You are on page 1of 13

Controversies in

Cardiovascular Medicine
Is Inhibition of Hypertrophy a
Good Therapeutic Strategy in
Ventricular Pressure Overload?

Inhibition of Hypertrophy Is a Good Therapeutic Strategy in


Ventricular Pressure Overload
Gabriele G. Schiattarella, MD; Joseph A. Hill, MD, PhD

H ypertrophic growth of ventricular myocytes is a hallmark


feature of numerous forms of cardiovascular disease.1
The hypertrophic process is complex, involving a vast array
to increases in workload demand, serving to minimize wall
stress and maintain contractile function. However, several
lines of evidence, preclinical and epidemiological, highlight
of structural, signaling, transcriptional, electrophysiological, the maladaptive features of chronic ventricular hypertro-
metabolic, and functional events within the growing cell.2,3 phy. Indeed, in many instances, suppression of load-induced
Downloaded from http://ahajournals.org by on February 27, 2024

Other cellular elements within the ventricle, fibroblasts, vas- growth is well tolerated.4 Furthermore, left ventricular hyper-
cular smooth muscle cells, and endothelium, also manifest trophy is among the most robust markers of increased risk for
intricate stress responses, resulting in fibrosis, inflammatory developing chronic heart failure.5 Therefore, we submit that
cell infiltration, endothelial dysfunction, and vascular stiff- suppression of load-induced ventricular hypertrophy warrants
ness. Current thinking holds that these events, the reaction of careful consideration as a therapeutic strategy.
the heart to a host of pathological stresses, provide short-term
benefit. However, if disease-related stress remains unchecked,
these remodeling events become maladaptive and predispose
Cardiomyocyte Hypertrophy: Comprehensive
to cardiovascular morbidity and mortality. Reprogramming of the Cell
Although evidence suggests that a small fraction of cells
Response by Crozatier and Ventura-Clapier within the ventricle are capable of re-entering the cell cycle,6–8
on p 1447 the majority of cardiomyocytes are postmitotic and, hence, do
Among the risks conferred by disease-related ventricu- not retain the ability to divide. Rather, they respond to stress
lar hypertrophy are ventricular tachyarrhythmia, predispos- by growing, shrinking, or dying. In the context of many dis-
ing to sudden cardiac death, and transition to heart failure. ease-related stresses, cardiac myocytes undergo hypertrophic
Ultimately, these events derive from wholesale reprogram- transformation, which entails significant cellular growth. In
ming and relative dedifferentiation of the cardiac myocyte, the setting of uncontrolled hypertension, for example, cardio-
coupled with similar events in other cell types. myocytes hypertrophy. Similarly, after myocardial infarction,
Conventional thinking holds that hypertrophic growth surviving cardiac myocytes in border and remote zones of tis-
of the myocardium is a compensatory response of the heart sue increase in size in response to increases in hemodynamic

The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.
From Departments of Internal Medicine (Cardiology) (G.G.S., J.A.H.) and Molecular Biology (J.A.H.), University of Texas Southwestern Medical
Center, Dallas, TX.
This article is Part I of a 2-part article. Part II appears on p 1448.
Correspondence to Joseph A. Hill, MD, PhD, Division of Cardiology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd,
NB11.200, Dallas, TX 75390–8573. E-mail joseph.hill@utsouthwestern.edu
(Circulation. 2015;131:1435-1447. DOI: 10.1161/CIRCULATIONAHA.115.013894.)
© 2015 American Heart Association, Inc.
Circulation is available at http://circ.ahajournals.org DOI: 10.1161/CIRCULATIONAHA.115.013894

1435
1436  Circulation  April 21, 2015

demand arising secondary to the loss of ventricular tissue. In and physiological hypertrophy differ in the signaling cascades
both cases, when the pressure overload state is persistent, the that drive the process. Some evidence suggests that the dis-
hypertrophic phenotype of the myocardium inexorably pro- tinct phenotypes do not derive simply from differences in the
gresses to a state of decompensation and clinical heart failure. duration of the stimulus, highlighting significant gaps in our
Mechanisms governing this transition from adaptive hypertro- understanding of these 2 remodeling responses.13 Other data
phy to maladaptive failure remain poorly understood. suggest that cell size is regulated by shared signaling path-
Hypertrophic transformation of the cardiomyocyte involves ways, but cell shape and sarcomeric organization are regu-
much more than simple cell growth. Rather, it entails a near- lated by distinct pathways.14 Current understanding does not
comprehensive retooling of multiple aspects of cellular archi- allow us to parse the effects of the myriad genes and pathways
tecture and machinery. One element of this process is relative involved, but it is thought that some confer benefit, whereas
dedifferentiation of the cell and reactivation of numerous others are maladaptive.
transcriptional, signaling, electrical, and metabolic events that Substantial evidence points to alterations in transmembrane
characterized the cell during development. As part of this, a Ca2+ fluxes, another central feature of pathological remodel-
wide range of transcriptional and posttranslational events ing, as a proximal trigger contributing to the pathogenesis
occur, including activation of a pattern of gene expression of hypertrophy and failure.15 These alterations perturb exci-
reminiscent of that observed during fetal development (fetal tation-contraction coupling, alter mitochondrial metabolism,
gene program). Indeed, the fetal gene program, which was and abnormally activate Ca2+-responsive signaling pathways.
extinguished shortly after birth, reignites rapidly in the setting Recent evidence indicates that β-myosin heavy chain is
of disease. induced by pressure overload in rodents in only a minor sub-
Based on the pattern of sarcomere reorganization, it is pos- population of smaller cardiac myocytes.16 The myocytes that
sible to distinguish 2 broad patterns of ventricular hypertro- hypertrophied after surgical constriction of the thoracic aorta
phy.9 Pressure stress provokes concentric hypertrophy, which express α-myosin heavy chain only. Thus, hypertrophic trans-
is characterized by recruitment of sarcomeres laid down formation may involve yet another layer of complexity in that
in parallel; on the contrary, excess volume elicits eccen- it manifests significant heterogeneity among myocytes.
tric hypertrophy in which cardiomyocytes respond with the
Cardiac Myocyte Death
Downloaded from http://ahajournals.org by on February 27, 2024

addition of sarcomeres in series.9 In both cases, increases in


wall thickness occur. (Concentric hypertrophy is marked by Cell death within the myocardium is characteristic of a num-
increases in wall thickness with relatively little change in ven- ber of cardiac diseases, and it can occur to some extent in car-
tricular volume; eccentric hypertrophy is marked by increases diac hypertrophy. The major types of cardiomyocyte death are
in both wall thickness and ventricular cavity size.) According necrosis and apoptosis, with the former occurring to a greater
to the pioneering stress-adaptation hypothesis by Grossman et extent. An emerging literature has demonstrated that necro-
al,10 these increases in wall thickness are an adaptive response; sis can occur as a result of a series of programmed events,
based on the law of Laplace, ventricular wall stress is pro- not just as simple catastrophic dismantling of the cell. Indeed,
portional to both ventricular pressure and cavity radius and programmed necrosis and apoptosis share a number of fea-
inversely proportional to ventricular wall thickness.11 Thus, tures and may represent different manifestations of a common
increases in wall thickness tend to lessen wall stress and mechanism termed necroptosis.17,18
thereby diminish oxygen demand. Both dying and hypertrophying cells often harbor signs
Myocyte growth is dictated by a delicate balance between of activated autophagy, an evolutionarily ancient process of
protein synthesis and protein degradation. In the setting of ordered recycling of intracellular contents.19 Whether activa-
elevated afterload, protein synthesis predominates, culminat- tion of the autophagic cascade reflects a cellular response to
ing in hypertrophic growth. However, it is important to recog- stress, serving to promote cell survival, or is a process con-
nize that hypertrophic remodeling is not a simple process of tributing to cell death and disease progression, is context
the addition of new sarcomeres. Rather, this highly dynamic dependent.20
cellular response involves intricate coordination of de novo
protein synthesis and organelle biogenesis, sarcomere remod- Fibrosis
eling, protein degradation, organelle breakdown, transcrip- Another hallmark feature of pathological hypertrophic remod-
tional reprogramming, and metabolic shifts. In many ways, eling is accumulation and deposition of excessive extracellular
the entire cellular architecture of the myocyte–frame, chassis, matrix.21 This surplus extracellular matrix, which constitutes
drive train, and engine–is retooled. tissue scar or fibrosis, perturbs electrical conduction, thereby
In contrast to disease-related triggers, physiological stresses, predisposing to rhythm disturbances. It also promotes dys-
such as endurance exercise and pregnancy, induce a hypertro- function of both mechanical contraction and relaxation. As
phic response characterized by normal or enhanced contractile a result, cardiac fibrosis contributes importantly to morbidity
function coupled with normal architecture and organization and mortality in cardiac hypertrophy. Indeed, the amount of
of cardiac structure.12 Beyond differences in growth triggers, fibrotic scar in the myocardium correlates directly with the
biological phenotypes, and clinical outcomes, pathological incidence of arrhythmias and sudden cardiac death.22–24
Schiattarella and Hill   Targeting Maladaptive Hypertrophy   1437

