Sei sulla pagina 1di 17

Heart Failure Compendium

Circulation Research Compendium on Heart Failure


Research Advances in Heart Failure: A Compendium
Epidemiology of Heart Failure
Genetic Cardiomyopathies Causing Heart Failure
Non-coding RNAs in Cardiac Remodeling and Heart Failure
Mechanisms of Altered Ca2+ Handling in Heart Failure
Cardiac Metabolism in Heart Failure: Implications Beyond ATP Production
Integrating the Myocardial Matrix Into Heart Failure Recognition and Management
Adrenergic Nervous System in Heart Failure: Pathophysiology and Therapy
Emerging Paradigms in Cardiomyopathies Associated With Cancer Therapies
Electromechanical Dyssynchrony and Resynchronization of the Failing Heart
Molecular Changes After Left Ventricular Assist Device Support for Heart Failure
Heart Failure Gene Therapy: The Path to Clinical Practice
Cell Therapy for Heart Failure: A Comprehensive Overview of Experimental and Clinical Studies, Current Challenges,
and Future Directions
Eugene Braunwald, Guest Editor

Cardiac Metabolism in Heart Failure


Implications Beyond ATP Production
Torsten Doenst, Tien Dung Nguyen, E. Dale Abel

Abstract: The heart has a high rate of ATP production and turnover that is required to maintain its continuous
mechanical work. Perturbations in ATP-generating processes may therefore affect contractile function directly.
Characterizing cardiac metabolism in heart failure (HF) revealed several metabolic alterations called metabolic
remodeling, ranging from changes in substrate use to mitochondrial dysfunction, ultimately resulting in ATP
deficiency and impaired contractility. However, ATP depletion is not the only relevant consequence of metabolic
remodeling during HF. By providing cellular building blocks and signaling molecules, metabolic pathways
control essential processes such as cell growth and regeneration. Thus, alterations in cardiac metabolism may
also affect the progression to HF by mechanisms beyond ATP supply. Our aim is therefore to highlight that
metabolic remodeling in HF not only results in impaired cardiac energetics but also induces other processes
implicated in the development of HF such as structural remodeling and oxidative stress. Accordingly, modulating
cardiac metabolism in HF may have significant therapeutic relevance that goes beyond the energetic aspect.
(Circ Res. 2013;113:709-724.)
Key Words: heart failure hypertrophy/remodeling metabolism mitochondria

H eart failure (HF) is a clinical syndrome in which myo-


cardial pump function is inadequate for maintaining and
supporting an individuals physiological requirements. The
when systolic function was impaired. Stages of cardiac hyper-
trophy without systolic dysfunction have mostly been called
compensated hypertrophy. More recently, it has become clear
clinical presentation is characterized by pulmonary conges- that HF symptoms can also exist despite relatively preserved
tion, dyspnea, and fatigue. Historically, HF was suspected systolic function. This condition is caused mostly by diastolic

Original received April 3, 2013; revision received June 3, 2013; accepted June 21, 2013.
From the Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich-Schiller-University Jena, Jena, Germany (T.D., T.D.N.); and Division of
Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT (E.D.A.).
Correspondence to Torsten Doenst, MD, Department of Cardiothoracic Surgery, Friedrich-Schiller-University Jena, Erlanger Allee 101 07747 Jena,
Germany. E-mail doenst@med.uni-jena.de
2013 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.113.300376

709
Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016
710Circulation ResearchAugust 30, 2013

ATP from ADP through reactions catalyzed by the cytosolic


Nonstandard Abbreviations and Acronyms
creatine kinase.6 Because of its continuous mechanical work,
AMPK AMP-activated protein kinase the heart has a high rate of ATP hydrolysis (0.5 mol/g wet
CoA coenzyme A weight per second). Accordingly, the high-energy phosphate
CPT carnitine palmitoyltransferase pool in the heart is relatively small and can be exhausted
DCA dichloroacetate within a few seconds. Therefore, cardiac work depends
ERR estrogen-related receptors strongly on ATP generation, and impairments in this process
ETC electron transport chain can rapidly induce contractile dysfunction. Approximately
FA fatty acid 70% to 90% of cardiac ATP is produced by the oxidation of
G6P glucose 6-phosphate
fatty acids (FAs). The remaining 10% to 30% comes from the
oxidation of glucose and lactate, as well as small amounts of
GLP glucagon-like peptide
ketone bodies and certain amino acids.2,7,8 However, it is also
GLUT glucose transporter
important to note that cardiac substrate selection in human
HBP hexosamine biosynthetic pathway
studies has been assessed mostly in the fasted state. In the fed
HF heart failure
state, when plasma levels of glucose and insulin rise, the con-
NRF nuclear respiratory factors
tribution of glucose use to cardiac ATP production increas-
PCr phosphocreatine
es. Figure1 is a schematic representation of well-described
PDH pyruvate dehydrogenase metabolic processes, focused on the generation of ATP from
PGC-1 peroxisome proliferatoractivated receptor coactivator-1 glucose and FAs.
PPAR peroxisome proliferatoractivated receptor
PPP pentose phosphate pathway FA Use
ROS reactive oxygen species
FA use can be divided into 3 steps: uptake into the cytosol,
transportation across the mitochondrial membrane, and oxi-
dation within the mitochondria. Although FAs can traverse
the plasma membrane (flip flop), their uptake is facilitated by
dysfunction, which may even be present in conditions histori- transport proteins including FA translocase (CD36) and the
cally called compensated hypertrophy. Thus, moving forward, plasma membrane FA-binding protein.9,10 In the cytosol, free
metabolic research will need to take this aspect into account. FAs are esterified to fatty acyl-coenzyme A (CoA), which is
It is possible that distinct changes in cardiac metabolism may either esterified to triglyceride or converted to long-chain ac-
mediate these 2 forms of cardiac dysfunction. For the sake ylcarnitine by carnitine palmitoyltransferase (CPT) I located
of simplicity and because most studies on cardiac metabo- in the outer mitochondrial membrane before entering the
lism in HF did not investigate diastolic function, we use the mitochondria.9,11 The turnover of the myocardial triglyceride
term compensated hypertrophy for all conditions associated pool is high and represents an important source of fatty acyl-
with normal systolic function and the term HF to indicate the CoA that subsequently is metabolized by the mitochondria.12
presence of systolic dysfunction. Metabolic changes that spe- Long-chain acylcarnitine is then transported into the mito-
cifically affect or contribute to diastolic dysfunction will be chondrial matrix and converted back to long-chain acyl-CoA
addressed if information is available. by CPT II. Acyl-CoA then enters -oxidation, generating
In this review, we focus primarily on metabolic adaptations acetyl-CoA and the reducing equivalents reduced nicotin-
in chronic HF. Acute changes such as those that develop after amide adenine dinucleotide (NADH) and reduced flavin ad-
myocardial stunning or ischemia/reperfusion have been ad- enine dinucleotide.
dressed elsewhere. To understand metabolic alterations in HF, Fatty acyl-CoA, particularly palmitoyl-CoA, can be used for
an overview of basic metabolic processes in the healthy heart de novo synthesis of ceramides, which has pleiotropic effects
is necessary. on cellular function.13,14 Incomplete FA oxidation, particularly
in skeletal muscle, may result in release of acylcarnitines from
Metabolism in the Normal Heart
mitochondria into the cytosol and subsequently into the circu-
Under normoxic conditions, >95% of ATP generated in the
lation. These acylcarnitines have been implicated in the patho-
heart is derived from oxidative phosphorylation in the mi-
physiology of insulin resistance.15
tochondria. The remaining 5% comes mainly from glycoly-
The reaction catalyzed by CPT I represents a critical regu-
sis and to a lesser extent from the citric acid cycle (Krebs
latory node that determines FA oxidation rates. In the normal
cycle).1,2 The heart uses 60% to 70% of generated ATP to
adult heart, the muscle isoform (CPT Ib) is predominant.16 In
fuel contraction and the remaining 30% to 40% for various
contrast, the expression of the liver isoform (CPT Ia), which
ion pumps, especially the Ca2+-ATPase in the sarcoplasmic
is expressed abundantly in the fetal heart, has been shown to
reticulum.3,4 The energy pool of the heart includes ATP (5
increase in the hypertrophied heart.17 The activity of CPT I is
mol/g wet weight) and phosphocreatine (PCr; 8 mol/g wet
inhibited by malonyl-CoA,18,19 which is generated by carbox-
weight), with the latter serving as an ATP transport and buf-
ylation of cytosolic acetyl-CoA by the enzyme acetyl-CoA
fer system.5 In the mitochondria, the high-energy phosphate
carboxylase. Malonyl-CoA is converted back to acetyl-CoA
bond in ATP can be transferred to creatine by mitochondrial
by malonyl-CoA decarboxylase (Figure 1).20,21 Although this
creatine kinase to form PCr. With a smaller molecular weight
pathway has been evaluated as a therapeutic target in the set-
than ATP, PCr can easily diffuse through the mitochondrial
ting of ischemic heart disease,22,23 recent evidence has also
membrane into the cytosol. Here, it can be used to generate

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


Doenst et al Cardiac Metabolism in Heart Failure 711

Figure 1. Schematic representation of classic pathways of cardiac metabolism. Substrates are transported across the extracellular
membrane into the cytosol and are metabolized in various ways. For oxidation, the respective metabolic intermediates (eg, pyruvate or acyl-
coenzyme A [CoA]) are transported across the inner mitochondrial membrane by specific transport systems. Once inside the mitochondrion,
substrates are oxidized or carboxylated (anaplerosis) and fed into the Krebs cycle for the generation of reducing equivalents (reduced
nicotinamide adenine dinucleotide [NADH]2; reduced flavin adenine dinucleotide [FADH]) and GTP. The reducing equivalents are used by
the electron transport chain to generate a proton gradient, which in turn is used for the production of ATP. This principal functionality can be
affected in various ways during heart failure (HF), thereby limiting ATP production or affecting cellular function in other ways (see text and
other figures for details). ACC indicates acetyl-CoA carboxylase; CPT, carnitine palmitoyltransferase; FAT, fatty acid transporter; G6P, glucose
6-phosphate; GLUT, glucose transporter; IMS, mitochondrial intermembrane space; MCD, malonyl-CoA decarboxylase; MPC, mitochondrial
pyruvate carrier; PDH, pyruvate dehydrogenase; and PDK, pyruvate dehydrogenase kinase. Illustration Credit: Ben Smith.

suggested that CPT I activity may be regulated via mecha- of its E1 subunit.30 The carboxylation of pyruvate is a major
nisms that are independent of changes in malonyl-CoA.24 anaplerotic pathway (see below).
Glucose Use Accessory Pathways of Glucose Metabolism
Glucose that is used by the heart either comes from uptake of Beside glycolysis, G6P may also be channeled into glycogen
exogenous glucose or is derived from glycogen stores. The synthesis or the pentose phosphate pathway (PPP). The PPP
heart has a relatively small glycogen pool (30 mol/g wet is an important source of reduced nicotinamide adenine di-
weight, which is 20% that of skeletal muscle), but rates of gly- nucleotide phosphate (NADPH), which plays a critical role
cogen turnover are high. In the heart, glycogen-derived glucose in regulating cellular oxidative stress and is required for lipid
may contribute 40% of glucose-mediated ATP production in synthesis31 and anaplerosis (see below). The key oxidative
rats.25 Glucose is transported into the cytosol by glucose trans- enzyme of the PPP is G6P dehydrogenase, which catalyzes
porters (GLUTs), including GLUT1 and GLUT4. Although the first reaction of the pathway to generate NADPH. In ad-
GLUT1 is the major glucose transporter in the fetal heart and dition to the PPP, a small amount of G6P can enter the hexos-
contributes to constitutive glucose uptake, in the adult heart, amine biosynthetic pathway (HBP), leading to the formation
GLUT4 is the predominant isoform and mediates the bulk of of UDPN-acetylglucosamine (GlcNAc), a monosaccharide
basal myocardial glucose uptake.26,27 donor for O-GlcNAcylation of various proteins.12 Because the
After uptake, free glucose is rapidly phosphorylated to HBP requires not only glucose but also acetyl-CoA and glu-
glucose 6-phosphate (G6P), which subsequently enters many tamine, it may serve as a metabolic sensor linking metabolic
metabolic pathways. Glycolysis represents the major pathway status to several cellular processes. Although flux through the
in glucose use. Glycolysis generates pyruvate, NADH, and PPP and HBP in the normal heart is relatively small, recent
a small amount of substrate-level ATP. This glycolytic ATP evidence has suggested a role for these pathways in the patho-
seems to be important for Ca2+ uptake into the sarcoplasmic physiology of heart disease (see below).
reticulum and diastolic relaxation.28,29 In the cytosol, pyruvate
can be converted to lactate. If transported into the mitochon- The Krebs Cycle and Anaplerosis
drial matrix, pyruvate undergoes oxidation to acetyl-CoA or Acetyl-CoA, the common end product of glucose and FA oxi-
carboxylation to form oxaloacetate or malate. Pyruvate oxida- dation, enters the citric acid cycle (also known as tricarboxylic
tion is catalyzed by the multienzyme complex pyruvate de- acid cycle or Krebs cycle) to generate GTP (or ATP), CO2, and
hydrogenase (PDH), the activity of which is highly regulated reducing equivalents NADH2.2 Because Krebs cycle interme-
by its products (acetyl-CoA, NADH) and by phosphorylation diates can be used for many biosynthetic pathways (eg, amino

