Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO.

3, 2017

ª 2017 THE AUTHORS. PUBLISHED BY ELSEVIER ON BEHALF OF THE AMERICAN ISSN 2452-302X

COLLEGE OF CARDIOLOGY FOUNDATION. THIS IS AN OPEN ACCESS ARTICLE UNDER http://dx.doi.org/10.1016/j.jacbts.2016.11.009

THE CC BY-NC-ND LICENSE (http://creativecommons.org/licenses/by-nc-nd/4.0/).

STATE-OF-THE-ART REVIEW

Metabolic Origins of Heart Failure


Adam R. Wende, PHD,a Manoja K. Brahma, PHD,a Graham R. McGinnis, PHD,b Martin E. Young, DPHILb

SUMMARY

For more than half a century, metabolic perturbations have been explored in the failing myocardium, highlighting a
reversion to a more fetal-like metabolic profile (characterized by depressed fatty acid oxidation and concomitant
increased reliance on use of glucose). More recently, alterations in ketone body and amino acid/protein metabolism
have been described during heart failure, as well as mitochondrial dysfunction and perturbed metabolic signaling
(e.g., acetylation, O-GlcNAcylation). Although numerous mechanisms are likely involved, the current review provides
recent advances regarding the metabolic origins of heart failure, and their potential contribution toward contractile
dysfunction of the heart. (J Am Coll Cardiol Basic Trans Science 2017;2:297–310) © 2017 The Authors. Published by
Elsevier on behalf of the American College of Cardiology Foundation. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

I n order to meet the exceptionally high metabolic


demands of continuous contractility, the heart
catabolizes an array of substrates. Indeed, the
heart has been termed a “metabolic omnivore,”
predictable shifts in metabolism, associated with
impaired signaling (e.g., Ca2þ, reactive oxygen spe-
cies [ROS]), energy insufficiency, and contractile
dysfunction. This review highlights recent insights
capable of consuming fatty acids (FAs), glucose, ke- regarding the metabolic basis of HF; for the contribu-
tone bodies, and amino acids (AA) for the replenish- tion of perturbed Ca 2þ homeostasis and ROS
ment of ATP. Central to achievement of this goal is signaling, the reader is directed to the papers by
metabolic flexibility, wherein the heart shifts reliance Brown and Griendling (1) and Bers (2).
from one substrate to another, in response to acute
perturbations in workload and/or substrate availabil- CONTRIBUTIONS OF
ity (including feeding-fasting and sleep-wake cycles, INDIVIDUAL SUBSTRATES
which occur on a daily basis). The importance of
metabolic flexibility is underscored by appreciation FATTY ACID METABOLISM. Fatty acid oxidation
for the fact that various substrates are more than (FAO) represents a significant fuel source for the
just a fuel for the heart, serving also as building myocardium, providing an estimated 50% to 70% of
blocks for numerous cellular components (e.g., mem- the ATP consumed during contraction (3). In com-
branes, proteins), cofactors, and signaling molecules. parison with carbohydrate use, rates of cardiac FAO
During various disease states, particularly heart fail- are relatively unaffected by acute changes in work-
ure (HF), cardiac metabolism is perturbed in a chronic load or energy demand (4,5). Cardiac FAO typically
manner, resulting in metabolic inflexibility. Thus, exhibits greater flexibility following changes in
HF is characterized by relatively permanent and substrate availability (6). Such observations could

From the aDepartment of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; and the bDepartment of
Medicine, University of Alabama at Birmingham, Birmingham, Alabama. This work was supported by the U.S. National Institutes
of Health grants HL106199, HL074259, HL123574, and HL122974 to Dr. Young and HL111322 and HL133011 to Dr. Wende and
American Heart Association grant 16POST270100009 to Dr. McGinnis. All other authors have reported that they have no
relationships relevant to the contents of this paper to disclose.
All authors attest they are in compliance with human studies committees and animal welfare regulations of the authors’
institutions and Food and Drug Administration guidelines, including patient consent where appropriate. For more information,
visit the JACC: Basic to Translational Science author instructions page.

Manuscript received August 9, 2016; revised manuscript received November 11, 2016, accepted November 15, 2016.
298 Wende et al. JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017

Metabolic Origins of Heart Failure JUNE 2017:297–310

ABBREVIATIONS indicate that FAO maintains baseline energy cardiomyopathic phenotype (20). Importantly,
AND ACRONYMS needs of the heart while matching rates of FA cardiac-restricted VLCAD deletion also results in
uptake with oxidation. If true, then cardiac contractile dysfunction, illustrating the importance of
AA = amino acid
FAO deficits could potentially precipitate normal cardiac FA metabolism (21). Similarly, genetic
BCAA = branched-chain amino
contractile dysfunction through energy deletion of lipoprotein lipase (LPL) (liberates FAs
acids
impairment and/or diversion of excess FAs from circulating lipoproteins), long chain acyl-CoA
FA = fatty acid
into signaling and/or “lipotoxic” pathways. synthetase-1 (ACSL1) (activates long-chain FAs for
FAO = fatty acid oxidation
This section reviews evidence supporting metabolism), and adipose triglyceride lipase (ATGL)
GCN2 = general control
these concepts. (liberates FAs from intracellular triglyceride stores)
nonderepressible 2
One of the most consistent metabolic per- result in concomitant decreases in cardiac FAO and
GLOX = glucose oxidation
turbations during HF is decreased use of FA, contractile function (22–24). It is noteworthy, how-
HBP = hexosamine
biosynthesis pathway
which has been observed in both animal and ever, that genetic mutations resulting in decreased
human studies (7–11). In doing so, the failing cardiac FAO do not always result in contractile
HF = heart failure
heart reverts to a fetal-like metabolic pro- dysfunction. For example, knockout of CD36 (FA
IR = insulin resistance
gram, reflected by a repression of various transporter) or PPAR-a /PGC-1a (transcription factors
LV = left ventricle
genes encoding core FAO pathway proteins promoting FAO/mitochondrial metabolism) results in
MI = myocardial infarction
(e.g., medium chain acyl-coenzyme A [CoA] decreased FAO without effects on basal contractility
PTM = post-translational
dehydrogenase, beta-hydroxyacyl-CoA dehy- (25–27). Possible explanations for the latter discrep-
modification
drogenase) and their upstream regulators ancies are that FAO is only modestly impaired; suffi-
ROS = reactive oxygen species
(e.g., PPAR- a , RXR-a , PGC-1 a ) (12,13). It has cient compensation from alternative substrate use
TauT = taurine transporter
been proposed that, acutely, this metabolic occurs; diversion of FAs species into lipotoxic path-
perturbation serves as an adaptation by promoting ways is limited; and/or a secondary stress is required
increased reliance on more energy-efficient fuels (in to elicit dysfunction (e.g., pressure overload, high-fat
terms of ATP per oxygen molecule consumed), which diet, and so forth).
may be particularly important in the setting of If acquired deficiencies in cardiac FAO were to
ischemic heart disease (14). Consistent with this contribute significantly to contractile dysfunction of
concept, attenuation of FAO is observed prior to the the failing myocardium, then normalization of the
onset of contractile dysfunction (e.g., induced by FAO deficit would be predicted to improve contrac-
pressure overload) (15). However, this metabolic tility. Both genetic and dietary strategies have been
reprogramming causes chronic dyssynchrony between used to address this concept. An important example
energy demand (which is increased), substrate avail- includes the study by Kolwicz et al. (28), wherein
ability (circulating FAs are typically increased), and selective deletion of acetyl-CoA carboxylase 2, an
use (i.e., FAO is decreased) during HF. In other words, enzyme that generates malonyl-CoA (a potent inhib-
a decrease in FAO rates could reduce ATP availability itor of b-oxidation), prevents pressure overload-
for contraction (if below the capacity of alternative induced depression of FAO and concomitantly main-
compensating pathways) concomitant with increased tains contractile function. Interestingly, feeding ro-
diversion of FA species into signaling/lipotoxic path- dents calorie-dense high-fat diets has been shown to
ways, culminating in impairment of contractility. Ev- preserve and even improve contractility in distinct
idence in support of this concept includes reports of models of HF (including pressure overload, myocar-
modest perturbations in markers of energy status in dial infarction, and hypertension), although not all
the failing myocardium, as well as accumulation of studies report this benefit (perhaps due to differences
lipotoxic markers (16–18). The latter, when elevated, in dietary composition, duration of feeding, and other
can contribute to cell death and cardiac remodeling. factors) (29–33). Observations such as these raise the
Energy deficiency versus l i p o t o x i c i t y . Both question of whether cardiac FAO impairment pri-
genetic and pharmacologic approaches have been marily leads to energy deficiency as opposed to lip-
used to address causal relationships between FAO otoxicity and signaling imbalance. However,
impairment and HF. Genetic studies revealed that strategies designed to cause a mismatch between FA
inborne errors of FAO, such as inherited deficiencies uptake and FAO (e.g., overexpression of FATP-1 or
in acyl-CoA dehydrogenases, can be associated with ACSL-1) invariably result in cardiomyopathy associ-
cardiomyopathy in humans; similar pathologies are ated with markers of lipotoxicity (34,35). In addition,
often recapitulated through targeted genetic manip- the failing heart is considered to be in a pro-lipotoxic
ulation in mouse models (19). One example includes environment (36). Furthermore, haploinsufficiency of
very-long-chain acyl-CoA dehydrogenase (VLCAD); mCPT1 increases susceptibility to pressure overload-
germline deletion results in energy impairment and a induced cardiac dysfunction through lipotoxic
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017 Wende et al. 299
JUNE 2017:297–310 Metabolic Origins of Heart Failure

