Вы находитесь на странице: 1из 12

The n e w e ng l a n d j o u r na l of m e dic i n e

review article

mechanisms of disease

The Failing Heart — An Engine Out of Fuel


Stefan Neubauer, M.D., F.R.C.P.

H
From the Department of Cardiovascular eart failure is an enormous medical and societal burden.1,2
Medicine, University of Oxford and John It is a common disease: more than 2% of the U.S. population, or almost
Radcliffe Hospital, Oxford, United King-
dom. Address reprint requests to Dr. Neu- 5 million people, are affected, and 30 to 40% of patients die from heart
bauer at the Department of Cardiovascu- failure within 1 year after receiving the diagnosis.3 Heart failure can be disabling,
lar Medicine, University of Oxford, John and it can severely reduce a patient’s quality of life. It consumes approximately 2%
Radcliffe Hospital, Oxford OX3 9DU,
United Kingdom, or at stefan.neubauer@ of the National Health Service budget in the United Kingdom, and in the United
cardiov.ox.ac.uk. States, the total annual cost of treatment for heart failure is approximately $28 bil-
lion. Moreover, the financial burden of heart failure will increase in coming de-
N Engl J Med 2007;356:1140-51.
Copyright © 2007 Massachusetts Medical Society. cades because of the aging population and the improved treatments of its causes.
Over the past 20 years, there has been considerable progress in the treatment
of chronic heart failure with angiotensin-converting–enzyme (ACE) inhibitors,4,5
aldosterone antagonists,6 beta-receptor blockers,7,8 and resynchronization therapy.9,10
Even with the very best of modern therapy, however, heart failure is still associated
with an annual mortality rate of 10%.10 The search for better treatments is one of
the major challenges in cardiology.
Chronic heart failure is multifactorial. There are many reasons why a human
heart can fail,11 but the available evidence suggests that the failing heart is an
engine out of fuel — that is, altered energetics play an important role in the mech-
anisms of heart failure. For this reason, the modulation of cardiac metabolism has
promise as a new approach to the treatment of heart failure.
This review describes cardiac energy metabolism, appraises the methods used
for its assessment, evaluates the role of impaired energy metabolism in heart fail-
ure, and gives options for metabolic therapy.

The Energy-S ta rvat ion H y p o the sis

The concept that the failing heart is an energy-starved engine that has run out of
fuel is decades old. It was proposed in 1939 by Herrmann and Decherd,12 who, in
their article entitled “The Chemical Nature of Heart Failure,” described a signifi-
cantly reduced creatine content in failing myocardium. Over the next 20 years, the
energy-depletion hypothesis was pursued by various groups,13-15 and today, energy
metabolism in the heart — myocardial energetics — is a topic of considerable inter-
est.16-24 A major reason for the attention to this topic is that any energy-sparing
treatment for heart failure such as beta-receptor blockers,7,8 ACE inhibitors,4,5 or
angiotensin II blockers25,26 improves the prognosis. The failing heart has been
compared to a weak and tired horse, and if this horse is nourished properly, it can
recover and work in the long term, albeit at a reduced level.27

C a r di ac Energy Me ta bol ism

Deprivation of cardiac energy has a major role in heart failure.18 The heart con-
sumes more energy than any other organ. It cycles about 6 kg of ATP every day —

1140 n engl j med 356;11  www.nejm.org  march 15, 2007

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

20 to 30 times its own weight. Each day, it beats atine is constantly lost from the heart by passive
about 100,000 times and pumps approximately diffusion across the sarcolemma.32 An impor­tant
10 tons of blood through the body. To acquire the function of the creatine kinase system is to act as
energy that is necessary to carry out its function, an energy buffer. When the energy demand out-
the heart converts chemical energy stored in fatty strips the energy supply, the phosphocreatine level
acids and glucose into the mechanical energy of falls, keeping ATP at a normal level, but the free
the actin–myosin interaction of myofibrils. Failure ADP level rises.29 The increased level of free ADP
to produce an adequate amount of energy causes inhibits the function of many intracellular en-
mechanical failure of the heart. zymes, causing failure of the heart’s contraction
mechanism. Thus, a metabolic derangement in the
Components of Cardiac Energy Metabolism cardiac myocyte can occur when phosphocreatine
Cardiac energy metabolism is complex (Fig. 1). levels fall and free ADP levels rise, even if ATP
The metabolic machinery has three main compo- levels remain unchanged.
nents. The first is substrate utilization — the use
of fuel that comes from food. This process entails Assessment of Cardiac Energy Metabolism
the cellular uptake of mainly free fatty acids and The various components of energy metabolism in
glucose, the breakdown of these components by the heart can be measured with the use of stan-
beta-oxidation and glycolysis, and the entry of the dard methods in myocardial specimens obtained
resulting intermediary metabolites into the Krebs during a biopsy or at the time of transplantation
cycle. The second component is oxidative phos- or in cardiac tissue from animals. The analysis of
phorylation — the production of energy by the ATP and phosphocreatine in tissue samples is
mitochondrial respiratory chain. The phosphory- problematic, however, because of the instability
lation of ADP by this mechanism produces the of these molecules.29 For this reason, the principal
high-energy phosphate compound ATP, which is method for measuring ATP and phosphocreatine
the direct source of energy for all energy-consum- is phosphorus-31 magnetic resonance (31P-MR)
ing reactions in the heart. The third component spectroscopy.33-36 This method can be used with
is ATP transfer and utilization — the transport of high-field magnets of up to 12.0 Tesla (a measure
energy to and its consumption by the heart’s of magnetic field strength) in rodents and with
motor, the myofibrils. This process entails an standard clinical magnetic resonance imaging
energy-transfer mechanism termed the creatine (MRI) systems (usually 1.5 Tesla) in humans. As
kinase energy shuttle.28-30 shown in Figure 2A, 31P-MR spectra yield peaks
for phosphocreatine and the three phosphorus
The Creatine Kinase System atoms of ATP (γ-ATP, α-ATP, and β-ATP) that are
In the third component of cardiac energy metabo- proportional to the concentrations of these me-
lism, ATP transfer and utilization, mitochondrial tabolites. The MRI system can obtain cine images
creatine kinase catalyzes the transfer of the high- of the heart at the same time for quantification
energy phosphate bond in ATP to creatine to form of cardiac function. The most powerful method
phosphocreatine and ADP. Phosphocreatine, a for assessing energy metabolism in heart failure
smaller molecule than ATP, rapid­ly diffuses from entails the in vivo assessment of turnover rates of
the mitochondria to the myofibrils, where myofi- glucose and fatty acids38-41 and rates of oxidative
brillar creatine kinase catalyzes the reformation phosphorylation42 and ATP transfer.34,36 An im-
of ATP from phosphocreatine. The free creatine, portant methodologic consideration is intracel-
formed by the removal of phosphate from phos- lular compartmentalization.43 Whether a cardiac
phocreatine, diffuses back to the mitochondria. myocyte functions normally cannot be determined
Creatine is produced by the liver and kidneys by measuring the average cellular level of ATP,
and transported to the heart, where it is taken up phosphocreatine, or ADP, but instead is deter-
by a specific plasma-membrane creatine trans- mined by their concentrations in the perimyofi-
porter31 against a 50-fold concentration gradient. brillar space and near the sarcoplasmic reticu-
Creatine kinase catalyzes the phosphorylation of lum and sarcolemmal ion pumps. No method is
about two thirds of the total creatine pool in the currently available to make such measurements;
heart to phosphocreatine, and the other one third therefore, they have to be extrapolated from glob-
remains as free creatine. A small amount of cre- al measurements.

n engl j med 356;11  www.nejm.org  march 15, 2007 1141

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Energy production: oxidative phosphorylation

∆µ H+
Transport of energy to and consumption
by the engine: ATP transfer and utilization
H+ H+ H+ H+

Creatine kinase energy shuttle


UCP
F0 ADP PCr ADP
ATP Pi
F
Respiratory synthase 1 Contractile
chain ADP work
I–IV ANT
Mitochondrion H2O
ATP
NAD O2 ATP Cr ATP
CKmito CKMM
NADH

CO2
Krebs cycle Acetyl-CoA Beta-oxidation

Pyruvate
Creatine
Inner membrane transporter
Outer membrane
Free fatty Triglycerides
acids
Sarcolemma Cr
Pyruvate Lactate

Glycolysis
Fatty acid
Glucose
transporter
Free fatty
GLUT acids

Fuel from food: substrate utilization Glucose

Figure 1. Cardiac Energy Metabolism.