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


712Circulation ResearchAugust 30, 2013

and nucleic acids), they are constantly removed from the cycle posttranslational modifications such as phosphorylation, acet-
and therefore need to be replaced. The replenishment of the ylation, or methylation.42
Krebs cycle intermediate pool through pathways independent AMP-activated protein kinase (AMPK) is another impor-
of acetyl-CoA is called anaplerosis.32,33 The carboxylation of tant mediator of metabolic adaptation that promotes increased
pyruvate to malate is an important anaplerotic reaction. This ATP production and inhibition of energy-consuming biosyn-
requires NADPH and therefore links anaplerosis to processes thetic pathways under conditions of hemodynamic stress or
that generate (PPP) or consume (lipogenesis, antioxidative de- ischemia. AMPK boosts ATP production by acutely stimu-
fense) NADPH. Anaplerosis is a crucial process in the heart, lating both FA oxidation and glycolysis while slowing down
where impairment rapidly causes contractile dysfunction.34 ATP-consuming processes such as lipid and protein synthesis.
Changes in anaplerotic pathways have recently been impli- Furthermore, AMPK may also improve cardiac energetics in a
cated in heart disease (see below). long-term manner by activating PGC-1, leading to increased
mitochondrial biogenesis and oxidative capacity.45
Oxidative Phosphorylation and the Generation of
Reactive Oxygen Species Metabolism Beyond ATP Production
The reducing equivalents (NADH2 and FADH) enter the elec- Metabolic intermediates may act as regulators of many path-
tron transport chain (ETC) for oxidative phosphorylation. A ways not directly related to ATP production. Table1 sum-
byproduct of this activity is the generation of reactive oxy- marizes classic metabolites that have been recognized as
gen species (ROS) arising mainly from complexes I and III.35 signal transducers. Although much of our discussion so far
Mitochondrial ROS production has been implicated in a wide has focused on substrate oxidation and ATP production in the
variety of cellular processes, ranging from protective mecha- heart, it is equally important to consider that cardiac metab-
nisms such as preconditioning to detrimental mechanisms olism also involves anabolic processes that are essential for
such as oxidative damage and activation of adverse ventricular maintaining cellular activities and promoting cell growth and
remodeling.36 proliferation.46,47 The development of HF is characterized by
profound changes in cardiac structure and function. Given the
Short-term Regulation of Cardiac Substrate Use fundamental role of metabolism in all of these processes, it
FA and glucose use is tightly linked and coregulated. Use of 1 is reasonable to assume that cardiac metabolism occupies a
substrate may directly inhibit the use of the other (the Randle central position in the pathophysiology of HF.
cycle).37 Complete metabolism of glucose is more oxygen ef-
ficient than that of FAs.38 Therefore, substrate selection and
the interaction between glucose and FA use in the heart have Table 1. Classic Metabolic Intermediates That Have Been
been considered highly relevant in cardiac disease.39 Substrate Recognized as Signal Transducers
use and selection in the heart are also modulated by hormones Metabolic Intermediates Regulatory Effects
such as insulin, glucagon-like peptides (GLPs), and catechol-
Fatty acids Activation of PPARs,48 modulation of ion channels
amines. Although important, these regulatory mechanisms lie by palmitoylation49
outside of the scope of this review.
Acylcarnitines Activation of Ca+ channels,50 induction of insulin
Long-term Regulation of Cardiac Metabolism resistance15
Expression levels of metabolic enzymes and their posttrans- Ceramides Activation of PP2A, PKCinhibition of insulin
lational modifications may strongly affect cardiac metabolic signaling,14 induction of mitochondrial and ER stress
and apoptosis51
activity. Peroxisome proliferatoractivated receptor (PPAR)
nuclear receptors are important transcriptional regulators Pyruvate Stimulation of mitochondrial biogenesis,52 regulation
of PGC-1 expression53
of FA use. Although PPAR is the predominant isoform in
the heart, PPAR and PPAR may also modulate FA me- Acetyl-CoA Induction of cell growth and proliferation by
promoting the acetylation of histones54
tabolism.11 Cardiac FA use is also regulated by the estrogen-
related receptor (ERR). In addition to sharing common Hexosamine Multiple cellular effects via O-GlcNAcylation of
regulatory proteins55
targets with PPAR, ERR stimulates the transcription
of genes involved in glucose metabolism and oxidative NAD(P)+/NAD(P)H Modulation of activity of metabolic enzymes,
numerous additional effects via regulation of redox
phosphorylation.40,41
state and sirtuins31,36
The transcriptional activities of PPARs and ERRs are po-
ROS Regulation of redox state, enzyme activity; high levels
tently induced by interacting with members of the PPAR-
induce apoptosis, hypertrophy, inflammation36,56
coactivator-1 (PGC-1) family. PGC-1 binds to and coacti-
AMP Diverse effects on metabolism and cell growth
vates PPAR and ERR, leading to increased capacity of FA
through activation of AMPK45
uptake and oxidation.42 Although less extensively studied,
BCAA Stimulation of protein synthesis57 and various
PGC-1 may also play an important role in the regulation
additional effects via activation of mTOR and
of cardiac energy metabolism.43 PGC-1 proteins are power- inhibition of autophagy58
ful activators of mitochondrial biogenesis. This is mediated
AMPK indicates AMP-activated protein kinase; BCAA, branched-chain amino
in part by induction of the nuclear respiratory factor 1 and acids; CoA, coenzyme A; ER, endoplasmic reticulum; GlcNA, N-acetylglucosamine;
the mitochondrial transcription factor A.44 The activity of mTOR, mechanistic target of rapamycin; PGC-1, peroxisome proliferatoractivated
PGC-1 is highly regulated by both its expression levels and receptor coactivator-1; PKC, protein kinase C; PPAR, peroxisome proliferator
activated receptor; PP2A, protein phosphatase; and ROS, reactive oxygen species.

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


Doenst et al Cardiac Metabolism in Heart Failure 713

Cardiac Metabolism in HF idiopathic dilated cardiomyopathy. It is unlikely that these dif-


There are 2 major points that should be considered in the inter- ferences can be explained by methodological differences. We
pretation of study results on metabolic alterations in HF. First, therefore suggest that in contrast to changes in FA oxidation,
metabolic phenotypes and their mechanisms differ between glucose oxidation does not correlate with contractile function
HF of different pathogeneses. Second, because the progres- in HF but that changes in glucose oxidation may depend on
sion to HF is often long and complex, the time point of assess- both the stage and the pathogenesis of HF.
ment (ie, compensated hypertrophy with or without diastolic
dysfunction versus manifest systolic dysfunction) influences
Anaplerosis in HF
Another factor that may influence the interpretation of glucose
the metabolic adaptations that are observed. Despite these ca-
oxidation results is the fact that pyruvate may be channeled
veats, findings in humans and animal models provide clear
into anaplerotic pathways (Figure 2B). In hypertrophied rat
evidence indicating severe metabolic alterations in the failing
hearts induced by aortic constriction, Sorokina et al17 showed
heart. Table2 summarizes reported changes in substrate use
that glucose oxidation by PDH is unchanged despite increased
and energy production in various models of HF.
glycolysis. More important, they identified an increase in
FA Use in HF anaplerotic flux into the Krebs cycle probably via pyruvate
Most studies reveal reduced cardiac FA use. FA uptake is re- carboxylation by malic enzyme.17 The increase in this alterna-
duced in concert with high-salt dietinduced HF65 and by rap- tive pathway of pyruvate use may account for the mismatch
id pacing.71 In agreement with these findings, studies in other between glycolysis and glucose oxidation that is commonly
models of HF also observed a reduction in mRNA and protein seen in models of pressure overloadinduced cardiac hyper-
expression of FA transporters when systolic dysfunction was trophy. This hypertrophy-associated anaplerotic change has
present.59,65,68,70 We found that FA oxidation rate and the ex- also been confirmed in mice with aortic constriction75 and in
pression of FA oxidation enzymes were already decreased in hyperthyroid rats.76 Thus, induction of anaplerotic pathways
early (compensated) stages of left ventricular hypertrophy.59 seems to be a hallmark of metabolic remodeling in cardiac
These results are also consistent with findings in spontaneous- hypertrophy. Considering that hypertrophic growth requires
ly hypertensive rats66,72 and in rats with abdominal aortic con- increased supply of amino acids and nucleic acids derived
striction63,64 but not with those in Dahl salt-sensitive rats65 and from precursors in the Krebs cycle, activation of anaplerotic
in rats with myocardial infarction.68 However, by the time that pathways may be required to maintain Krebs cycle function.
systolic dysfunction manifests, there is clear evidence from However, this compensatory mechanism might also have un-
studies in animal models and human subjects that myocardial favorable consequences (Figures 2B and 3 for illustration).
FA oxidation is decreased. Figure2A summarizes the fate of For example, increased flux of pyruvate through anaplerotic
FAs in HF and how these changes may lead to decreased ATP pathways reduces its availability for oxidation by the PDH
production. complex. As a result, pyruvate oxidation might not sufficiently
compensate for the impaired FA oxidation, leading to ener-
Glucose Use in HF getic inefficiency of the Krebs cycle.17 Furthermore, increased
In contrast to FA use, data on cardiac glucose use are less con- flux through malic enzyme may also affect cardiac function by
sistent (Table 2). In the presence of systolic dysfunction, car- consuming NADPH,77 which is required for triglyceride syn-
diac glucose uptake was found to be decreased in mice after thesis and for defending against oxidative stress. Therefore,
aortic constriction,61 unchanged in rats with myocardial infarc- the role of anaplerotic changes, particularly in relation to the
tion,69 and increased in Dahl salt-sensitive rats.65 In compen- nature of cardiac hypertrophy, remains incompletely under-
sated hypertrophy, induced by abdominal aortic constriction, stood. This knowledge gap is attributable in part to the techni-
glycolysis was modestly increased without changes in glu- cal challenges inherent in measuring anaplerosis.
cose oxidation.62,63 By assessing substrate oxidation at various
time points after aortic constriction in rats, we observed that Mitochondrial Biogenesis and Function in HF
cardiac glucose oxidation tended to increase initially but was The above-described alterations in substrate oxidation and
unchanged relative to controls in the phase of compensated hy- anaplerosis occur at the level of mitochondria. It is therefore
pertrophy and ultimately decreased when systolic dysfunction not surprising that many studies have described significant
occurred.59 The impaired glucose oxidation that parallels sys- mitochondrial changes in HF (Table 2). In rat hearts with sys-
tolic dysfunction might be attributable in part to mitochondrial tolic dysfunction induced by aortic constriction, we identified
dysfunction, reduced expression of genes involved in glycoly- abnormal mitochondrial morphology, reduced mitochondrial
sis and glucose oxidation, or decreased abundance of the PDH volume density, and altered levels of most ETC proteins,
complex.65,74 However, in infarcted rat hearts, we observed no with the majority being decreased.60 Mitochondrial proteomic
change in glucose oxidation rate despite manifest systolic dys- remodeling in HF has also been demonstrated in mice with
function.69 Interestingly, spontaneously hypertensive rats, at pressure overload, which is characterized primarily by altered
the cardiac hypertrophy stage and before HF, were found to abundance of proteins involved in ETC and substrate metabo-
have higher glucose oxidation rates66 or increased flux through lism.74 These results suggest that perturbed mitochondrial bio-
the PDH complex67 relative to control rats. In addition, Osorio genesis is an important feature of pressure overloadinduced
et al71 showed increased glucose oxidation rates in failing dog HF. Consistent with this notion, expression of PGC-1 is de-
hearts induced by rapid pacing, and Dvila-Romn et al73 creased in systolic HF, as has been shown by us43,60 and oth-
demonstrated higher total rates of glucose use in patients with ers.78,79 There is limited evidence of mitochondrial alterations