Extracellular matrix deposition and fibrosis arise through for heterogeneity of APD prolongation has been reported in
the action of cardiac fibroblasts. These cells, the most abun- a model of pacing-induced heart failure in dogs where APD
dant cell type in the myocardium, proliferate in response prolongation in midmyocardial cells was substantially greater
to pathological stress. Furthermore, they differentiate into than in subepicardial cells.39
myofibroblasts, thereby gaining the capacity to contract and Alterations in Ca2+ handling contribute to both hypertrophic
secrete collagen I, collagen III, and fibronectin.25 Fibrosis signaling and electrical remodeling. For example, L-type Ca2+
can be categorized as reactive (perivascular or interstitial) or current (ICa,L) is a major mechanism of Ca2+ influx in cardiac
replacement, occurring at the site of an eliminated myocyte. myocytes. As a general rule, ICa,L density correlates inversely
Myofibroblasts derive from activated, resident fibroblasts, but with disease progression; in models of mild-to-moderate
may also originate from adult epicardial cells26 and circulat- hypertrophy, ICa,L is often increased, whereas in severe hyper-
ing, collagen-secreting, bone marrow–derived cells.27,28 Both trophy and failure, ICa,L often manifests significant declines.
individual myofibroblasts and collagenous septa within the Importantly, because membrane impedance is relatively high
tissue facilitate and propagate the arrhythmic phenotype of during phase 2 of the action potential, small changes in ICa,L
the hypertrophied heart.29–31 can have significant effects on action potential morphology
Cardiac fibrosis is an independent and predictive risk factor and duration. Furthermore, entry of small amounts of extra-
for heart failure development in the setting of ischemic or non- cellular Ca2+ triggers release of much larger amounts of Ca2+
ischemic cardiomyopathy.32–34 Importantly, strong evidence from intracellular stores. As a consequence, modest increases
indicates that cardiac fibrosis, long held to be irreversible, in inward Ca2+ flux are amplified within the cell.
may regress under certain conditions.21,35 However, although Electrical activity within the myocardium hinges critically
several signaling pathways have been implicated in fibrogen- on electrotonic cell-cell coupling, such that depolarization in
esis, tailored therapeutic approaches targeting cardiac fibrosis one cell is transmitted seamlessly to neighboring cells. This
remain elusive.21 coupling is mediated through gap junctions, such as connexin
43, which can become disorganized in the hypertrophied or
Electrical Remodeling failing heart, disrupting normal impulse conduction.40
Patients with left ventricular hypertrophy are at increased risk It is worth remembering that the atria are also touched by
remodeling events in cardiac hypertrophy and failure. Reduced
Downloaded from http://ahajournals.org by on February 27, 2024

of malignant arrhythmia, which contributes significantly to


morbidity and mortality. Indeed, arrhythmia, especially ven- contractility, development of fibrosis, and chamber enlarge-
tricular tachyarrhythmia, is a major cause of death in patients ment can occur, leading to heterogeneity of conduction veloc-
with cardiac hypertrophy or failure. Underlying mechanisms, ity41 and propensity to atrial fibrillation.42 This arrhythmia, in
collectively termed electrical remodeling, encompass altera- turn, eliminates the component of ventricular filling provided
tions in multiple electrogenic transport and signaling processes by atrial contraction. Furthermore, it promotes decreases in
within the cardiac myocyte. Although numerous insights have the atrial myocyte effective refractory period and shortened
emerged in elucidating the molecular pathogenesis of cardiac APD, which together promote sustained atrial fibrillation.43
hypertrophy,3,36 our understanding of mechanisms underlying
the myriad facets of electrical remodeling remains limited. As Metabolic Remodeling
a result, pharmacological treatment of hypertrophy-associated The metabolic demands of the myocardium are exceptionally
arrhythmias lacks efficacy, and device-based therapy has high. As a continuously working pump, the heart consumes
emerged as a widely used surrogate. robust quantities of ATP, more than any other organ in the
The action potential phenotype of ventricular myocyte body, and myocardial energy reserves are remarkably low,
hypertrophy is characterized by delayed repolarization lead- sufficient for less than 10 contractions. The cardiomyocyte
ing to prolongation of action potential duration (APD). This derives ATP largely from fatty acid oxidation. That being said,
derives, at least in part, from distorted transmembrane electric the heart is a metabolic omnivore that can flexibly burn fuel
currents.2,15,37 Indeed, a wide range of alterations in myocyte derived from a wide range of sources. In the end, cardiomyo-
ion channels and electrogenic ion transporters contribute to cytes must generate energy continuously, consuming nutrients
APD prolongation.2,15,37 Delayed recovery of excitability, constantly and without interruption.44
in turn, predisposes to early and late afterdepolarizations. Among the characteristic changes occurring with cardiac
Hypertrophy is also associated with myocardial fibrosis (vide hypertrophy is a shift in energy substrate use.45–47 This meta-
supra), altered electrotonic coupling among cells, slowed con- bolic remodeling response entails upregulation of glucose
duction, and dispersion of refractoriness, which together pro- uptake and glycolysis, whereas β-oxidation of fatty acids is
mote re-entrant arrhythmias. reduced. Approximate numbers are that glycolysis accounts
This electrical remodeling response is heterogeneous within for 10% of ATP production in the normal heart and 20% in the
the ventricle. In the setting of excessive afterload, such as in hypertrophied heart. Conversely, ATP production from fatty
severe transverse aortic constriction-induced heart failure, acid metabolism drops from 70% to 50%. These shifts are
APD is prolonged more in subepicardial ventricular myo- consistent with the overarching process of cellular dediffer-
cytes than in subendocardial myocytes.38 Additional evidence entiation in the pathologically stressed myocardium, because
1438  Circulation  April 21, 2015

the shift in metabolism mimics the metabolic program in fetal therapies using selective approaches targeting specific cellu-
myocardium. lar and molecular elements of pathological cardiac remodel-
The impact of the partial switch to a fetal program of glu- ing and heart failure could be elucidated. Recently, a novel
cose metabolism from preferential use of lipid remains uncer- connection among autophagy, afterload stress, and inflam-
tain. Arguably the greatest impact derives from improvements mation was reported, where a lack of autophagy-mediated
in oxygen efficiency.48 However, concerns have been raised removal of mitochondrial DNA promoted an inflamma-
because of evidence that cardiomyocytes cultured in high- tory response contributing to depressed cardiac contractile
glucose media are dysfunctional.49 performance.70
To accomplish the synthesis of macromolecules and
organelles required for cardiomyocyte hypertrophic growth, Vascular Remodeling
exogenous nutrients, such as glucose, cannot be metabolized The vasculature that supplies the ventricle itself remod-
exclusively for ATP production. Rather, metabolic intermedi- els in both physiological and pathological hypertrophy.
ates must be channeled to support anabolic pathways. Here, Hypertrophic growth of the heart involves parallel increases
interplay between the plasticity of metabolic pathways and in myocardial mass and vascular supply, the latter occurring
protein quality control is critical to providing intermediate through proliferation of endothelium, smooth muscle cells,
metabolites that feed into the tricarboxylic acid cycle for ATP and blood vessels.71,72 Indeed, it is possible to distinguish 2
production, as well as serving to promote macromolecule syn- interdependent phenomena involving heart vessels. The first is
thesis.50 Overall, currently available information implicates remodeling of pre-existing vasculature with modifications of
metabolic reprogramming in the hypertrophied heart as an endothelium, smooth muscle cells, and interstitial matrix, and
ultimately maladaptive response.51 the second is de novo myocardial angiogenesis involving an
intricate network of molecules secreted from cardiomyocytes,
Inflammation leukocytes, and fibroblasts (reviewed in References 73 and 74).
Activation of immune mechanisms participates in ventricu- Paracrine signaling through mediators released by endothelial
lar remodeling, contributing to long-term cardiac injury in cells may modify vascular smooth muscle cell function and
certain contexts. In the case of heart failure, a variety of extracellular matrix components.75,76 Also, in the setting of
hypertension, vascular smooth muscle proliferates and hyper-
Downloaded from http://ahajournals.org by on February 27, 2024

inflammatory molecules and pathways are activated.52–55 In


preclinical models, pressure overload triggers myocardial trophies, culminating in vascular wall thickening. In some set-
inflammation, as demonstrated by increased expression of tings, flow reserve is compromised.
several proinflammatory cytokines and leukocyte infiltration Dysfunction of vascular endothelial cells occurring in
within the myocardium.56–58 This and other evidence, both in pressure overload–induced heart failure may represent a
animals and patients, highlights the role of inflammation as crucial node in vascular remodeling occurring in pathologi-
an important component of pathological hypertrophy and cal hypertrophy.77,78 Indeed, one model holds that capillary
points to the relationship between a proinflammatory state growth in pathological hypertrophy does not keep up with
and load-induced ventricular remodeling.59–62 Interestingly, myocyte growth, leading to inadequate oxygen diffusion
myocardial cytokine levels, a surrogate marker of inflam- capacity. Along those lines, it has also been postulated that
mation, are often higher in patients with elevated afterload an imbalance in the capillary:cardiomyocyte ratio contrib-
and preserved left ventricular function compared with those utes to the transition from compensatory hypertrophy to
patients with reduced ejection fraction.63 These data chal- decompensated heart failure.74 Consistent with this notion,
lenge the concept that cytokine activity and an inflammatory enhancing angiogenesis in a model of afterload stress can
response are necessarily detrimental in chronic pressure be protective.79 In fact, therapeutic strategies aimed to
overload. restore the capillary network in heart failure have been
A crucial element within the inflammatory response evaluated in both preclinical models and in humans.80–83
observed in models of pressure overload involves macro- Despite some favorable findings, therapeutic myocardial
phages that infiltrate ventricular tissue, triggering myocardial angiogenesis has failed to achieve clinical significance, at
expression of nuclear factor-кB and inflammatory cytokines.64 least partly because these approaches typically involve the
One of the mechanisms whereby macrophages are involved delivery of genetic material or growth factors with atten-
includes actions of macrophage-derived microRNA-155, dant complications.
which functions as a modulator of cardiomyocyte hypertrophy
via paracrine mechanisms.65 Functional Role of Pathological Hypertrophy
These observations, coupled with the effects of anti- Three stages of hypertrophic transformation of the heart were
inflammatory therapies in animals, have prompted efforts initially proposed by Meerson.84 This model emphasizes that
to translate these insights into patients through clinical tri- the duration of pressure overload dictates the progression of
als.56,66 However, to date, anti-inflammatory therapies for events, including hypertrophic growth and ultimately ven-
heart failure in humans have disappointed.67–69 Looking tricular systolic function. In this model, short-term hypertro-
to the future, the possible role of novel anti-inflammatory phy represents a beneficial event that serves to normalize wall
Schiattarella and Hill   Targeting Maladaptive Hypertrophy   1439