Energy metabolism in the heart has three components. The first is substrate utilization (outlined in red), the cellular uptake of substrates
and their breakdown by beta-oxidation and glycolysis; these processes result in the formation of acetyl coenzyme A (CoA),COLOR which
FIGURE
is fed
Draft 7 02/22/07
into the Krebs cycle and produces NADH and carbon dioxide (CO2). The second component is oxidative phosphorylation (outlined in
Author Neubauer
blue), the production of energy. Respiratory-chain complexes I through IV transfer electrons from NADH to oxygen, Fig #
thereby1
creating a
proton electrochemical gradient (Δμ H+) across the inner mitochondrial membrane as well as NAD and water. This Title gradient Cardiacdrives the
Energy Metabolism
F1, F0 ATP synthase, which produces ATP by phosphorylating ADP. Uncoupling proteins (UCPs) cause mitochondria ME to produce
Hogan heat
DE to and Schwartz
rather than ATP. The third component is energy transfer and utilization (outlined in green), the transport of energy consumption
Artist KMK
by myofibrillar ATPase and other ATP-consuming reactions, such as sarcolemmal and sarcoplasmic reticulum ion pumps. AUTHORATP PLEASEtransfer
NOTE:
is achieved by the creatine kinase energy shuttle. Creatine, which is not produced in the heart, is taken up by theFigure
creatine transporter.
has been redrawn and type has been reset
Please check carefully
GLUT denotes glucose transporter, Pi inorganic phosphate, ANT adenine nucleotide translocase, PCr phosphocreatine, Cr free creatine,
Issue date 03/15/07
CKmito mitochondrial creatine kinase isoenzyme, and CKMM myofibrillar creatine kinase isoenzyme.

lism: substrate utilization, oxidative phosphoryla-


Der a nge men t of Energy tion, and high-energy phosphate metab­olism.
Me ta bol ism in He a r t Fa ilur e
The changes in cardiac energy metabolism in Substrate Utilization
heart failure are shown in Figure 3, which sum- Substrate utilization can become limiting for car-
marizes the findings in animal models19,44-57 and diac function in heart failure as a result of reduced
clinical studies of heart failure.58-68 Changes occur substrate uptake, oxidation, or both. This may
in all three components of cardiac energy metabo- also occur as a result of the change in the relative

1142 n engl j med 356;11  www.nejm.org  march 15, 2007

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

A B
DCM, PCr:ATP <1.0 50

40 PCr:ATP <1.6

Total Mortality (%)


30
DCM, PCr:ATP <1.6 P=0.034
20

10 PCr:ATP >1.6

DCM, PCr:ATP >1.6 0


0 500 1000 1500
Follow-up (days)

2,3-DPG C

Normal Cardiac Function


PCr

α β-ATP
γ
PDE

Normal Heart Dilated Cardiomyopathy


10 0 −10 −20
Relative Magnetic Resonance Frequency (ppm)

Figure 2. The Phosphocreatine:ATP Ratio in Heart Failure.


Panel A shows cardiac 31P-MR spectra in (from bottom to top) a healthy subject, a patient with dilated cardiomyop-
athy (DCM) and a normal phosphocreatine (PCr):ATP ratio (>1.6; 1.6 was the median of the ratio), a patient with
DCM and a reduced PCr:ATP ratio ICM (<1.6),AUTHOR:
and a patient with DCM and a RETAKE
Neubuer severely reduced
1st PCr:ATP ratio (<1.0). The
patient with the severely reduced ratio 2nd
REG Fdied 7 days2 after
FIGURE: of 3 undergoing magnetic resonance examination. 2,3-DPG de-
3rd
notes 2,3-diphosphoglycerate, PDECASE phosphodiesters, and γ, α, and β phosphorus Revised
atoms of ATP. Panel B shows a
Kaplan–Meier life-table analysis ofEMail
mortality in two groupsLine of patients
4-C with DCM:
SIZE one with a higher PCr:ATP ratio
ARTIST:
and one with a lower ratio. Patients with an initiallyts low ratio
H/Thad anH/T
increased33p9
mortality over the study period (aver-
Enon
age follow-up, 2.5 years). Data are from Neubauer et al.37 Panel ComboC shows short-axis cine MRI scans of a normal
heart and the severely dilated heart of a patient with
AUTHOR, DCM.PLEASE NOTE:
Figure has been redrawn and type has been reset.
Please check carefully.

contributions of fatty acids (60JOB: to 90%) and glu-


35611 separate the changes
ISSUE: 03-15-06 in the metabolic pathway
cose (10 to 40%) to ATP synthesis. Studies of sub- capacities that are inherent in the heart muscle
strate utilization in heart failure have yielded con- from the indirect changes in the myocardium that
flicting results, but most indicate that fatty acid are due to the altered metabolic milieu.19
utilization, which is unchanged or slightly in-
creased in early heart failure,19,44 is substantial­ly Oxidative Phosphorylation
decreased in advanced heart failure.45 Changes Impaired oxidative phosphorylation can reduce
in glucose utilization are also inconsistent, but cardiac function by providing an insufficient sup­
many studies show that it is increased early in ply of ATP to cardiac myocytes. In heart failure,
heart failure.46,47 In advanced heart failure, insu- cardiac mitochondria have structural abnormali-
lin resistance develops in the myocardium, and ties and are probably increased in number.49 The
most studies have shown a decline in glucose activity of electron transport–chain complexes
utilization.58-60 However, the interpretation of and ATP synthase capacity are reduced50,61,69; the
these results is complicated by the substantial in- regulation of oxidative phosphorylation by the
creases in the concentrations of plasma free fatty phosphate acceptors ADP, AMP, and creatine is
acids, glucose, and insulin that are common in impaired41; and the levels of uncoupling proteins
heart failure. These increases make it difficult to (which cause mitochondria to produce heat rather