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


714Circulation ResearchAugust 30, 2013

Table 2. Changes in Substrate Oxidation and Mitochondrial Function in Various Models of Cardiac Hypertrophy and Failure
Morphological and Functional Mitochondrial Changes
Models of Heart Disease Characteristics FA Metabolism Glucose Metabolism and Energy Status Reference
Aortic constriction in rats Compensated hypertrophy FAO and mRNA expression of GO decreased in State-3 respiration initially 59
without/with diastolic genes regulating FAO decreased systolic dysfunction increased but decreased in
dysfunction; systolic in compensated hypertrophy and systolic dysfunction
dysfunction systolic dysfunction
Aortic constriction in rats Hypertrophy with diastolic and FAO, mRNA, and protein GO decreased, Disorganized mitochondrial 60
systolic dysfunction expression of FAO enzymes protein expression of cristae, mitochondrial
decreased PDH complex increased volume density, and levels
of most ETC proteins
decreased, state-3
respiration decreased
Aortic constriction in mice Hypertrophy with systolic FAO unchanged Glycolysis and GO Mitochondrial dysfunction, 43
dysfunction increased decreased respiration and
ATP content
Aortic constriction in mice Hypertrophy with diastolic FAO unchanged GU, glycogen synthesis, 61
and systolic dysfunction glucose, and lactate oxidation
decreased
Abdominal aortic Mild, compensated FAO and mRNA expression of Glycolysis increased 62
constriction in rats hypertrophy genes regulating FAO unchanged modestly
Abdominal aortic Mild, compensated FAO and mRNA expression of GO unchanged 63
constriction in rats hypertrophy genes regulating FAO decreased
Abdominal aortic Hypertrophy, heart function FAO decreased Glycolysis increased, GO 64
constriction in rats ex vivo unchanged unchanged
Dahl salt-sensitive rats Compensated hypertrophy; FA uptake and mRNA GU increased, mRNA mRNA and protein 65
systolic dysfunction expression of genes expression of genes regulating expression of genes
regulating FA use decreased glycolysis and GO decreased; regulating mitochondrial
in systolic dysfunction pentose phosphate pathway biogenesis decreased in
flux increased in systolic systolic dysfunction; PCr/
dysfunction ATP ratio decreased
SHRs (Sprague-Dawley Hypertrophy, heart function FAO lower (note that some strains GO higher 66
rats as controls) ex vivo unchanged of SHRs have a mutation in
CD36, which could independently
modulate FAO)
Spontaneously Hypertrophy, minor differences Flux through PDH PCr/ATP ratio not different 67
hypertensive rats (Wistar in diastolic and systolic higher
rats as controls) functions
Myocardial infarction Compensated mRNA expression of genes mRNA and protein expression 68
in rats hypertrophy; systolic regulating FA use decreased of GLUT1 increased in systolic
dysfunction in systolic dysfunction dysfunction
Myocardial infarction No hypertrophy, left FAO and mRNA expression of GU and GO unchanged mRNA expression of genes 69
in rats ventricular dilatation, genes regulating FAO decreased regulating mitochondrial
systolic dysfunction biogenesis decreased
Myocardial infarction Mild hypertrophy, FA use decreased, protein 70
in rats systolic dysfunction expression of fatty acid
transporters decreased
Pacing-induced HF in dogs No hypertrophy, left ventricular FA uptake and oxidation, activity GO increased 71
dilatation, systolic dysfunction of CPT I, and MCAD decreased
Patients with HF with IDCM Systolic dysfunction mRNA expression of MCAD, 72
or ischemic heart disease LCAD lower
Patients with HF with IDCM Hypertrophy, systolic FA use lower Glucose use higher 73
dysfunction
Patients with HHD, AS, Hypertrophy with normal PCr/ATP ratio not different 5
or DCM systolic function in patients among groups; PCr content
with HHD and AS; left lower in AS and DCM; ATP
ventricular dilatation and systolic content lower in DCM
dysfunction in patients with DCM
AS indicates aortic stenosis; CPT, carnitine palmitoyltransferase; DCM, dilated cardiomyopathy; ETC, electron transport chain; FA, fatty acid; FAO, fatty acid oxidation;
GLUT, glucose transporter; GO, glucose oxidation; GU, glucose uptake; HF, heart failure; HHD, hypertensive heart disease; IDCM, idiopathic dilated cardiomyopathy; LCAD,
long-chain acyl-CoA dehydrogenase; MCAD, medium-chain acyl-CoA dehydrogenase; PCr, phosphocreatine; PDH, pyruvate dehydrogenase; and SHR, spontaneously
hypertensive rat.
Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016
Doenst et al Cardiac Metabolism in Heart Failure 715

Figure 2. Alterations in substrate metabolism and mitochondria during the development of heart failure (HF). Bold lines indicate
pathways reported to be activated. Thin lines represent pathways reported decreased. The question marks imply unknown changes or
inconsistent observations. A, Fatty acid oxidation is impaired in cardiac hypertrophy and failure, leading to reduced ATP production.
B, Most evidence suggests that glucose oxidation is unchanged in compensated hypertrophy and decreased in HF, but discrepancies
exist. In contrast, several nonATP-generating pathways of glucose metabolism (HBP, PPP, anaplerosis) are induced. C, Increased
generation of mitochondrial reactive oxygen species (ROS; perhaps because of changes in the electron transport chain) causes direct
mitochondrial damage, which may further increase mitochondrial ROS production to create a vicious cycle. Mitochondrial damage results
in ATP deficiency. Mitochondrial ROS may also cause oxidative damage to other cellular components and may contribute to adverse
structural remodeling. BCAA indicates branched-chain amino acid; CoA, coenzyme A; CPT, carnitine palmitoyltransferase; FAT, fatty
acid transporter; G6P, glucose 6-phosphate; GLUT, glucose transporter; HBP, hexosamine biosynthetic pathway; IMS, mitochondrial
intermembrane space; PPP, pentose phosphate pathway; and TG, triglyceride. Illustration Credit: Ben Smith.

in other models of HF. In failing rat hearts caused by myocar- mitochondrial biogenesis and its signaling in cardiac hyper-
dial infarction, we found normal mRNA expression of PGC- trophy and failure are still incompletely understood, available
1. However, the expression of p38 mitogenactivated protein evidence supports the concept that cardiac mitochondria are
kinase, which could modulate PGC-1 activity, was reduced.69 affected early and undergo progressive remodeling during the
In patients with HF of diverse pathogeneses, Karamanlidis et development of cardiac hypertrophy and failure.
al80 observed decreased mitochondrial DNA content accompa- Mitochondrial dysfunction as evidenced by decreased mi-
nied by reductions in mitochondrial DNAencoded proteins. tochondrial respiration and reduced ATP production is consis-
Of note, these changes were associated with increased abun- tently observed when systolic dysfunction occurs in various
dance of PGC-1 protein but decreased expression of ERR models of HF.59,82,83 Interestingly, we and others found that mi-
and mitochondrial transcription factor A at both the mRNA tochondrial respiration in pressure-overloaded hearts initially
and protein levels.80 These findings suggest that mitochondrial increased and did not decrease until systolic dysfunction de-
biogenesis signaling is depressed in advanced HF and that veloped.59 Considering that mitochondrial damage is probably
posttranslational modulation of PGC-1 may play a role. an early event in the progression to HF, this biphasic response
Little is known about the regulation of mitochondrial of mitochondrial respiratory capacity might reflect unknown
biogenesis in compensated hypertrophy. We found a signifi- compensatory mechanisms that can sustain oxidative phos-
cant decline in mRNA expression of PGC-1, ERR, and phorylation during the stage of compensated hypertrophy but
mitochondrial transcription factor A at times when left ven- are lost as cardiac dysfunction develops.
tricular ejection fraction was still preserved (unpublished
observations), suggesting repressed mitochondrial biogen- Cardiac Energetics in HF
esis pathways in compensated hypertrophy. In contrast, in Mitochondrial dysfunction in advanced HF has been linked to
hypertrophied mouse hearts with normal function induced impaired myocardial energetics. Studies in animal models and
by long-term angiotensin II infusion, Dai et al81 reported in- humans have reported a decrease in the PCr/ATP ratio, ATP con-
creased mRNA expression of PGC-1, ERR, and mitochon- tent, and the ATP flux through creatine kinase in advanced HF
drial transcription factor A. However, these hearts exhibited with reduced ejection fraction. Given that sufficient ATP supply
increased mitochondrial damage and mitophagy.81 Therefore, is essential for normal cardiac function, this change has been
the induction of mitochondrial biogenic signaling could rep- suggested to be responsible for the transition to systolic dys-
resent a compensatory mechanism triggered by mitochon- function.6,84 If this were true, it would be reasonable to hypoth-
drial injury and loss. Collectively, although the regulation of esize that ATP depletion should be detectable in compensated

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


716Circulation ResearchAugust 30, 2013

hypertrophy just before the onset of systolic dysfunction. Kato to impaired calcium handling91 or modification of sarcomeric
et al65 showed a 12% decrease in the PCr/ATP ratio in com- proteins,92 which have all been reviewed by others.
pensated hypertrophy in Dahl salt-sensitive rats. In contrast,
Dodd et al67 found that both absolute content of high-energy Mitochondrial ROS and Its Link to Cardiac
phosphate metabolites and the PCr/ATP ratio were normal in Metabolism in HF
the hypertrophied heart of spontaneously hypertensive rats. In Although metabolic and structural alterations in the fail-
hearts of patients with hypertensive heart disease with nor- ing heart have been increasingly well characterized, little is
mal ejection fraction, the PCr/ATP ratio was found to be de- known about the mechanisms that drive the remodeling pro-
creased,85 but the absolute content of ATP and PCr has been cess in HF. Increased cardiac ROS levels have been implicated
shown to be preserved.5,86 Thus, data in hypertrophied hearts in HF, but the role of ROS in the pathogenesis of HF was ques-
with normal systolic function do not support a model in which tioned as a result of disappointing outcomes of antioxidant in-
impaired ATP-producing capacity might be an antecedent to terventions in human studies.93,94 However, in a recent animal
the transition to decompensated HF. It is possible that the lack study, Dai et al81 demonstrated that angiotensin II increased
of a clear conclusion could be attributable in part to method- mitochondrial ROS, leading to mitochondrial damage, activa-
ological limitations in using PCr/ATP ratios to quantify myo- tion of mitogen-activated protein kinases, and finally cardiac
cardial energetics. However, an alternative conclusion is that hypertrophy and fibrosis.81 Another study by the same group
decreased levels of high-energy phosphates do not cause HF also showed that mitochondria-targeted antioxidant treatment
per se but represent an adaptation to decreased pump function. but not nontargeted ROS scavenging with N-acetyl cysteine
In advanced HF, there is consistent evidence of decreased ameliorated cardiomyopathy induced by chronic angiotensin
myocardial ATP availability, but it remains unclear whether II infusion.95 Of note, mouse hearts in this model of pres-
the reduction in ATP abundance contributes to contractile dys- sure overload presented mild diastolic dysfunction without
function. Approaches that would directly enhance myocardial changes in left ventricular ejection fraction,81 indicating an
ATP abundance in the failing heart are required to address this early stage of compensated hypertrophy. These results sug-
question. In a recent study, the xanthine oxidase inhibitor al- gest that increased mitochondrial ROS could be an early event
triggering structural remodeling and mitochondrial defects in
lopurinol was shown to acutely improve the relative and abso-
hypertensive heart disease. Figure 2C illustrates the genera-
lute concentrations of myocardial high-energy phosphates and
tion of mitochondrial ROS and its potential impact on energy
ATP flux through creatine kinase in the failing human heart.
metabolism and other pathological processes.
Nevertheless, whether cardiac function changed in response
The regulation of cellular ROS homeostasis is complex and
to this energetic improvement was unknown.87 Furthermore,
is only partially understood. In the mitochondria, ROS are pro-
in a retrospective study in a large cohort of patients with HF,
duced mainly from the ETC particularly from complexes I and
Struthers et al88 found no beneficial effects of long-term al-
III. Therefore, changes in the ETC may favor electron leakage
lopurinol treatment on cardiovascular mortality. Of note, be-
and consequently ROS formation.36 The elevation in ROS pro-
cause allopurinol may exert complex systemic effects, these
duction may further affect the function of the ETC, leading to
negative results can neither confirm nor refute a potential
a vicious cycle (Figures 2C and 3). Studies in various tissues
role of energetic impairment in HF pathogenesis. Although
have linked FA use to increased ROS levels.96,97 In diabetic car-
the correlation between cardiac energy status and survival diomyopathy, ROS have also been associated with myocardial
in patients with HF is striking,89 this correlation could also lipid accumulation, lipotoxicity, and decreased cardiac effi-
represent the worsening mitochondrial function that occurs in ciency on the basis of mitochondrial uncoupling.98 The failing
concert with progression of HF severity. Although this alter- heart is believed to be subjected to higher levels of free FAs,
native hypothesis remains to be disproved, studies on cardiac probably as a result of increased lipolysis.39 In addition, models
energetics in HF are attractive because they may provide not of HF have been associated with myocardial lipid accumula-
only mechanistic insights and potential new therapeutic strate- tion.99,100 As a result, FA use has been considered unfavorable
gies but also valuable prognostic information. for the stressed heart because of increasing ROS production.39
Normal cardiac function requires the coordinated regula- However, in animal models with a cardiac-specific loss of
tion and interaction of several physiological and signaling acetyl-CoA carboxylase-2, increased myocardial FA use did
systems in addition to energy supply. In rats developing HF at- not adversely affect left ventricular remodeling after pressure
tributable to pressure overload, we calculated the ATP amount overload. This might represent a scenario of complete sub-
generated from substrate oxidation and found that the ratio strate oxidation without increased generation of mitochondrial
of cardiac power to ATP was consistently reduced as soon as ROS.75 Furthermore, cardiac FA oxidation is depressed during
cardiac hypertrophy developed.59 This result provides indi- HF progression, and there is little direct evidence in HF models
rect evidence for inefficient transduction from ATP into me- that FA use may increase ROS production. Thus, the causes of
chanical power. Before the onset of systolic dysfunction, the increased mitochondrial ROS in the progression to HF remain
heart undergoes severe structural changes (eg, hypertrophy, unclear and warrant further investigation.
fibrosis, dilatation). It is therefore likely that such profound
remodeling of the contractile apparatus may involve mecha- Changes in the PPP in HF
nisms that significantly affect cardiac function independently The regulation of the cellular redox environment is tightly
of ATP supply. Potential mechanisms range from changes in linked to substrate metabolism via the PPP as an important
the extracellular matrix (myocardial fibrosis, inflammation)90 source of NADPH. NADPH is required for the generation of