pathways (37). Similarly, germline VLCAD deletion GLUCOSE METABOLISM. Use of cardiac glucose (i.e.,
increases cardiac lipotoxicity during high-fat feeding glycolysis and glucose oxidation [GLOX]), is impor-
(38). Collectively, these studies suggest that impaired tant for the developing (fetal) heart, during which
cardiac FAO could lead to cardiac dysfunction not time glucose delivery is high, and oxygen availability
only through energy deficiency but also through is relatively low (45). Soon after birth, use of glucose
lipotoxicity. decreases concomitant with increased dietary FA and
M e d i a t o r s o f d e p r e s s e d F A O d u r i n g H F . Various oxygen delivery. However, the healthy adult heart
mechanisms have been proposed as mediators of car- has the capacity to increase reliance on use of glucose
diac FAO impairment during HF. These include in response to both physiologic (e.g., exercise) and
transcription-based mechanisms, post-translational pathologic (e.g., ischemia) stresses. During HF,
modifications (PTMs), mitochondrial dysfunction, metabolic flexibility is lost, which may be due in part
cofactor availability, and substrate competition. With to cardiac insulin resistance (IR); complex alterations
regard to transcription-based mechanisms, particular in insulin signaling within cardiomyocytes during HF
attention has been given to multiple PPAR isoforms have been reviewed recently (46). It has been sug-
(particularly PPAR-a ); upon complex formation with gested that, under some conditions (e.g., hemody-
RXR-a , FAs (ligand), and coactivators (e.g., PGC-1 a ), namic stress), IR may actually protect the heart by
PPAR-a induces a number of genes encoding known FA reducing fuel toxicity (47). However, as HF pro-
transporters and core b-oxidation enzymes (13). gresses, an uncoupling between glycolysis and GLOX
Importantly, during HF, cardiac levels of PPAR-a , RXR- ensues, potentially contributing to cellular dysfunc-
a, and PGC-1a have been shown to decrease, to varying tion (48). Interestingly, circulating glucose levels
degrees, associated with decreased expression of tend to be elevated during both acute and chronic HF.
target genes (8,12). Furthermore, PPAR-a activity may For example, elevated serum glucose levels at the
be repressed even more through PTMs (39). The time of hospital admission for acute HF syndromes,
enzymes involved in FA metabolism also undergo independent of diabetes status, are associated with
PTMs during HF, such as acetylation; this PTM poten- higher mortality (49,50). Chronically, HF is also
tially promotes FAO in the heart both directly (i.e., associated with peripheral IR. Whether these pertur-
acetylation and activation of b -oxidation enzymes) bations in cardiac and/or whole-body glucose ho-
and indirectly (i.e., acetylation and inhibition of meostasis contribute to the pathogenesis of HF is still
pyruvate dehydrogenase, and therefore impairment of under debate (46). The purpose of this subsection
glucose oxidation) (40,41). It is noteworthy that car- (Figure 1) is to review current knowledge regarding
diac FAO capacity is extremely high, such that the use of glucose during HF; we will focus primarily
enzymes involved in b -oxidation must be inhibited on this independent of diabetes status, as the latter
markedly in order to impact FAO flux. This is exem- has been reviewed extensively elsewhere (51–53).
plified by VLCAD and CPT1b heterozygous knockout Whether perturbations in use of cardiac glucose are
mouse hearts, which exhibit no baseline phenotype adaptive or maladaptive during HF appears to depend
despite a 50% loss in enzymatic activity (21,37); on the underlying stress (i.e., ischemic versus non-
homozygous cardiomyocyte-specific knockout of ischemic), as well as duration (i.e., acute versus
VLCAD also has no significant effects on cardiac FAO chronic). In the context of hypertension-induced
rates, likely due to compensation by other acyl-CoA dilated cardiomyopathy, glucose use is increased,
dehydrogenase isoforms (e.g., LCAD) (21). In contrast, with a predominate augmentation of glucose uptake
cardiac FAO is affected by cofactor availability (e.g., and glycolysis, and a concomitant uncoupling of
carnitine, CoA) and/or mitochondrial function, which glycolytic flux from GLOX (48,54). There is mounting
is decreased in the failing myocardium (42–44); evidence that the remodeling of glucose metabolism
decreased carnitine would attenuate mitochondrial FA is one of the initial changes driving the heart to hy-
uptake, and decreased CoA would attenuate b-oxida- pertrophy and could act as an early marker of disease
tion, whereas limited mitochondrial electron transfer progression (55). Acutely enhancing glucose meta-
would attenuate dehydrogenases in the b -oxidation bolism in the setting of ischemic injury and ventric-
spiral. Furthermore, increased reliance on alternative ular fibrillation may be protective (56,57).
substrates (e.g., glucose and ketone bodies, as dis- Glucose transport during H F . Regulation of
cussed below) during HF would attenuate FAO through glucose uptake into the cardiomyocyte is regulated by
established allosteric and cofactor limitation mecha- members of the solute carrier family 2A (SLC2A),
nisms. In the failing myocardium, all the above- which encode the GLUT proteins (54,58). Of the 12
referenced mechanisms likely contribute to attenu- SLC2A genes expressed in human and rodent cardiac
ated FAO. tissue, three are predominantly expressed in the
300 Wende et al. JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017

Metabolic Origins of Heart Failure JUNE 2017:297–310

F I G U R E 1 Glucose Contributions to Myocardial Dysfunction During Heart Failure

Increased glucose uptake channels carbon into the polyol, PPP, and HBP pathways; this likely contributes to mitochondrial dysfunction,
genetic reprogramming, and impaired calcium handling during heart failure. HBP ¼ hexosamine biosynthesis; PPP ¼ pentose phosphate
pathway; ROS ¼ reactive oxygen species.

myocardium: GLUT1, GLUT4, and GLUT8 (59). Of Conversely, cardiomyocyte-specific GLUT4 ablation
these, GLUT1 and GLUT4 have received extensive decreased functional recovery in response to
attention, in part due to observations that both pro- ischemic injury (66). Although loss of GLUT8 has been
teins are decreased in failing human hearts (60). Such explored in the context of diet-induced obesity (67), a
observations are consistent with repressed insulin- direct role during HF remains to be explored. Studies
mediated glucose uptake (primarily through GLUT4) of these transporters, as well as those of the other
during HF, potentially secondary to chronic activa- cardiac-enriched GLUTs (GLUT3, GLUT10, and
tion of GRK2 and IR (61). However, during HF, basal GLUT12) will continue to provide crucial insight into
rates of glucose uptake and glycolysis are elevated the contribution of glucose uptake and metabolism
(and even exceed rates of glucose oxidation) (62). One during the progression of HF.
possible explanation for this apparent disconnect is An emerging area has focused on non–GLUT-
increased GLUT translocation or activity in an insulin- mediated glucose transport through the sodium-
independent manner. A number of cardiomyocyte- glucose cotransporters (SGLT), especially given that
specific gain- and loss-of-function models have been empagliflozin (an SGLT2 inhibitor) decreased HF inci-
used in an attempt to address this question, as well as dence in diabetic patients (68). The mechanism of this
the importance of glucose use during HF. protection is likely multifaceted, including lowering of
A number of studies have interrogated the impor- both glucose and sodium, as well as influences on
tance of various GLUT isoforms in the maintenance of glomerular filtration and the cardiorenal axis (69).
cardiac function. Lifelong GLUT1 overexpression Interestingly, SGLT1 is induced in the heart during
protects against pressure overload-induced contrac- both diabetic and ischemic cardiomyopathy (70), and
tile dysfunction (63), whereas acute GLUT1 augmen- phlorizin (another SGLT inhibitor) decreased cardiac
tation partially rescues disease progression (64), glucose uptake and directly affected tolerance of the
suggesting that enhanced glucose uptake is protec- heart to ischemia (71). Future studies will undoubtedly
tive in this setting. However, cardiomyocyte-specific reveal important insights regarding the effects of SGLT
ablation of GLUT1 did not exacerbate pressure- inhibitors on cardiac metabolism and protection.
overload-induced dysfunction (perhaps due to suffi- P o l y o l p a t h w a y . Augmented glucose uptake and
cient glucose uptake by other GLUT isoforms) (65). glycolytic flux, particularly when in excess of GLOX,
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017 Wende et al. 301
JUNE 2017:297–310 Metabolic Origins of Heart Failure