n engl j med 356;11  www.nejm.org  march 15, 2007 1143

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

than ATP) may be increased.70 These changes re-


Normal Heart Failure
sult in a substantial reduction of oxygen consump-
tion and energy production in the failing myo-
A cardium.
Fatty acid utilization
High-Energy Phosphate Metabolism
Glucose utilization Impaired ATP transfer and utilization may limit
contractile function by means of a decrease in the
PPARα average ATP concentration, a reduction in the
ATP transfer capacity through creatine kinase so
B that insufficient high-energy phosphate bonds are
Respiratory-chain complexes transported from the mitochondria to the myo-
fibrils, or an increase in the concentration of
ATP synthase free ADP.
Myocardial ATP levels remain normal (at ap-
Oxygen consumption
proximately 10 mmol per liter) until the advanced
Uncoupling proteins stages of heart failure, when they decrease by no
more than 30 to 40%.65,66,71 The average ATP
PCG-1α levels remain far above those required for ATP-
consuming reactions such as myosin–ATPase, and
C do not limit contractile function in heart failure.
However, both phosphocreatine and total creatine
ATP
levels decrease at earlier stages and to a greater
Phosphocreatine extent (by 30 to 70%).66,67 Down-regulation of the
creatine transporter function contributes to the
Total creatine reduced total creatine, and thus phosphocreatine,
levels in heart failure.72,73
Creatine transporter
There are profound changes in the creatine
Total creatine kinase activity
kinase system in heart failure.67,74-78 Mitochon-
drial creatine kinase activity may be reduced to
CKmito as little as 20% of normal activity, and myofibril-
lar creat­ine kinase activity can decrease by up to
CKMM 50% as compared with normal values. The losses
of high-energy phosphates and creatine kinase
ATP transfer
activity cause a severe decline in ATP trans-
Free ADP fer53,54,79,80 — that is, a decrease in energy flux
within the cell — and thus a reduction in energy
Figure 3. Changes in Cardiac Energy Metabolism in Heart Failure. delivery to the myofibrils by up to 71%.81 This
In patients with heart failure, changes in substrate utilization (Panel A) in- metabolic abnormality may contribute to contrac-
AUTHOR:
clude initial up-regulation
ICM and Neubauer
subsequent reductionRETAKEof glucose1stutilization tile dysfunction and particularly to the loss of
FIGURE: 3 of 3 2nd
and a decreaseREGin Ffatty acid utilization, in part mediated by down-regulation
3rd inotropic reserve that is characteristic of the myo-
of peroxisomeCASEproliferator–activated receptor α (PPARα). Oxidative
Revised phos- cardium in heart failure.
phorylation changes
EMail (Panel B) are characterized by decreased
Line 4-C SIZE energy pro-
duction, with reductions ARTIST: ts H/T
in oxygen consumption H/Tand respiratory-chain and
When the failing heart is stimulated with
Enon 22p3
ATP synthase activity, in part mediatedCombo by down-regulation of PPARα coacti- catecholamines, thereby causing high-workload
vator 1α (PCG-1α). ChangesAUTHOR, PLEASEphosphate
in high-energy NOTE: metabolism (Panel C) conditions, the free ADP concentration increases
Figure has been redrawn and type has been reset.
include a severely impaired creatine kinase
Please check energy-transfer mechanism, in-
carefully. to a value that is approximately twice that in
creased free ADP levels, and, in advanced heart failure, reduced ATP content. normal myocardium.82 The increase of free ADP
Free ADP isJOB:
calculated
35611 from the creatine kinase equilibrium assumption:
ISSUE: 03-15-06
ADP = ([ATP] × [creatine]) ÷ ([phosphocreatine] × [H+] × Keq), where H+ is the
in the relevant microcompartments (the perimyo-
intracellular hydrogen ion concentration and Keq is the equilibrium constant fibrillar microcompartment and the microcom-
of the creatine kinase reaction. CKmito denotes mitochondrial creatine kinase partments near the sarcoplasmic reticulum and
isoenzyme, and CKMM myofibrillar creatine kinase isoenzyme. sarcolemmal ion pumps) during high-workload
conditions limits the contractile reserve of the fail­

1144 n engl j med 356;11  www.nejm.org  march 15, 2007

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

ing heart, and this reduction in inotropic reserve Nuclear-Receptor Transcription Factors
is manifested clinically as dyspnea on exertion. Several nuclear-receptor transcription factors are
Most of the evidence concerning the derange- activated by lipid metabolites in a manner analo-
ment of myocardial energetics in heart failure in gous to the activation of nuclear receptors by ste-
humans is based on studies with 31P-MR spec- roid hormones. These transcription factors rapidly
troscopy. This method can be used to determine couple gene expression with a changing substrate
the ratio of phosphocreatine to ATP, which is a milieu, and they typically require coactivator pro-
powerful index of the energetic state of the heart. teins for their action. Among these transcription
The creatine kinase reaction equilibrium favors factors, the most widely studied are the nuclear
ATP synthesis over phosphocreatine synthesis by receptors of the peroxisome proliferator–activated
a factor of approximately 100. Therefore, when- receptor (PPAR) family, which comprises three
ever the demand for ATP outstrips ATP synthe- isoforms: PPARα, PPARβ, and PPARγ. All three
sis, phosphocreatine levels decline first, and ATP affect cardiac lipid metabolism, but the primary
decreases only when phosphocreatine is substan- regulator appears to be PPARα, which controls the
tially depleted. In chronic heart failure, however, expression of enzymes directly involved in fatty
a second mechanism comes into play: the total acid oxidation. In cardiac hypertrophy in both
creatine level falls, and this reduction further animal models88 and humans,89,90 the expres­sion
decreases the phosphocreatine:ATP ratio62,63,68 of PPARα is decreased in proportion to the de-
(Fig. 2). Myocardial phosphocreatine:ATP ratios pression of fatty acid utilization. For this reason,
are reduced in heart failure, and they correlate the down-regulation of PPARα is thought to be
with New York Heart Association classes63 and the main mechanism underlying the switch in sub­
with indexes of systolic83 and diastolic84 function. strate utilization from fatty acids to glucose. This
One study of 39 patients with dilated cardiomyop­ switch is typical of the hypertrophied heart.
athy indicated that the phosphocreatine:ATP ratio A nuclear-receptor coactivator, PPARγ coacti-
may be a stronger predictor of both total mortal- vator-1 (also known as PCG-1α), is a master regu-
ity and mortality attributable to cardiovascular lator of metabolic function in mitochondria. It
disease than functional or clinical indexes37 activates multiple genes that are responsible for
(Fig. 2B), but this finding requires confirmation fatty acid uptake and oxidation and for oxidative
in larger clinical trials. phosphorylation.87 These genes include PPARα and
Hypertrophic cardiomyopathy is an exemplar PPARβ and nuclear respiratory factors 1 and 2.
of myocardial energy depletion.85 In patients with Experimental studies suggest that the inhibition
hypertrophic cardiomyopathy, the cardiac phos- of PCG-1α,91,92 probably as a direct consequence
phocreatine:ATP ratio is reduced through a range of high plasma catecholamine levels,93 leads to
of specific mutations that affect the sarcomere, down-regulation of mitochondrial gene expres-
whether or not left ventricular hypertrophy is sion.20 In this way, it contributes to the impair-
present.86 Because the abnormalities in energetics ment of oxidative phosphorylation in the failing
are an early and integral part of the primary heart heart. The development of heart failure is acceler-
muscle disease, one can infer that the compro- ated by PCG-1α deficiency,93 suggesting that this
mise of myocardial energetics has a causal role coactivator may have a cardioprotective function.
in hypertrophic cardiomyopathy. Despite these advances, more work is needed
to fully understand which changes in metabolic
signaling are adaptive, maladaptive, or both (de-
Mol ecul a r R egul at or s
of Energy Me ta bol ism pending on the stage of heart failure). Further-
more, the molecular regulators of changes in
The energy demands of the heart vary widely dur- creatine transport and creatine kinase expression
ing cardiac development and with physiologic or in heart failure are unknown.
abnormal stress. Energy production must be close­
ly coupled with energy demand, but the heart has gene-K no ckou t Model s a nd
little capacity for substrate storage. However, there L os s- of -F unc t ion Mu tat ions
are mechanisms that induce the expression of
genes that encode the molecular regulators of The causal role of altered energetics in heart fail-
energy metabolism.87 ure has been controversial for decades, and this