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


Doenst et al Cardiac Metabolism in Heart Failure 717

cytosolic ROS, which, at low levels, are involved in prolifera- pathways that initiate cardiac remodeling, additional signal-
tion and survival signaling. Conversely, NADPH also main- ing pathways linked to the HBP may also promote favorable
tains the pool of reduced antioxidants such as glutathione that effects. Further studies are therefore needed that focus on the
are crucial defenses against oxidative stress.36 Hence, the PPP characterization and targeted modulation of intracellular sig-
may play a dual role in the regulation of redox homeostasis. naling pathways that are regulated by HBP, in the context of
Although much effort has been made to characterize glycoly- clinical and experimental models of HF.
sis and glucose oxidation, little is known about the regulation
of the PPP in HF. In dogs subjected to pacing-induced HF, Autophagy and Its Link to Cardiac Metabolism
increased superoxide levels were attributed to increased activ- in HF
ity of G6P dehydrogenase, the key oxidative enzyme of the Metabolic remodeling in HF is also associated with changes
PPP.101 If the effects of superoxide are indeed damaging, this in autophagy. Autophagy is a highly conserved process by
activation of the PPP in the failing heart could be considered which organelles and large cellular components are degraded.
detrimental. However, G6P dehydrogenasedeficient mice The products of autophagy (amino acids, FAs, sugars, and nu-
developed higher oxidative stress and worsened contractile cleosides) may then be channeled into both energy-generating
function after myocardial infarction or aortic constriction.102 and -biosynthesis pathways. Under normal conditions, basal
In Dahl salt-sensitive rats, Kato et al65 also found that the flux autophagy is crucial by eliminating damaged organelles and
through the PPP progressively increased during the develop- misfolded proteins. In states of nutrient deprivation such as
ment of HF. Importantly, they demonstrated that treatment starvation or ischemia, autophagic activity is increased, which
with dichloroacetate (DCA) further increased this flux, which may support cell function by mobilizing endogenous nutrition-
was associated with improved cardiac function.65 Taken to- al sources.111 In models of HF including pressure overload112
gether, the PPP is activated in HF models. Although superox- and myocardial infarction,113 autophagy has also been shown
ide production may consequently increase, currently available to be induced. Mechanisms for the activation of autophagy in
data support the notion that higher flux through the PPP in HF are less clear. A number of changes observed in the hy-
HF may represent a compensatory mechanism whose further pertrophied and failing heart such as energy depletion, AMPK
activation could be of therapeutic relevance. activation,114 mitochondrial ROS overproduction, and damaged
mitochondria are strong mediators of autophagy in various set-
Changes in the HBP in HF tings.111,115 However, the potential contribution of these events
The HBP, which is linked to metabolism of glucose, FAs, and to autophagic activation in HF remains to be verified.
amino acids, has been implicated in various models of heart Autophagy has also been implicated in ventricular remodel-
disease. The HBP and HBP-dependent O-GlcNAcylation are ing. In an elegant study, Cao et al116 provided evidence indi-
induced in the diabetic heart, which may result in increased cating that the activation of autophagy, although degradative
apoptosis,103 mitochondrial dysfunction,104 and impaired Ca2+ in nature, is essential for cardiac hypertrophy. Because cell
cycling.105 Recently, the role of the HBP in HF has also re- growth requires stimulation of biosynthesis pathways, these
ceived special interest. By using various models of HF, includ- data may seem paradoxical at first glance. However, they are
ing aortic constriction, myocardial infarction, and hypertensive supported by earlier studies showing that blunting of protein
rats, Lunde et al106 demonstrated that global O-GlcNAcylation degradation mediated by the ubiquitin-proteasome system
was increased by 40% in hypertrophied and failing hearts. may attenuate cardiac hypertrophy.117,118 Together, these data
They also confirmed these findings in patients with aortic ste- indicate that cardiac hypertrophy and remodeling are dynamic
nosis.106 In endothelial cells, increased flux through the HBP reconstructive processes in which degradation of certain cel-
has been attributed to overproduction of mitochondrial super- lular components by autophagy and the ubiquitin-proteasome
oxide.107 Considering that mitochondrial ROS formation also system is an important antecedent that might stimulate the
increases in cardiac hypertrophy,81 mitochondrial ROS might biosynthesis of new structures.
represent a potential mechanism for activation of the HBP in Because of the essential role of autophagy in ventricular
the heart. remodeling, one might expect that inhibiting autophagy may
Whereas the induction of O-GlcNAcylation in the progres- prevent cardiac hypertrophy and failure. However, studies in
sion of HF is well described, less is known about the functional knockout models with suppressed autophagy have delivered
relevance of these changes. In a cardiomyocyte model of hy- inconsistent results.119,120 Given the highly complex regulation
pertrophy, Facundo et al108 found that increased flux through of autophagy, it is reasonable to assume that specific changes in
the HBP and the resulting increase in O-GlcNAcylation were the induction or targeting of autophagy but not the autophagic
responsible for hypertrophic growth via activation of the nu- flux itself may affect functional outcome. For example, Oka
clear factor of activated T cells. This finding is highly relevant et al121 recently demonstrated in mice with aortic constriction
because it indicates that induction of the HBP is an early event that mitochondrial DNA that escapes from autophagy causes
that triggers myocardial remodeling. Because nuclear factor of cardiac inflammation and failure. Therefore, the regulation
activated T cells signaling has been associated with pathologi- and the role of autophagy in HF remain to be fully elucidated.
cal hypertrophy,109 one might consider this activation of the Because both the induction of autophagy and the processing
HBP to be detrimental. However, reducing O-GlcNAcylation of autophagic products are linked to metabolism, the relation-
in mice with myocardial infarction exacerbated ventricular ship between metabolic remodeling in HF and the regulation
dysfunction.110 Thus, although the HBP activates signaling of autophagy is an attractive target for future studies.

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


718Circulation ResearchAugust 30, 2013

Structural remodeling Metabolic remodeling

Glucose Fatty acids

Hypertrophy,
Fibrosis ? O-Glyc H

G O
P
NADPH
Pyruvate
O

synthesis
Acetyl- CoA

Protein
A
Oxidative
stress
Krebs
Inflammation Amino acids
degradation cycle

?
Protein
mitochondrial

NADH2
DNA release

ROS ETC
UPS

Mitochondrial
Mitophagy Autophagy
damage

ATP ATP
depletion
?

Figure 3. Overview of metabolic remodeling and proposed mechanisms linking it to other processes in the progression to heart
failure (HF). Metabolic pathways are blue. Bold lines indicate pathways/processes that are increased or dominant. Thin lines represent
pathways/processes that are decreased. The question marks imply unknown causes/effects. In general, metabolic remodeling in cardiac
hypertrophy and failure is characterized by a shift away from energy production to activation of biosynthetic pathways required for
structural remodeling processes such as ventricular hypertrophy and fibrosis. Particularly, fatty acid oxidation is decreased and may not
be sufficiently compensated given the lack of increase in glucose oxidation. These alterations and further mitochondrial defects result
in ATP depletion. Instead of being oxidized, pyruvate may be preferentially used for anaplerosis to maintain Krebs cycle moieties, which
might be increasingly channeled into protein synthesis. Hypertrophic mediators such as mitogen-activated protein kinases (MAPKs)
and nuclear factor of activated T cell (NFAT) are activated as a result of increased mitochondrial reactive oxygen species (ROS) and flux
through the hexosamine biosynthetic pathway (HBP), respectively. Overproduction of mitochondrial ROS causes oxidative damage.
Although the flux through the pentose phosphate pathway (PPP) is increased, antioxidative defense might be inadequate, attributable
to the consumption of NADPH by the anaplerotic malic enzyme. Mitochondrial damage and ATP depletion may stimulate autophagy.
Increased activity of autophagy and the ubiquitin-proteasome system (UPS) may contribute to hypertrophy by providing amino acids and
other metabolites. Increase in mitophagy may trigger myocardial inflammation by releasing mitochondrial DNA. A indicates anaplerosis;
CoA, coenzyme A; ETC, electron transport chain; G, glycolysis; H, HBP; O, oxidation; and P, PPP.

Figure 3 schematically illustrates the interaction between to reduce cardiac FA use, and some studies revealed bene-
metabolic and structural remodeling of the heart in the pro- ficial effects.122 However, the concept of inhibiting FA use
gression to HF. Metabolic remodeling in HF is characterized has now been challenged by contradictory results of stud-
by defects in energy production and changes in metabolic path- ies in animal models of HF subjected to high-fat diets.123
ways that are involved in the regulation of essential cellular For example, Raher et al124 showed that high-fat diet in
functions. The figure attempts to illustrate that the inability mice induced myocardial insulin resistance and exacerbat-
to produce sufficient ATP is only one aspect of metabolic re- ed left ventricular remodeling caused by aortic constric-
modeling. This model promotes the hypothesis that during HF, tion. In contrast, Okere et al125 reported decreased cardiac
other nonATP-producing processes, which might play a rela- hypertrophy and improved contractile function after treat-
tively small role in the nonstressed heart, may gain more im- ment with high-fat diet in hypertensive Dahl salt-sensitive
portance and directly affect contractile function, as illustrated rats. In addition, a`cute inhibition of FA oxidation in a
in Figure 4. This schematic also provides a road map toward population of patients with end-stage HF led to significant
identifying targets that could be translated to HF therapeutics. worsening of left ventricular function.126 The plethora of
studies on the modulation of cardiac FA use and its effects
Potential Metabolic Therapy for HF on contractile function has been extensively reviewed and
Modulation of Cardiac FA Metabolism discussed elsewhere.122,123,127 Although a mechanistic link
Modulation of cardiac FA use has been a target of meta- between cardiac FA metabolism and contractile function
bolic therapy in HF for the past decade. The failing heart remains controversial because some therapeutic strategies
has been suggested to be in a state of FA overload, suffer- targeting cardiac FA use have suggested functional con-
ing from increased oxygen wastage and oxidative stress sequences of manipulating this pathway, additional inves-
or from accumulation of cardiotoxic lipid derivatives.39 tigation of therapies that modulate FA metabolism in the
Consequently, many pharmacological approaches sought failing heart is warranted.