enhances diverting of glucose moieties into signaling genetic (90) manipulation studies suggest that per-
pathways. This includes the polyol pathway. turbations in the use of ketone bodies can play a role
Although primarily implicated in diabetic complica- in cardiac health and disease. The heart readily uses
tions, overexpression of aldose reductase, the first ketone bodies such that their oxidation is typically
step in this pathway, results in an age-related decline increased in proportion to their delivery (91). Ace-
in heart function and exacerbated ischemic injury toacetate (AcAc) and b -hydroxybutyrate (OHB) are
(72). Further studies are warranted in order to eluci- the primary ketone bodies that can be metabolized,
date fully the importance of the polyol pathway in the which are synthesized by the liver during periods of
pathogenesis of HF. elevated FA availability, including fasting, prolonged
P e n t o s e p h o s p h a t e p a t h w a y . The pentose phos- exercise, ketogenic diets, uncontrolled type 1 dia-
phate pathway (PPP) is important for NADPH and betes, and HF (92). In the last case, multiple studies
ribose-5-phosphate generation. In a canine model of have consistently shown that HF patients with no
congestive HF, post-prandial glycemic levels were history of diabetes present with elevated levels of
sufficient to increase PPP flux. When this was pre- systemic ketone bodies (93,94). During HF, elevated
vented, cardiac GLOX and stroke work were normal- norepinephrine levels secondary to increased sym-
ized (73). Furthermore, in a genetic mouse model of pathetic outflow likely promote ketogenesis by
dilated cardiomyopathy that progressed to HF, non- increasing FA supply through lipolysis in adipose
oxidative glucose pathways such as the PPP and tissue (95). The strength of these associations is such
glycogen synthesis were increased (74). The PPP af- that exhaled acetone (indication of ketoacidosis) is a
fects ROS balance not only through NADPH but also predictive biomarker for severity of HF (96–98). It is
through regulation by pyridine nucleotides (75). noteworthy, however, that a recent study reported
Interestingly, glucose-6-phosphate dehydrogenase reduced circulating ketone body levels in HF patients
(G6PD) (first enzyme of the PPP) deficiency is associ- with reduced ejection fraction relative to that in HF
ated with cardiac disease progression; however, mice patients with preserved ejection fraction and non-HF
with G6PD deletion have shown both protective and controls (99). Severity of HF and other comorbidities
deleterious effects on cardiac function (76), suggest- associated with the patients recruited in these studies
ing the need for further study. could potentially account for these discrepancies.
Whether altered myocardial ketone body meta-
H e x o s a m i n e b i o s y n t h e s i s p a t h w a y . The hexos-
bolism contributes to the pathogenesis of HF was
amine biosynthesis pathway (HBP) requires input from
recently investigated in rodents and humans
glucose, AAs, FAs, and nucleotides, resulting in the
(100,101). These studies reported increased expres-
end product uridine diphosphate N-acetylglucos-
sion of ketone body metabolism enzymes (D-
amine (UDP-GlcNAc). This molecule in turn is used to
b-hydroxybutyrate dehydrogenase (BDH1) and succi-
regulate nearly all aspects of cell physiology through
nyl-CoA:3-oxoacid CoA transferase (SCOT), reduced
the PTM of serine and threonine residues by the addi-
intermediary metabolites of ketone body catabolism,
tion of an O-linked N-acetylglucosamine (O-GlcNAc)
as well as increased ketone body oxidation (stable
(77).This O-GlcNAc modification is elevated in both
isotope measurements) in the failing heart. An
hypertrophy and HF (78) and has both adaptive and
important question relates to whether increased use of
maladaptive contributions to cardiac function (79,80).
ketone body during HF is adaptive or maladaptive. In
Specifically, increased O-GlcNAc is protective
support of an adaptive role, cardiomyocyte-specific
following acute ischemic injury; whereas, during HF,
deletion of SCOT led to adverse cardiac remodeling
elevated O-GlcNAc may contribute to contractile and
following pressure overload (90). However, these
mitochondrial dysfunction. O-GlcNAc likely impacts a
studies do not address whether normalization of
number of cardiac processes, including transcription,
ketone body use, particularly in the setting of HF, is
epigenetics, metabolism, and Ca 2þ handling. In the last
beneficial or detrimental (as opposed to ablation
case, O-GlcNAc can regulate SERC2A, CaMKII, and
during compensated hypertrophy). Interestingly,
STIM1 (81–84), either increasing or decreasing calcium
elevated ketone body availability would be antici-
sensitivity, depending on the duration and disease
pated to repress both FAO and GLOX in the heart
state (e.g., ischemic or nonischemic).
through substrate competition, thus resulting in a
KETONE BODY METABOLISM. Compared with FA and metabolic signature reminiscent of advanced HF.
glucose metabolism, current knowledge of the role of Furthermore, ketone bodies serve as signaling
altered ketone body metabolism during HF is rela- molecules, acting through both extracellular
tively limited. However, a growing body of evidence receptors (e.g., GPR41) and intracellular inhibitors of
from pharmacological (85,86), dietary (86–89), and histone deacetylases (HDACs) (85,102), which in turn
302 Wende et al. JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017

Metabolic Origins of Heart Failure JUNE 2017:297–310

F I G U R E 2 Myocardial Ketone Body Metabolism in the Failing Heart

Elevated levels of b-OHB during heart failure provide excess acetyl-CoA for acetylation reactions and inhibit deacetylation at the same time.
Increased ketone bodies also likely compete for the oxidation of fatty acids and glucose and potentially activate cell surface receptors.
CoA ¼ coenzyme A.

could influence cardiac processes (Figure 2). yielded novel insights. This subsection (Figure 3)
The mechanisms by which ketone body uses enzymes highlights the contribution that AA metabolism per-
are induced during HF also remain unanswered. turbations potentially play in the cause of HF.
Future studies interrogating these questions are A m i n o a c i d s d u r i n g H F . Both AA availability and use
warranted, which will aid in understanding the are influenced by HF. For example, profiling plasma
role of ketone body metabolism in the pathogenesis of AAs (and their derivatives) from HF patients using
HF. high-performance liquid chromatography revealed
AMINO ACID METABOLISM. Compared with FAs and that the circulating levels of almost one-half of the
glucose, AAs quantitatively contribute to a lesser de- species assessed (17 of 41) were altered in HF, the ma-
gree to ATP generation in the heart. However, AAs play jority of which were increased (15 of 17) (103).
essential roles in myocardial function that extend Furthermore, a subset of these AAs, including gluta-
beyond energy, such as synthesis of protein, meta- mate and monoethanolamine (a serine derivative),
bolic and signaling intermediates, and cofactors. In negatively correlated with ejection fraction in HF
the last case, notable AA derivatives include L -carni- patients (with trends for phenylalanine and tyrosine as
tine (from lysine and methionine), CoQ10 (from tyro- well), suggesting higher circulating AAs were indica-
sine and mevalonate), and taurine (from methionine tive of worsening cardiac function. This is likely due to
or cysteine), which play important roles in cardiac accelerated protein breakdown in skeletal muscle,
processes (e.g., metabolism, redox biology, and which serves as an AA reservoir during HF (104).
calcium homeostasis). Furthermore, it has been esti- Despite increased demand for AAs in the heart, there is
mated that the mammalian heart renews all cellular evidence of AA accumulation in the failing myocar-
components within a 30-day period (50), illustrating a dium, as shown by metabolomic analysis of failing
significant demand on AA availability for protein and mouse myocardium accumulation of AAs, consistent
cofactor synthesis. Moreover, protein turnover is with the notion that AA catabolism was compromised
accelerated in the heart during periods of remodeling, (105). Transcriptomic analysis in mice shows that
such as hypertrophic growth. Significant efforts genes associated with AA catabolism are down-
have been made to increase our understanding of regulated during compensated hypertrophy and overt
AA metabolism perturbations during HF and have failure (105).
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017 Wende et al. 303
JUNE 2017:297–310 Metabolic Origins of Heart Failure

F I G U R E 3 Perturbations in Amino Acid Metabolism During Heart Failure

During heart failure, intracardiac branched chain amino acids are increased while taurine levels are decreased, leading to impairments in
autophagy, mitochondrial function, and calcium homeostasis. BCAA ¼ branched-chain amino acids; BCKA ¼ branched chain alpha-keto acids;
LAT ¼ large neutral amino acid transporter; mTOR ¼ mammalian target of rapamycin; TauT ¼ taurine transporter; TCA ¼ tricarboxylic acid.