n engl j med 356;11  www.nejm.org  march 15, 2007 1145

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

controversy remains unsettled. A promising way Direct manipulation of substrate utilization is


to obtain definitive answers is through studies of feasible with the use of partial inhibitors of fatty
genetically manipulated mice with selective knock­ acid oxidation or carnitine palmitoyl transferase 1
out (loss of function) of genetic components of inhibitors. These compounds have complex types
the metabolic machinery or of single-gene inborn of action,19,21,23 but they all partially inhibit fatty
errors of metabolism in humans. Table 1 lists the acid utilization and promote glucose utilization.
genetic abnormalities that have been studied in Whether the suppression of fatty acid oxidation is
mouse models94-103 and humans,104-108 along with beneficial or detrimental in heart failure is high­
the corresponding metabolic and functional car- ly controversial, and the cause or stage of heart
diac phenotypes. The deletion of a variety of genes failure may dictate the outcome of this kind of
that encode specific metabolic components re- treatment. Regardless of the theoretical argu-
lated to substrate utilization, oxidative phosphory- ments, a number of recent proof-of-principle clini­
lation, and high-energy phosphates causes a loss cal studies have suggested that partial inhibition
of contractile reserve, overt heart failure, cardiac of fatty acid oxidation is promising. For example,
hypertrophy, tachyarrhythmias, or bradyarrhyth- treatment with trimetazidine, an inhibitor of fatty
mias. These genetic studies show that a fully inte- acid oxidation, improved left ventricular function
grated metabolic machine is important for nor- over a period of 6 months in elderly patients113;
mal cardiac function and that selective ablation an 18-month study confirmed this finding in pa­
of components of energy metabolism can cause tients with heart failure due to a previous myo-
early or advanced heart failure. cardial infarction.114 Small, single-center, and
The strength of these genetic studies, however, thus far unconfirmed studies have shown that in
is also their weakness, because chronic heart patients with heart failure of ischemic or non­
failure is multifactorial and entails many mecha- ischemic origin, 2 months of treatment with the
nisms other than those controlled by the single fatty acid oxidation inhibitor perhexiline115 or
gene under study. Furthermore, we do not under- 3 months of treatment with the carnitine palmi-
stand how a gene encoding a highly conserved toyl transferase 1 inhibitor etoxomir116 improved
protein with a central role in cardiac energetics the left ventricular ejection fraction. The results
can be deleted and yet not result in overt heart of these small studies have to be interpreted cau-
failure (Table 1). Whether and to what extent ad- tiously. Some were not conducted under random-
aptations occur in response to the deletion of an ized, blinded, or placebo-controlled conditions,
essential metabolic component are unknown. and others included patients with angina, which
may in part explain the beneficial effect of in-
hibiting fatty acid oxidation. Nevertheless, they
Impl ic at ions for the T r e atmen t
of He a r t Fa ilur e provide support for the results of studies of the
effects of such inhibitors in animal models of
ACE inhibitors, diuretics, and beta-blockers may heart failure.117,118
have indirect metabolic effects on the heart,39,63,109 The effects of PPAR activators on cardiac sub-
but they do not directly affect energy metabo- strate utilization are complex. They include direct
lism. Could energy metabolism be a specific tar- up-regulation of fatty acid oxidation and its in-
get for therapy in patients with heart failure? direct down-regulation through reduced plasma
lipid levels. The findings that heart failure devel-
Modulation of Substrate Utilization ops in mice that overexpress PPARα119 and that
A promising strategy for metabolic intervention in PPAR activators may have beneficial,120 adverse,121
heart failure is to modulate substrate utilization. or no122 effects in animal models of heart failure
In a study of eight patients with heart failure, indicate that the actions of these compounds in
intracoronary infusion of pyruvate improved car- the failing heart need to be better understood
diac function in the short term,110 and in a dog before larger clinical trials can be considered.
model of heart failure, an increase in glucose uti-
lization by glucagon-like peptide 1 improved left Modulation of Oxidative Phosphorylation
ventricular function.111 In addition, in a mouse A second strategy to metabolic therapy in heart
model of heart failure, transgenic overexpression failure is direct stimulation of oxidative phosphor-
of glucose transporter 1 prevented the develop- ylation. Currently, however, there are no effective
ment of left ventricular dysfunction.112 stimulators of oxidative phosphorylation. Even so,

1146 n engl j med 356;11  www.nejm.org  march 15, 2007

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

Table 1. Mouse Gene-Knockout Models and Human Inborn Errors of Metabolism and Their Cardiac Phenotypes.*

Genetic Abnormality Metabolic Abnormality Cardiac Phenotype


Mouse gene-knockout models
Long-chain and very-long-chain acyl–CoA Inhibition of very-long-chain and long- Very-long-chain acyl–CoA dehydrogenase: ven-
dehydrogenase chain fatty acid beta-oxidation tricular tachycardia, severe bradycardia94
Long-chain acyl–CoA dehydrogenase: cardio-
myopathy, sudden death95
PPARα Substrate utilization switch from fatty Reduced contractile reserve and depletion of
­acids to glucose and lactate cardiac energy stores during inotropic chal-
lenge96
Glucose transporter 4 Increased basal but abolished insulin- Cardiac hypertrophy97
stimulated glucose transport
PCG-1α Reduced oxidative phosphorylation and Loss of contractile reserve98
fatty acid oxidation
Heart-specific Tfam, a nuclear-encoded Reduced activity of respiratory-chain Cardiac hypertrophy, dilatation, heart failure,
mitochondrial DNA replication complexes, reduced fatty acid oxida- conduction defects99
transcription factor tion, increased glucose utilization
Adenine nucleotide translocase 1 Impaired ADP-stimulated mitochondrial Cardiac hypertrophy100
respiration
Mitochondrial and myofibrillar CK iso­ Loss of mitochondrial CK, loss of mito- Hypertrophy and dilatation, impaired contrac-
enzymes chondrial and myofibrillar CK tile reserve, shortened diffusion distances
between mitochondria and myofibrils101,102
Guanidino acetate methyl transferase Deficient creatine biosynthesis, accumu- Loss of inotropic reserve, increased susceptibil-
lation of precursor guanidino acetate, ity to ischemia and reperfusion injury103
CK reaction velocity 1%
Human inborn errors of metabolism
Systemic carnitine deficiency Defective carnitine biosynthesis, trans- Dilated cardiomyopathy, cardiac arrest, cardio-
membrane transport, intestinal up- megaly — oral carnitine therapy reverses
take or tubular reabsorption phenotype104
Malonyl carboxylase deficiency Elevation of malonyl–CoA, a potent inhibi- Cardiomyopathy, decreased contractility, heart
tor of carnitine palmitoyl transferase 1 failure105
Carnitine palmitoyl transferase 2 Impaired mitochondrial acyl–CoA trans- Cardiac hypertrophy, fatal cardiomegaly,
deficiency port ­dysrhythmias106
Short-chain, medium-chain, long-chain, Dysfunction of enzymes of fatty acid beta- Short-chain acyl–CoA dehydrogenase: mild left
and very-long-chain acyl–CoA oxidation ventricular dysfunction, biatrial hypertrophy107
­dehydrogenase Medium-chain acyl–CoA dehydrogenase:
­cardiac involvement rare107
Long-chain and very-long-chain acyl–CoA
­dehydrogenase: severe dilated or hypertro-
phic cardiomyopathy107
Kearns–Sayre syndrome, MELAS, Barth syn- Various deficiencies of respiratory-chain Dilated cardiomyopathy, hypertrophic cardio-
drome: mutations of mitochondrial complexes myopathy, conduction defects, ventricular
DNA; Leigh’s syndrome: mutation ectopy107,108
of mitochondrial or nuclear DNA

