additional pathways using glu-
is described in detail in Chapter 5. Briefly, PDH cose as substrate: the pentose phosphate path-
activity is limited by its product acetyl-CoA as way and the hexosamine biosynthesis pathway.
well as by NADH generated in the CAC. The They are not necessary for the generation of ATP
enzyme activity is also inhibited by phosphory- but promote formation of metabolically relevant
lation through pyruvate dehydrogenase kinase metabolites for DNA synthesis and antioxida-
(PDK). Elevated acetyl-CoA levels and increased tive defense as well as mechanisms of cell sig-
conversion of ADP to ATP leads to activation of naling.
PDK and hence to phosphorylation and inactiva-
tion of PDH. In contrast, elevated concentrations
of pyruvate, ADP, and pyrophosphate lead to ac- PENTOSE PHOSPHATE PATHWAY
tivation of pyruvate dehydrogenase phosphatase,
resulting in dephosphorylation of PDH thereby The pentose phosphate pathway (PPP) is an
activating it [15]. alternative way of glucose use. It consists of
Under ischemic conditions, pyruvate is not an aerobic and an anaerobic part. For that rea-
decarboxylated but reduced to lactate by lac- son, the PPP can act as a pathway or a cycle
tate dehydrogenase (LDH) and NADH that was both at the same time. Its aerobic part leads to
built in the previous GAPDH reaction. Therefore ribulose-5-phosphate, carbon dioxide (CO2),
increasing amounts of lactate act as feedback in- and reduced nicotinamide adenine dinucleotide
hibitor of glycolysis at the level of GAPDH. In phosphate (NADPH). One molecule of ribulose-
the absence of oxygen, the glycolytic production 5-phosphate and two molecules of NADPH
of lactate is often referred to as “anaerobic gly- are produced out of one molecule of glucose.
colysis” although the reaction steps do not differ Subsequently, following anaerobic transforma-
from those in the presence of oxygen. The fate tion of ribulose-5-phosphate delivers no energy
of the end product differs – i­nstead of oxidation but new glucose-6-phosphate. This can reenter
to acetyl-CoA, pyruvate is reduced to lactate. other glycolytic pathways such as glycolysis
Importantly, lactate is a normal (and even pre- or the HBP (see later in the chapter). In total,
ferred) substrate for cardiac energy metabolism a series of PPP reactions cycle 6 molecules of
under aerobic conditions. Especially in times of glucose-6-phosphate to 5 molecules of glucose-
high skeletal muscle activity (e.g., during exer- 6-­phosphate, 12 NADPH and 6 CO2 [18]. Fig-
cise), lactate utilization by the heart increases ure 4.4 shows a scheme of all reactions within
proportionally to its appearance in plasma the PPP. NADPH is mainly used for fatty acid
through increased skeletal muscle contraction synthesis, pyruvate oxidation to malate, and the
[16]. Therefore, LDH oxidizes lactate to pyru- reduction of glutathione. Ribulose-5-phosphate,
vate in a NAD+-­dependent reaction. the product of the aerobic part of PPP is easily
Increasing amounts of NADH and/or ATP converted to ribose-5-phosphate, which is used
act as negative allosteric effectors controlling the for synthesis of nucleotides and nucleic acids.
flux through glycolysis [1,2]. Furthermore, me- Hence, the PPP links carbohydrate and fatty
tabolites of the CAC are able to regulate glycoly- acid metabolism, anaplerosis, nucleotide syn-
sis. Higher rates of fatty acid oxidation lead to thesis, and antioxidative defense depending on
accumulation of citric acid, the first intermediate the individual need of a cell’s metabolism.
of CAC. This acts as a negative allosteric inhibi- The PPP as well as glycolysis and the hex-
tor of the glycolytic enzyme PFK and is hence osamine biosynthesis pathway use glucose-6-­
able to slow glycolytic breakdown [17]. phosphate. This substrate is oxidized twice by

 
46 4.  Metabolic Pathways and Cycles

FIGURE 4.4  Reaction steps of the pentose phosphate pathway.