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


Doenst et al Cardiac Metabolism in Heart Failure 719

Figure 4. Simplified illustration of the


relationship between cardiac metabolism and
contractile function, reflecting the role of ATP
production and nonATP-producing processes.
In the normal heart under steady-state conditions
(A), the main metabolic task is to produce ATP
for contractile function. Alternative pathways
such as the pentose phosphate pathway (PPP),
the hexosamine biosynthetic pathway (HBP),
the activation of autophagy, or the production of
reactive oxygen species (ROS) play a minor role. In
the progression to heart failure (B), ATP-producing
capacity is reduced. In addition, the alternative
pathways may be activated to various degrees.
Such activation may provide additional metabolic
mechanisms influencing contractile function that
could represent potential new metabolic targets for
therapy.

Modulation of Cardiac Glucose Metabolism of DCA stimulated myocardial lactate consumption, which
A small number of studies suggest that direct modulation of glu- was accompanied by decreased oxygen consumption and in-
cose use could also be beneficial. In mice subjected to pressure creased cardiac work. However, not all human studies have
overload, life long cardiac-specific overexpression of GLUT1 in- demonstrated short-term benefit,132 and long-term clinical
creased glucose uptake and glycolysis and improved cardiac ener- trials have never been performed in HF, perhaps in part be-
getics and function.128 Although the enhanced ATP production may cause neuropathy has been reported when DCA was used
account for the improved functional outcome in these mice, possi- long term in cases of inherited mitochondrial disorders.133
ble changes in accessory pathways of glucose metabolism such as In Dahl salt-sensitive rats with hypertension, DCA treat-
ment reduced ventricular hypertrophy and improved cardiac
the PPP and the HBP could also play a role but were not evaluated.
function and survival, which was associated with increased
Because the increase in glucose uptake in this model preceded the
myocardial glucose uptake and energy reserves. Of note,
induction of pressure overload, it remains to be determined wheth-
treatment with DCA also enhanced flux through the PPP,
er increasing glucose uptake after the onset of pressure overload or which was shown to be the mechanism by which DCA re-
before the transition to HF will have any effect. duced oxidative damage and apoptosis in cardiomyocytes.65
Among the limited pharmacological strategies to stimu- Interestingly, long-term DCA treatment also increased flux
late glucose use directly in HF, DCA has been the most wide- through PDH and reduced cardiac hypertrophy in rats with
ly studied compound. DCA indirectly activates the PDH hyperthyroidism.76 Collectively, these results suggest that
complex and thus glucose oxidation, which is considered augmenting cardiac glucose use could represent a poten-
the mechanism of cardioprotection in ischemia/reperfusion tial strategy to treat HF, which not only may improve car-
models.129,130 The use of DCA to treat HF has been tested diac energetics but also may attenuate oxidative stress and
in some animal models and humans. In patients with ad- structural remodeling. However, additional studies with less
vanced HF, Bersin et al131 showed that a 30-minute infusion toxic agents are required.

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


720Circulation ResearchAugust 30, 2013

Modulation of Cardiac Anaplerosis thioredoxin.141 Because ROS can lead to mitochondrial dam-
As described above, anaplerotic flux through pyruvate carbox- age and cardiac structural remodeling,81 inhibition of ROS
ylation is elevated in compensated hypertrophy, which may af- signaling by AMPK could potentially attenuate these patho-
fect cardiac lipogenesis and antioxidant defense mechanisms logical processes. Similarly, AMPK plays an important role
by consuming NADPH. In hypertrophied rat hearts, Pound in the regulation of autophagy, which has been implicated in
et al77 demonstrated that acute activation of PDH by DCA ventricular hypertrophy116 and inflammation.121 Given that
competed for pyruvate and partially reduced its flux through ventricular remodeling may occur early in the development of
malic enzyme, which led to normalization of the myocardial HF, studies examining the impact of early AMPK activation
triglyceride pool and improved contractile function. These re- on pathological remodeling are therefore warranted.
sults may suggest therapeutic relevance of inhibiting pyruvate AMPK can be activated by a number of drugs. A long-
carboxylation in cardiac hypertrophy. Furthermore, they pro- used agonist is 5-aminoimidazole-4-carboxamide 1--D-
vide an additional protective mechanism beyond the energetic ribofuranoside which activates AMPK in various tissues,
aspect of improving cardiac glucose oxidation. However, the including the heart, by forming the AMP-mimetic ZMP
role of long-term inhibition of anaplerotic pathways in cardiac (5-aminoimidazole-4-carboxamide 1--D-ribofuranoside mo-
hypertrophy and failure remains to be elucidated. nophosphate).45 Although 5-aminoimidazole-4-carboxamide
Cardiac function may also be enhanced by targeting other 1--D-ribofuranoside has been used extensively in animal
anaplerotic routes. If the induction of anaplerosis through studies, its role in humans is still unclear.142 The antidiabetic
pyruvate carboxylation is a compensatory mechanism to drug metformin has also been shown to activate AMPK in the
maintain Krebs cycle moieties,17 early supplementation with heart. Although metformin was initially contraindicated in
an anaplerotic substrate may allow pyruvate to be oxidized advanced HF attributable to the risk of lactic acidosis, recent
by PDH. We found that feeding aortic-constricted rats with studies have shown beneficial effects of early administration
of metformin in models of HF.83,143,144 Metformin is believed to
triheptanoin-enriched diets, which may exert anaplerotic ef-
activate AMPK by inhibiting complex I, leading to increased
fects by providing the Krebs cycle with propionyl-CoA, at-
cellular AMP levels.145,146 However, complex Iindependent
tenuated ventricular hypertrophy and diastolic dysfunction.
mechanisms have also been proposed.147 Because metformin
Importantly, these effects were accompanied by increased
may also act independently of AMPK,148,149 caution is warrant-
cardiac glucose oxidation.134 Although the mechanistic role of
ed when attributing treatment effects of metformin to AMPK
anaplerosis in these treatments remains to be substantiated,
activation.
the results again point to anaplerotic pathways as a potential
therapeutic target that may modulate metabolic and structural
Activation of Cardiac GLP-1 Receptors
remodeling in HF. GLP-1 is an incretin hormone that is secreted by neuro-
endocrine cells in the gut in response to feeding. Once se-
AMPK Activation creted, GLP-1 is rapidly degraded by the enzyme dipeptidyl
Cardiac hypertrophy and failure are associated with increased
peptidase-4. Dipeptidyl peptidase-4 inhibitors and GLP-1
activity of AMPK, which may be attributable to increased agonists are now commonly used to treat diabetes mellitus
AMP/ATP ratio, ROS, or Ca2+ load.135 Pharmacological inter- because increased GLP-1 or activation of its receptors in the
ventions that further increase AMPK activity in HF models cell or gastrointestinal tract stimulates insulin secretion
have resulted in reduced ventricular remodeling and improved and delays gastric emptying, thereby improving metabolic
cardiac function.83,135 These findings suggest that the activation homeostasis.150
of AMPK in HF may be adaptive and amenable to pharma- GLP-1 receptors are expressed in the heart. Activating
cological manipulation. Activation of AMPK induces a wide cardiac GLP-1 receptors in models of ischemia/reperfusion
range of effects that coordinately improve cardiac energetics have consistently shown beneficial effects in terms of reduced
and counteract ventricular remodeling. For example, AMPK infarct size and improved contractile function.151 In pacing-
activation may acutely stimulate both glucose and FA oxida- induced HF in dogs, Nikolaidis et al152 found a significant im-
tion, thereby enhancing energy production. This mechanism provement in ventricular function after a 48-hour infusion of
has been shown to exist in normal hearts or after ischemia and GLP-1, which was associated with increased cardiac glucose
reperfusion136138 and may apply to HF. Activation of AMPK uptake and insulin sensitivity. In spontaneously hypertensive
could also increase energy metabolism by increasing PGC-1 rats receiving a long-term infusion of GLP-1, Poornima et al153
activity,139 which would induce genes of FA use. Furthermore, also reported improved survival and cardiac function. Because
a PGC-1mediated increase in mitochondrial biogenesis GLP-1 treatment increased insulin secretion in these rats, it is
could potentially compensate for mitochondrial damage and unclear whether GLP-1 or insulin accounted for these effects.
loss in HF. This potential mechanism has been suggested in In contrast, in our rat model of pressure overload, we also ob-
mice with myocardial infarction,83 but further studies in other served improved functional outcome and preserved cardiac
models of HF are warranted. glucose oxidation without changes in insulin levels,154 which
Activation of AMPK also alleviates ventricular remodel- suggest insulin-independent effects of GLP-1. There is also
ing in terms of hypertrophy, myocardial fibrosis, and inflam- evidence that GLP-1 might mediate cardiovascular effects via
mation.83,140 Various potential mechanisms may account for GLP-1 receptor independent mechanisms.155
these beneficial effects. AMPK reduces ROS-related oxida- Although the protective effects of GLP-1 have been dem-
tive stress in endothelial cells by inducing the antioxidant onstrated by numerous studies, less is known about the

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


Doenst et al Cardiac Metabolism in Heart Failure 721

underlying molecular mechanisms. In HF models, the protec- 11. Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS, Stanley WC.

Myocardial fatty acid metabolism in health and disease. Physiol Rev.
tive effects of GLP-1 are mostly associated with improved
2010;90:207258.
glucose use. However, the mechanisms by which GLP-1 in- 12. Banke NH, Wende AR, Leone TC, ODonnell JM, Abel ED, Kelly DP,
fluences glucose metabolism and the relationship between Lewandowski ED. Preferential oxidation of triacylglyceride-derived fatty
changes in glucose metabolism and cardiac function in this acids in heart is augmented by the nuclear receptor PPARalpha. Circ Res.
2010;107:233241.
context are incompletely understood. 13. Bikman BT, Summers SA. Ceramides as modulators of cellular and

whole-body metabolism. J Clin Invest. 2011;121:42224230.
Conclusions 14. Chavez JA, Summers SA. A ceramide-centric view of insulin resistance.
HF is associated with profound changes in cardiac metabo- Cell Metab. 2012;15:585594.
15. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O,
lism. Metabolic remodeling in HF is characterized by the Bain J, Stevens R, Dyck JR, Newgard CB, Lopaschuk GD, Muoio DM.
decreased cardiac energy production that may result from pro- Mitochondrial overload and incomplete fatty acid oxidation contribute to
gressive impairments in substrate use and mitochondrial bio- skeletal muscle insulin resistance. Cell Metab. 2008;7:4556.
16. Cook GA, Edwards TL, Jansen MS, Bahouth SW, Wilcox HG, Park
genesis and function. In addition to ATP deficiency, metabolic EA. Differential regulation of carnitine palmitoyltransferase-I gene iso-
remodeling involves changes in metabolic pathways that regu- forms (CPT-I alpha and CPT-I beta) in the rat heart. J Mol Cell Cardiol.
late essential, nonATP-generating cellular processes such as 2001;33:317329.
growth, redox homeostasis, and autophagy. Therefore, modu- 17. Sorokina N, ODonnell JM, McKinney RD, Pound KM, Woldegiorgis
G, LaNoue KF, Ballal K, Taegtmeyer H, Buttrick PM, Lewandowski
lating cardiac metabolism may also affect these critical pro- ED. Recruitment of compensatory pathways to sustain oxidative flux
cesses and improve cardiac function by mechanisms beyond with reduced carnitine palmitoyltransferase I activity characterizes in-
ATP production. Nevertheless, the exact mechanisms linking efficiency in energy metabolism in hypertrophied hearts. Circulation.
2007;115:20332041.
metabolic changes to other pathological processes in HF are 18. McGarry JD, Mills SE, Long CS, Foster DW. Observations on the affinity
still poorly understood. Future mechanistic investigations are for carnitine, and malonyl-CoA sensitivity, of carnitine palmitoyltransfer-
therefore needed to decipher this complex network and to im- ase I in animal and human tissues: demonstration of the presence of malo-
prove the effectiveness of metabolic therapies for HF. nyl-CoA in non-hepatic tissues of the rat. Biochem J. 1983;214:2128.
19. Zammit VA, Fraser F, Orstorphine CG. Regulation of mitochondrial outer-
membrane carnitine palmitoyltransferase (CPT I): role of membrane-to-
Acknowledgment pology. Adv Enzyme Regul. 1997;37:295317.
20. Kim YS, Kolattukudy PE. Purification and properties of malonyl-CoA
We thank Dr Michael Schwarzer for help in reviewing the vast litera-
decarboxylase from rat liver mitochondria and its immunological com-
ture and the many discussions. parison with the enzymes from rat brain, heart, and mammary gland. Arch
Biochem Biophys. 1978;190:234246.
Sources of Funding 21. Hamilton C, Saggerson ED. Malonyl-CoA metabolism in cardiac myo-
cytes. Biochem J. 2000;350(pt 1):6167.
T. Doenst is supported by grant DO602/9-1 from the Deutsche 22. Dyck JR, Hopkins TA, Bonnet S, Michelakis ED, Young ME, Watanabe
Forschungsgemeinschaft. E.D. Abel is supported by grant M, Kawase Y, Jishage K, Lopaschuk GD. Absence of malonyl coenzyme
R01HL108379 from the National Institutes of Health. A decarboxylase in mice increases cardiac glucose oxidation and protects
the heart from ischemic injury. Circulation. 2006;114:17211728.
23. Ussher JR, Lopaschuk GD. Targeting malonyl CoA inhibition of mito-
Disclosures chondrial fatty acid uptake as an approach to treat cardiac ischemia/reper-
None. fusion. Basic Res Cardiol. 2009;104:203210.
24. Zhou L, Huang H, Yuan CL, Keung W, Lopaschuk GD, Stanley WC.
Metabolic response to an acute jump in cardiac workload: effects on mal-
References onyl-CoA, mechanical efficiency, and fatty acid oxidation. Am J Physiol
1. Ingwall JS. ATP and the Heart. Norwell, Massachusetts: Kluwer Academic Heart Circ Physiol. 2008;294:H954H960.
Publishers; 2002. 25. Henning SL, Wambolt RB, Schnekess BO, Lopaschuk GD, Allard