Considerable interest has been placed on branched- cofactors are often found to be depleted, including
chain AA (BCAA; leucine, isoleucine, valine) meta- taurine. The importance of taurine in the heart is
bolism during HF. Branched chain alpha-keto acids supported by studies using taurine-deficient mice
(BCKA; product after initial step of BCAA catabolism) (induced by genetic ablation of the taurine transporter
are elevated within the myocardium in HF patients [TauT]) and rats (TauT inhibition with b-alanine),
(106). Furthermore, subunits of the branched chain resulting in cardiomyopathy (107). Taurine deficiency
alpha-keto acid dehydrogenase (BCKD) complex, is characterized by reduced glucose and FAO in iso-
which is responsible for subsequent catabolism of lated perfused rat hearts (108), reduced mitochondrial
BCKAs, are transcriptionally repressed. These findings complex I and III activity, and increased ROS pro-
have been replicated in mice during transaortic duction in cardiomyocytes (109). Taurine deficiency is
constriction-induced HF, where pharmacologic acti- also associated with aberrant Ca2þ homeostasis and
vation of BCKD normalized BCAA catabolism, pre- signaling, involving alterations in phospholamban
vented BCKA accumulation, and protected against and SERCA2 (110). Taurine supplementation has been
cardiac dysfunction (106). These findings suggest that shown to be efficacious during HF, eliciting improve-
an imbalance between BCAA availability and use dur- ments in left ventricular (LV) function (111) and exer-
ing HF may contribute to contractile dysfunction and cise capacity (112).
that normalization of this balance may be a novel, Various studies suggest that AA supplementation
efficacious therapeutic strategy. increases functional capacity and quality of life in pa-
Consistent with impairment of appropriate AA use tients with chronic, stable HF (113). For example,
and metabolism by the failing myocardium, various mixed AA supplementation increased functional
304 Wende et al. JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017

Metabolic Origins of Heart Failure JUNE 2017:297–310

exercise capacity (V O2 peak, exercise time during and organelles and, therefore, myocardial quality
exercise test, 6-min walk test) in humans with chronic control. While target-specific forms of autophagy
HF (114,115). Similarly, BCAA supplementation pre- exist, including mitophagy (mitochondria), glyco-
served cardiac function during high-salt-induced HF phagy (glycogen), and lipophagy (lipids), this sub-
in Dahl salt-sensitive rats (a physiological model of section focuses on macroautophagy (hereafter
hypertension leading to HF) (116). These somewhat referred to as autophagy).
counterintuitive observations (i.e., beneficial effects Observational evidence (e.g., electron microscopy)
of AA supplementation during HF, when AA avail- in humans with ischemic and dilated cardiomyopa-
ability appears to exceed capacity of the myocardium thy, as well as congestive HF, suggests that auto-
to metabolize them) may be explained by extracardiac phagy might be induced in the stressed myocardium
effects. For instance, BCAA supplementation represses (123,124). Paired sampling of cardiac tissue during
skeletal muscle cachexia (i.e., muscle wasting), and LV assist device (LVAD) implantation or explantation
previous studies suggest that the degree of cachexia is indicates autophagy markers are increased during HF
a strong independent risk factor for mortality during and are reduced following mechanical unloading
HF and significantly reduces survival (117). (125). However, difficulties with measuring autopha-
Amino acids regulate signaling during HF. Various gic flux in static samples can hinder interpretation of
AAs (and their derivatives) function as signaling observational results in clinical human studies,
molecules. This is particularly true for BCAAs, which necessitating the use of animal models. Experimental
activate the mammalian target of rapamycin evidence in mice subjected to pressure overload
(mTOR), a modulator of various anabolic (i.e., protein indicated transient activation of autophagy, which is
synthesis) and catabolic (i.e., autophagy [discussed elevated within hours of transaortic constriction in
in the next section]) pathways. Aberrant mTOR mice, returning to subbasal levels within days
signaling has been implicated in the progression of (126,127). Interestingly, diminishing myocardial
HF (118,119). In mice, mTOR is activated by pressure autophagy in cardiomyocyte-specific Beclin-1 þ/ mice
overload and pharmacologic inhibition (with rapa- partially rescued myocardial function following
mycin) improves contractile function of the decom- pressure overload. Conversely, inducing autophagy
pensated myocardium (120). However, ablation of in cardiomyocytes (through overexpression of
mTOR complex-1 signaling (through genetic deletion Beclin-1) significantly increased mortality and cardiac
of Raptor) prevents compensated hypertrophy remodeling following pressure overload (126), sug-
following pressure overload (121), resulting in a rapid gesting autophagy is maladaptive during cardiac
transition to HF and increased mortality. These stress. However, genetic disruption of myocardial
findings suggest mTOR activation may be adaptive autophagy through cardiomyocyte-specific deletion
during the initial compensated phase but maladap- of the autophagy-related gene-5 (ATG5) exacerbated
tive during overt failure. It is noteworthy that hypertrophy and remodeling during pressure over-
BCAAs also appear to affect cellular processes in an load (128). One possible explanation for these seem-
mTOR-independent manner, potentially through ingly opposing observations is related to the manner
eukaryotic initiation factor-2-alpha (eIF2- a ) kinase in which autophagy is disrupted: if inhibited later in
general control nonderepressible-2 (GCN2). GCN2 is the process (as opposed to initiated), autophago-
activated by noncharged tRNA during AA starvation somes will accumulate within the myocardium, thus
(particularly BCAA depletion), which leads to eIF2 a impairing cellular function. Consistent with this
phosphorylation and repression of translation. Inter- concept, doxorubicin-induced cardiomyopathy is
estingly, when exposed to pressure overload, GCN2 characterized by an imbalance in autophagy initiation
null mice are protected against contractile dysfunc- versus completion, resulting in accumulation of
tion (122), raising the possibility that BCAA supple- autophagasomes and dysfunction of cardiomyocytes
mentation may afford some protection during HF (129).
through GCN2 inhibition. The ubiquitin proteasome system also plays a
Autophagy and the ubiquitin proteasome critical role in protein turnover. Accumulation of
s y s t e m d u r i n g H F . Turnover of cellular components ubiquitinated proteins has been consistently
(such as proteins) is essential for maintenance of observed across studies investigating human HF
function, especially in terminally differentiated cells samples (123,125,130,131). This accumulation could
with limited capacity for renewal, such as car- result from an imbalance between the activity of
diomyocytes. Several cellular processes, including ubiquitin ligases, de-ubiquitinating enzymes, and the
autophagy and the ubiquitin proteasome system, are proteasome. In the last case, studies assessing pro-
critical for the turnover or degradation of proteins teasome activity have produced inconsistent results.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017 Wende et al. 305
JUNE 2017:297–310 Metabolic Origins of Heart Failure

C ENTR AL I LL U STRA T I O N Hypothetical Model for the Metabolic Origins of Heart Failure

Wende, A.R. et al. J Am Coll Cardiol Basic Trans Science. 2017;2(3):297–310.

ADP ¼ adenosine diphosphate; AMP ¼ adenosine monophosphate; AMPK ¼ adenosine monophosphate kinase; ATP ¼ adenosine triphos-
phate; BCAA ¼ branched-chain amino acids; BCKA ¼ branched chain keto acid; BCKDH ¼ branched chain keto acid dehydrogenase;
CoA ¼ coenzyme A; FA ¼ fatty acid; FAO ¼ fatty acid oxidation; KB ¼ ketone body; mTOR ¼ mammalian target of rapamycin.

For example, Birks et al. (131) reported increased 20S METABOLIC DYS-SYNCHRONY DURING HF:
proteasome chymotrypsin-like activity, whereas Day AN ENGINE FLOODED WITH FUEL?
et al. (132) showed chymotrypsin-like and caspase-
like proteasome activities were reduced. Interest- Previous sections have outlined macronutrient
ingly, proteasome activity increases in patients metabolic perturbations during HF, the potential
after LVAD implantation (130). Additional studies mechanisms leading to their occurrence, and their
are required to elucidate fully the contribution of potential contribution to contractile dysfunction of
perturbed ubiquitin proteasome system function in the heart. Here, we propose a unifying hypothesis
the cause of HF. for the metabolic origins of HF, based on the concept
306 Wende et al. JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017

Metabolic Origins of Heart Failure JUNE 2017:297–310

F I G U R E 4 Increased Circulating Levels of Various Substrates During Heart Failure

ANP ¼ atrial natriuretic peptide; BNP ¼ brain natriuretic peptide; FA ¼ fatty acid; FAO ¼ fatty acid oxidation; GLOX ¼ glucose oxidation; KB ¼
ketone body; other abbreviation as in Figure 2.