the ­NADP+-dependent glucose-6-­phosphate the conversion of ribulose-5-phosphate to


dehydrogenase to 6-phospho-glucon-d-­lacton intermediates of glycolysis. So the PPP may
­
as intermediate and by gluconolactonase rather be seen as a cycle instead of a linear
to ­6-phosphogluconate. In the next step, pathway. Isomerization and epimerization of
6-phosphogluconate is converted to ribu-
­ ribulose-5-phosphate allow the formation of
lose-5-phosphate by NADP+-dependent ribose-5-phosphate and xylolose-5-­phosphate.
6-phosphogluconate dehydrogenase. Hereby,
­ The enzyme transketolase catalyzes their re-
3-keto 6-phosphogluconate occurs as an un- action to seduheptulose-7-­ phosphate and
stable intermediate. These last three reactions glycerinaldehyde-­3-phosphate. The latter two
of the PPP result in two molecules of NADPH. build erythrose-4-phosphate and fructose-
The following anaerobic part of PPP allows 6-phosphate, catalyzed by transaldolase. A ­final

 
Hexosamine biosynthesis pathway 47
reaction of erythrose-4-phosphate and another small flux of glucose into PPP and HBP [19].
molecule of xylolose-5-phosphate form fruc- Metabolite supply of these alternate pathways is
tose-6-phosphate and glycerinaldehyde-­ 3- enhanced when glycolysis is limited as it occurs
phosphate, which may directly enter glycolysis. with increasing amounts of free fatty acids. The
HBP has importance in glucose metabolism and is
activated not only by increasing but also rapidly
HEXOSAMINE BIOSYNTHESIS decreasing glucose concentrations or changing
PATHWAY intracellular calcium levels [20]. A detailed over-
view of all reaction steps within the HBP is given
The hexosamine biosynthesis pathway (HBP) in Fig. 4.5.
is another branch for glucose utilization. In the Initially, glucose-6-phosphate is converted to
normal working healthy heart, there is only a fructose-6-phosphate, which is not utilized by

FIGURE 4.5  Reaction steps of the hexosamine biosynthesis pathway.

 
48 4.  Metabolic Pathways and Cycles

PFK as in glycolysis but is ­rather reacting with AMINO ACIDS


the amino-donor glutamine to glucosamine-
6-phosphate. This reaction is d ­riven by the Besides fatty acids and glucose, the heart is
rate-limiting enzyme glucosamine-6-­phosphate also able to use amino acids as substrates for
synthase/glutamine-fructose-6-­phosphate ami- energy production [27], although amino acids
notransferase (GFAT). Due to its short half-life are the primary building blocks for protein syn-
of less than an hour GFAT mainly seems to be thesis. For use as substrate, they are either taken
regulated by its gene expression. It is also con- up by the cell or generated by protein degrada-
trolled by allosteric inhibition and/or ­covalent tion. The major site of amino acid degradation
modification [21]. In the next step, ­acetyl-­CoA in mammals is the liver. However, the branched
is transferred to glucosamine-6-­ phosphate re- chain amino acids (BCAA) valine, leucine, and
sulting in N-­acetylglucosamine-6-phosphate. isoleucine are also metabolized in the heart mus-
After isomerization N-acetyl­glucosa­mine-1- cle. First, the amino group is removed in a deam-
phosphate and UTP react to the final product ination reaction that is driven by ­transaminases.
UDP-N-­acetylglucosamine (UDP-GlcNAc). The The carbon skeletons of the 20 amino acids are
end product UDP-GlcNAc plays an important transformed into only 7 molecules: acetyl-CoA,
role in reversible posttranslational protein mod- acetoacetyl-CoA (ketogenic amino acids), and
ification and may have special impact in signal pyruvate, a-ketoglutarate, succinyl-CoA, fuma-
transduction [22]. The linkage of UDP-GlcNAc rate, and oxaloacetate (glucogenic amino acids)
to proteins is called glycosylation and takes [28]. These carbon skeletons of the resulting
place in the Golgi apparatus. It is mediated by a-ketoacids enter metabolic pathways as precur-
O-GlcNAc transferase (OGT) that catalyzes the sors to glucose or CAC intermediates (see later
O-linked binding of UDP-GlcNAc to mainly hy- in the chapter).
droxyl oxygen of serine and threonine residues The BCAA valine, leucine, and isoleucine
but also of tyrosine, hydroxylysine, or hydroxy- are of major interest with regard to regulation
proline side chains of proteins [23]. Glycosyl- of cardiac metabolism and the development of
ation of proteins leads to formation of various common diseases such as insulin resistance,
protein alterations such as proteoglycans and diabetes, and cardiovascular diseases [29]. The
glycoproteins. The removal of O-GlcNAc from BCAA leucine, and the metabolites of leucine,
proteins is driven by O-­GlcNAcase (OGA). In valine, and isoleucine have been shown to in-
contrast to protein modifications, such as phos- hibit protein degradation and accelerated pro-
phorylations, glycosylation is mediated by only tein synthesis in the heart and may play a role in
two enzymes OGT and OGA. For that reason, it the regulation of protein turnover [30].
is less specific than phosphorylation, which in
turn is mediated by dozens of particular kinases
and ­phosphatases [24]. Cellular O-GlcNAc lev- CITRIC ACID CYCLE
els are increased by stressors such as heat, free
radicals, and ultraviolet (UV) radiation [25]. The CAC marks the center of interconnected
Levels are chronically elevated in response to energy providing pathways and cycles. In the
diabetes, impaired Ca2+ handling, and in heart mitochondrial matrix, enzymes of the CAC
failure [26]. A special focus of recent work was (also known as Krebs cycle or tricarboxylic acid
on cellular signaling pathways and transcrip- cycle -TCA) produce the reducing equivalents
tional regulation. However, there is also grow- NADH and FADH2. They deliver electrons to
ing evidence that O-GlcNAcylation may play a complexes of mitochondrial electron-transport
role in epigenetic regulation [20,24]. chain, which builds up a proton gradient that