variable. Future work will determine whether this 4-tiered


classification conveys independent information regarding
prognosis or therapy.
Despite these caveats, the presence of left ventricular
hypertrophy, per se, is unequivocally associated with adverse
cardiovascular outcomes independent of the underlying dis-
ease-related cause.5 As noted earlier, whereas ventricular
hypertrophy may be beneficial in the short term to mini-
mize wall stress, in the long term it promotes progression to
heart failure and other cardiovascular disorders. Numerous
epidemiological studies have clearly demonstrated that left
ventricular hypertrophy is not benign but rather represents
a major risk factor for cardiovascular morbidity and mortal-
ity, more robust than other conventional risk factors.5,100–103
Substudies from the Framingham Heart Study have dem-
onstrated marked increases in coronary heart disease, heart
failure, and sudden cardiac death associated with left ven-
tricular hypertrophy, revealed by electrocardiographic and
echocardiographic approaches.104–106
Together these data clearly identify left ventricular
hypertrophy as an important risk factor of cardiovascular
disease. However, it is important to highlight that these
Figure 1. Canonical 3-stage model of hypertrophic transforma- observations are strictly correlative, and no mechanism(s)
tion of the heart. This model posits short-term hypertrophy as a
beneficial event and long-term hypertrophy as detrimental. LVH of hypertrophy-dependent increase in risk can be inferred.
indicates left ventricular hypertrophy. Our knowledge of potential mechanism(s) by which ven-
tricular hypertrophy confers increased cardiovascular risk
Downloaded from http://ahajournals.org by on February 27, 2024

stress, whereas prolonged hypertrophy is detrimental, provok- is limited by the fact that transition from the early, com-
ing increased oxygen consumption and cardiomyocyte death pensatory stage of hypertrophy to the maladaptive phase is
(Figure 1). poorly characterized.
Considerable evidence, from both preclinical and clini-
cal contexts, indicates that chronic, unremitting stress, as Is Load-Induced Hypertrophy
occurs in hypertension or valvular heart disease, leads inevi- Ever Truly Compensatory?
tably to systolic dysfunction.85–89 Moreover, the Framingham Traditionally, ventricular hypertrophy has been viewed as
Heart Study established an association between ventricular an obligatory initial response to pathological stress; only
hypertrophy and increased cardiac mortality.5 However, the with time does it transition to a disease-promoting event.
inevitability of the transition from hypertrophy to failure Consistent with this view, the term compensatory connotes
has been questioned. Drazner and colleagues90,91 have iden- an adaptive role of hypertrophy as a consequence of pressure
tified 3 challenges to this point of view: 1) in animal models stress based on the premise that it helps normalize ventricu-
of pressure overload, hypertrophy can be blocked without lar wall stress and myocardial oxygen demand. However,
development of heart failure92–94; 2) in humans, concentric evidence from genetic and pharmacological studies in
hypertrophy does not uniformly progress to failure in the mouse models of pressure overload have demonstrated that
absence of myocardial infarction95–97; and 3) some hyper- hypertrophic growth is not universally required to preserve
tensive subjects develop dilated cardiomyopathy apparently cardiac function.92,94
without antecedent concentric hypertrophy and without Shortly after the report107 that calcineurin is capable of
clinical evidence of myocardial infarction.98 These impor- triggering a robust cardiac growth response, we set out to
tant caveats are based on the notion that the time course of determine whether calcineurin signaling is required for
transition to heart failure in patients, if it occurs, is rela- afterload-induced growth of the heart. Mice were exposed
tively uniform and can be captured within the time span of to thoracic aortic constriction and injected daily with cyclo-
epidemiological studies. sporine or vehicle, delivered in an investigator-blinded
In an effort to address these issues, magnetic resonance manner. After 3 weeks, hearts were evaluated by echocar-
imaging data from the Dallas Heart Study have been used diography and by postmortem gravimetric analyses. In the
to propose a 4-tiered classification of left ventricular hyper- animals exposed to cyclosporine, the load-induced growth
trophy.99 This scheme seeks to overcome the limitations of response was abolished, leading us to conclude that cal-
the concentric versus eccentric hypertrophy model by incor- cineurin activation was, in fact, required for load-induced
porating left ventricle end-diastolic volume as a categorical cardiac hypertrophy in this context. Very surprising to us,
1440  Circulation  April 21, 2015

however, was the fact that ventricular size and function by Therefore, the concept of transition from a short-term com-
echo were normal in the cyclosporine-treated animals. This pensatory response to maladaptation points to cardiac hyper-
was true despite the imposition of 70 to 80 mm Hg of after- trophy as a novel therapeutic target.
load stress, which was persistently present until the end of
the entire study; ventricular volumes and contractile param-
Translational Studies in Large Animals
eters were normal, and the animals behaved normally. This
Our understanding of mechanisms that govern the transi-
astonishing finding was published,92 and we estimate that
tion between compensated ventricular hypertrophy and heart
this observation, which often goes unnoticed in the report-
failure remains incomplete. This stems partly from a lack of
ing of studies, has been replicated independently at least
longitudinal analyses of ventricular structure and function
100 times since then.
in patients with increased afterload (eg, aortic stenosis and
These findings, demonstrating that load-induced hyper-
arterial hypertension). To date, most evidence that provides
trophy is not always required to maintain ventricular size
insight into the effects of suppressing cardiac hypertrophy on
and performance, raise the prospect that the hypertrophic
cardiac function derives from studies in rodents.110 However,
growth response may be a relevant target for therapeutic
because of intrinsic differences in contractile performance,
targeting.4 It is important, however, to recognize that these
Ca2+ handling, myosin isoform distributions, heart rate, and
observations derive largely from genetically engineered
life span between rodents and humans, these models do not
mice and, therefore, are based on models with important
faithfully recapitulate human disease. In this regard, large ani-
limitations. Heart rate in humans is substantially slower
mal models could be more informative.
than that in mice, and left ventricular ejection fraction is
Pressure overload–induced cardiac hypertrophy has been
lower. In addition, surgical banding of the aorta triggers an
studied in nonhuman primates, where left ventricular hyper-
acute increase in pressure stress on the left ventricle and
trophy and myocardial fibrosis comparable to that seen in
therefore does not mimic conditions of chronic, progres-
patients with aortic stenosis are found.111 Studies to evaluate
sively increasing hemodynamic load as occurs in humans.
Most preclinical studies in mice were conducted over a 3- molecular mechanisms and pathophysiological adaptations in
to 4-week period, which amounts to ≈3 years in humans; pressure overload–induced heart failure have been conducted
perhaps blocking hypertrophy for a longer period of time primarily in dogs.112–114 In view of the considerable remaining
Downloaded from http://ahajournals.org by on February 27, 2024

would be harmful. Unfortunately, the critical study in large gaps in our understanding of this biology, additional explora-
animals has not been performed to date. tion of the role of compensatory hypertrophy in large animal
models is warranted.

How Can All This Be?


Increases in afterload elicit early changes in myocyte biology How Might Hypertrophy Be Targeted?
across a wide range of mechanisms. Frank-Starling–related Clinical management of pathological left ventricular hyper-
adaptations ensue, triggered by stretch. In addition, contractil- trophy currently focuses on the underlying growth cues (eg,
ity continues to increase for >10 to 15 minutes after the ini- hypertension and valve disease) and typically involves a
tial stretch, a response initially described more than 100 years wide spectrum of pharmacological agents that have shown
ago by Gleb von Anrep and now termed the Anrep effect.108 safety and efficacy in reducing hypertrophy. These pharma-
Initially attributed to increases in circulating catecholamines cological agents are mostly directed against the crucial neu-
released by the adrenal gland, the Anrep effect was later dem- rohormonal axes activated in response to stress. Adrenergic
onstrated in isolated ventricular myofilaments and was attrib- and renin-angiotensin-aldosterone system (RAAS) path-
uted to increases in myofilament calcium sensitivity.109 Thus, ways represent 2 mechanisms recruited to increase con-
the Anrep effect, a response that remains poorly characterized, tractility in the early phases of stress, becoming deleterious
is a rapid, procontractile response that occurs in the setting of in the chronic context. β-Adrenergic receptor blockers,
abrupt increases in afterload. It is possible that this response, angiotensin-converting enzyme inhibitors, and angiotensin
coupled with hypertrophic growth, represents a load adapta- receptor blockers are effective in reducing left ventricular
tion intrinsic to cardiac muscle. mass and are associated with a favorable clinical outcome
Initial, rapid mechanisms whereby cardiac muscle responds in clinical settings.115,116
to increased load are, however, insufficient to maintain car- Pharmacotherapy to reduce blood pressure can lead to
diac contractility for an extended period if the inciting stress regression of ventricular hypertrophy.36 Although all antihy-
is not eliminated. Therefore, increases in cardiac mass ensue pertensive drugs promote hypertrophy regression to some
but ultimately lead to untoward consequences. Indeed, with extent, current evidence suggests that the RAAS-targeting
persistent pressure stress, the heart undergoes apparently drugs are the most efficacious at regressing ventricular
irreversible decompensation, resulting in chamber dilatation hypertrophy. Based on robust evidence that regression of
and reduced systolic function. This maladaptive hypertro- hypertrophy is independently associated with improved cardio-
phy, as emerges when the inciting stress is not abated, repre- vascular outcome, numerous clinical trials have documented
sents a common feature in virtually all forms of heart failure. the favorable impact of antihypertensive drug therapy. For
Schiattarella and Hill   Targeting Maladaptive Hypertrophy   1441

Table. Novel Compounds and Targets With Confirmed or example, the Losartan Intervention for Endpoint Reduction in
Potential Antihypertrophic Activity a Hypertension study reported a greater reduction in left ven-
Target(s)/Mechanism(s) of tricular mass index in the losartan-treated cohort compared
Variable Action References (PMID) with an atenolol-based regimen.117 Several other trials reached
HDAC inhibitors a similar conclusion, pointing to a class effect of RAAS-
Apicidin Class I HDACs 18697792 targeting antihypertensive agents at promoting hypertrophy
Vorinostat Class I, II HDACs 20139990-17211407 regression.118–121 Together, these data highlight the role of cur-
Romidepsin Class I, II HDACs 20139990-21699444 rent therapies targeting chronic neurohormonal activation in
Trichostatin A Pan-HDACs 16380549-16735673- the prevention of heart failure and limiting progression of ven-
12975471 tricular hypertrophy, lending further support to the notion that
Valproic acid Class I HDACs (weak) 16380549 inhibiting ventricular hypertrophy is an attractive therapeutic
Scriptaid Pan-HDACs 16735673 option in patients.
SK-7041 Pan-HDACs 16380549 All of these trials were conducted using compounds tar-
Prohypertrophic microRNAs as therapeutic targets geting cardiomyocyte cell-surface receptors. More recently,
miR-199a Hif1α/sirtuin 1 20458739
efforts have focused on antihypertrophic effects of molecules
that act inside the cardiomyocyte, targeting crucial signal-
miR-199b Dyrk1a 21102440
ing cascades that alter gene expression and protein function.
miR-208a THRAP1 and myostatin 19726871-21900086
These agents include histone deacetylase (HDAC) inhibitors,
miR-23a MuRF1 19574461
a wide spectrum of prohypertrophic microRNAs (reviewed in
miR-499 DRP1 22752967
Reference 122), and several other small molecules (reviewed in
miR-21 Sprouty2 and spry1 17234972-19043405
Reference 123; Table).
miR-24 E2F2 23307820-19748357
Our group has focused on reversible protein acetylation as
miR-27b PPAR-γ 21844895 a tractable means of governing cardiomyocyte growth,124–127
miR-350 MAPK11/14 and MAPK8/9 23000971 autophagy,126 and disease responsiveness.128 Among those
miR-195 HMGA1 17108080-25100012 studies, we have noted that inhibition of HDACs is capable
miR-221 p27 22275134 of blunting load-induced growth.127 Beyond that, HDAC
Downloaded from http://ahajournals.org by on February 27, 2024