* Many of these deficiency models and syndromes also have extracardiac manifestations not listed here. CoA denotes coenzyme A, CK cre-
atine kinase, and MELAS mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes.

increasing PCG-1α activity as a means of up-regu- Manipulation of High-Energy Phosphate


lating oxidative phosphorylation enzymes may be Metabolites
a promising approach.93 An alternative is to re- A third strategy for metabolic intervention is the
duce free fatty acid levels, which should repress direct manipulation of high-energy phosphate
mitochondrial uncoupling proteins, thereby in- stores, their availability, or the efficiency of their
creasing ATP synthesis. utilization. Creatine and phosphocreatine levels

n engl j med 356;11  www.nejm.org  march 15, 2007 1147

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

can be augmented by increasing the creatine suggest new molecular targets for therapeutic in-
transporter function.123 Although massive in- tervention. New metabolic modulator compounds
creases in the creatine transporter function are need to be developed by academia and industry.
detrimental (because a substantially supranormal Proof-of-principle clinical studies may use the myo­
creatine level increases the free ADP level), future cardial phosphocreatine:ATP ratio to monitor the
studies will show whether reversing the decrease early energetic response of the heart to metabolic
in creatine and phosphocreatine levels by moder- therapy, and this method may provide a surro-
ate stimulation of creatine transporter activity is gate marker of long-term prognostic effects. Final­
beneficial in heart failure. Finally, it may be fea- ly, large-scale clinical trials will have to prove or
sible to improve the myofibrillar efficiency of disprove the clinical efficacy of metabolic modu-
ATP utilization with new calcium-sensitizing124 lators. There is substantial hope that such a com-
or myosin activator compounds. bined effort will lead to new therapies targeted at
cardiac energetics. These therapies may improve
C onclusions the symptoms and prognosis of patients with the
life-threatening illness of chronic heart failure.
Metabolic therapy is a promising new avenue for Supported by grants from the British Heart Foundation and
the treatment of heart failure, and suitable targets the Medical Research Council, London.
for therapy are substrate utilization, oxidative Dr. Neubauer reports serving as a consultant to GlaxoSmith-
Kline and receiving grant support from Siemens Medical Solu-
phosphorylation, and the availability of high- tions. No other potential conflict of interest relevant to this article
energy phosphates. A multipronged effort is need- was reported.
ed to fully investigate this concept. Experimental I thank Kieran Clarke, Joanne S. Ingwall, Rong Tian, and Hugh
Watkins for their review of the manuscript and helpful sugges-
studies will, for example, further clarify the mech- tions and all the colleagues, research fellows, students, and tech-
anisms leading to energetic derangement and will nicians who have worked on cardiac energetics with me.

References
1. Heart disease and stroke statistics: al. Cardiac-resynchronization therapy with 19. Stanley WC, Recchia FA, Lopaschuk
2005 update. Dallas: American Heart As- or without an implantable defibrillator in GD. Myocardial substrate metabolism in
sociation, 2005. advanced chronic heart failure. N Engl J the normal and failing heart. Physiol Rev
2. Dayer M, Cowie MR. Heart failure: Med 2004;350:2140-50. 2005;85:1093-129.
diagnosis and healthcare burden. Clin 10. Cleland JGF, Daubert J-C, Erdmann E, 20. Ventura-Clapier R, Garnier A, Veksler
Med 2004;4:13-8. et al. The effect of cardiac resynchroniza- V. Energy metabolism in heart failure.
3. McMurray JJ, Pfeffer MA. Heart failure. tion on morbidity and mortality in heart J Physiol 2004;555:1-13.
Lancet 2005;365:1877-89. failure. N Engl J Med 2005;352:1539-49. 21. Morrow DA, Givertz MM. Modulation
4. The CONSENSUS Trial Study Group. 11. Mann DL, Bristow MR. Mechanisms of myocardial energetics: emerging evi-
Effects of enalapril on mortality in severe and models in heart failure: the biome- dence for a therapeutic target in cardio-
congestive heart failure: results of the Co- chanical model and beyond. Circulation vascular disease. Circulation 2005;112:
operative North Scandinavian Enalapril 2005;111:2837-49. [Erratum, Circulation 3218-21.
Survival Study (CONSENSUS). N Engl J 2005;112(4):e75.] 22. Lopaschuk GD, Rebeyka IM, Allard
Med 1987;316:1429-35. 12. Herrmann G, Decherd GM. The chem- MF. Metabolic modulation: a means to
5. Pfeffer MA, Braunwald E, Moyé LA, et ical nature of heart failure. Ann Intern mend a broken heart. Circulation 2002;
al. Effect of captopril on mortality and Med 1939;12:1233-44. 105:140-2.
morbidity in patients with left ventricular 13. Wollenberger A. On the energy-rich 23. Essop MF, Opie LH. Metabolic therapy
dysfunction after myocardial infarction: phosphate supply of the failing heart. Am for heart failure. Eur Heart J 2004;25:
results of the Survival and Ventricular En- J Physiol 1947;150:733-6. 1765-8.
largement Trial. N Engl J Med 1992;327: 14. Olson RE, Schwartz WB. Myocardial 24. van Bilsen M, Smeets PJ, Gilde AJ,
669-77. metabolism in congestive heart failure. van der Vusse GJ. Metabolic remodelling
6. Pitt B, Zannad F, Remme WJ, et al. Medicine (Baltimore) 1951;30:21-41. of the failing heart: the cardiac burn-out
The effect of spironolactone on morbidity 15. Olson RE. Myocardial metabolism in syndrome? Cardiovasc Res 2004;61:218-
and mortality in patients with severe heart congestive heart failure. J Chronic Dis 26.
failure. N Engl J Med 1999;341:709-17. 1959;9:442-64. 25. Cohn JN, Tognoni G. A randomized
7. CIBIS Investigators and Committees. 16. Ingwall JS, Weiss RG. Is the failing trial of the angiotensin-receptor blocker
A randomized trial of β-blockade in heart heart energy starved? On using chemical valsartan in chronic heart failure. N Engl
failure: the Cardiac Bisoprolol Insuffi- energy to support cardiac function. Circ J Med 2001;345:1667-75.
ciency Study (CIBIS). Circulation 1994;90: Res 2004;95:135-45. 26. Pfeffer MA, Swedberg K, Granger CB,
1765-73. 17. Taegtmeyer H. Metabolism — the lost et al. Effects of candesartan on mortality
8. Packer M, Bristow MR, Cohn JN, et al. child of cardiology. J Am Coll Cardiol and morbidity in patients with chronic
The effect of carvedilol on morbidity and 2000;36:1386-8. heart failure: the CHARM-Overall pro-
mortality in patients with chronic heart 18. Idem. Cardiac metabolism as a target gramme. Lancet 2003;362:759-66.
failure. N Engl J Med 1996;334:1349-55. for the treatment of heart failure. Circula- 27. Katz AM. Heart failure: pathophysiol-
9. Bristow MR, Saxon LA, Boehmer J, et tion 2004;110:894-6. ogy, molecular biology and clinical man-