 
Citric acid cycle 49
TABLE 4.3 Catalytic Efficiency of the Citric Acid Cycle
Metabolic step Reducing equivalent ATP output

Isocitrate → a-Ketoglutarate NAD → NADH + H


+ +
2.7
a-Ketoglutarate → Succinyl-CoA NAD → NADH + H
+ +
2.7
Succinyl-CoA → Succinate Substrate chain phosphorylation
GDP + P → GTP 1.0
Succinate → Fumarate FAD → FADH2 1.6
Malate → Oxaloacetate NAD → NADH + H
+ +
2.7
Sum 10.7

drives ATP production. This key process links with the result of the first reducing equivalent
glycolysis, fatty acid oxidation, and also amino NADH. Decarboxylation of oxalosuccinate
acid oxidation. In contrast to glycolysis and fatty leads to formation of a-ketoglutarate and CO2.
acid oxidation, which can be described as linear In the next step, a-ketoglutarate dehydroge-
pathways, cycles such as the CAC are energeti- nase catalyzes the oxidation and carboxylation
cally more efficient. ATP output of a full cycle is of a-­ketoglutarate to succinate by formation of
shown in Table 4.3 [31,32]. The centrality of the succinyl-CoA as an unstable intermediate. This
Krebs cycle for metabolism is remarkable. It not reaction is comparable to the pyruvate dehy-
only connects all catabolic substrate oxidation drogenase reaction and leads to formation of
pathways to the respiratory chain, but also rep- one molecule of high-energetic GTP. Efficiency
resents the source of biosynthetic products for of a-ketoglutarate dehydrogenase is crucial and
many anabolic processes [33]. Thus, although controls the substrate flux through the com-
the cycle does not lose intermediates (i.e., moi- plete CAC [34]. Further oxidation of succinate
eties) by its own reactions, it still needs to be re- by FAD+-dependent succinate dehydrogenase
plenished continuously (anaplerosis, see later in results in the reducing equivalent FADH2 and
the chapter). fumarate, which is then hydrogenated to ma-
The CAC stepwise catabolizes one molecule late by fumarase. In the “last” step of the CAC,
of acetyl-CoA, the high-energetic end product of the NAD+-dependent malate dehydrogenase
glycolysis and b-oxidation into two molecules converts malate to oxaloacetate and another
of carbon dioxide and the reducing equivalents NADH. Consequently, oxaloacetate may react
NADH and FADH2. A schematic overview of the with another molecule of acetyl-CoA and start a
full CAC is shown in Fig. 4.6. Initially, acetyl- new round of the cycle.
CoA reacts with the cycle intermediate oxaloac- The CAC as the pivotal element of energy
etate. This reaction is driven by citrate synthase, metabolism is strictly regulated. The cycle is
which is the key enzyme of the cycle and may triggered by ADP, inorganic phosphate, and
also be seen as a marker of mitochondrial activ- calcium while it is inhibited by NADH and ATP.
ity. The product citrate is dehydrated to the un- Additionally, most CAC enzymes are regulated
stable intermediate cis-aconitate, which is sub- by their educts and products in a kinetic man-
sequently hydrated to isocitrate. Both reactions ner. For example, succinate dehydrogenase is
are catalyzed by aconitase. In the third step of inhibited by oxaloacetate but activated by suc-
the CAC, NAD+-dependent isocitrate dehydro- cinate. Although the amount of the intermedi-
genase transforms isocitrate to oxalosuccinate ates of the CAC is strictly regulated, they also