that the failing heart is oversupplied with macronu- Collectively, the failing myocardium is in an envi-
trients, leading to an imbalance in fuel availability ronment rich in fuels (Figure 4).
and use and subsequent accumulation of key meta- The myocardium has a high capacity and preference
bolic intermediates that worsen contractile function for use of ketone body, which attenuates use of other
of the heart (Central Illustration). The rationale for substrates; elevated use of ketone body concomitant
this model will be discussed. with decreased total CoA in the failing myocardium
During HF, the myocardium is undoubtedly in a (42) will limit mitochondria-free CoA for FAO, pyru-
state of dys-synchrony with regard to energy demand vate oxidation, and BCAA metabolism. One strategy to
and ATP generation. Accordingly, compensatory liberate CoA for continued oxidative metabolism in-
mechanisms attempt to regain synchrony through volves exchanging the CoA with carnitine, and sub-
decreasing workload and increasing metabolism. sequent generation of acetyl-carnitine (as observed
For example, increased atrial natriuretic peptide/ during HF) (100,101,136). However, diminished
brain natriuretic peptide secretions promote natri- carnitine levels in the failing heart (137) will attenuate
uresis, thus reducing workload (133). Elevation of FAO capacity further. Impairment of FAO in the face of
these cardiokines, as well as various cytokines (e.g., elevated circulating FAs would promote diversion of
tumor necrosis factor [TNF]- a ) and sympathetic tone, FA species into signaling and lipotoxic pathways.
also serve to signal fuel mobilization during HF, Elevated use of ketone body would also limit the ac-
enhancing adipocyte lipolysis (releasing FAs), hepatic tivity status of pyruvate dehydrogenase; in the face of
gluconeogenesis (releasing glucose), and skeletal elevated glucose uptake, an uncoupling between
muscle proteolysis (releasing AAs, including BCAAs) glycolysis and glucose oxidation ensues. Similarly,
(104,134,135). These fuels become available not only impairment of the BCKD due to cofactor perturbations
to the heart (for ATP generation) but also to extrac- and/or PTM, coupled with increased circulating
ardiac tissues, including the liver; increased FA BCAAs, will lead to accumulation of BCKA and mito-
availability promotes ketogenesis, thereby elevating chondrial dysfunction. The latter amplifies metabolic
circulating ketone bodies in HF subjects (95–98). dyssynchrony further due to activation of
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017 Wende et al. 307
JUNE 2017:297–310 Metabolic Origins of Heart Failure

mechanisms designed to promote cardiomyocyte example, nicotinic acid, an antilipolytic agent, has
substrate uptake in the face of energy deficit (e.g., been proposed to be beneficial during ischemic heart
AMPK activation promoting GLUT1/4 and CD36 disease (138); attenuation of lipolysis would reduce
translocation for glucose and FA uptake, respec- FAs available for ketogenesis and lipotoxicity. Simi-
tively). Importantly, during diabetes, dyssynchrony larly, inhibition of hepatic FAO would attenuate
between fuel availability and use will be amplified ketogenesis; this may contribute to the benefit of
further, due to greater levels of circulating FAs, ke- FAO inhibitors, such as trimetazidine, in the setting
tone bodies, glucose, and BCAAs. In other words, the of HF (139–141). However, some FAO inhibitors, such
failing myocardium can be considered an engine as etomoxir, appear to have detrimental effects due
flooded with fuel. to hepatic toxicity (142). According to our model,
limited availability of specific cofactors (e.g., carni-
CONCLUSIONS tine and CoA) during HF would exacerbate metabolic
dyssynchrony Interestingly, several studies suggest
According to the model described above (Central that carnitine supplementation has benefits during
Illustration), strategies designed to regain synchrony HF (143); whether pantothenate (precursor for CoA
among energy demand, substrate availability, and biosynthesis) supplementation has benefit during HF
substrate use would be beneficial during HF. Estab- is currently unknown. Promotion of oxidation of in-
lished and emerging HF therapeutics include dividual substrates, such as pyruvate and BCKA, also
b-blockers and valsartan-sacubitril. Both treatments appears to be beneficial in animal models (106);
focus primarily on reduction of workload, which in whether this translates to the clinical setting is
turn would help regain synchrony due to attenuation currently unknown. However, caution should be
of energy demand. In addition, b -blockers help regain taken to promote the use of a single substrate in the
synchrony further through inhibition of lipolysis, presence of excess FA availability, as this in turn
thus decreasing substrate supply (FAs, and likely could result in further inhibition of FAO, and poten-
ketone bodies). In contrast, through promotion of tially lipotoxicity.
lipolysis, valsartan-saculcitril therapy has the poten-
tial to negatively affect metabolic synchrony, aug- ADDRESS FOR CORRESPONDENCE: Dr. Martin E.
menting substrate supply further. Although no Young, Division of Cardiovascular Diseases, Department of
pharmacological strategy has been taken to specif- Medicine, University of Alabama at Birmingham, 703 19th
ically attenuate ketogenesis in the setting of HF, Street South, ZRB 308, Birmingham, Alabama 35294.
some strategies may influence this indirectly. For E-mail: meyoung@uab.edu.

REFERENCES

1. Brown DI, Griendling KK. Regulation of signal 7. Kato T, Niizuma S, Inuzuka Y, et al. Analysis of 12. Sack MN, Rader TA, Park S, Bastin J,
transduction by reactive oxygen species in the metabolic remodeling in compensated left ven- McCune SA, Kelly DP. Fatty acid oxidation enzyme
cardiovascular system. Circ Res 2015;116:531–49. tricular hypertrophy and heart failure. Circ Heart gene expression is downregulated in the failing
Fail 2010;3:420–30. heart. Circulation 1996;94:2837–42.
2. Bers DM. Cardiac sarcoplasmic reticulum cal-
cium leak: basis and roles in cardiac dysfunction. 8. Osorio JC, Stanley WC, Linke A, et al. Impaired 13. Barger P, Kelly D. PPAR signaling in the control
Annu Rev Physiol 2014;76:107–27. myocardial fatty acid oxidation and reduced pro- of cardiac energy metabolism. Trends Cardiovasc
tein expression of retinoid X receptor-alpha in Med 2000;10:238–45.
3. Lopaschuk GD, Ussher JR, Folmes CD,
pacing-induced heart failure. Circulation 2002;
Jaswal JS, Stanley WC. Myocardial fatty acid 14. Lopaschuk G, Belke D, Gamble J, Itoi T,
106:606–12.
metabolism in health and disease. Physiol Rev Schönekess B. Regulation of fatty acid oxidation in
2010;90:207–58. 9. Davila-Roman VG, Vedala G, Herrero P, et al. the mammalian heart in health and disease. Bio-
Altered myocardial fatty acid and glucose meta- chim Biophys Acta 1994;1213:263–76.
4. Allard M, Schonekess B, Henning S, English D, bolism in idiopathic dilated cardiomyopathy. J Am
Lopaschuk G. Contribution of oxidative meta- Coll Cardiol 2002;40:271–7. 15. Taegtmeyer H, Overturf ML. Effects of mod-
bolism and glycolysis to ATP production in hy- erate hypertension on cardiac function and meta-
10. Yazaki Y, Isobe M, Takahashi W, et al. Assess-
pertrophied hearts. Am J Physiol 1994;267: bolism in the rabbit. Hypertension 1988;11:
H742–50. ment of myocardial fatty acid metabolic abnor- 416–26.
malities in patients with idiopathic dilated
5. Goodwin G, Taylor C, Taegtmeyer H. Regulation cardiomyopathy using 123I BMIPP SPECT: corre- 16. Beer M, Seyfarth T, Sandstede J, et al. Abso-
of energy metabolism of the heart during acute lation with clinicopathological findings and clinical lute concentrations of high-energy phosphate
increase in heart work. J Biol Chem 1998;273: course. Heart 1999;81:153–9. metabolites in normal, hypertrophied, and failing
29530–9. human myocardium measured noninvasively with
11. Heather LC, Cole MA, Lygate CA, et al. Fatty
(31)P-SLOOP magnetic resonance spectroscopy.
6. Wisneski JA, Gertz EW, Neese RA, Mayr M. acid transporter levels and palmitate oxidation
J Am Coll Cardiol 2002;40:1267–74.
Myocardial metabolism of free fatty acids. Studies rate correlate with ejection fraction in the
with 14C-labeled substrates in humans. J Clin infarcted rat heart. Cardiovasc Res 2006;72: 17. Hirsch GA, Bottomley PA, Gerstenblith G,
Invest 1987;79:359–66. 430–7. Weiss RG. Allopurinol acutely increases adenosine
308 Wende et al. JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017