2. Opie LH. Heart Physiology: From Cell to Circulation. Philadelphia, PA: MF. Contribution of glycogen to aerobic myocardial glucose utilization.
Lippincott Williams & Wilkins; 2004. Circulation. 1996;93:15491555.
3. Gibbs CL. Cardiac energetics. Physiol Rev. 1978;58:174254. 26. Aerni-Flessner L, Abi-Jaoude M, Koenig A, Payne M, Hruz PW. GLUT4,
4. Suga H. Ventricular energetics. Physiol Rev. 1990;70:247277. GLUT1, and GLUT8 are the dominant GLUT transcripts expressed in the
5. Beer M, Seyfarth T, Sandstede J, Landschtz W, Lipke C, Kstler H, von murine left ventricle. Cardiovasc Diabetol. 2012;11:63.
Kienlin M, Harre K, Hahn D, Neubauer S. Absolute concentrations of 27. Abel ED. Glucose transport in the heart. Front Biosci. 2004;9:201215.
high-energy phosphate metabolites in normal, hypertrophied, and failing 28. Entman ML, Bornet EP, Van Winkle WB, Goldstein MA, Schwartz A.
human myocardium measured noninvasively with (31)P-SLOOP magnet- Association of glycogenolysis with cardiac sarcoplasmic reticulum, II:
ic resonance spectroscopy. J Am Coll Cardiol. 2002;40:12671274. effect of glycogen depletion, deoxycholate solubilization and cardiac isch-
6. Neubauer S. The failing heart: an engine out of fuel. N Engl J Med. emia: evidence for a phorphorylase kinase membrane complex. J Mol Cell
2007;356:11401151. Cardiol. 1977;9:515528.
7. Wisneski JA, Gertz EW, Neese RA, Gruenke LD, Craig JC. Dual carbon- 29. Kusuoka H, Marban E. Mechanism of the diastolic dysfunction induced
labeled isotope experiments using D-[6-14C] glucose and L-[1,2,3-13C3] by glycolytic inhibition. Does adenosine triphosphate derived from gly-
lactate: a new approach for investigating human myocardial metabolism colysis play a favored role in cellular Ca2+ homeostasis in ferret myocar-
during ischemia. J Am Coll Cardiol. 1985;5:11381146. dium? J Clin Invest. 1994;93:12161223.
8. Gertz EW, Wisneski JA, Stanley WC, Neese RA. Myocardial substrate 30. Stanley WC, Recchia FA, Lopaschuk GD. Myocardial substrate metabo-
utilization during exercise in humans: dual carbon-labeled carbohydrate lism in the normal and failing heart. Physiol Rev. 2005;85:10931129.
isotope experiments. J Clin Invest. 1988;82:20172025. 31. Ussher JR, Jaswal JS, Lopaschuk GD. Pyridine nucleotide regulation of
9. van der Vusse GJ, van Bilsen M, Glatz JF. Cardiac fatty acid uptake and cardiac intermediary metabolism. Circ Res. 2012;111:628641.
transport in health and disease. Cardiovasc Res. 2000;45:279293. 32. Kornberg HL. The regulation of anaplerotic enzymes in E. coli. Bull Soc
10. Glatz JF, Luiken JJ, Bonen A. Involvement of membrane-associated pro- Chim Biol (Paris). 1967;49:14791490.
teins in the acute regulation of cellular fatty acid uptake. J Mol Neurosci. 33. Des Rosiers C, Labarthe F, Lloyd SG, Chatham JC. Cardiac anaplerosis in
2001;16:123132; discussion 151. health and disease: food for thought. Cardiovasc Res. 2011;90:210219.

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


722Circulation ResearchAugust 30, 2013

34. Russell RR 3rd, Taegtmeyer H. Changes in citric acid cycle flux and 61. Zhabyeyev P, Gandhi M, Mori J, Basu R, Kassiri Z, Clanachan A,

anaplerosis antedate the functional decline in isolated rat hearts utilizing Lopaschuk GD, Oudit GY. Pressure-overload-induced heart failure in-
acetoacetate. J Clin Invest. 1991;87:384390. duces a selective reduction in glucose oxidation at physiological afterload.
35. Murphy MP. How mitochondria produce reactive oxygen species. Biochem Cardiovasc Res. 2013;97:676685.
J. 2009;417:113. 62. Degens H, de Brouwer KF, Gilde AJ, Lindhout M, Willemsen PH, Janssen
36. Burgoyne JR, Mongue-Din H, Eaton P, Shah AM. Redox signaling in car- BJ, van der Vusse GJ, van Bilsen M. Cardiac fatty acid metabolism is
diac physiology and pathology. Circ Res. 2012;111:10911106. preserved in the compensated hypertrophic rat heart. Basic Res Cardiol.
37. Randle PJ. Regulatory interactions between lipids and carbohydrates: the glu- 2006;101:1726.
cose fatty acid cycle after 35 years. Diabetes Metab Rev. 1998;14:263283. 63. Akki A, Smith K, Seymour AM. Compensated cardiac hypertrophy is
38. Mjos OD. Effect of free fatty acids on myocardial function and oxygen characterised by a decline in palmitate oxidation. Mol Cell Biochem.
consumption in intact dogs. J Clin Invest. 1971;50:13861389. 2008;311:215224.
39. Opie LH, Knuuti J. The adrenergic-fatty acid load in heart failure. J Am 64. Allard MF, Schnekess BO, Henning SL, English DR, Lopaschuk GD.
Coll Cardiol. 2009;54:16371646. Contribution of oxidative metabolism and glycolysis to ATP production in
40. Huss JM, Torra IP, Staels B, Gigure V, Kelly DP. Estrogen-related re- hypertrophied hearts. Am J Physiol. 1994;267:H742H750.
ceptor alpha directs peroxisome proliferator-activated receptor alpha sig- 65. Kato T, Niizuma S, Inuzuka Y, Kawashima T, Okuda J, Tamaki Y, Iwanaga
naling in the transcriptional control of energy metabolism in cardiac and Y, Narazaki M, Matsuda T, Soga T, Kita T, Kimura T, Shioi T. Analysis
skeletal muscle. Mol Cell Biol. 2004;24:90799091. of metabolic remodeling in compensated left ventricular hypertrophy and
41. Wende AR, Huss JM, Schaeffer PJ, Gigure V, Kelly DP. PGC-1alpha heart failure. Circ Heart Fail. 2010;3:420430.
coactivates PDK4 gene expression via the orphan nuclear receptor 66. Christe ME, Rodgers RL. Altered glucose and fatty acid oxidation

ERRalpha: a mechanism for transcriptional control of muscle glucose me- in hearts of the spontaneously hypertensive rat. J Mol Cell Cardiol.
tabolism. Mol Cell Biol. 2005;25:1068410694. 1994;26:13711375.
42. Rowe GC, Jiang A, Arany Z. PGC-1 coactivators in cardiac development 67. Dodd MS, Ball DR, Schroeder MA, Le Page LM, Atherton HJ, Heather
and disease. Circ Res. 2010;107:825838. LC, Seymour AM, Ashrafian H, Watkins H, Clarke K, Tyler DJ. In vivo
43. Riehle C, Wende AR, Zaha VG, et al. PGC-1beta deficiency accelerates alterations in cardiac metabolism and function in the spontaneously hyper-
the transition to heart failure in pressure overload hypertrophy. Cir Res. tensive rat heart. Cardiovasc Res. 2012;95:6976.
2011;109:783793 68. Rosenblatt-Velin N, Montessuit C, Papageorgiou I, Terrand J, Lerch R.
44. Huss JM, Kelly DP. Mitochondrial energy metabolism in heart failure: a Postinfarction heart failure in rats is associated with upregulation of GLUT-
question of balance. J Clin Invest. 2005;115:547555. 1 and downregulation of genes of fatty acid metabolism. Cardiovasc Res.
45. Zaha VG, Young LH. AMP-activated protein kinase regulation and bio- 2001;52:407416.
logical actions in the heart. Circ Res. 2012;111:800814. 69. Amorim PA, Nguyen TD, Shingu Y, Schwarzer M, Mohr FW, Schrepper
A, Doenst T. Myocardial infarction in rats causes partial impairment in in-
46. Cai L, Tu BP. Driving the cell cycle through metabolism. Annu Rev Cell
sulin response associated with reduced fatty acid oxidation and mitochon-
Dev Biol. 2012;28:5987.
drial gene expression. J Thorac Cardiovasc Surg. 2010;140:11601167.
47. Ward PS, Thompson CB. Signaling in control of cell growth and metabo-
70. Heather LC, Cole MA, Lygate CA, Evans RD, Stuckey DJ, Murray