 
50 4.  Metabolic Pathways and Cycles

FIGURE 4.6  Reactions steps of the citric acid cycle.

represent the basic building blocks for many ANAPLEROSIS


synthesis processes. The pool of CAC interme-
diates is therefore continuously “in change” Anaplerosis was first mentioned by
and needs to be replenished if synthesis pro- ­ ornberg in 1966 [35,36]. He described the
K
cesses have prevalence – a process that is called mechanism as pathways to recover a meta-
anaplerosis. bolic cycle. In conditions of catabolic oxidation

 
The respiratory chain 51
concentrations of CAC intermediates are very come to a lack of NADPH which is necessary
low (10−5−10−4 mol/L) [1]. No cycle interme- for fatty acid synthesis and defense of reactive
diates are either removed or built. However, oxygen species. A second focus of anaplerosis
besides substrate oxidation, intermediates of is on transamination reactions driven by trans-
the CAC act as starting points of anabolic path- aminases. An example of such transaminations
ways. The intermediates serve for fatty acid bio- is the generation of a-ketoglutarate from the
synthesis (citrate), heme biosynthesis (succinyl- amino acid glutamate. Also oxaloacetate can
CoA), gluconeogenesis (oxaloacetate), and be refilled. It is generated by aspartate [39]. Al-
biosynthesis of nonessential amino acids (a-­ though odd chain fatty acids are of lower im-
ketoglutarate and oxaloacetate) [37]. Depletion portance for the generation of ATP, they may
of CAC intermediates leads to a rapid decline of serve for anaplerotic reactions. Here, the CAC
contractile function of the heart [15]. It has been intermediate succinyl-CoA can be regenerated
shown that impairment of anaplerotic path- from propionyl-CoA, the three-carbon residue
ways rapidly causes contractile dysfunction of odd chain fatty acid oxidation. Most nota-
[19,38]. Changes in anaplerotic pathways have bly, during muscle contraction another anaple-
recently been implicated in heart disease. As a rotic reaction is important: the conversion of
mechanism to maintain steady state, anaplero- aspartate to ammonia and fumarate in a GTP-
sis becomes necessary in case of loss of carbon consuming reaction [46]. Increasing amounts of
intermediates of the CAC [39,40]. In contrast to fumarate are able to replenish CAC intermedi-
skeletal muscle, where concentrations of inter- ates, especially in conditions of high efflux such
mediates of the CAC are not totally stable and as exercise and fasting.
for example increase immediately during exer- Many of the anaplerotic reactions discussed
cise [41–43], strong interactions between me- earlier, only lead to small increases in CAC inter-
tabolites and enzymes ­maintain a steady state mediates, especially the net synthesis of malate
in the heart. Here, metabolites such as pyruvate from pyruvate is kinetically very unfavorable
that are able to undergo various reactions and [47]. For that reason, transamination reactions
that act as substrate for different enzymes are [41,45] in parallel with the generation of fuma-
essential. They allow the flexibility of mov- rate by the purine nucleotide cycle might be
ing from ATP generation (conversion of py- the most important anaplerotic reactions with
ruvate to acetyl-CoA) to the replenishment of regard to the amount of intermediate that is re-
cycle intermediates. Pyruvate as a link between plenished in the CAC.
glycolysis and the CAC plays an important
­
role in anaplerosis. The most typical anaple-
rotic reaction is the carboxylation of pyruvate THE RESPIRATORY CHAIN
to oxaloacetate, which is catalyzed by the ATP-
dependent pyruvate carboxylase. This reaction In a final step, all the energy so far converted
is initiated by elevated amounts of acetyl-­CoA, and not used for anabolic processes is trans-
the allosteric activator, as well as by exercise ferred to the respiratory chain. Here, the reduc-
[44,45] and ensures a sufficient supply of oxa- ing equivalents (NADH to Complex I and FADH
loacetate to run the CAC. Another possible car- to Complex II) are handing off their electrons
boxylation of pyruvate produces malate by the and protons to be passed on to oxygen, result-
NADP+-dependent malic enzyme. This reaction ing in water production. The energy released is
might be more important in the opposite direc- used to generate a proton gradient across the in-
tion (decarboxylation of malate to pyruvate) ner mitochondrial membrane. The release of this
because during anaplerotic reactions it may gradient through Complex V (the F0F1-ATPase)