miR-212/132 FoxO3 23011132 inhibitors can promote regression of ventricular hypertro-


miR-25 SERCA2a 24670661 phy despite the persistent presence of afterload stress126 (and
miR-155 Socs1 23956210 with preservation–even improvement–in contractile perfor-
Other compounds mance.) HDAC inhibitors, 4 of which are US Food and Drug
CPG-ODN c274 TLR9 agonist 23638055 Administration approved as third-line therapy for Sézary syn-
Curcumin P300 inhibitor 18292809 drome, may emerge as a novel means of targeting ventricu-
FTY720 Sphingolipid/NFAT inhibitor 23753531 lar hypertrophy. Indeed, we have speculated that it might be
BAY94-8862 MR antagonist 22791416 possible to use HDAC inhibitors to sculpt the hypertrophied
Cinaciguat sGC stimulator 22778174 ventricle in patients with heart failure with preserved ejection
LCl699 Aldosterone synthase inhibitor 21986283 fraction, trimming left ventricular wall thickness progres-
Resveratrol Antioxidant 23784505 sively, to afford clinical benefit.
MTP-131 Antioxidant 21620606
Comprehensive understanding of molecular events
involved in maladaptive cardiac hypertrophy and the
TRV120027 AT1 receptor 22891045
remodeling that culminates in decompensated heart fail-
LCZ696 Angiotensin/neprilysin 23731190
inhibitor ure is the first step toward developing novel treatments
 HA-1077 ROCK 14500337-15840407-
with clinical potential. This is especially true for patients
(Fasudil) 15096457 with heart failure with preserved ejection fraction, most of
Y-27632 ROCK 12623300-12176125 whom harbor ventricular hypertrophy, which contributes
Sildenafil PDE5 25139994 to symptoms and clinical outcomes,129 and for which evi-
Ruboxistaurin PKCβ 15878171
dence-based therapy is lacking. Although these pathways
manifest redundancy in their effects, hypertrophy often
AT1 indicates angiotensin II receptor type 1; DRP1, dynamin-related protein
1; Dyrk1A, dual specificity tyrosine-phosphorylation-regulated 1A; E2F2, E2F remains present in models in which 1 pathway is sup-
transcription factor 2; FoxO3, forkhead box O3; HDAC, histone deacetylase; Hif1α, pressed, suggesting that serial pharmacological brakes may
hypoxia inducible factor1α; HMGA1, high mobility group A1; MAPK, mitogen- be required for clinical gain.
activated protein kinase; MR, mineralocorticoid receptor; MuRF1, muscle RING-finger
protein-1; NFAT, nuclear factor of activated T cells; PDE5, phosphodiesterase type 5;
PKCβ, protein kinase Cβ; PPAR, peroxisome proliferator–activated receptor; ROCK, A Note of Caution
rho-associated protein kinase; SERCA2a, sarcoplasmic reticulum Ca2+ ATPase; Considerable evidence points to a progression of remodel-
Socs1, suppressor of cytokine signaling 1; THRAP1, thyroid hormone receptor– ing events in which stress elicits the following: 1) a hyper-
associated protein 1; TLR9, toll-like receptor 9 ; and sGC, soluble guanylyl cyclase. trophic growth response which has beneficial features; 2)
1442  Circulation  April 21, 2015

maladaptive hypertrophy, which is a clear-cut marker for transformation of the heart under conditions of disease-related
untoward events; and 3) the clinical syndrome of heart fail- stress is similar to many other processes in biology. There has
ure. However, assuming this model has validity, little is been significant evolutionary pressure to promote organismal
known regarding specific markers of these different phases survival until procreation can occur and young can be fostered
of disease pathogenesis or transition points separating to independence. However, our species did not evolve to live 9
them. decades! In the settings of life-threatening stress, activation of
A large number of preclinical studies have demonstrated that both the β-adrenergic cascade and the RAAS axis occurs, and
it is possible to blunt load-induced hypertrophy, even in the set- the end result is enhanced cardiac performance and improved
ting of persistent afterload stress, without affecting contractile survival. These pathways allow for survival in the setting of
function.4,92,94 These studies, then, have delineated a strategy sublethal injury, for example. However, chronic activation of
wherein one might target maladaptive hypertrophy and thus these processes, life saving in the short term, clearly conveys
obviate the untoward consequences of continued progression markedly enhanced risk in the long term. Indeed, chronic
of this process. Consistently, in both preclinical studies and suppression of those evolutionarily ancient neurohumoral
clinical trials, inhibition of cardiac hypertrophic growth usually responses is fundamental to present day, evidence-based ther-
results in the amelioration of left ventricular dysfunction.5,9,130 apy for heart failure. Again, a large number of studies have
Caution is warranted. Not all forms of pathological car- demonstrated robust benefits from therapeutic strategies in
diac hypertrophy can be blocked without provoking ven- which evolutionary adaptations to stress are blocked.
tricular dysfunction.131–134 Although the great majority of We suggest that ventricular hypertrophy is the same. It pro-
studies (again, exclusively in rodents to date) demonstrate vides short-term benefit and long-term harm (Figure 2). If this
that the load-induced growth response can be inhibited model holds true, then suppression of ventricular hypertrophy,
without untoward effects, there are examples where hyper- a therapeutic target currently not under widespread consider-
trophy elicited by a specific signaling cascade appears to be ation, emerges as a novel and potentially important target for
required.135–137 Also, ventricular mass alone may not provide consideration of drug development going forward.
the full picture of the myriad remodeling events involved in
heart growth.138 Therefore, further work is required to deter- Summary and Perspective
mine whether accompanying alterations (eg, contractile func-
Downloaded from http://ahajournals.org by on February 27, 2024

Heart failure is defined as a syndrome in which cardiac out-


tion and coronary hemodynamics) associated with ventricular put is unable to meet the metabolic needs of peripheral tis-
hypertrophy can be reversed by treatment and in which cat- sues. In this setting, when cardiac output is depressed, the
egory of patients. myocardium has a limited repertoire of responses, and, as a
We wish to highlight that, whereas cardiac hypertrophy in result, so does the treating physician. The heart is capable of
response to pathological stimuli manifests common charac- responding in 4 ways: increases in 1) heart rate, 2) ventricu-
teristics irrespective of the triggering stress, it is likely that lar filling, 3) contractility, and 4) mass. Clinically, we have
several subtypes of pathological hypertrophy exist. Thus, a full spectrum of means to regulate heart rate and optimize
whereas efforts to block the inciting stimuli are warranted, we ventricular filling pressures. Contractility is a mechanism
do not know whether all forms of maladaptive hypertrophy that we target routinely in the acute setting, but no safe and
should be prevented. effective therapies are available that promote contractility
chronically. This leaves ventricular hypertrophy as the final
Stress-Induced Cardiac Growth: A Model frontier of heart failure therapy, a target that has never been
How is it that hypertrophy can be both beneficial and del- developed and that, in fact, seems counterintuitive on initial
eterious? We propose a model in which hypertrophic consideration.

Figure 2. Model of the hypertrophic response to


toxic stress. We propose that hypertrophic transfor-
mation of the heart under stress is similar to other
biological processes, which evolved to provide
short-term benefit. These include activation of the
β-adrenergic and renin-angiotensin-aldosterone
axes (RAAS). Cornerstone therapy for heart failure
(HF Rx) involves interruption of these responses,
and we suggest that judicious suppression of
hypertrophy may provide similar benefit.
Schiattarella and Hill   Targeting Maladaptive Hypertrophy   1443

That said, it is critical to recognize that hypertrophic J. Evidence for cardiomyocyte renewal in humans. Science. 2009;324:98–
102. doi: 10.1126/science.1164680.
transformation of the ventricle is just that, a transformation
7. Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN,
involving cellular dedifferentiation and comprehensive repro- Sadek HA. Transient regenerative potential of the neonatal mouse heart.
gramming of the cardiac myocyte and other cellular elements Science. 2011;331:1078–1080. doi: 10.1126/science.1200708.
within the ventricle. Although increases in myocyte size and 8. Steinhauser ML, Lee RT. Regeneration of the heart. EMBO Mol Med.
2011;3:701–712. doi: 10.1002/emmm.201100175.
consequent increases in ventricular mass are hallmark fea- 9. Lorell BH, Carabello BA. Left ventricular hypertrophy: pathogenesis,
tures, a wide range of additional events occur in these stressed detection, and prognosis. Circulation. 2000;102:470–479.
cells. Here, we present an overview of the large body of robust 10. Grossman W, Jones D, McLaurin LP. Wall stress and patterns of hyper-
epidemiological and preclinical data pointing to the untoward trophy in the human left ventricle. J Clin Invest. 1975;56:56–64. doi:
10.1172/JCI108079.
consequences of hypertrophic transformation of the myocar- 11. BURCH GE, RAY CT, CRONVICH JA. Certain mechanical peculiarities
dium. These data, at the very least, raise the prospect of target- of the human cardiac pump in normal and diseased states. Circulation.
ing hypertrophy therapeutically. 1952;5:504–513.
In recent years, significant strides have been achieved in 12. Weeks KL, McMullen JR. The athlete’s heart vs. the failing heart: can
signaling explain the two distinct outcomes? Physiology (Bethesda).
our understanding of, and therapeutic targeting of, patho- 2011;26:97–105. doi: 10.1152/physiol.00043.2010.
logical hypertrophic remodeling. We view hypertrophic 13. Perrino C, Naga Prasad SV, Mao L, Noma T, Yan Z, Kim HS, Smithies
transformation as a fundamental, potentially indispensable O, Rockman HA. Intermittent pressure overload triggers hypertrophy-
step in the myocardial response to pressure overload, which independent cardiac dysfunction and vascular rarefaction. J Clin Invest.
2006;116:1547–1560. doi: 10.1172/JCI25397.
is coupled with significant detrimental consequences. This 14. Bass GT, Ryall KA, Katikapalli A, Taylor BE, Dang ST, Acton ST,
response may be beneficial in the short term, but when main- Saucerman JJ. Automated image analysis identifies signaling pathways
tained chronically it becomes significantly detrimental. It is regulating distinct signatures of cardiac myocyte hypertrophy. J Mol Cell
Cardiol. 2012;52:923–930. doi: 10.1016/j.yjmcc.2011.11.009.
analogous to the responses of β-adrenergic and RAAS acti-
15. Hill JA. Electrical remodeling in cardiac hypertrophy. Trends Cardiovasc
vation, which we now go to great lengths to block and for Med. 2003;13:316–322.
which suppression seemed counterintuitive at first. We argue 16. Lopez JE, Myagmar BE, Swigart PM, Montgomery MD, Haynam S,
that hypertrophy is comparable, and patients may benefit Bigos M, Rodrigo MC, Simpson PC. β-Myosin heavy chain is induced
by pressure overload in a minor subpopulation of smaller mouse cardiac
from judicious attenuation of this chronic, long-term hyper- myocytes. Circ Res. 2011;109:629–638.
trophic response.
Downloaded from http://ahajournals.org by on February 27, 2024

17. Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G. Molecular


mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol
Cell Biol. 2010;11:700–714. doi: 10.1038/nrm2970.
Acknowledgments 18. Whelan RS, Kaplinskiy V, Kitsis RN. Cell death in the pathogen-
We thank members of the Hill laboratory for constructive comments. esis of heart disease: mechanisms and significance. Annu Rev Physiol.
2010;72:19–44. doi: 10.1146/annurev.physiol.010908.163111.
19. Yang Z, Klionsky DJ. Eaten alive: a history of macroautophagy. Nat Cell
Sources of Funding Biol. 2010;12:814–822. doi: 10.1038/ncb0910-814.
20. Lavandero S, Chiong M, Rothermel BA, Hill JA. Autophagy in cardiovas-
This work was supported by grants from the National Institutes of cular biology. J Clin Invest. 2015;125:55–64. doi: 10.1172/JCI73943.
Health (HL-120732 and HL-100401), American Heart Association 21. Rockey DC, Bell DP, Hill JA. Fibrosis: a common pathways of organ
(14SFRN20510023), Cancer Prevention & Research Institute of injury and failure. N Engl J Med. 2015;372:1138–1149.
Texas (RP110486P3), Leducq Foundation (11CVD04), and the 22. Yan AT, Shayne AJ, Brown KA, Gupta SN, Chan CW, Luu TM, Di Carli
Sostegno Territoriale alle Attività di Ricerca program (University of MF, Reynolds HG, Stevenson WG, Kwong RY. Characterization of the peri-
Naples Federico II). infarct zone by contrast-enhanced cardiac magnetic resonance imaging is
a powerful predictor of post-myocardial infarction mortality. Circulation.
2006;114:32–39. doi: 10.1161/CIRCULATIONAHA.106.613414.
Disclosures 23. Assomull RG, Prasad SK, Lyne J, Smith G, Burman ED, Khan M,
None. Sheppard MN, Poole-Wilson PA, Pennell DJ. Cardiovascular magnetic
resonance, fibrosis, and prognosis in dilated cardiomyopathy. J Am Coll
Cardiol. 2006;48:1977–1985. doi: 10.1016/j.jacc.2006.07.049.
References 24. Wu KC, Weiss RG, Thiemann DR, Kitagawa K, Schmidt A, Dalal D, Lai
1. Hill JA, Olson EN. Cardiac plasticity. N Engl J Med. 2008;358:1370– S, Bluemke DA, Gerstenblith G, Marbán E, Tomaselli GF, Lima JA. Late
1380. doi: 10.1056/NEJMra072139. gadolinium enhancement by cardiovascular magnetic resonance heralds
2. Hill JA, Olson EN. Muscle: Fundamental Biology and Mechanisms of an adverse prognosis in nonischemic cardiomyopathy. J Am Coll Cardiol.
Disease. Waltham, MA: Academic Press; 2012:1528. 2008;51:2414–2421. doi: 10.1016/j.jacc.2008.03.018.
3. Burchfield JS, Xie M, Hill JA. Pathological ventricular remodeling: 25. Spinale FG. Myocardial matrix remodeling and the matrix metalloprotein-
mechanisms: part 1 of 2. Circulation. 2013;128:388–400. doi: 10.1161/ ases: influence on cardiac form and function. Physiol Rev. 2007;87:1285–
CIRCULATIONAHA.113.001878. 1342. doi: 10.1152/physrev.00012.2007.
4. Frey N, Katus HA, Olson EN, Hill JA. Hypertrophy of the heart: a new 26. Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson EG. Evidence
therapeutic target? Circulation. 2004;109:1580–1589. doi: 10.1161/01. that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest.
CIR.0000120390.68287.BB. 2002;110:341–350. doi: 10.1172/JCI15518.
5. Levy D, Garrison RJ, Savage DD, Kannel WB, Castelli WP. Prognostic 27. Quan TE, Cowper S, Wu SP, Bockenstedt LK, Bucala R. Circulating fibro-
implications of echocardiographically determined left ventricular mass in cytes: collagen-secreting cells of the peripheral blood. Int J Biochem Cell
the Framingham Heart Study. N Engl J Med. 1990;322:1561–1566. doi: Biol. 2004;36:598–606. doi: 10.1016/j.biocel.2003.10.005.
10.1056/NEJM199005313222203. 28. Haudek SB, Xia Y, Huebener P, Lee JM, Carlson S, Crawford JR, Pilling
6. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider F, D, Gomer RH, Trial J, Frangogiannis NG, Entman ML. Bone marrow-
Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, Frisén derived fibroblast precursors mediate ischemic cardiomyopathy in
1444  Circulation  April 21, 2015

mice. Proc Natl Acad Sci U S A. 2006;103:18284–18289. doi: 10.1073/ 50. Wang ZV, Hill JA. Protein quality control and metabolism: bidirec-
pnas.0608799103. tional control in the heart. Cell Metab. 2015;21:215–226. doi: 10.1016/j.
29. Spach MS, Boineau JP. Microfibrosis produces electrical load variations due cmet.2015.01.016.
to loss of side-to-side cell connections: a major mechanism of structural heart 51. Kolwicz SC Jr, Purohit S, Tian R. Cardiac metabolism and its interac-
disease arrhythmias. Pacing Clin Electrophysiol. 1997;20(2 pt 2):397–413. tions with contraction, growth, and survival of cardiomyocytes. Circ Res.
30. Miragoli M, Gaudesius G, Rohr S. Electrotonic modulation of cardiac 2013;113:603–616. doi: 10.1161/CIRCRESAHA.113.302095.
impulse conduction by myofibroblasts. Circ Res. 2006;98:801–810. doi: 52. Aukrust P, Gullestad L, Lappegård KT, Ueland T, Aass H, Wikeby L,
10.1161/01.RES.0000214537.44195.a3. Simonsen S, Frøland SS, Mollnes TE. Complement activation in patients
31. Miragoli M, Salvarani N, Rohr S. Myofibroblasts induce ectopic activ- with congestive heart failure: effect of high-dose intravenous immuno-
ity in cardiac tissue. Circ Res. 2007;101:755–758. doi: 10.1161/ globulin treatment. Circulation. 2001;104:1494–1500.
CIRCRESAHA.107.160549. 53. Diwan A, Tran T, Misra A, Mann DL. Inflammatory mediators and the
32. Weidemann F, Herrmann S, Störk S, Niemann M, Frantz S, Lange V, Beer failing heart: a translational approach. Curr Mol Med. 2003;3:161–182.
M, Gattenlöhner S, Voelker W, Ertl G, Strotmann JM. Impact of myocardial 54. Soejima H, Irie A, Fukunaga T, Oe Y, Kojima S, Kaikita K, Kawano H,
fibrosis in patients with symptomatic severe aortic stenosis. Circulation. Sugiyama S,Yoshimura M, Kishikawa H, NishimuraY, Ogawa H. Osteopontin
2009;120:577–584. doi: 10.1161/CIRCULATIONAHA.108.847772. expression of circulating T cells and plasma osteopontin levels are increased
33. Iles L, Pfluger H, Lefkovits L, Butler MJ, Kistler PM, Kaye DM, Taylor in relation to severity of heart failure. Circ J. 2007;71:1879–1884.
AJ. Myocardial fibrosis predicts appropriate device therapy in patients 55. Caforio AL, Mahon NG, Baig MK, Tona F, Murphy RT, Elliott PM,
with implantable cardioverter-defibrillators for primary prevention of sud- McKenna WJ. Prospective familial assessment in dilated cardio-
den cardiac death. J Am Coll Cardiol. 2011;57:821–828. doi: 10.1016/j. myopathy: cardiac autoantibodies predict disease development in
jacc.2010.06.062. asymptomatic relatives. Circulation. 2007;115:76–83. doi: 10.1161/
34. Dweck MR, Joshi S, Murigu T, Alpendurada F, Jabbour A, Melina G, CIRCULATIONAHA.106.641472.
Banya W, Gulati A, Roussin I, Raza S, Prasad NA, Wage R, Quarto C, 56. Gullestad L, Aukrust P. Review of trials in chronic heart failure show-
Angeloni E, Refice S, Sheppard M, Cook SA, Kilner PJ, Pennell DJ, ing broad-spectrum anti-inflammatory approaches. Am J Cardiol.
Newby DE, Mohiaddin RH, Pepper J, Prasad SK. Midwall fibrosis is an 2005;95(11A):17C–23C, 38C. doi: 10.1016/j.amjcard.2005.03.008.
independent predictor of mortality in patients with aortic stenosis. J Am 57. Frantz S, Bauersachs J, Ertl G. Post-infarct remodelling: contribution of
Coll Cardiol. 2011;58:1271–1279. doi: 10.1016/j.jacc.2011.03.064. wound healing and inflammation. Cardiovasc Res. 2009;81:474–481. doi:
35. Berry JM, Le V, Rotter D, Battiprolu PK, Grinsfelder B, Tannous P, 10.1093/cvr/cvn292.
Burchfield JS, Czubryt M, Backs J, Olson EN, Rothermel BA, Hill JA. 58. Cave AC, Brewer AC, Narayanapanicker A, Ray R, Grieve DJ, Walker
Reversibility of adverse, calcineurin-dependent cardiac remodeling. Circ S, Shah AM. NADPH oxidases in cardiovascular health and disease.
Res. 2011;109:407–417. doi: 10.1161/CIRCRESAHA.110.228452. Antioxid Redox Signal. 2006;8:691–728. doi: 10.1089/ars.2006.8.691.
36. Xie M, Burchfield JS, Hill JA. Pathological ventricular remodeling: 59. Nicoletti A, Michel JB. Cardiac fibrosis and inflammation: interaction with
therapies: part 2 of 2. Circulation. 2013;128:1021–1030. doi: 10.1161/ hemodynamic and hormonal factors. Cardiovasc Res. 1999;41:532–543.
CIRCULATIONAHA.113.001879. 60. Ammarguellat FZ, Gannon PO, Amiri F, Schiffrin EL. Fibrosis, matrix
Downloaded from http://ahajournals.org by on February 27, 2024