lism. Cold Spring Harb Perspect Biol. 2012;4:a006783.
AJ, Neubauer S, Clarke K. Fatty acid transporter levels and palmitate
48. Hihi AK, Michalik L, Wahli W. PPARs: transcriptional effectors of fatty
oxidation rate correlate with ejection fraction in the infarcted rat heart.
acids and their derivatives. Cell Mol Life Sci. 2002;59:790798.
Cardiovasc Res. 2006;72:430437.
49. Shipston MJ. Ion channel regulation by protein palmitoylation. J Biol
71. Osorio JC, Stanley WC, Linke A, Castellari M, Diep QN, Panchal AR,
Chem. 2011;286:87098716.
Hintze TH, Lopaschuk GD, Recchia FA. Impaired myocardial fatty acid
50. Spedding M, Mir AK. Direct activation of Ca2+ channels by palmitoyl car-
oxidation and reduced protein expression of retinoid X receptor-alpha in
nitine, a putative endogenous ligand. Br J Pharmacol. 1987;92:457468.
pacing-induced heart failure. Circulation. 2002;106:606612.
51. Wang G, Silva J, Krishnamurthy K, Tran E, Condie BG, Bieberich E.
72. Sack MN, Rader TA, Park S, Bastin J, McCune SA, Kelly DP. Fatty acid
Direct binding to ceramide activates protein kinase Czeta before the for-
oxidation enzyme gene expression is downregulated in the failing heart.
mation of a pro-apoptotic complex with PAR-4 in differentiating stem
Circulation. 1996;94:28372842.
cells. J Biol Chem. 2005;280:2641526424. 73. Dvila-Romn VG, Vedala G, Herrero P, de las Fuentes L, Rogers JG, Kelly
52. Wilson L, Yang Q, Szustakowski JD, Gullicksen PS, Halse R. Pyruvate DP, Gropler RJ. Altered myocardial fatty acid and glucose metabolism in
induces mitochondrial biogenesis by a PGC-1 alpha-independent mecha- idiopathic dilated cardiomyopathy. J Am Coll Cardiol. 2002;40:271277.
nism. Am J Physiol Cell Physiol. 2007;292:C1599C1605. 74. Dai DF, Hsieh EJ, Liu Y, Chen T, Beyer RP, Chin MT, MacCoss MJ,
53. Philp A, Perez-Schindler J, Green C, Hamilton DL, Baar K. Pyruvate sup- Rabinovitch PS. Mitochondrial proteome remodelling in pressure over-
presses PGC1alpha expression and substrate utilization despite increased load-induced heart failure: the role of mitochondrial oxidative stress.
respiratory chain content in C2C12 myotubes. Am J Physiol Cell Physiol. Cardiovasc Res. 2012;93:7988.
2010;299:C240C250. 75. Kolwicz SC Jr, Olson DP, Marney LC, Garcia-Menendez L, Synovec RE,
54. Cai L, Sutter BM, Li B, Tu BP. Acetyl-CoA induces cell growth and prolif- Tian R. Cardiac-specific deletion of acetyl CoA carboxylase 2 prevents
eration by promoting the acetylation of histones at growth genes. Mol Cell. metabolic remodeling during pressure-overload hypertrophy. Circ Res.
2011;42:426437. 2012;111:728738.
55. Ngoh GA, Facundo HT, Zafir A, Jones SP. O-GlcNAc signaling in the 76. Atherton HJ, Dodd MS, Heather LC, Schroeder MA, Griffin JL, Radda
cardiovascular system. Circ Res. 2010;107:171185. GK, Clarke K, Tyler DJ. Role of pyruvate dehydrogenase inhibition in
56. Bulua AC, Simon A, Maddipati R, Pelletier M, Park H, Kim KY, Sack MN, the development of hypertrophy in the hyperthyroid rat heart: a combined
Kastner DL, Siegel RM. Mitochondrial reactive oxygen species promote magnetic resonance imaging and hyperpolarized magnetic resonance
production of proinflammatory cytokines and are elevated in TNFR1- spectroscopy study. Circulation. 2011;123:25522561.
associated periodic syndrome (TRAPS). J Exp Med. 2011;208:519533. 77. Pound KM, Sorokina N, Ballal K, Berkich DA, Fasano M, Lanoue KF,
57. Chua B, Siehl DL, Morgan HE. Effect of leucine and metabolites of Taegtmeyer H, ODonnell JM, Lewandowski ED. Substrate-enzyme com-
branched chain amino acids on protein turnover in heart. J Biol Chem. petition attenuates upregulated anaplerotic flux through malic enzyme in
1979;254:83588362. hypertrophied rat heart and restores triacylglyceride content: attenuating
58. Huang Y, Zhou M, Sun H, Wang Y. Branched-chain amino acid metabo- upregulated anaplerosis in hypertrophy. Circ Res. 2009;104:805812.
lism in heart disease: an epiphenomenon or a real culprit? Cardiovasc Res. 78. Garnier A, Fortin D, Delomnie C, Momken I, Veksler V, Ventura-Clapier
2011;90:220223. R. Depressed mitochondrial transcription factors and oxidative capacity in
59. Doenst T, Pytel G, Schrepper A, Amorim P, Frber G, Shingu Y, Mohr rat failing cardiac and skeletal muscles. J Physiol. 2003;551:491501.
FW, Schwarzer M. Decreased rates of substrate oxidation ex vivo predict 79. Arany Z, Novikov M, Chin S, Ma Y, Rosenzweig A, Spiegelman BM.
the onset of heart failure and contractile dysfunction in rats with pressure Transverse aortic constriction leads to accelerated heart failure in mice
overload. Cardiovasc Res. 2010;86:461470. lacking PPAR-gamma coactivator 1alpha. Proc Natl Acad Sci U S A.
60. Bugger H, Schwarzer M, Chen D, Schrepper A, Amorim PA, Schoepe 2006;103:1008610091.
M, Nguyen TD, Mohr FW, Khalimonchuk O, Weimer BC, Doenst T. 80. Karamanlidis G, Nascimben L, Couper GS, Shekar PS, del Monte F, Tian
Proteomic remodelling of mitochondrial oxidative pathways in pressure R. Defective DNA replication impairs mitochondrial biogenesis in human
overload-induced heart failure. Cardiovasc Res. 2010;85:376384. failing hearts. Circ Res. 2010;106:15411548.

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


Doenst et al Cardiac Metabolism in Heart Failure 723

81. Dai DF, Johnson SC, Villarin JJ, Chin MT, Nieves-Cintrn M, Chen T, 101. Gupte SA, Levine RJ, Gupte RS, Young ME, Lionetti V, Labinskyy V,
Marcinek DJ, Dorn GW 2nd, Kang YJ, Prolla TA, Santana LF, Rabinovitch Floyd BC, Ojaimi C, Bellomo M, Wolin MS, Recchia FA. Glucose-6-
PS. Mitochondrial oxidative stress mediates angiotensin II-induced car- phosphate dehydrogenase-derived NADPH fuels superoxide production
diac hypertrophy and Galphaq overexpression-induced heart failure. Circ in the failing heart. J Mol Cell Cardiol. 2006;41:340349.
Res. 2011;108:837846. 102. Hecker PA, Lionetti V, Ribeiro RF Jr, Rastogi S, Brown BH, OConnell
82. Faerber G, Barreto-Perreia F, Schoepe M, Gilsbach R, Schrepper A, KA, Cox JW, Shekar KC, Gamble DM, Sabbah HN, Leopold JA, Gupte
Schwarzer M, Mohr FW, Hein L, Doenst T. Induction of heart failure SA, Recchia FA, Stanley WC. Glucose 6-phosphate dehydrogenase de-
by minimally invasive aortic constriction in mice: reduced peroxi- ficiency increases redox stress and moderately accelerates the develop-
some proliferator-activated receptor gamma coactivator levels and ment of heart failure. Circ Heart Fail. 2013;6:118126.
mitochondrial dysfunction. J Thorac Cardiovasc Surg. 2011;141: 103. Rajamani U, Essop MF. Hyperglycemia-mediated activation of the hex-
492500, 500.e1. osamine biosynthetic pathway results in myocardial apoptosis. Am J
83. Gundewar S, Calvert JW, Jha S, Toedt-Pingel I, Ji SY, Nunez D,
Physiol Cell Physiol. 2010;299:C139C147.
Ramachandran A, Anaya-Cisneros M, Tian R, Lefer DJ. Activation of 104. Makino A, Suarez J, Gawlowski T, Han W, Wang H, Scott BT, Dillmann
AMP-activated protein kinase by metformin improves left ventricular WH. Regulation of mitochondrial morphology and function by
function and survival in heart failure. Circ Res. 2009;104:403411. O-GlcNAcylation in neonatal cardiac myocytes. Am J Physiol Regul
84. Rosca MG, Tandler B, Hoppel CL. Mitochondria in cardiac hypertrophy Integr Comp Physiol. 2011;300:R1296R1302.
and heart failure. J Mol Cell Cardiol. 2013;55:3141. 105. Clark RJ, McDonough PM, Swanson E, Trost SU, Suzuki M, Fukuda
85. Lamb HJ, Beyerbacht HP, van der Laarse A, Stoel BC, Doornbos J, van M, Dillmann WH. Diabetes and the accompanying hyperglycemia
der Wall EE, de Roos A. Diastolic dysfunction in hypertensive heart impairs cardiomyocyte calcium cycling through increased nuclear
disease is associated with altered myocardial metabolism. Circulation. O-GlcNAcylation. J Biol Chem. 2003;278:4423044237.
1999;99:22612267. 106. Lunde IG, Aronsen JM, Kvaly H, Qvigstad E, Sjaastad I, Tnnessen
86. Okada M, Mitsunami K, Inubushi T, Kinoshita M. Influence of aging or T, Christensen G, Grnning-Wang LM, Carlson CR. Cardiac O-GlcNAc
left ventricular hypertrophy on the human heart: contents of phosphorus signaling is increased in hypertrophy and heart failure. Physiol Genomics.
metabolites measured by 31P MRS. Magn Reson Med. 1998;39:772782. 2012;44:162172.
87. Hirsch GA, Bottomley PA, Gerstenblith G, Weiss RG. Allopurinol acutely 107. Du XL, Edelstein D, Rossetti L, Fantus IG, Goldberg H, Ziyadeh F, Wu
increases adenosine triphospate energy delivery in failing human hearts. J, Brownlee M. Hyperglycemia-induced mitochondrial superoxide over-
J Am Coll Cardiol. 2012;59:802808. production activates the hexosamine pathway and induces plasminogen
88. Struthers AD, Donnan PT, Lindsay P, McNaughton D, Broomhall J,
activator inhibitor-1 expression by increasing Sp1 glycosylation. Proc
MacDonald TM. Effect of allopurinol on mortality and hospitalisa- Natl Acad Sci U S A. 2000;97:1222212226.
tions in chronic heart failure: a retrospective cohort study. Heart. 108. Facundo HT, Brainard RE, Watson LJ, Ngoh GA, Hamid T, Prabhu SD,
2002;87:229234. Jones SP. O-GlcNAc signaling is essential for NFAT-mediated transcrip-
89. Neubauer S, Horn M, Cramer M, Harre K, Newell JB, Peters W, Pabst T, tional reprogramming during cardiomyocyte hypertrophy. Am J Physiol
Ertl G, Hahn D, Ingwall JS, Kochsiek K. Myocardial phosphocreatine- Heart Circ Physiol. 2012;302:H2122H2130.
to-ATP ratio is a predictor of mortality in patients with dilated cardiomy- 109. Wilkins BJ, Dai YS, Bueno OF, Parsons SA, Xu J, Plank DM, Jones
opathy. Circulation. 1997;96:21902196. F, Kimball TR, Molkentin JD. Calcineurin/NFAT coupling participates
90. Dez J, Gonzlez A, Lpez B, Querejeta R. Mechanisms of disease: patho- in pathological, but not physiological, cardiac hypertrophy. Circ Res.
logic structural remodeling is more than adaptive hypertrophy in hyper- 2004;94:110118.
tensive heart disease. Nat Clin Pract Cardiovasc Med. 2005;2:209216. 110. Watson LJ, Facundo HT, Ngoh GA, Ameen M, Brainard RE, Lemma
91. Marks AR. Calcium cycling proteins and heart failure: mechanisms and KM, Long BW, Prabhu SD, Xuan YT, Jones SP. O-linked -N-
therapeutics. J Clin Invest. 2013;123:4652. acetylglucosamine transferase is indispensable in the failing heart. Proc
92. Hamdani N, Kooij V, van Dijk S, Merkus D, Paulus WJ, Remedios CD, Natl Acad Sci U S A. 2010;107:1779717802.
Duncker DJ, Stienen GJ, van der Velden J. Sarcomeric dysfunction in 111. Rabinowitz JD, White E. Autophagy and metabolism. Science.

heart failure. Cardiovasc Res. 2008;77:649658. 2010;330:13441348.
93. Yusuf S, Dagenais G, Pogue J, Bosch J, Sleight P. Vitamin E supple- 112. Rothermel BA, Hill JA. Autophagy in load-induced heart disease. Circ
mentation and cardiovascular events in high-risk patients: the Heart Res. 2008;103:13631369.
Outcomes Prevention Evaluation Study Investigators. N Engl J Med. 113. Kanamori H, Takemura G, Goto K, Maruyama R, Tsujimoto A, Ogino
2000;342:154160. A, Takeyama T, Kawaguchi T, Watanabe T, Fujiwara T, Fujiwara H,
94. Lonn E, Bosch J, Yusuf S, Sheridan P, Pogue J, Arnold JM, Ross C, Seishima M, Minatoguchi S. The role of autophagy emerging in postin-
Arnold A, Sleight P, Probstfield J, Dagenais GR; HOPE and HOPE-TOO farction cardiac remodelling. Cardiovasc Res. 2011;91:330339.
Trial Investigators. Effects of long-term vitamin E supplementation on 114. Tian R, Musi N, DAgostino J, Hirshman MF, Goodyear LJ. Increased
cardiovascular events and cancer: a randomized controlled trial. JAMA. adenosine monophosphate-activated protein kinase activity in rat hearts
2005;293:13381347. with pressure-overload hypertrophy. Circulation. 2001;104:16641669.
95. Dai DF, Chen T, Szeto H, Nieves-Cintrn M, Kutyavin V, Santana LF, 115. Huang J, Lam GY, Brumell JH. Autophagy signaling through reactive
Rabinovitch PS. Mitochondrial targeted antioxidant peptide ameliorates oxygen species. Antioxid Redox Signal. 2011;14:22152231.
hypertensive cardiomyopathy. J Am Coll Cardiol. 2011;58:7382. 116. Cao DJ, Wang ZV, Battiprolu PK, Jiang N, Morales CR, Kong Y,

96. Inoguchi T, Li P, Umeda F, Yu HY, Kakimoto M, Imamura M, Aoki T, Rothermel BA, Gillette TG, Hill JA. Histone deacetylase (HDAC) in-
Etoh T, Hashimoto T, Naruse M, Sano H, Utsumi H, Nawata H. High glu- hibitors attenuate cardiac hypertrophy by suppressing autophagy. Proc
cose level and free fatty acid stimulate reactive oxygen species produc- Natl Acad Sci U S A. 2011;108:41234128.
tion through protein kinase Cdependent activation of NAD(P)H oxidase 117. Depre C, Wang Q, Yan L, Hedhli N, Peter P, Chen L, Hong C, Hittinger
in cultured vascular cells. Diabetes. 2000;49:19391945. L, Ghaleh B, Sadoshima J, Vatner DE, Vatner SF, Madura K. Activation
97. Schnfeld P, Wojtczak L. Fatty acids decrease mitochondrial genera- of the cardiac proteasome during pressure overload promotes ventricular
tion of reactive oxygen species at the reverse electron transport but in- hypertrophy. Circulation. 2006;114:18211828.
crease it at the forward transport. Biochim Biophys Acta. 2007;1767: 118. Hedhli N, Depre C. Proteasome inhibitors and cardiac cell growth.