 
52

 
4.  Metabolic Pathways and Cycles

FIGURE 4.7  Complexity of cardiac metabolic pathways and cycles.


REFERENCES 53
is used to generate ATP from ADP and inorganic the understanding of cardiac metabolism and
phosphate. The function of the respiratory chain the investigation of disease. Here, we have il-
is described in detail together with its assess- lustrated a summary of individual reactions
ment in Chapter 3 of this book. and some regulatory aspects for the key path-
ways and cycles focusing on cardiac energy me-
tabolism. We have not described the respiratory
PUTTING IT ALL TOGETHER chain in this chapter, as it is described in detail
in Chapter 3.
The functional interaction of all metabolic
pathways and cycles described in this chapter
is summarized in Fig. 4.7. The pathways and References
cycles function for the permanent supply of [1] Stanley WC, Recchia FA, Lopaschuk GD. Myocardial
energy (i.e., glycogen breakdown, glycolysis, substrate metabolism in the normal and failing heart.
b-oxidation), the generation of metabolites for Physiol Rev 2005;85(3):1093–129.
synthesis processes, and replenishment of the [2] Sharma N, Okere IC, Duda MK, Chess DJ, O’Shea KM,
Stanley WC. Potential impact of carbohydrate and fat
CAC (i.e., PPP and anaplerosis), and metabolic
intake on pathological left ventricular hypertrophy.
regulation (i.e., HBP). The high-energy demand ­Cardiovasc Res 2007;73(2):257–68.
of cardiac contractile function is primarily cov- [3] Lopaschuk GD, Belke DD, Gamble J, Itoi T, Schonekess
ered by oxidative phosphorylation of acetyl- BO. Regulation of fatty acid oxidation in the mamma-
CoA derived from b-oxidation and to a lesser lian heart in health and disease. Biochim Biophys Acta
1994;1213(3):263–76.
amount from glycolysis. Within the CAC, ace-
[4] Lewandowski ED, Kudej RK, White LT, O’Donnell JM,
tyl-CoA results in the formation NADH and Vatner SF. Mitochondrial preference for short chain fatty
FADH2 delivering their protons to Complexes I acid oxidation during coronary artery constriction.
and II of mitochondrial electron-transport chain ­Circulation 2002;105(3):367–72.
to finally run ATP synthase. The PPP generates [5] Berg JM, Tymoczko JL, Stryer L. Biochemistry. 5th ed.
New York: W. H. Freeman; 2002. Section 22.2, The Uti-
ribose to build DNA and RNA elements and
lization of Fatty Acids as Fuel Requires Three Stages of
NADPH that is an important player in cellular Processing.
defense of reactive oxygen species. The hexos- [6] Kodde IF, van der Stok J, Smolenski RT, de Jong JW.
amine pathway generates O-GlcNAc that leads ­Metabolic and genetic regulation of cardiac energy sub-
to glycosylated proteins, which contribute to strate preference. Comp Biochem Physiol A Mol Integr
Physiol 2007;146(1):26–39.
the regulation of the complex interplay of all
[7] Abel ED. Glucose transport in the heart. Front Biosci
metabolic pathways and cycles. 2004;9:201–15.
[8] Depre C, Vanoverschelde JL, Taegtmeyer H. Glucose for
the heart. Circulation 1999;99(4):578–88.
SUMMARY [9] Depre C, Rider MH, Hue L. Mechanisms of control of
heart glycolysis. Eur J Biochem 1998;258(2):277–90.
[10] Pierce GN, Philipson KD. Binding of glycolytic enzymes
Cardiac metabolism is a highly concerted to cardiac sarcolemmal and sarcoplasmic reticular mem-
plethora of chemical reactions leading to the branes. J Biol Chem 1985;260(11):6862–70.
conversion of substrates for energy production [11] Weiss J, Hiltbrand B. Functional compartmentation of
in the form of ATP to sustain cell function and glycolytic versus oxidative metabolism in isolated rab-
bit heart. J Clin Invest 1985;75(2):436–47.
allow contraction and also for the synthesis of
[12] Berg JM, Tymoczko JL, Stryer L. Biochemistry. 5th ed.
building blocks to allow for growth, repair, and New York: W. H. Freeman; 2002. Section 16.1 Glycolysis
regeneration. Knowing the key pathways and is an Energy-Conversion Pathway in Many Organisms.
cycles of metabolism is a basic requirement for [13] Bricker DK, Taylor EB, Schell JC, Orsak T, Boutron