1148 n engl j med 356;11  www.nejm.org  march 15, 2007

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

agement. Philadelphia: Lippincott Wil- ing and expression for mitochondrial mem­ tional and bioenergetic consequences of
liams & Wilkins, 2000. brane proteins during left ventricular re- postinfarction left ventricular remodeling
28. Bessman SP, Geiger PJ. Transport of modeling and contractile failure after in a new porcine model: MRI and 31 P-MRS
energy in muscle: the phosphorylcreatine myocardial infarction. J Am Coll Cardiol study. Circulation 1996;94:1089-100.
shuttle. Science 1981;211:448-52. 2000;36:282-7. 57. Zhang J, Toher C, Erhard M, et al. Re-
29. Ingwall JS. ATP and the heart. Nor- 43. Gudbjarnason S, Mathes P, Ravens lationships between myocardial bioener-
well, MA: Kluwer Academic, 2002. KG. Functional compartmentation of ATP getic and left ventricular function in hearts
30. Wallimann T, Wyss M, Brdiczka D, and creatine phosphate in heart muscle. with volume-overload hypertrophy. Circu-
Nicolay K, Eppenberger HM. Intracellular J Mol Cell Cardiol 1970;1:325-39. lation 1997;96:334-43.
compartmentation, structure and function 44. Chandler MP, Kerner J, Huang H, et al. 58. Kalsi KK, Smolenski RT, Pritchard RD,
of creatine kinase isoenzymes in tissues Moderate severity heart failure does not Khaghani A, Seymour AM, Yacoub MH.
with high and fluctuating energy demands: involve a downregulation of myocardial Energetics and function of the failing hu-
the ‘phosphocreatine circuit’ for cellular fatty acid oxidation. Am J Physiol Heart man heart with dilated or hypertrophic
energy homeostasis. Biochem J 1992;281: Circ Physiol 2004;287:H1538-H1543. cardiomyopathy. Eur J Clin Invest 1999;29:
21-40. 45. Osorio JC, Stanley WC, Linke A, et al. 469-77.
31. Guimbal C, Kilimann MWA. A Na(+)- Impaired myocardial fatty acid oxidation 59. Razeghi P, Young ME, Alcorn JL,
dependent creatine transporter in rabbit and reduced protein expression of retinoid Moravec CS, Frazier OH, Taegtmeyer H.
brain, muscle, heart, and kidney: cDNA X receptor-alpha in pacing-induced heart Metabolic gene expression in fetal and
cloning and functional expression. J Biol failure. Circulation 2002;106:606-12. failing human heart. Circulation 2001;104:
Chem 1993;268:8418-21. 46. Remondino A, Rosenblatt-Velin N, 2923-31.
32. Wyss M, Wallimann T. Creatine me- Montessuit C, et al. Altered expression of 60. Taylor M, Wallhaus TR, Degrado TR,
tabolism and the consequences of cre- proteins of metabolic regulation during et al. An evaluation of myocardial fatty
atine depletion in muscle. Mol Cell Bio- remodeling of the left ventricle after myo- acid and glucose uptake using PET with
chem 1994;133-134:51-66. cardial infarction. J Mol Cell Cardiol [18F]fluoro-6-thia-heptadecanoic acid and
33. Garlick PB, Radda GK, Seeley PJ. Phos- 2000;32:2025-34. [18F]FDG in patients with congestive
phorus NMR studies on perfused heart. 47. Nascimben L, Ingwall JS, Lorell BH, heart failure. J Nucl Med 2001;42:55-62.
Biochem Biophys Res Commun 1977;74: et al. Mechanisms for increased glycolysis 61. Quigley AF, Kapsa RM, Esmore D,
1256-62. in the hypertrophied rat heart. Hyperten- Hale G, Byrne E. Mitochondrial respira-
34. Ingwall JS. Phosphorus nuclear mag- sion 2004;44:662-7. tory chain activity in idiopathic dilated
netic resonance spectroscopy of cardiac 48. Murakami Y, Zhang Y, Cho YK, et al. cardiomyopathy. J Card Fail 2000;6:47-55.
and skeletal muscles. Am J Physiol 1982; Myocardial oxygenation during high work 62. Hardy CJ, Weiss RG, Bottomley PA,
242:H729-H744. states in hearts with postinfarction re- Gerstenblith G. Altered myocardial high-
35. Bottomley PA. MR spectroscopy of the modeling. Circulation 1999;99:942-8. energy phosphate metabolites in patients
human heart: the status and the chal- 49. Ide T, Tsutsui H, Hayashidani S, et al. with dilated cardiomyopathy. Am Heart J
lenges. Radiology 1994;191:593-612. Mitochondrial DNA damage and dysfunc- 1991;122:795-801.
36. Neubauer S. Cardiac magnetic reso- tion associated with oxidative stress in 63. Neubauer S, Krahe T, Schindler R, et
nance spectroscopy. In: Lardo AC, Fayad failing hearts after myocardial infarction. al. 31P magnetic resonance spectroscopy
ZA, Chronos NAF, Fuster V, eds. Cardio- Circ Res 2001;88:529-35. in dilated cardiomyopathy and coronary
vascular magnetic resonance: established 50. Marin-Garcia J, Goldenthal MJ, Moe artery disease: altered cardiac high-ener-
and emerging applications. London: Mar- GW. Abnormal cardiac and skeletal muscle gy phosphate metabolism in heart failure.
tin Dunitz, 2003:39-60. mitochondrial function in pacing-induced Circulation 1992;86:1810-8.
37. Neubauer S, Horn M, Cramer M, et al. cardiac failure. Cardiovasc Res 2001;52: 64. Regitz V, Fleck E. Myocardial adenine
Myocardial phosphocreatine-to-ATP ratio 103-10. nucleotide concentrations and myocardial
is a predictor of mortality in patients with 51. Pool PE, Spann JF Jr, Buccino RA, norepinephrine content in patients with
dilated cardiomyopathy. Circulation 1997; Sonnenblick EH, Braunwald E. Myocardial heart failure secondary to idiopathic di-
96:2190-6. high energy phosphate stores in cardiac lated or ischemic cardiomyopathy. Am J
38. Vitale GD, deKemp RA, Ruddy TD, hypertrophy and heart failure. Circ Res Cardiol 1992;69:1574-80.
Williams K, Beanlands RS. Myocardial 1967;21:365-73. 65. Starling RC, Hammer DF, Altschuld
glucose utilization and optimization of 52. Bittner V, Reeves RC, Digerness SB, RA. Human myocardial ATP content and
(18)F-FDG PET imaging in patients with Caulfield JB, Pohost GM. 31P NMR spec- in vivo contractile function. Mol Cell Bio-
non-insulin-dependent diabetes mellitus, troscopy in chronic adriamycin cardiotox- chem 1998;180:171-7.
coronary artery disease, and left ventric- icity. Magn Reson Med 1991;17:69-81. 66. Beer M, Seyfarth T, Sandstede J, et al.
ular dysfunction. J Nucl Med 2001;42: 53. Nascimben L, Friedrich J, Liao R, Pau- Absolute concentrations of high-energy
1730-6. letto P, Pessina AC, Ingwall JS. Enalapril phosphate metabolites in normal, hyper-
39. Wallhaus TR, Taylor M, DeGrado TR, treatment increases cardiac performance trophied, and failing human myocardium
et al. Myocardial free fatty acid and glu- and energy reserve via the creatine kinase measured noninvasively with (31)P-SLOOP
cose use after carvedilol treatment in pa- reaction in myocardium of Syrian myo- magnetic resonance spectroscopy. J Am
tients with congestive heart failure. Cir- pathic hamsters with advanced heart fail- Coll Cardiol 2002;40:1267-74.
culation 2001;103:2441-6. ure. Circulation 1995;91:1824-33. 67. Nascimben L, Ingwall JS, Pauletto P,
40. Davila-Roman VG, Vedala G, Herrero 54. Neubauer S, Horn M, Naumann A, et et al. Creatine kinase system in failing
P, et al. Altered myocardial fatty acid and al. Impairment of energy metabolism in and nonfailing human myocardium. Cir-
glucose metabolism in idiopathic dilated intact residual myocardium of rat hearts culation 1996;94:1894-901.
cardiomyopathy. J Am Coll Cardiol 2002; with chronic myocardial infarction. J Clin 68. Conway MA, Allis J, Ouwerkerk R, Ni-
40:271-7. Invest 1995;95:1092-100. ioka T, Rajagopalan B, Radda GK. Detec-
41. Lewandowski ED. Cardiac carbon 13 55. Zhang J, Merkle H, Hendrich K, et al. tion of low phosphocreatine to ATP ratio
magnetic resonance spectroscopy: on the Bioenergetic abnormalities associated with in failing hypertrophied human myocar-
horizon or over the rainbow? J Nucl Car- severe left ventricular hypertrophy. J Clin dium by 31P magnetic resonance spectros-
diol 2002;9:419-28. Invest 1993;92:993-1003. copy. Lancet 1991;338:973-6.
42. Ning XH, Zhang J, Liu J, et al. Signal- 56. Zhang J, Wilke N, Wang Y, et al. Func- 69. Casademont J, Miro O. Electron trans-