10321040. Cardiovasc Res. 2010;85:321329.
98. Boudina S, Abel ED. Diabetic cardiomyopathy revisited. Circulation. 119. Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya
2007;115:32133223. S, Mizote I, Matsumura Y, Asahi M, Nishida K, Hori M, Mizushima N,
99. Sharma S, Adrogue JV, Golfman L, Uray I, Lemm J, Youker K, Noon Otsu K. The role of autophagy in cardiomyocytes in the basal state and in
GP, Frazier OH, Taegtmeyer H. Intramyocardial lipid accumulation response to hemodynamic stress. Nat Med. 2007;13:619624.
in the failing human heart resembles the lipotoxic rat heart. FASEB J. 120. Zhu H, Tannous P, Johnstone JL, Kong Y, Shelton JM, Richardson JA, Le
2004;18:16921700. V, Levine B, Rothermel BA, Hill JA. Cardiac autophagy is a maladaptive
100. Krishnan J, Suter M, Windak R, Krebs T, Felley A, Montessuit C, response to hemodynamic stress. J Clin Invest. 2007;117:17821793.
Tokarska-Schlattner M, Aasum E, Bogdanova A, Perriard E, Perriard JC, 121. Oka T, Hikoso S, Yamaguchi O, Taneike M, Takeda T, Tamai T, Oyabu J,
Larsen T, Pedrazzini T, Krek W. Activation of a HIF1alpha-PPARgamma Murakawa T, Nakayama H, Nishida K, Akira S, Yamamoto A, Komuro I,
axis underlies the integration of glycolytic and lipid anabolic pathways in Otsu K. Mitochondrial DNA that escapes from autophagy causes inflam-
pathologic cardiac hypertrophy. Cell Metab. 2009;9:512524. mation and heart failure. Nature. 2012;485:251255.

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


724Circulation ResearchAugust 30, 2013

122. Lionetti V, Stanley WC, Recchia FA. Modulating fatty acid oxidation in protein kinase inhibition of acetyl-CoA carboxylase. J Biol Chem.
heart failure. Cardiovasc Res. 2011;90:202209. 1995;270:1751317520.
123. Stanley WC, Dabkowski ER, Ribeiro RF Jr, OConnell KA. Dietary fat 139. Jger S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated pro-
and heart failure: moving from lipotoxicity to lipoprotection. Circ Res. tein kinase (AMPK) action in skeletal muscle via direct phosphorylation
2012;110:764776. of PGC-1alpha. Proc Natl Acad Sci U S A. 2007;104:1201712022.
124. Raher MJ, Thibault HB, Buys ES, Kuruppu D, Shimizu N, Brownell AL, 140. McGaffin KR, Witham WG, Yester KA, Romano LC, ODoherty RM,
Blake SL, Rieusset J, Kaneki M, Derumeaux G, Picard MH, Bloch KD, McTiernan CF, ODonnell CP. Cardiac-specific leptin receptor deletion ex-
Scherrer-Crosbie M. A short duration of high-fat diet induces insulin resis- acerbates ischaemic heart failure in mice. Cardiovasc Res. 2011;89:6071.
tance and predisposes to adverse left ventricular remodeling after pressure 141. Li XN, Song J, Zhang L, LeMaire SA, Hou X, Zhang C, Coselli JS, Chen
overload. Am J Physiol Heart Circ Physiol. 2008;295:H2495H2502. L, Wang XL, Zhang Y, Shen YH. Activation of the AMPK-FOXO3 path-
125. Okere IC, Chess DJ, McElfresh TA, Johnson J, Rennison J, Ernsberger way reduces fatty acid-induced increase in intracellular reactive oxygen
P, Hoit BD, Chandler MP, Stanley WC. High-fat diet prevents cardiac species by upregulating thioredoxin. Diabetes. 2009;58:22462257.
hypertrophy and improves contractile function in the hypertensive Dahl 142. Newman MF, Ferguson TB, White JA, Ambrosio G, Koglin J, Nussmeier
salt-sensitive rat. Clin Exp Pharmacol Physiol. 2005;32:825831. NA, Pearl RG, Pitt B, Wechsler AS, Weisel RD, Reece TL, Lira A,
126. Tuunanen H, Engblom E, Naum A, Ngren K, Hesse B, Airaksinen KE, Harrington RA; RED-CABG Steering Committee and Investigators.
Nuutila P, Iozzo P, Ukkonen H, Opie LH, Knuuti J. Free fatty acid deple- Effect of adenosine-regulating agent acadesine on morbidity and mor-
tion acutely decreases cardiac work and efficiency in cardiomyopathic tality associated with coronary artery bypass grafting: the RED-CABG
heart failure. Circulation. 2006;114:21302137. randomized controlled trial. JAMA. 2012;308:157164.
127. Jaswal JS, Keung W, Wang W, Ussher JR, Lopaschuk GD. Targeting fatty 143. Cittadini A, Napoli R, Monti MG, Rea D, Longobardi S, Netti PA, Walser
acid and carbohydrate oxidationa novel therapeutic intervention in the M, Sam M, Aimaretti G, Isgaard J, Sacc L. Metformin prevents the
ischemic and failing heart. Biochim Biophys Acta. 2011;1813:13331350. development of chronic heart failure in the SHHF rat model. Diabetes.
128. Liao R, Jain M, Cui L, DAgostino J, Aiello F, Luptak I, Ngoy S, 2012;61:944953.
Mortensen RM, Tian R. Cardiac-specific overexpression of GLUT1 pre- 144. Aguilar D, Chan W, Bozkurt B, Ramasubbu K, Deswal A. Metformin use
vents the development of heart failure attributable to pressure overload in and mortality in ambulatory patients with diabetes and heart failure. Circ
mice. Circulation. 2002;106:21252131. Heart Fail. 2011;4:5358.
145. El-Mir MY, Nogueira V, Fontaine E, Avret N, Rigoulet M, Leverve X.
129. Liu Q, Docherty JC, Rendell JC, Clanachan AS, Lopaschuk GD. High
Dimethylbiguanide inhibits cell respiration via an indirect effect targeted
levels of fatty acids delay the recovery of intracellular pH and cardiac
on the respiratory chain complex I. J Biol Chem. 2000;275:223228.
efficiency in post-ischemic hearts by inhibiting glucose oxidation. J Am
146. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre
Coll Cardiol. 2002;39:718725.
J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ, Moller DE.
130. Ussher JR, Wang W, Gandhi M, Keung W, Samokhvalov V, Oka T, Wagg
Role of AMP-activated protein kinase in mechanism of metformin ac-
CS, Jaswal JS, Harris RA, Clanachan AS, Dyck JR, Lopaschuk GD.
tion. J Clin Invest. 2001;108:11671174.
Stimulation of glucose oxidation protects against acute myocardial in-
147. Ouyang J, Parakhia RA, Ochs RS. Metformin activates AMP kinase
farction and reperfusion injury. Cardiovasc Res. 2012;94:359369.
through inhibition of AMP deaminase. J Biol Chem. 2011;286:111.
131. Bersin RM, Wolfe C, Kwasman M, Lau D, Klinski C, Tanaka K,

148. Foretz M, Hbrard S, Leclerc J, Zarrinpashneh E, Soty M, Mithieux G,
Khorrami P, Henderson GN, de Marco T, Chatterjee K. Improved hemo-
Sakamoto K, Andreelli F, Viollet B. Metformin inhibits hepatic gluco-
dynamic function and mechanical efficiency in congestive heart failure neogenesis in mice independently of the LKB1/AMPK pathway via a
with sodium dichloroacetate. J Am Coll Cardiol. 1994;23:16171624. decrease in hepatic energy state. J Clin Invest. 2010;120:23552369.
132. Lewis JF, DaCosta M, Wargowich T, Stacpoole P. Effects of dichlo- 149. Xiao H, Ma X, Feng W, Fu Y, Lu Z, Xu M, Shen Q, Zhu Y, Zhang Y.
roacetate in patients with congestive heart failure. Clin Cardiol. Metformin attenuates cardiac fibrosis by inhibiting the TGFbeta1-Smad3
1998;21:888892. signalling pathway. Cardiovasc Res. 2010;87:504513.
133. Pfeffer G, Majamaa K, Turnbull DM, Thorburn D, Chinnery PF.
150. Drucker DJ. The biology of incretin hormones. Cell Metab.

Treatment for mitochondrial disorders. Cochrane Database Syst Rev. 2006;3:153165.
2012;4:CD004426. 151. Ravassa S, Zudaire A, Dez J. GLP-1 and cardioprotection: from bench to
134. Nguyen TD, Shingu Y, Amorim PA, Schwarzer M, Doenst T. Triheptanoin bedside. Cardiovasc Res. 2012;94:316323.
diet alleviates hypertrophy and diastolic dysfunction and preserves car- 152. Nikolaidis LA, Elahi D, Hentosz T, Doverspike A, Huerbin R, Zourelias
diac glucose oxidation in rats with pressure overload. Clin Res Cardiol. L, Stolarski C, Shen YT, Shannon RP. Recombinant glucagon-like pep-
2013;102(suppl 1):V577. tide-1 increases myocardial glucose uptake and improves left ventricular
135. Beauloye C, Bertrand L, Horman S, Hue L. AMPK activation, a pre- performance in conscious dogs with pacing-induced dilated cardiomy-
ventive therapeutic target in the transition from cardiac injury to heart opathy. Circulation. 2004;110:955961.
failure. Cardiovasc Res. 2011;90:224233. 153. Poornima I, Brown SB, Bhashyam S, Parikh P, Bolukoglu H, Shannon
136. Russell RR 3rd, Bergeron R, Shulman GI, Young LH. Translocation of RP. Chronic glucagon-like peptide-1 infusion sustains left ventricular
myocardial GLUT-4 and increased glucose uptake through activation of systolic function and prolongs survival in the spontaneously hyperten-
AMPK by AICAR. Am J Physiol. 1999;277:H643H649. sive, heart failure-prone rat. Circ Heart Fail. 2008;1:153160.
137. Luiken JJ, Coort SL, Willems J, Coumans WA, Bonen A, van der Vusse 154. Nguyen TD, Shingu Y, Amorim PA, Schwarzer M, Doenst T. Chronic
GJ, Glatz JF. Contraction-induced fatty acid translocase/CD36 transloca- glp-1 treatment preserves diastolic function and improves survival in rats
tion in rat cardiac myocytes is mediated through AMP-activated protein with pressure overload. Eur Heart J. 2011;32:783, P4391.
kinase signaling. Diabetes. 2003;52:16271634. 155. Ban K, Noyan-Ashraf MH, Hoefer J, Bolz SS, Drucker DJ, Husain M.
138. Kudo N, Barr AJ, Barr RL, Desai S, Lopaschuk GD. High rates of fatty Cardioprotective and vasodilatory actions of glucagon-like peptide 1 re-
acid oxidation during reperfusion of ischemic hearts are associated with ceptor are mediated through both glucagon-like peptide 1 receptor-de-
a decrease in malonyl-CoA levels due to an increase in 5-AMP-activated pendent and -independent pathways. Circulation. 2008;117:23402350.

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016


Cardiac Metabolism in Heart Failure: Implications Beyond ATP Production
Torsten Doenst, Tien Dung Nguyen and E. Dale Abel

Circ Res. 2013;113:709-724


doi: 10.1161/CIRCRESAHA.113.300376
Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright 2013 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circres.ahajournals.org/content/113/6/709

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the
Editorial Office. Once the online version of the published article for which permission is being requested is
located, click Request Permissions in the middle column of the Web page under Services. Further information
about this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation Research is online at:


http://circres.ahajournals.org//subscriptions/

Downloaded from http://circres.ahajournals.org/ by guest on July 23, 2016

Potrebbero piacerti anche