Metabolic Origins of Heart Failure JUNE 2017:297–310

triphosphate energy delivery in failing human improves contractile function in the hypertensive resistance improves metabolic and contractile ef-
hearts. J Am Coll Cardiol 2012;59:802–8. dahl salt-sensitive rat. Clin Exp Pharmacol Physiol ficiency in stressed rat heart. FASEB J 2012;26:
2005;32:825–31. 3118–26.
18. Sharma S, Adrogue J, Golfman L, et al. Intra-
myocardial lipid accumulation in the failing human 32. Raher MJ, Thibault HB, Buys ES, et al. A short 48. Doenst T, Pytel G, Schrepper A, et al.
heart resembles the lipotoxic rat heart. FASEB J duration of high-fat diet induces insulin resistance Decreased rates of substrate oxidation ex vivo
2004;18:1692–700. and predisposes to adverse left ventricular predict the onset of heart failure and contractile
remodeling after pressure overload. Am J Physiol dysfunction in rats with pressure overload. Car-
19. Abdurrachim D, Luiken JJ, Nicolay K, Glatz JF,
Heart Circ Physiol 2008;295:H2495–502. diovasc Res 2010;86:461–70.
Prompers JJ, Nabben M. Good and bad conse-
quences of altered fatty acid metabolism in heart 33. Stanley WC, Dabkowski ER, Ribeiro RF Jr., 49. Helfand BK, Maselli NJ, Lessard DM, et al.
failure: evidence from mouse models. Cardiovasc O’Connell KA. Dietary fat and heart failure: moving Elevated serum glucose levels and survival after
Res 2015;106:194–205. from lipotoxicity to lipoprotection. Circ Res 2012; acute heart failure: a population-based perspec-
110:764–76. tive. Diab Vasc Dis Res 2015;12:119–25.
20. Exil VJ, Roberts RL, Sims H, et al. Very-long-
chain acyl-coenzyme a dehydrogenase deficiency 34. Chiu HC, Kovacs A, Blanton RM, et al. Trans- 50. Sud M, Wang X, Austin PC, et al. Presentation
in mice. Circ Res 2003;93:448–55. genic expression of fatty acid transport protein 1 in blood glucose and death, hospitalization, and

21. Xiong D, He H, James J, et al. Cardiac-specific the heart causes lipotoxic cardiomyopathy. Circ future diabetes risk in patients with acute heart
VLCAD deficiency induces dilated cardiomyopathy Res 2005;96:225–33. failure syndromes. Eur Heart J 2015;36:924–31.
and cold intolerance. Am J Physiol Heart Circ 35. Chiu HC, Kovacs A, Ford DA, et al. A novel 51. Dei Cas A, Khan SS, Butler J, et al. Impact of
Physiol 2014;306:H326–38. mouse model of lipotoxic cardiomyopathy. J Clin diabetes on epidemiology, treatment, and out-
22. Noh HL, Okajima K, Molkentin JD, Homma S, Invest 2001;107:813–22. comes of patients with heart failure. J Am Coll
Goldberg IJ. Acute lipoprotein lipase deletion in Cardiol HF 2015;3:136–45.
36. Opie LH, Knuuti J. The adrenergic-fatty acid
adult mice leads to dyslipidemia and cardiac load in heart failure. J Am Coll Cardiol 2009;54: 52. Sung MM, Hamza SM, Dyck JRB. Myocardial
dysfunction. Am J Physiol Endocrinol Metab 1637–46. metabolism in diabetic cardiomyopathy: potential
2006;291:E755–60. therapeutic targets. Antioxid Redox Signal 2015;
37. He L, Kim T, Long Q, et al. Carnitine
23. Ellis JM, Mentock SM, Depetrillo MA, et al. 22:1606–30.
palmitoyltransferase-1b deficiency aggravates
Mouse cardiac acyl coenzyme a synthetase 1 pressure overload-induced cardiac hypertrophy 53. Wende AR. Post-translational modifications of
deficiency impairs Fatty Acid oxidation and in- caused by lipotoxicity. Circulation 2012;126: the cardiac proteome in diabetes and heart failure.
duces cardiac hypertrophy. Mol Cell Biol 2011;31: 1705–16. Proteomic Clin Appl 2016;10:25–38.
1252–62.
38. Tucci S, Flogel U, Hermann S, Sturm M, 54. Shao D, Tian R. Glucose transporters in cardiac
24. Kienesberger PC, Pulinilkunnil T, Nagendran J, Schafers M, Spiekerkoetter U. Development and metabolism and hypertrophy. Compr Physiol 2015;
et al. Early structural and metabolic cardiac pathomechanisms of cardiomyopathy in very 6:331–51.
remodelling in response to inducible adipose tri- long–chain acyl-CoA dehydrogenase deficient
55. Kundu BK, Zhong M, Sen S, Davogustto G,
glyceride lipase ablation. Cardiovasc Res 2013;99: (VLCAD(/) mice. Biochim Biophys Acta 2014;
Keller SR, Taegtmeyer H. Remodeling of glucose
442–51. 1842:677–85.
metabolism precedes pressure overload-induced
25. Kuang M, Febbraio M, Wagg C, Lopaschuk GD, 39. Oka S, Zhai P, Yamamoto T, et al. Peroxisome left ventricular hypertrophy: review of a hypoth-
Dyck JR. Fatty acid translocase/CD36 deficiency proliferator activated receptor-alpha association esis. Cardiology 2015;130:211–20.
does not energetically or functionally compromise with silent information regulator 1 suppresses
56. Azam MA, Wagg CS, Massé S, et al. Feeding
hearts before or after ischemia. Circulation 2004; cardiac fatty acid metabolism in the failing heart.
the fibrillating heart: dichloroacetate improves
109:1550–7. Circ Heart Fail 2015;8:1123–32.
cardiac contractile dysfunction following VF. Am J
26. Lehman JJ, Boudina S, Banke NH, et al. The 40. Alrob OA, Sankaralingam S, Ma C, et al. Physiol Heart Circ Physiol 2015;309:H1543–53.
transcriptional coactivator PGC-1alpha is essential Obesity-induced lysine acetylation increases car-
57. Grossman AN, Opie LH, Beshansky JR,
for maximal and efficient cardiac mitochondrial diac fatty acid oxidation and impairs insulin sig-
Ingwall JS, Rackley CE, Selker HP. Glucose-insulin-
fatty acid oxidation and lipid homeostasis. Am J nalling. Cardiovasc Res 2014;103:485–97.
potassium revived. Circulation 2013;127:1040–8.
Physiol Heart Circ Physiol 2008;295:H185–96.
41. Jing E, O’Neill BT, Rardin MJ, et al. Sirt3 reg-
58. Joost HG, Bell GI, Best JD, et al. Nomenclature
27. Luptak I, Balschi JA, Xing Y, Leone TC, ulates metabolic flexibility of skeletal muscle
of the GLUT/SLC2A family of sugar/polyol trans-
Kelly DP, Tian R. Decreased contractile and through reversible enzymatic deacetylation. Dia-
port facilitators. Am J Physiol Endocrinol Metab
metabolic reserve in peroxisome proliferator- betes 2013;62:3404–17.
2002;282:E974–6.
activated receptor-alpha-null hearts can be
42. Reibel DK, Uboh CE, Kent RL. Altered coen-
rescued by increasing glucose transport and utili- 59. Aerni-Flessner L, Abi-Jaoude M, Koenig A,
zyme A and carnitine metabolism in pressure-
zation. Circulation 2005;112:2339–46. Payne M, Hruz PW. GLUT4, GLUT1, and GLUT8 are
overload hypertrophied hearts. Am J Physiol
the dominant GLUT transcripts expressed in the
28. Kolwicz SC Jr., Olson DP, Marney LC, Garcia- 1983;244:H839–43.
murine left ventricle. Cardiovasc Diabetol 2012;11:
Menendez L, Synovec RE, Tian R. Cardiac-specific
43. Regitz V, Shug AL, Fleck E. Defective 63.
deletion of acetyl CoA carboxylase 2 prevents
myocardial carnitine metabolism in congestive
metabolic remodeling during pressure-overload 60. Razeghi P, Young ME, Alcorn JL, Moravec CS,
heart failure secondary to dilated cardiomyopathy
hypertrophy. Circ Res 2012;111:728–38. Frazier OH, Taegtmeyer H. Metabolic gene
and to coronary, hypertensive and valvular heart
expression in fetal and failing human heart.
29. Okere IC, Young ME, McElfresh TA, et al. Low diseases. Am J Cardiol 1990;65:755–60.
Circulation 2001;104:2923–31.
carbohydrate/high-fat diet attenuates cardiac hy-
44. Lee CF, Tian R. Mitochondrion as a target for
pertrophy, remodeling, and altered gene expres- 61. Ciccarelli M, Chuprun JK, Rengo G, et al.
heart failure therapy—role of protein lysine acet-
sion in hypertension. Hypertension 2006;48: G protein-coupled receptor kinase 2 activity im-
ylation. Circ J 2015;79:1863–70.
1116–23. pairs cardiac glucose uptake and promotes insulin
45. Depré C, Vanoverschelde J-LJ, Taegtmeyer H. resistance after myocardial ischemia. Circulation
30. Berthiaume JM, Young ME, Chen X,
Glucose for the heart. Circulation 1999;99: 2011;123:1953–62.
McElfresh TA, Yu X, Chandler MP. Normalizing the
578–88.
metabolic phenotype after myocardial infarction: 62. Bishop SP, Altschuld RA. Increased glycolytic
Impact of subchronic high fat feeding. J Mol Cell 46. Riehle C, Abel ED. Insulin signaling and heart metabolism in cardiac hypertrophy and congestive
Cardiol 2012;53:125–33. failure. Circ Res 2016;118:1151–69. failure. Am J Physiol 1970;218:153–9.