n engl j med 356;11  www.nejm.org  march 15, 2007 1149

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

port chain defects in heart failure. Heart 84. Lamb HJ, Beyerbacht HP, van der 98. Arany Z, He H, Lin J, et al. Transcrip-
Fail Rev 2002;7:131-9. Laarse A, et al. Diastolic dysfunction in tional coactivator PGC-1 alpha controls the
70. Murray AJ, Anderson RE, Watson GC, hypertensive heart disease is associated energy state and contractile function of
Radda GK, Clarke K. Uncoupling proteins with altered myocardial metabolism. Cir- cardiac muscle. Cell Metab 2005;1:259-71.
in human heart. Lancet 2004;364:1786-8. culation 1999;99:2261-7. 99. Hansson A, Hance N, Dufour E, et al.
71. Shen W, Asai K, Uechi M, et al. Pro- 85. Ashrafian H, Redwood C, Blair E, A switch in metabolism precedes increased
gressive loss of myocardial ATP due to a Watkins H. Hypertrophic cardiomyopa- mitochondrial biogenesis in respiratory
loss of total purines during the develop- thy: a paradigm for myocardial energy chain-deficient mouse hearts. Proc Natl
ment of heart failure in dogs: a compen- depletion. Trends Genet 2003;19:263-8. Acad Sci U S A 2004;101:3136-41.
satory role for the parallel loss of creatine. 86. Crilley JG, Boehm EA, Blair E, et al. 100. Graham BH, Waymire KG, Cottrell
Circulation 1999;100:2113-8. Hypertrophic cardiomyopathy due to sar- B, Trounce IA, MacGregor GR, Wallace
72. Ten Hove M, Chan S, Lygate C, et al. comeric gene mutations is characterized DC. A mouse model for mitochondrial
Mechanisms of creatine depletion in by impaired energy metabolism irrespec- myopathy and cardiomyopathy resulting
chronically failing rat heart. J Mol Cell tive of the degree of hypertrophy. J Am from a deficiency in the heart/muscle iso-
Cardiol 2005;38:309-13. Coll Cardiol 2003;41:1776-82. form of the adenine nucleotide transloca-
73. Neubauer S, Remkes H, Spindler M, et 87. Huss JM, Kelly DP. Nuclear receptor tor. Nat Genet 1997;16:226-34.
al. Downregulation of the Na(+)-creatine signaling and cardiac energetics. Circ Res 101. Nahrendorf M, Spindler M, Hu K, et
cotransporter in failing human myocar- 2004;95:568-78. al. Creatine kinase knockout mice show
dium and in experimental heart failure. 88. Barger PM, Brandt JM, Leone TC, left ventricular hypertrophy and dilatation,
Circulation 1999;100:1847-50. Weinheimer CJ, Kelly DP. Deactivation of but unaltered remodeling post-myocardial
74. Vatner DE, Ingwall JS. Effects of mod- peroxisome proliferator-activated receptor- infarction. Cardiovasc Res 2005;65:419-27.
erate pressure overload cardiac hypertro- alpha during cardiac hypertrophic growth. 102. Kaasik A, Veksler V, Boehm E, Novo-
phy on the distribution of creatine kinase J Clin Invest 2000;105:1723-30. tova M, Minajeva A, Ventura-Clapier R.
isozymes. Proc Soc Exp Biol Med 1984;175: 89. Sack MN, Rader TA, Park S, Bastin J, Energetic crosstalk between organelles:
5-9. McCune SA, Kelly DP. Fatty acid oxidation architectural integration of energy pro-
75. Ingwall JS, Kramer MF, Fifer MA, et enzyme gene expression is downregulat- duction and utilization. Circ Res 2001;89:
al. The creatine kinase system in normal ed in the failing heart. Circulation 1996; 153-9.
and diseased human myocardium. N Engl 94:2837-42. 103. ten Hove M, Lygate CA, Fischer A, et
J Med 1985;313:1050-4. 90. Karbowska J, Kochan Z, Smolenski RT. al. Reduced inotropic reserve and increased
76. Laser A, Ingwall JS, Tian R, et al. Re- Peroxisome proliferator-activated receptor susceptibility to cardiac ischemia/reper-
gional biochemical remodeling in non- alpha is downregulated in the failing hu- fusion injury in phosphocreatine-deficient
infarcted tissue of rat heart post-myocar- man heart. Cell Mol Biol Lett 2003;8:49-53. guanidinoacetate-N-methyltransferase-
dial infarction. J Mol Cell Cardiol 1996;28: 91. Garnier A, Fortin D, Delomenie C, knockout mice. Circulation 2005;111:2477-
1531-8. Momken I, Veksler V, Ventura-Clapier R. 85.
77. Khuchua ZA, Ventura-Clapier R, Kuz­ Depressed mitochondrial transcription fac­ 104. Chapoy PR, Angelini C, Brown WJ,
netsov AV, Grishin MN, Saks VA. Altera- tors and oxidative capacity in rat failing Stiff JE, Shug AL, Cederbaum SD. System-
tions in the creatine kinase system in the cardiac and skeletal muscles. J Physiol ic carnitine deficiency — a treatable in-
myocardium of cardiomyopathic hamsters. 2003;551:491-501. herited lipid-storage disease presenting
Biochem Biophys Res Commun 1989;165: 92. Finck BN, Kelly DP. PGC-1 coactiva- as Reye’s syndrome. N Engl J Med 1980;
748-57. tors: inducible regulators of energy metab­ 303:1389-94.
78. De Sousa E, Veksler V, Minajeva A, et olism in health and disease. J Clin Invest 105. Matalon R, Michaels K, Kaul R, et al.
al. Subcellular creatine kinase alterations: 2006;116:615-22. Malonic aciduria and cardiomyopathy.
implications in heart failure. Circ Res 93. Arany Z, Novikov M, Chin S, Ma Y, J Inherit Metab Dis 1993;16:571-3.
1999;85:68-76. Rosenzweig A, Spiegelman BM. Trans- 106. Hug G, Bove KE, Soukup S. Lethal
79. Ye Y, Gong G, Ochiai K, Liu J, Zhang J. verse aortic constriction leads to acceler- neonatal multiorgan deficiency of carni-
High-energy phosphate metabolism and ated heart failure in mice lacking PPAR- tine palmitoyltransferase II. N Engl J Med
creatine kinase in failing hearts: a new gamma coactivator 1alpha. Proc Natl Acad 1991;325:1862-4.
porcine model. Circulation 2001;103:1570- Sci U S A 2006;103:10086-91. 107. Guertl B, Noehammer C, Hoefler G.
6. 94. Exil VJ, Roberts RL, Sims H, et al. Metabolic cardiomyopathies. Int J Exp
80. Weiss RG, Gerstenblith G, Bottomley Very-long-chain acyl-coenzyme A dehydro- Pathol 2000;81:349-72.
PA. ATP flux through creatine kinase in genase deficiency in mice. Circ Res 2003; 108. Russell LK, Finck BN, Kelly DP.
the normal, stressed, and failing human 93:448-55. Mouse models of mitochondrial dysfunc-
heart. Proc Natl Acad Sci U S A 2005;102: 95. Kurtz DM, Rinaldo P, Rhead WJ, et al. tion and heart failure. J Mol Cell Cardiol
808-13. Targeted disruption of mouse long-chain 2005;38:81-91.
81. Liao R, Nascimben L, Friedrich J, acyl-CoA dehydrogenase gene reveals cru- 109. Hugel S, Horn M, de Groot M, et al.
Gwathmey JK, Ingwall JS. Decreased en- cial roles for fatty acid oxidation. Proc Effects of ACE inhibition and beta-recep-
ergy reserve in an animal model of dilated Natl Acad Sci U S A 1998;95:15592-7. tor blockade on energy metabolism in rats
cardiomyopathy: relationship to contrac- 96. Luptak I, Balschi JA, Xing Y, Leone postmyocardial infarction. Am J Physiol
tile performance. Circ Res 1996;78:893- TC, Kelly DP, Tian R. Decreased contrac- 1999;277:H2167-H2175.
902. tile and metabolic reserve in peroxisome 110. Hermann HP, Pieske B, Schwar-
82. Liu J, Wang C, Murakami Y, et al. Mito- proliferator-activated receptor-alpha-null zmuller E, Keul J, Just H, Hasenfuss G.
chondrial ATPase and high-energy phos- hearts can be rescued by increasing glu- Haemodynamic effects of intracoronary
phates in failing hearts. Am J Physiol cose transport and utilization. Circulation pyruvate in patients with congestive heart
Heart Circ Physiol 2001;281:H1319-H1326. 2005;112:2339-46. failure: an open study. Lancet 1999;353:
83. Neubauer S, Horn M, Pabst T, et al. 97. Abel ED, Kaulbach HC, Tian R, et al. 1321-3.
Contributions of 31P-magnetic resonance Cardiac hypertrophy with preserved con- 111. Nikolaidis LA, Elahi D, Hentosz T, et
spectroscopy to the understanding of di- tractile function after selective deletion of al. Recombinant glucagon-like peptide-1
lated heart muscle disease. Eur Heart J GLUT4 from the heart. J Clin Invest 1999; increases myocardial glucose uptake and
1995;16:Suppl O:115-8. 104:1703-14. improves left ventricular performance in