37. Wang Y, Hill JA. Electrophysiological remodeling in heart failure. J Mol metalloproteinases, and inflammation in the heart of DOCA-salt hyperten-
Cell Cardiol. 2010;48:619–632. doi: 10.1016/j.yjmcc.2010.01.009. sive rats: role of ET(A) receptors. Hypertension. 2002;39(2 pt 2):679–684.
38. Wang Y, Cheng J, Joyner RW, Wagner MB, Hill JA. Remodeling of 61. Hein S, Arnon E, Kostin S, Schönburg M, Elsässer A, Polyakova V, Bauer
early-phase repolarization: a mechanism of abnormal impulse conduc- EP, Klövekorn WP, Schaper J. Progression from compensated hypertrophy
tion in heart failure. Circulation. 2006;113:1849–1856. doi: 10.1161/ to failure in the pressure-overloaded human heart: structural deterioration
CIRCULATIONAHA.106.615682. and compensatory mechanisms. Circulation. 2003;107:984–991.
39. Akar FG, Rosenbaum DS. Transmural electrophysiological heterogene- 62. Kuwahara F, Kai H, Tokuda K, Takeya M, Takeshita A, Egashira K,
ities underlying arrhythmogenesis in heart failure. Circ Res. 2003;93:638– Imaizumi T. Hypertensive myocardial fibrosis and diastolic dysfunction:
645. doi: 10.1161/01.RES.0000092248.59479.AE. another model of inflammation? Hypertension. 2004;43:739–745. doi:
40. Smith JH, Green CR, Peters NS, Rothery S, Severs NJ. Altered patterns of 10.1161/01.HYP.0000118584.33350.7d.
gap junction distribution in ischemic heart disease: an immunohistochemi- 63. Vanderheyden M, Paulus WJ, Voss M, Knuefermann P, Sivasubramanian
cal study of human myocardium using laser scanning confocal micros- N, Mann D, Baumgarten G. Myocardial cytokine gene expression is
copy. Am J Pathol. 1991;139:801–821. higher in aortic stenosis than in idiopathic dilated cardiomyopathy. Heart.
41. Li D, Fareh S, Leung TK, Nattel S. Promotion of atrial fibrillation by heart fail- 2005;91:926–931. doi: 10.1136/hrt.2004.035733.
ure in dogs: atrial remodeling of a different sort. Circulation. 1999;100:87–95. 64. Frantz S, Nahrendorf M. Cardiac macrophages and their role in ischaemic
42. Sanders P, Morton JB, Davidson NC, Spence SJ, Vohra JK, Sparks PB, heart disease. Cardiovasc Res. 2014;102:240–248. doi: 10.1093/cvr/cvu025.
Kalman JM. Electrical remodeling of the atria in congestive heart failure: 65. Heymans S, Corsten MF, Verhesen W, Carai P, van Leeuwen RE, Custers
electrophysiological and electroanatomic mapping in humans. Circulation. K, Peters T, Hazebroek M, Stöger L, Wijnands E, Janssen BJ, Creemers EE,
2003;108:1461–1468. doi: 10.1161/01.CIR.0000090688.49283.67. Pinto YM, Grimm D, Schürmann N, Vigorito E, Thum T, Stassen F, Yin X,
43. Pang H, Ronderos R, Pérez-Riera AR, Femenía F, Baranchuk A. Mayr M, de Windt LJ, Lutgens E, Wouters K, de Winther MP, Zacchigna
Reverse atrial electrical remodeling: a systematic review. Cardiol J. S, Giacca M, van Bilsen M, Papageorgiou AP, Schroen B. Macrophage
2011;18:625–631. microRNA-155 promotes cardiac hypertrophy and failure. Circulation.
44. Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS, Stanley WC. 2013;128:1420–1432. doi: 10.1161/CIRCULATIONAHA.112.001357.
Myocardial fatty acid metabolism in health and disease. Physiol Rev. 66. Mann DL. Targeted anticytokine therapy and the failing heart. Am J Cardiol.
2010;90:207–258. doi: 10.1152/physrev.00015.2009. 2005;95(11A):9C–16C, 38C. doi: 10.1016/j.amjcard.2005.03.007.
45. Sambandam N, Lopaschuk GD, Brownsey RW, Allard MF. Energy metab- 67. Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT, Anti TNFTACHFI.
olism in the hypertrophied heart. Heart Fail Rev. 2002;7:161–173. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chi-
46. Allard MF. Energy substrate metabolism in cardiac hypertrophy. Curr meric monoclonal antibody to tumor necrosis factor-α, in patients with moder-
Hypertens Rep. 2004;6:430–435. ate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive
47. Wang ZV, Ferdous A, Hill JA. Cardiomyocyte autophagy: meta- Heart Failure (ATTACH) trial. Circulation. 2003;107:3133–3140.
bolic profit and loss. Heart Fail Rev. 2013;18:585–594. doi: 10.1007/ 68. Mann DL, McMurray JJ, Packer M, Swedberg K, Borer JS, Colucci WS, Djian
s10741-012-9350-y. J, Drexler H, Feldman A, Kober L, Krum H, Liu P, Nieminen M, Tavazzi L, van
48. Korvald C, Elvenes OP, Myrmel T. Myocardial substrate metabolism Veldhuisen DJ, Waldenstrom A, Warren M, Westheim A, Zannad F, Fleming T.
influences left ventricular energetics in vivo. Am J Physiol Heart Circ Targeted anticytokine therapy in patients with chronic heart failure: results of
Physiol. 2000;278:H1345–H1351. the Randomized Etanercept Worldwide Evaluation (RENEWAL). Circulation.
49. Davidoff AJ, Davidson MB, Carmody MW, Davis ME, Ren J. Diabetic 2004;109:1594–1602. doi: 10.1161/01.CIR.0000124490.27666.B2.
cardiomyocyte dysfunction and myocyte insulin resistance: role of glu- 69. Heymans S, Hirsch E, Anker SD, Aukrust P, Balligand JL, Cohen-Tervaert
cose-induced PKC activity. Mol Cell Biochem. 2004;262:155–163. JW, Drexler H, Filippatos G, Felix SB, Gullestad L, Hilfiker-Kleiner D,
Schiattarella and Hill   Targeting Maladaptive Hypertrophy   1445

Janssens S, Latini R, Neubauer G, Paulus WJ, Pieske B, Ponikowski 88. Milani RV, Lavie CJ, Mehra MR, Ventura HO, Kurtz JD, Messerli FH.
P, Schroen B, Schultheiss HP, Tschöpe C, Van Bilsen M, Zannad F, Left ventricular geometry and survival in patients with normal left ven-
McMurray J, Shah AM. Inflammation as a therapeutic target in heart fail- tricular ejection fraction. Am J Cardiol. 2006;97:959–963. doi: 10.1016/j.
ure? A scientific statement from the Translational Research Committee of amjcard.2005.10.030.
the Heart Failure Association of the European Society of Cardiology. Eur 89. Milani RV, Drazner MH, Lavie CJ, Morin DP, Ventura HO. Progression
J Heart Fail. 2009;11:119–129. doi: 10.1093/eurjhf/hfn043. from concentric left ventricular hypertrophy and normal ejection fraction
70. Oka T, Hikoso S, Yamaguchi O, Taneike M, Takeda T, Tamai T, Oyabu J, to left ventricular dysfunction. Am J Cardiol. 2011;108:992–996. doi:
Murakawa T, Nakayama H, Nishida K, Akira S, Yamamoto A, Komuro I, 10.1016/j.amjcard.2011.05.038.
Otsu K. Mitochondrial DNA that escapes from autophagy causes inflamma- 90. Drazner MH. The progression of hypertensive heart disease. Circulation.
tion and heart failure. Nature. 2012;485:251–255. doi: 10.1038/nature10992. 2011;123:327–334. doi: 10.1161/CIRCULATIONAHA.108.845792.
71. Anversa P, Levicky V, Beghi C, McDonald SL, Kikkawa Y. Morphometry 91. Krishnamoorthy A, Brown T, Ayers CR, Gupta S, Rame JE, Patel PC,
of exercise-induced right ventricular hypertrophy in the rat. Circ Res. Markham DW, Drazner MH. Progression from normal to reduced left
1983;52:57–64. ventricular ejection fraction in patients with concentric left ventricular
72. Anversa P, Capasso JM, Ricci R, Sonnenblick EH, Olivetti G. hypertrophy after long-term follow-up. Am J Cardiol. 2011;108:997–
Morphometric analysis of coronary capillaries during physiologic myo- 1001. doi: 10.1016/j.amjcard.2011.05.037.
cardial growth and induced cardiac hypertrophy: a review. Int J Microcirc 92. Hill JA, Karimi M, Kutschke W, Davisson RL, Zimmerman K, Wang
Clin Exp. 1989;8:353–363. Z, Kerber RE, Weiss RM. Cardiac hypertrophy is not a required com-
73. Heusch G, Libby P, Gersh B, Yellon D, Böhm M, Lopaschuk G, Opie L. pensatory response to short-term pressure overload. Circulation.
Cardiovascular remodelling in coronary artery disease and heart failure. 2000;101:2863–2869.
Lancet. 2014;383:1933–1943. doi: 10.1016/S0140-6736(14)60107-0. 93. Hill JA, Rothermel B, Yoo KD, Cabuay B, Demetroulis E, Weiss RM,
74. Oka T, Akazawa H, Naito AT, Komuro I. Angiogenesis and cardiac hyper- Kutschke W, Bassel-Duby R, Williams RS. Targeted inhibition of cal-
trophy: maintenance of cardiac function and causative roles in heart failure. cineurin in pressure-overload cardiac hypertrophy: preservation of sys-
Circ Res. 2014;114:565–571. doi: 10.1161/CIRCRESAHA.114.300507. tolic function. J Biol Chem. 2002;277:10251–10255. doi: 10.1074/jbc.
75. Weber KT, Janicki JS, Shroff SG, Pick R, Chen RM, Bashey RI. Collagen M110722200.
remodeling of the pressure-overloaded, hypertrophied nonhuman primate 94. Esposito G, Rapacciuolo A, Naga Prasad SV, Takaoka H, Thomas SA,
myocardium. Circ Res. 1988;62:757–765. Koch WJ, Rockman HA. Genetic alterations that inhibit in vivo pressure-
76. Perry GJ, Wei CC, Hankes GH, Dillon SR, Rynders P, Mukherjee R, overload hypertrophy prevent cardiac dysfunction despite increased wall
Spinale FG, Dell’Italia LJ. Angiotensin II receptor blockade does not stress. Circulation. 2002;105:85–92.
improve left ventricular function and remodeling in subacute mitral regur- 95. Levy D, Garrison RJ, Savage DD, Kannel WB, Castelli WP. Left ventric-
gitation in the dog. J Am Coll Cardiol. 2002;39:1374–1379. ular mass and incidence of coronary heart disease in an elderly cohort:
77. Bauersachs J, Widder JD. Endothelial dysfunction in heart failure. the Framingham Heart Study. Ann Intern Med. 1989;110:101–107.
Pharmacol Rep. 2008;60:119–126. 96. Mehta SK, Rame JE, Khera A, Murphy SA, Canham RM, Peshock RM,
de Lemos JA, Drazner MH. Left ventricular hypertrophy, subclinical ath-
Downloaded from http://ahajournals.org by on February 27, 2024