31. Okere IC, Chess DJ, McElfresh TA, et al. High- 47. Harmancey R, Lam TN, Lubrano GM, 63. Liao R, Jain M, Cui L, et al. Cardiac-specific
fat diet prevents cardiac hypertrophy and Guthrie PH, Vela D, Taegtmeyer H. Insulin overexpression of GLUT1 prevents the
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017 Wende et al. 309
JUNE 2017:297–310 Metabolic Origins of Heart Failure

development of heart failure attributable to 78. Lunde IG, Aronsen JM, Kvaløy H, et al. Cardiac 93. Janardhan A, Chen J, Crawford PA. Altered
pressure overload in mice. Circulation 2002;106: O-GlcNAc signaling is increased in hypertrophy systemic ketone body metabolism in advanced
2125–31. and heart failure. Physiol Genomics 2012;44: heart failure. Tex Heart Inst J 2011;38:533–8.
162–72.
64. Pereira RO, Wende AR, Olsen C, et al. Induc- 94. Lommi J, Koskinen P, Naveri H, Harkonen M,
ible overexpression of GLUT1 prevents mitochon- 79. Marsh SA, Collins HE, Chatham JC. Protein Kupari M. Heart failure ketosis. J Intern Med 1997;
drial dysfunction and attenuates structural O-GlcNAcylation and cardiovascular (patho)phys- 242:231–8.
remodeling in pressure overload but does not iology. J Biol Chem 2014;289:34449–56.
95. Packer M. Pathophysiology of chronic heart
prevent left ventricular dysfunction. J Am Heart
80. Dassanayaka S, Jones SP. O-GlcNAc and the failure. Lancet 1992;340:88–92.
Assoc 2013;2:e000301.
cardiovascular system. Pharmacol Ther 2014;142: 96. Kupari M, Lommi J, Ventila M, Karjalainen U.
65. Pereira RO, Wende AR, Olsen C, et al. GLUT1 62–71. Breath acetone in congestive heart failure. Am J
deficiency in cardiomyocytes does not accelerate
81. Erickson JR, Pereira L, Wang L, et al. Diabetic Cardiol 1995;76:1076–8.
the transition from compensated hypertrophy to
hyperglycaemia activates CaMKII and arrhythmias 97. Marcondes-Braga FG, Gutz IG, Batista GL,
heart failure. J Mol Cell Cardiol 2014;72:95–103.
by O-linked glycosylation. Nature 2013;502: et al. Exhaled acetone as a new biomaker of heart
66. Tian R, Abel ED. Responses of GLUT4- 372–6. failure severity. Chest 2012;142:457–66.
deficient hearts to ischemia underscore the
82. Zhu-Mauldin X, Marsh SA, Zou L, 98. Yokokawa T, Sugano Y, Shimouchi A, et al.
importance of glycolysis. Circulation 2001;103:
Marchase RB, Chatham JC. Modification of STIM1 Exhaled acetone concentration is related to he-
2961–6.
by O-linked N-acetylglucosamine (O-GlcNAc) at- modynamic severity in patients with non-ischemic
67. DeBosch BJ, Chen Z, Finck BN, Chi M, tenuates store-operated calcium entry in chronic heart failure. Circ J 2016;80:1178–86.
Moley KH. Glucose transporter-8 (GLUT8) medi- neonatal cardiomyocytes. J Biol Chem 2012;287:
ates glucose intolerance and dyslipidemia in high- 99. Zordoky BN, Sung MM, Ezekowitz J, et al.
39094–106.
fructose diet-fed male mice. Mol Endocrinol 2013; Metabolomic fingerprint of heart failure with
27:1887–96. 83. Ramirez-Correa G, Ma J, Slawson C, et al. preserved ejection fraction. PLoS One 2015;10:
Removal of abnormal myofilament O-GlcNAcylation e0124844.
68. Heerspink HJ, Perkins BA, Fitchett DH,
restores Ca2þ sensitivity in diabetic cardiac muscle.
Husain M, Cherney DZ. Sodium glucose cotrans- 100. Aubert G, Martin OJ, Horton JL, et al. The
Diabetes 2015;64:3573–87.
porter 2 inhibitors in the treatment of diabetes failing heart relies on ketone bodies as a fuel.
mellitus: cardiovascular and kidney effects, po- 84. Ngoh GA, Watson LJ, Facundo HT, Jones SP. Circulation 2016;133:698–705.
tential mechanisms, and clinical applications. Cir- Augmented O-GlcNAc signaling attenuates oxida-
101. Bedi KC Jr., Snyder NW, Brandimarto J, et al.
culation 2016;134:752–72. tive stress and calcium overload in car-
Evidence for intramyocardial disruption of lipid
diomyocytes. Amino Acids 2011;40:895–911.
69. Pham D, Albuquerque Rocha N, McGuire DK, metabolism and increased myocardial ketone uti-
Neeland IJ. Impact of empagliflozin in patients 85. Kimura I, Inoue D, Maeda T, et al. Short-chain lization in advanced human heart failure. Circula-
with diabetes and heart failure. Trends Cardiovasc fatty acids and ketones directly regulate sympa- tion 2016;133:706–16.
Med 2016;27:144–51. thetic nervous system via G protein-coupled re-
102. Shimazu T, Hirschey MD, Newman J, et al.
ceptor 41 (GPR41). Proc Natl Acad Sci U S A 2011;
70. Banerjee SK, McGaffin KR, Pastor-Soler NM, Suppression of oxidative stress by beta-
108:8030–5.
Ahmad F. SGLT1 is a novel cardiac glucose trans- hydroxybutyrate, an endogenous histone deace-
porter that is perturbed in disease states. Car- 86. Valayannopoulos V, Bajolle F, Arnoux JB, et al. tylase inhibitor. Science 2013;339:211–4.
diovasc Res 2009;84:111–8. Successful treatment of severe cardiomyopathy in
103. Hakuno D, Hamba Y, Toya T, Adachi T. Plasma
glycogen storage disease type III With D, L-3-
71. Kashiwagi Y, Nagoshi T, Yoshino T, et al. amino acid profiling identifies specific amino acid
hydroxybutyrate, ketogenic and high-protein
Expression of SGLT1 in human hearts and impair- associations with cardiovascular function in pa-
diet. Pediatr Res 2011;70:638–41.
ment of cardiac glucose uptake by phlorizin during tients with systolic heart failure. PLoS One 2015;
ischemia-reperfusion injury in mice. PLoS One 87. Al-Zaid NS, Dashti HM, Mathew TC, Juggi JS. 10:e0117325.
2015;10:e0130605. Low carbohydrate ketogenic diet enhances cardiac 104. Pasini E, Aquilani R, Dioguardi FS,
tolerance to global ischaemia. Acta Cardiol 2007; D’Antona G, Gheorghiade M, Taegtmeyer H.
72. Son N-H, Ananthakrishnan R, Yu S, et al. Car-
62:381–9. Hypercatabolic syndrome: molecular basis and
diomyocyte aldose reductase causes heart failure
and impairs recovery from ischemia. PLoS One 88. Liu J, Wang P, Zou L, et al. High-fat, low- effects of nutritional supplements with amino
2012;7:e46549. carbohydrate diet promotes arrhythmic death acids. Am J Cardiol 2008;101:11E–5E.

73. Vimercati C, Qanud K, Mitacchione G, et al. and increases myocardial ischemia-reperfusion 105. Lai L, Leone TC, Keller MP, et al. Energy
Beneficial effects of acute inhibition of the injury in rats. Am J Physiol Heart Circ Physiol metabolic reprogramming in the hypertrophied
oxidative pentose phosphate pathway in the 2014;307:H598–608. and early stage failing heart: a multisystems
failing heart. Am J Physiol Heart Circ Physiol 2014; 89. Wang P, Tate JM, Lloyd SG. Low carbohydrate approach. Circ Heart Fail 2014;7:1022–31.
306:H709–17. diet decreases myocardial insulin signaling and 106. Sun H, Olson KC, Gao C, et al. Catabolic
74. Burke MA, Chang S, Wakimoto H, et al. increases susceptibility to myocardial ischemia. defect of branched-chain amino acids promotes
Molecular profiling of dilated cardiomyopathy that Life Sci 2008;83:836–44. heart failure. Circulation 2016;133:2038–49.
progresses to heart failure. JCI Insight 2016;1: 90. Schugar RC, Moll AR, Andre d’Avignon D, 107. Ito T, Kimura Y, Uozumi Y, et al. Taurine
e86898. Weinheimer CJ, Kovacs A, Crawford PA. depletion caused by knocking out the taurine
75. Ussher JR, Jaswal JS, Lopaschuk GD. Pyridine Cardiomyocyte-specific deficiency of ketone body transporter gene leads to cardiomyopathy with
nucleotide regulation of cardiac intermediary metabolism promotes accelerated pathological cardiac atrophy. J Mol Cell Cardiol 2008;44:
metabolism. Circ Res 2012;111:628–41. remodeling. Mol Metab 2014;3:754–69. 927–37.