1150 n engl j med 356;11  www.nejm.org  march 15, 2007

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.
Mechanisms of Disease

conscious dogs with pacing-induced dilat­ 116. Schmidt-Schweda S, Holubarsch C. ing and failure after experimental
ed cardiomyopathy. Circulation 2004;110: First clinical trial with etomoxir in pa- myocardial infarction. Circulation 2002;
955-61. tients with chronic congestive heart fail- 106:3126-32.
112. Liao R, Jain M, Cui L, et al. Cardiac- ure. Clin Sci (Lond) 2000;99:27-35. 121. Lygate CA, Hulbert K, Monfared M,
specific overexpression of GLUT1 pre- 117. Lionetti V, Linke A, Chandler MP, et Cole MA, Clarke K, Neubauer S. The
vents the development of heart failure at- al. Carnitine palmitoyl transferase-I inhi- PPARgamma-activator rosiglitazone does
tributable to pressure overload in mice. bition prevents ventricular remodeling and not alter remodeling but increases mor-
Circulation 2002;106:2125-31. delays decompensation in pacing-induced tality in rats post-myocardial infarction.
113. Vitale C, Wajngaten M, Sposato B, et heart failure. Cardiovasc Res 2005;66:454- Cardiovasc Res 2003;58:632-7.
al. Trimetazidine improves left ventricu- 61. 122. Frantz S, Hu K, Widder J, et al. Per-
lar function and quality of life in elderly 118. Chandler MP, Stanley WC, Morita H, oxisome proliferator activated-receptor
patients with coronary artery disease. Eur et al. Short-term treatment with ranola- agonism and left ventricular remodeling
Heart J 2004;25:1814-21. zine improves mechanical efficiency in in mice with chronic myocardial infarc-
114. Di Napoli P, Taccardi AA, Barsotti A. dogs with chronic heart failure. Circ Res tion. Br J Pharmacol 2004;141:9-14.
Long term cardioprotective action of tri­ 2002;91:278-80. 123. Wallis J, Lygate CA, Fischer A, et al.
metazidine and potential effect on the 119. Finck BN, Han X, Courtois M, et al. Supranormal myocardial creatine and
inflammatory process in patients with A critical role for PPARalpha-mediated li- phosphocreatine concentrations lead to
ischaemic dilated cardiomyopathy. Heart potoxicity in the pathogenesis of diabetic cardiac hypertrophy and heart failure: in-
2005;91:161-5. cardiomyopathy: modulation by dietary fat sights from creatine transporter-overex-
115. Lee L, Campbell R, Scheuermann- content. Proc Natl Acad Sci U S A 2003; pressing transgenic mice. Circulation 2005;
Freestone M, et al. Metabolic modulation 100:1226-31. 112:3131-9.
with perhexiline in chronic heart failure: 120. Shiomi T, Tsutsui H, Hayashidani S, 124. Ng TM. Levosimendan, a new cal-
a randomized, controlled trial of short- et al. Pioglitazone, a peroxisome prolif- cium-sensitizing inotrope for heart fail-
term use of a novel treatment. Circulation erator-activated receptor-gamma ago- ure. Pharmacotherapy 2004;24:1366-84.
2005;112:3280-8. nist, attenuates left ventricular remodel- Copyright © 2007 Massachusetts Medical Society.

posting presentations at medical meetings on the internet


Posting an audio recording of an oral presentation at a medical meeting on the
Internet, with selected slides from the presentation, will not be considered prior
publication. This will allow students and physicians who are unable to attend the
meeting to hear the presentation and view the slides. If there are any questions
about this policy, authors should feel free to call the Journal’s Editorial Offices.

n engl j med 356;11  www.nejm.org  march 15, 2007 1151

Downloaded from www.nejm.org by SHALINI MANCHANDA MD on May 22, 2007 .


Copyright © 2007 Massachusetts Medical Society. All rights reserved.

Вам также может понравиться