78. Marti CN, Gheorghiade M, Kalogeropoulos AP, Georgiopoulou VV,


Quyyumi AA, Butler J. Endothelial dysfunction, arterial stiffness, and erosclerosis, and inflammation. Hypertension. 2007;49:1385–1391. doi:
heart failure. J Am Coll Cardiol. 2012;60:1455–1469. doi: 10.1016/j. 10.1161/HYPERTENSIONAHA.107.087890.
jacc.2011.11.082. 97. Heidland UE, Strauer BE. Left ventricular muscle mass and elevated
79. Friehs I, Moran AM, Stamm C, Choi YH, Cowan DB, McGowan FX, heart rate are associated with coronary plaque disruption. Circulation.
del Nido PJ. Promoting angiogenesis protects severely hypertrophied 2001;104:1477–1482.
hearts from ischemic injury. Ann Thorac Surg. 2004;77:2004–2011. doi: 98. Drazner MH. The transition from hypertrophy to failure: how
10.1016/j.athoracsur.2003.11.003. certain are we? Circulation. 2005;112:936–938. doi: 10.1161/
80. Pearlman JD, Hibberd MG, Chuang ML, Harada K, Lopez JJ, Gladstone CIRCULATIONAHA.105.558734.
SR, Friedman M, Sellke FW, Simons M. Magnetic resonance mapping 99. Khouri MG, Peshock RM, Ayers CR, de Lemos JA, Drazner MH. A
demonstrates benefits of VEGF-induced myocardial angiogenesis. Nat 4-tiered classification of left ventricular hypertrophy based on left ven-
Med. 1995;1:1085–1089. tricular geometry: the Dallas Heart Study. Circ Cardiovasc Imaging.
81. Hariawala MD, Horowitz JR, Esakof D, Sheriff DD, Walter DH, Keyt B, 2010;3:164–171. doi: 10.1161/CIRCIMAGING.109.883652.
Isner JM, Symes JF. VEGF improves myocardial blood flow but produces 100. Artham SM, Lavie CJ, Milani RV, Patel DA, Verma A, Ventura HO.
EDRF-mediated hypotension in porcine hearts. J Surg Res. 1996;63:77– Clinical impact of left ventricular hypertrophy and implications for
82. doi: 10.1006/jsre.1996.0226. regression. Prog Cardiovasc Dis. 2009;52:153–167. doi: 10.1016/j.
82. Mack CA, Patel SR, Schwarz EA, Zanzonico P, Hahn RT, Ilercil A, pcad.2009.05.002.
Devereux RB, Goldsmith SJ, Christian TF, Sanborn TA, Kovesdi I, 101. Koren MJ, Devereux RB, Casale PN, Savage DD, Laragh JH. Relation of
Hackett N, Isom OW, Crystal RG, Rosengart TK. Biologic bypass with the left ventricular mass and geometry to morbidity and mortality in uncom-
use of adenovirus-mediated gene transfer of the complementary deoxyri- plicated essential hypertension. Ann Intern Med. 1991;114:345–352.
bonucleic acid for vascular endothelial growth factor 121 improves myo- 102. Aronow WS, Epstein S, Koenigsberg M, Schwartz KS. Usefulness
cardial perfusion and function in the ischemic porcine heart. J Thorac of echocardiographic left ventricular hypertrophy, ventricular tachy-
Cardiovasc Surg. 1998;115:168–176. cardia and complex ventricular arrhythmias in predicting ventricular
83. Losordo DW, Vale PR, Symes JF, Dunnington CH, Esakof DD, Maysky fibrillation or sudden cardiac death in elderly patients. Am J Cardiol.
M, Ashare AB, Lathi K, Isner JM. Gene therapy for myocardial an- 1988;62:1124–1125.
giogenesis: initial clinical results with direct myocardial injection of 103. Casale PN, Devereux RB, Milner M, Zullo G, Harshfield GA, Pickering
phVEGF165 as sole therapy for myocardial ischemia. Circulation. TG, Laragh JH. Value of echocardiographic measurement of left ven-
1998;98:2800–2804. tricular mass in predicting cardiovascular morbid events in hypertensive
84. Meerson FZ. Compensatory hyperfunction of the heart and cardiac insuf- men. Ann Intern Med. 1986;105:173–178.
ficiency. Circ Res. 1962;10:250–258. 104. Kannel WB, Gordon T, Castelli WP, Margolis JR. Electrocardiographic
85. Gunther S, Grossman W. Determinants of ventricular function in pres- left ventricular hypertrophy and risk of coronary heart disease: the
sure-overload hypertrophy in man. Circulation. 1979;59:679–688. Framingham study. Ann Intern Med. 1970;72:813–822.
86. Huber D, Grimm J, Koch R, Krayenbuehl HP. Determinants of ejection 105. Lavie CJ, Ventura HO, Messerli FH. Left ventricular hypertrophy: its rela-
performance in aortic stenosis. Circulation. 1981;64:126–134. tionship to obesity and hypertension. Postgrad Med. 1992;91:131–132, 135.
87. Krayenbuehl HP, Hess OM, Ritter M, Monrad ES, Hoppeler H. Left ventricu- 106. Lee M, Gardin JM, Lynch JC, Smith VE, Tracy RP, Savage PJ, Szklo
lar systolic function in aortic stenosis. Eur Heart J. 1988;9(suppl E):19–23. M, Ward BJ. Diabetes mellitus and echocardiographic left ventricular
1446  Circulation  April 21, 2015

function in free-living elderly men and women: the Cardiovascular study evaluating regression of ventricular enlargement (preserve) trial.
Health Study. Am Heart J. 1997;133:36–43. Circulation. 2001;104:1248–1254.
107. Molkentin JD, Lu JR, Antos CL, Markham B, Richardson J, Robbins J, 121. Solomon SD, Appelbaum E, Manning WJ, Verma A, Berglund T,
Grant SR, Olson EN. A calcineurin-dependent transcriptional pathway Lukashevich V, Cherif Papst C, Smith BA, Dahlöf B; Aliskiren in Left
for cardiac hypertrophy. Cell. 1998;93:215–228. Ventricular Hypertrophy (ALLAY) Trial Investigators. Effect of the di-
108. Cingolani HE, Pérez NG, Cingolani OH, Ennis IL. The Anrep effect: 100 rect Renin inhibitor aliskiren, the angiotensin receptor blocker losartan,
years later. Am J Physiol Heart Circ Physiol. 2013;304:H175–H182. doi: or both on left ventricular mass in patients with hypertension and left
10.1152/ajpheart.00508.2012. ventricular hypertrophy. Circulation. 2009;119:530–537. doi: 10.1161/
109. Kentish JC, Wrzosek A. Changes in force and cytosolic Ca2+ concentra- CIRCULATIONAHA.108.826214.
tion after length changes in isolated rat ventricular trabeculae. J Physiol. 122. Thum T, Condorelli G. Long noncoding RNAs and MicroRNAs in car-
1998;506 (pt 2):431–444. diovascular pathophysiology. Circ Res. 2015;116:751–762. doi: 10.1161/
110. Patten RD, Hall-Porter MR. Small animal models of heart failure: CIRCRESAHA.116.303549.
development of novel therapies, past and present. Circ Heart Fail. 123. McKinsey TA, Kass DA. Small-molecule therapies for cardiac hypertro-
2009;2:138–144. doi: 10.1161/CIRCHEARTFAILURE.108.839761. phy: moving beneath the cell surface. Nat Rev Drug Discov. 2007;6:617–
111. Weber KT, Janicki JS, Pick R, Abrahams C, Shroff SG, Bashey RI, Chen 635. doi: 10.1038/nrd2193.
RM. Collagen in the hypertrophied, pressure-overloaded myocardium. 124. Berry JM, Cao DJ, Rothermel BA, Hill JA. Histone deacetylase inhibi-
Circulation. 1987;75(1 pt 2):I40–I47. tion in the treatment of heart disease. Expert Opin Drug Saf. 2008;7:53–
112. Nagatomo Y, Carabello BA, Coker ML, McDermott PJ, Nemoto S, 67. doi: 10.1517/14740338.7.1.53.
Hamawaki M, Spinale FG. Differential effects of pressure or volume 125. Xie M, Hill JA. HDAC-dependent ventricular remodeling. Trends
overload on myocardial MMP levels and inhibitory control. Am J Physiol Cardiovasc Med. 2013;23:229–235. doi: 10.1016/j.tcm.2012.12.006.
Heart Circ Physiol. 2000;278:H151–H161. 126. Cao DJ, Wang ZV, Battiprolu PK, Jiang N, Morales CR, Kong Y,
113. Alyono D, Anderson RW, Parrish DG, Dai XZ, Bache RJ. Alterations Rothermel BA, Gillette TG, Hill JA. Histone deacetylase (HDAC) inhibi-
of myocardial blood flow associated with experimental canine left ven- tors attenuate cardiac hypertrophy by suppressing autophagy. Proc Natl
tricular hypertrophy secondary to valvular aortic stenosis. Circ Res. Acad Sci U S A. 2011;108:4123–4128. doi: 10.1073/pnas.1015081108.
1986;58:47–57. 127. Kong Y, Tannous P, Lu G, Berenji K, Rothermel BA, Olson EN, Hill
114. Nguyen TN, Chagas AC, Glantz SA. Left ventricular adaptation to JA. Suppression of class I and II histone deacetylases blunts pressure-
gradual renovascular hypertension in dogs. Am J Physiol. 1993;265(1 pt overload cardiac hypertrophy. Circulation. 2006;113:2579–2588. doi:
2):H22–H38. 10.1161/CIRCULATIONAHA.106.625467.
115. Kjeldsen SE, Dahlöf B, Devereux RB, Julius S, Aurup P, Edelman J, 128. Xie M, Kong Y, Tan W, May H, Battiprolu PK, Pedrozo Z, Wang
Beevers G, de Faire U, Fyhrquist F, Ibsen H, Kristianson K, Lederballe- ZV, Morales C, Luo X, Cho G, Jiang N, Jessen ME, Warner JJ,
Pedersen O, Lindholm LH, Nieminen MS, Omvik P, Oparil S, Snapinn Lavandero S, Gillette TG, Turer AT, Hill JA. Histone deacetylase in-
S, Wedel H; LIFE (Losartan Intervention for Endpoint Reduction) Study hibition blunts ischemia/reperfusion injury by inducing cardiomyo-
Group. Effects of losartan on cardiovascular morbidity and mortality in cyte autophagy. Circulation. 2014;129:1139–1151. doi: 10.1161/
Downloaded from http://ahajournals.org by on February 27, 2024