76. Hecker PA, Leopold JA, Gupte SA, Recchia FA, 91. Stowe KA, Burgess SC, Merritt M, Sherry AD, 108. Schaffer SW, Shimada-Takaura K, Jong CJ,
Stanley WC. Impact of glucose-6-phosphate de- Malloy CR. Storage and oxidation of long-chain Ito T, Takahashi K. Impaired energy metabolism of
hydrogenase deficiency on the pathophysiology of fatty acids in the C57/BL6 mouse heart as the taurine deficient heart. Amino Acids 2016;48:
cardiovascular disease. Am J Physiol Heart Circ measured by NMR spectroscopy. FEBS Lett 2006; 549–58.
Physiol 2013;304:H491–500. 580:4282–7.
109. Jong CJ, Azuma J, Schaffer S. Mechanism
77. Hardivillé S, Hart GW. Nutrient regulation of 92. Cotter DG, Schugar RC, Crawford PA. Ketone underlying the antioxidant activity of taurine:
signaling, transcription, and cell physiology by body metabolism and cardiovascular disease. Am J prevention of mitochondrial oxidant production.
O-GlcNAcylation. Cell Metab 2014;20:208–13. Physiol Heart Circ Physiol 2013;304:H1060–76. Amino Acids 2012;42:2223–32.
310 Wende et al. JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 3, 2017

Metabolic Origins of Heart Failure JUNE 2017:297–310

110. Ramila KC, Jong CJ, Pastukh V, Ito T, 122. Lu Z, Xu X, Fassett J, et al. Loss of the significance, diagnostic and prognostic value. Can
Azuma J, Schaffer SW. Role of protein phosphor- eukaryotic initiation factor 2alpha kinase general J Physiol Pharmacol 2011;89:587–91.
ylation in excitation-contraction coupling in control nonderepressible 2 protects mice from
134. Lafontan M, Moro C, Berlan M, Crampes F,
taurine deficient hearts. Am J Physiol Heart Circ pressure overload-induced congestive heart fail-
Sengenes C, Galitzky J. Control of lipolysis by
Physiol 2015;308:H232–9. ure without affecting ventricular hypertrophy.
natriuretic peptides and cyclic GMP. Trends
Hypertension 2014;63:128–35.
111. Azuma J, Sawamura A, Awata N. Usefulness of Endocrinol Metab 2008;19:130–7.
taurine in chronic congestive heart failure and its 123. Hein S, Arnon E, Kostin S, et al. Progression
135. Tenenbaum A, Fisman EZ. Impaired glucose
prospective application. Jpn Circ J 1992;56:95–9. from compensated hypertrophy to failure in the
metabolism in patients with heart failure: patho-
pressure-overloaded human heart: structural
112. Beyranvand MR, Khalafi MK, Roshan VD, physiology and possible treatment strategies. Am
deterioration and compensatory mechanisms. Cir-
Choobineh S, Parsa SA, Piranfar MA. Effect of J Cardiovasc Drugs 2004;4:269–80.
culation 2003;107:984–91.
taurine supplementation on exercise capacity of 136. Rame JE. Metabolic staging in human heart
patients with heart failure. J Cardiol 2011;57: 124. Knaapen MW, Davies MJ, De Bie M, Haven AJ,
failure: circulating acylcarnitines and the failing
333–7. Martinet W, Kockx MM. Apoptotic versus auto-
heart’s energetic signature. J Am Coll Cardiol
phagic cell death in heart failure. Cardiovasc Res
113. Carubelli V, Castrini AI, Lazzarini V, 2016;67:300–2.
2001;51:304–12.
Gheorghiade M, Metra M, Lombardi C. Amino acids 137. Atar D, Spiess M, Mandinova A, Cierpka H,
and derivatives, a new treatment of chronic heart 125. Kassiotis C, Ballal K, Wellnitz K, et al. Markers
Noll G, Luscher TF. Carnitine—from cellular
failure? Heart Fail Rev 2015;20:39–51. of autophagy are downregulated in failing human
mechanisms to potential clinical applications in
heart after mechanical unloading. Circulation
heart disease. Eur J Clin Invest 1997;27:973–6.
114. Aquilani R, Viglio S, Iadarola P, et al. Oral 2009;120:S191–7.
amino acid supplements improve exercise capac- 138. Folmes CD, Clanachan AS, Lopaschuk GD.
126. Zhu H, Tannous P, Johnstone JL, et al. Car-
ities in elderly patients with chronic heart failure. Fatty acid oxidation inhibitors in the management
diac autophagy is a maladaptive response to he-
Am J Cardiol 2008;101:104E–10E. of chronic complications of atherosclerosis. Curr
modynamic stress. J Clin Invest 2007;117:1782–93.
Atheroscler Rep 2005;7:63–70.
115. Lombardi C, Carubelli V, Lazzarini V, et al.
127. Shirakabe A, Zhai P, Ikeda Y, et al. Drp1-
Effects of oral amino acid supplements on func- 139. Lopatin YM, Rosano GM, Fragasso G, et al.
dependent mitochondrial autophagy plays a pro-
tional capacity in patients with chronic heart fail- Rationale and benefits of trimetazidine by acting
tective role against pressure overload-induced
ure. Clin Med Insights 2014;8:39–44. on cardiac metabolism in heart failure. Int J Cardiol
mitochondrial dysfunction and heart failure. Cir-
2016;203:909–15.
116. Tanada Y, Shioi T, Kato T, Kawamoto A, culation 2016;133:1249–63.
Okuda J, Kimura T. Branched-chain amino acids 140. Zhang L, Lu Y, Jiang H, et al. Additional use
128. Nakai A, Yamaguchi O, Takeda T, et al. The
ameliorate heart failure with cardiac cachexia in of trimetazidine in patients with chronic heart
role of autophagy in cardiomyocytes in the basal
rats. Life Sci 2015;137:20–7. failure: a meta-analysis. J Am Coll Cardiol 2012;
state and in response to hemodynamic stress. Nat
59:913–22.
117. Anker SD, Ponikowski P, Varney S, et al. Med 2007;13:619–24.
Wasting as independent risk factor for mortality in 141. Fragasso G, Salerno A, Lattuada G, et al. Ef-
129. Li DL, Wang ZV, Ding G, et al. Doxorubicin
chronic heart failure. Lancet 1997;349:1050–3. fect of partial inhibition of fatty acid oxidation by
blocks cardiomyocyte autophagic flux by inhibiting
trimetazidine on whole body energy metabolism in
118. Shiojima I, Sato K, Izumiya Y, et al. Disruption lysosome acidification. Circulation 2016;133:
patients with chronic heart failure. Heart 2011;97:
of coordinated cardiac hypertrophy and angio- 1668–87.
1495–500.
genesis contributes to the transition to heart 130. Predmore JM, Wang P, Davis F, et al. Ubiq-
failure. J Clin Invest 2005;115:2108–18. uitin proteasome dysfunction in human hypertro- 142. Holubarsch CJ, Rohrbach M, Karrasch M,
phic and dilated cardiomyopathies. Circulation et al. A double-blind randomized multicentre
119. Riehle C, Wende AR, Sena S, et al. Insulin
2010;121:997–1004. clinical trial to evaluate the efficacy and safety of
receptor substrate signaling suppresses neonatal
two doses of etomoxir in comparison with placebo
autophagy in the heart. J Clin Invest 2013;123: 131. Birks EJ, Latif N, Enesa K, et al. Elevated in patients with moderate congestive heart failure:
5319–33. p53 expression is associated with dysregulation
the ERGO (etomoxir for the recovery of glucose
120. McMullen JR, Sherwood MC, Tarnavski O, of the ubiquitin-proteasome system in dilated
oxidation) study. Clin Sci 2007;113:205–12.
et al. Inhibition of mTOR signaling with rapamycin cardiomyopathy. Cardiovasc Res 2008;79:
472–80. 143. Ferrari R, Merli E, Cicchitelli G, Mele D, Fucili A,
regresses established cardiac hypertrophy induced
Ceconi C. Therapeutic effects of L–carnitine and
by pressure overload. Circulation 2004;109: 132. Day SM, Divald A, Wang P, et al. Impaired
propionyl-L-carnitine on cardiovascular diseases: a
3050–5. assembly and post-translational regulation of 26S
review. Ann N Y Acad Sci 2004;1033:79–91.
proteasome in human end-stage heart failure. Circ
121. Shende P, Plaisance I, Morandi C, et al. Car-
Heart Fail 2013;6:544–9.
diac raptor ablation impairs adaptive hypertrophy,
alters metabolic gene expression, and causes heart 133. Ghosh N, Haddad H. Atrial natriuretic KEY WORDS amino acids, fatty acids,
failure in mice. Circulation 2011;123:1073–82. peptides in heart failure: pathophysiological glucose, heart failure, ketone bodies

You might also like