 
54 4.  Metabolic Pathways and Cycles

A, Chen Y-C, Cox JE, Cardon CM, Van Vranken JG, hypertrophy and heart failure. Physiol Genomics 2012;
Dephoure N, Redin C, Boudina S, Gygi SP, Brivet M, 44:162–72.
Thummel CS, Rutter J. A mitochondrial pyruvate carrier [27] Chatham JC. Lactate – the forgotten fuel. J Physiol
required for pyruvate uptake in yeast, drosophila, and 2002;542(Pt 2):333.
humans. Science 2012;337(6090):96–100. [28] Berg JM, Tymoczko JL, Stryer L. Biochemistry. 5th ed.
[14] Herzig S, Raemy E, Montessuit S, Veuthey J-L, Zamboni New York: W H Freeman; 2002. Section 23.5, Carbon
N, Westermann B, Kunji ERS, Martinou J-C. Identifica- Atoms of Degraded Amino Acids Emerge as Major
­
tion and functional expression of the mitochondrial py- Metabolic Intermediates.
ruvate carrier. Science 2012;337(6090):93–6. [29] Newgard CB. Interplay between lipids and branched-
[15] Hansford RG, Cohen L. Relative importance of pyru- chain amino acids in development of insulin resistance.
vate dehydrogenase interconversion and feed-back in- Cell Metab 2012;15(5):606–14.
hibition in the effect of fatty acids on pyruvate oxidation [30] Chua B, Siehl DL, Morgan HE. Effect of leucine and
by rat heart mitochondria. Arch Biochem Biophys 1978; metabolites of branched chain amino acids on protein
191(1):65–81. turnover in heart. J Biol Chem 1979;254(17):8358–62.
[16] Drake AJ, Haines JR, Noble MI. Preferential uptake of [31] Brandt U, Der Citratzyklus – Abbau von Acetyl-CoA zu
lactate by the normal myocardium in dogs. Cardiovasc CO2 und H2O. In: Heinrich PC, Müller M, Graeve L, edi-
Res 1980;14(2):65–72. tors. Löffler/Petrides Biochemie und Pathobiochemie.
[17] Garland PB, Randle PJ, Newsholme EA. Citrate as an in- Berlin, Heidelberg: Springer; 2014. p. 226–34.
termediary in the inhibition of phosphofructokinase in [32] Berg JM, Tymoczko JL, Stryer L. Biochemistry. 5th ed.
rat heart muscle by fatty acids, ketone bodies, pyruvate, New York: W. H. Freeman; 2002. Section 17.1, The Citric
diabetes and starvation. Nature 1963;200(4902):169–70. Acid Cycle Oxidizes Two-Carbon Units. The Citric Acid
[18] Baldwin JE, Krebs H. The evolution of metabolic cycles. Cycle Oxidizes Two-Carbon Units.
Nature 1981;291(5814):381–2. [33] Doenst T, Nguyen TD, Abel ED. Cardiac metabolism in
[19] Doenst T, Nguyen TD, Abel ED. Cardiac metabolism in heart failure: implications beyond ATP production. Circ
heart failure: implications beyond ATP production. Circ Res 2013;113(6):709–24.
Res 2013;113(6):709–24. [34] Cooney GJ, Taegtmeyer H, Newsholme EA. Tricarbox-
[20] Chatham JC, Marchase RB. Protein O-GlcNAcylation: a ylic acid cycle flux and enzyme activities in the isolated
critical regulator of the cellular response to stress. Curr working rat heart. Biochem J 1981;200(3):701–3.
Signal Transd Ther 2010;5(1):49–59. [35] Kornberg H. Anaplerotic sequences and their role in
[21] Karason K, Sjöström L, Wallentin I, Peltonen M. Impact metabolism. Essays Biochem 1966;2:1–31.
of blood pressure and insulin on the relationship be- [36] Owen OE, Kalhan SC, Hanson RW. The key role of