patients with isolated systolic hypertension and left ventricular hypertro- CIRCULATIONAHA.113.002416.
phy: a Losartan Intervention for Endpoint Reduction (LIFE) substudy. 129. Klapholz M, Maurer M, Lowe AM, Messineo F, Meisner JS, Mitchell J,
JAMA. 2002;288:1491–1498. Kalman J, Phillips RA, Steingart R, Brown EJ Jr, Berkowitz R, Moskowitz
116. Chobanian AV, Bakris GL, Black HR, Cushman WC, Green LA, Izzo JL R, Soni A, Mancini D, Bijou R, Sehhat K, Varshneya N, Kukin M, Katz
Jr, Jones DW, Materson BJ, Oparil S, Wright JT Jr, Roccella EJ; National SD, Sleeper LA, Le Jemtel TH; New York Heart Failure Consortium.
Heart, Lung, and Blood Institute Joint National Committee on Prevention, Hospitalization for heart failure in the presence of a normal left ventricu-
Detection, Evaluation, and Treatment of High Blood Pressure; National lar ejection fraction: results of the New York Heart Failure Registry. J Am
High Blood Pressure Education Program Coordinating Committee. Coll Cardiol. 2004;43:1432–1438. doi: 10.1016/j.jacc.2003.11.040.
The Seventh Report of the Joint National Committee on Prevention, 130. Berenji K, Drazner MH, Rothermel BA, Hill JA. Does load-induced ven-
Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 tricular hypertrophy progress to systolic heart failure? Am J Physiol Heart
report. JAMA. 2003;289:2560–2572. doi: 10.1001/jama.289.19.2560. Circ Physiol. 2005;289:H8–H16. doi: 10.1152/ajpheart.01303.2004.
117. Devereux RB, Dahlöf B, Gerdts E, Boman K, Nieminen MS, 131. Hirota H, Chen J, Betz UA, Rajewsky K, Gu Y, Ross J Jr, Müller W,
Papademetriou V, Rokkedal J, Harris KE, Edelman JM, Wachtell K. Chien KR. Loss of a gp130 cardiac muscle cell survival pathway is a
Regression of hypertensive left ventricular hypertrophy by losartan com- critical event in the onset of heart failure during biomechanical stress.
pared with atenolol: the Losartan Intervention for Endpoint Reduction Cell. 1999;97:189–198.
in Hypertension (LIFE) trial. Circulation. 2004;110:1456–1462. doi: 132. Rogers JH, Tamirisa P, Kovacs A, Weinheimer C, Courtois M, Blumer
10.1161/01.CIR.0000141573.44737.5A. KJ, Kelly DP, Muslin AJ. RGS4 causes increased mortality and reduced
118. Mathew J, Sleight P, Lonn E, Johnstone D, Pogue J, Yi Q, Bosch J, cardiac hypertrophy in response to pressure overload. J Clin Invest.
Sussex B, Probstfield J, Yusuf S; Heart Outcomes Prevention Evaluation 1999;104:567–576. doi: 10.1172/JCI6713.
(HOPE) Investigators. Reduction of cardiovascular risk by regression 133. Badorff C, Ruetten H, Mueller S, Stahmer M, Gehring D, Jung F, Ihling
of electrocardiographic markers of left ventricular hypertrophy by C, Zeiher AM, Dimmeler S. Fas receptor signaling inhibits glycogen
the angiotensin-converting enzyme inhibitor ramipril. Circulation. synthase kinase 3β and induces cardiac hypertrophy following pressure
2001;104:1615–1621. overload. J Clin Invest. 2002;109:373–381. doi: 10.1172/JCI13779.
119. Gosse P, Sheridan DJ, Zannad F, Dubourg O, Guéret P, Karpov Y, de 134. Brancaccio M, Fratta L, Notte A, Hirsch E, Poulet R, Guazzone S, De
Leeuw PW, Palma-Gamiz JL, Pessina A, Motz W, Degaute JP, Chastang Acetis M, Vecchione C, Marino G, Altruda F, Silengo L, Tarone G,
C. Regression of left ventricular hypertrophy in hypertensive patients Lembo G. Melusin, a muscle-specific integrin β1-interacting protein, is
treated with indapamide SR 1.5 mg versus enalapril 20 mg: the LIVE required to prevent cardiac failure in response to chronic pressure over-
study. J Hypertens. 2000;18:1465–1475. load. Nat Med. 2003;9:68–75. doi: 10.1038/nm805.
120. Devereux RB, Palmieri V, Sharpe N, De Quattro V, Bella JN, de Simone 135. Bueno OF, De Windt LJ, Tymitz KM, Witt SA, Kimball TR, Klevitsky R,
G, Walker JF, Hahn RT, Dahlöf B. Effects of once-daily angiotensin- Hewett TE, Jones SP, Lefer DJ, Peng CF, Kitsis RN, Molkentin JD. The
converting enzyme inhibition and calcium channel blockade-based an- MEK1-ERK1/2 signaling pathway promotes compensated cardiac hyper-
tihypertensive treatment regimens on left ventricular hypertrophy and trophy in transgenic mice. EMBO J. 2000;19:6341–6350. doi: 10.1093/
diastolic filling in hypertension: the prospective randomized enalapril emboj/19.23.6341.
Schiattarella and Hill   Targeting Maladaptive Hypertrophy   1447

136. Oh H, Taffet GE, Youker KA, Entman ML, Overbeek PA, Michael the induction of physiological, but not pathological, cardiac hypertro-
LH, Schneider MD. Telomerase reverse transcriptase promotes cardiac phy. Proc Natl Acad Sci U S A. 2003;100:12355–12360. doi: 10.1073/
muscle cell proliferation, hypertrophy, and survival. Proc Natl Acad Sci pnas.1934654100.
U S A. 2001;98:10308–10313. doi: 10.1073/pnas.191169098. 138. Hill JA. Hypertrophic reprogramming of the left ventricle: transla-
137. McMullen JR, Shioi T, Zhang L, Tarnavski O, Sherwood MC, Kang PM, tion to the ECG. J Electrocardiol. 2012;45:624–629. doi: 10.1016/j.
Izumo S. Phosphoinositide 3-kinase(p110alpha) plays a critical role for jelectrocard.2012.08.003.

Response to Schiattarella and Hill


Bertrand Crozatier, MD, PhD; Renée Ventura-Clapier, PhD
We agree with many points presented in the review of Sciattarella and Hill, particularly with their description of the abnor-
malities present in the hypertrophied myocardium. We also acknowledge that ventricular hypertrophy has been identified as a
cardiovascular risk factor. However, this does not imply that hypertrophy, per se, has to be blocked. This management of the
hypertrophy process has proved to be beneficial in a number of studies, but we also presented examples of detrimental effects
of this procedure. Sciattarella and Hill were cautious to say that it cannot be applied to all forms of hypertrophy. We could thus
agree with them, but we fully disagree when they state that chronically maintaining hypertrophy is analogous to an activation
of β-adrenergic and renin-angiotensin-aldosterone systems. We specifically believe the opposite. As shown in our review, an
increased contractility is present in the ventricle that is not allowed to increase its wall thickness. Hypercontractility is necessary
for maintaining a normal ventricular function when wall stress is increased. It is likely that, in patients with pre-existing patholo-
gies such as coronaropathies, a prolonged increased contractility will induce a myocardial ischemia and will be detrimental.
Alternative strategies directed toward the cause of hypertrophy, such as ventricular assistance devices or renin-angiotensin-
aldosterone system and β-adrenergic inhibition, indirectly lead to a regression of hypertrophy. New therapeutic approaches such
as those presented in our article that would reorchestrate the different constitutive elements of the ventricles could be promising
approaches. They would better protect cardiomyocytes than a direct blockade of hypertrophy.
Downloaded from http://ahajournals.org by on February 27, 2024

You might also like