tween body fat and left ventricular structure. Eur Heart anaplerosis and cataplerosis for citric acid cycle func-
J 2003;24(16):1500–5. tion. J Biol Chem 2002;277(34):30409–12.
[22] Kreppel LK, Blomberg MA, Hart GW. Dynamic gly- [37] Gray LR, Tompkins SC, Taylor EB. Regulation of pyru-
cosylation of nuclear and cytosolic proteins. Cloning vate metabolism and human disease. Cell Mol Life Sci
and characterization of a unique O-GlcNAc transferase 2014;71(14):2577–604.
with multiple tetratricopeptide repeats. J Biol Chem [38] Russell RR, Taegtmeyer H. Changes in citric acid cycle
1997;272(14):9308–15. flux and anaplerosis antedate the functional decline in
[23] Torres CR, Hart GW. Topography and polypeptide isolated rat hearts utilizing acetoacetate. J Clin Invest
distribution of terminal N-acetylglucosamine residues 1991;87(2):384–90.
on the surfaces of intact lymphocytes. Evidence for [39] Des Rosiers C, Labarthe F, Lloyd SG, Chatham JC.
­O-linked GlcNAc. J Biol Chem 1984;259(5):3308–17. Cardiac anaplerosis in health and disease: food for
­
[24] McLarty JL, Marsh SA, Chatham JC. Post-translational thought. Cardiovasc Res 2011;90:210–9.
protein modification by O-linked N-acetyl-glucosamine: [40] Wessells RJ, Fitzgerald E, Cypser JR, Tatar M, Bodmer R.
its role in mediating the adverse effects of diabetes on Insulin regulation of heart function in aging fruit flies.
the heart. Life Sci 2012;92:621–7. Nat Genet 2004;36(12):1275–81.
[25] Zachara NE, Hart GW. O-GlcNAc a sensor of cellu- [41] Sahlin K, Katz A, Broberg S. Tricarboxylic acid cycle
lar state: the role of nucleocytoplasmic glycosylation intermediates in human muscle during prolonged
in modulating cellular function in response to nutri- ­exercise. Am J Physiol 1990;259(5 Pt 1):C834–41.
tion and stress. Biochim Biophys Acta 2004;1673(1–2): [42] Russell RR, Taegtmeyer H. Pyruvate carboxylation
13–28. ­prevents the decline in contractile function of rat hearts
[26] Lunde IG, Aronsen JM, Kvaløy H, Qvigstad E, Sjaastad oxidizing acetoacetate. Am J Physiol 1991;261:H1756–62.
I, Tønnessen T, Christensen G, Grønning-Wang LM, [43] Gibala MJ, MacLean DA, Graham TE, Saltin B. Tricar-
Carlson CR. Cardiac O-GlcNAc signaling is increased in boxylic acid cycle intermediate pool size and estimated

 
REFERENCES 55
cycle flux in human muscle during exercise. Am during brief dynamic exercise. J Physiol 1997;502(3):
J  Physiol 1998;275(2):E235–42. 703–13.
[44] Lancha AH Jr, Recco MB, Curi R. Pyruvate carboxylase [46] Lowenstein JM. The purine nucleotide cycle revised. Int
activity in the heart and skeletal muscles of the rat. Evi- J Sports Med 1990;11(S 2):S37–46.
dence for a stimulating effect of exercise. Biochem Mol [47] Lowenstein J, Tornheim K. Ammonia production in
Biol Int 1994;32(3):483–9. muscle: the purine nucleotide cycle. Science 1971;
[45] Gibala MJ, MacLean DA, Graham TE, Saltin B. 171(3969):397–400.
Anaplerotic processes in human skeletal muscle

 

Vous aimerez peut-être aussi