Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

Contents lists available at ScienceDirect

Journal of Molecular and Cellular Cardiology

journal homepage: www.elsevier.com/locate/yjmcc

Review article

Physiological and pathological cardiac hypertrophy


Ippei Shimizu a,b,⁎, Tohru Minamino a,⁎⁎
a
Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
b
Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan

a r t i c l e i n f o a b s t r a c t

Article history: The heart must continuously pump blood to supply the body with oxygen and nutrients. To maintain the high
Received 29 November 2015 energy consumption required by this role, the heart is equipped with multiple complex biological systems that
Received in revised form 10 May 2016 allow adaptation to changes of systemic demand. The processes of growth (hypertrophy), angiogenesis, and met-
Accepted 1 June 2016
abolic plasticity are critically involved in maintenance of cardiac homeostasis. Cardiac hypertrophy is classified as
Available online 2 June 2016
physiological when it is associated with normal cardiac function or as pathological when associated with cardiac
Keywords:
dysfunction. Physiological hypertrophy of the heart occurs in response to normal growth of children or during
Cardiac hypertrophy pregnancy, as well as in athletes. In contrast, pathological hypertrophy is induced by factors such as prolonged
Heart failure and abnormal hemodynamic stress, due to hypertension, myocardial infarction etc. Pathological hypertrophy is
Angiogenesis associated with fibrosis, capillary rarefaction, increased production of pro-inflammatory cytokines, and cellular
Akt dysfunction (impairment of signaling, suppression of autophagy, and abnormal cardiomyocyte/non-cardiomyo-
Autophagy cyte interactions), as well as undesirable epigenetic changes, with these complex responses leading to maladap-
Inflammation tive cardiac remodeling and heart failure. This review describes the key molecules and cellular responses
Epigenetic modification
involved in physiological/pathological cardiac hypertrophy.
MicroRNA
© 2016 Elsevier Ltd. All rights reserved.
Metabolism

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
2. Classification of heart failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
3. Various types of cardiac hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
4. Mechanism of physiological cardiac hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
4.1. Mechanosensors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248
4.2. Thyroid hormone/thyroid hormone receptor signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248
4.3. Insulin-like growth factor-1/insulin-like growth factor-1 receptor/Akt signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248
4.4. Insulin/insulin receptor (IR)/Akt signaling—as a pathway mediating physiological hypertrophy . . . . . . . . . . . . . . . . . . . . . 248
4.5. Akt is a key molecule for cardiac hypertrophy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
4.6. Role of sirtuins in modulating Akt signaling. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
5. Mechanisms of pathological cardiac hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
5.1. Calcineurin-nuclear factor of activated T cells (NFAT) signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
5.2. β-adrenergic receptor signaling and Ca2 +/calmodulin-dependent kinase II signaling . . . . . . . . . . . . . . . . . . . . . . . . . . 250
5.3. cGMP/protein kinase G signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
5.4. Protein kinase C and mitogen-activated protein kinase signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
5.5. Insulin/insulin receptor (IR)/Akt signaling—as a pathway mediating pathological hypertrophy . . . . . . . . . . . . . . . . . . . . . . 251
6. Modifiers of cardiac hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
6.1. Endogenous factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
6.1.1. Autophagy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
6.1.2. Modification of DNA and histones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252

⁎ Correspondence to: I. Shimizu, Department of Cardiovascular Biology and Medicine, Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and
Dental Sciences, 1-757 Asahimachidori, Chuo-ku, Niigata 951-8510, Japan.
⁎⁎ Correspondence to: T. Minamino, Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachidori,
Chuo-ku, Niigata 951-8510, Japan.
E-mail addresses: ippeishimizu@yahoo.co.jp (I. Shimizu), t_minamino@yahoo.co.jp (T. Minamino).

http://dx.doi.org/10.1016/j.yjmcc.2016.06.001
0022-2828/© 2016 Elsevier Ltd. All rights reserved.
246 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

6.1.3. MicroRNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252


6.1.4. Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
6.2. Exogenous factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
6.2.1. Fibroblasts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
6.2.2. Endothelial cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 254
6.2.3. Immune cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 254
6.3. Endogenous and exogenous factors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
6.3.1. Cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
7. Angiogenic response in physiological versus pathological cardiac hypertrophy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
8. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
Disclosure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 257

1. Introduction It is estimated that 50% of heart failure patients have preserved LV


ejection fraction and described as HFpEF. In addition to preserved LV
Mammalian cardiomyocytes generally exit the cell cycle soon after ejection fraction, concentric remodeling and diastolic LV dysfunction
birth, so most cardiomyocytes are terminally differentiated in adults are characteristically observed in HFpEF patients. Overweight/obesity,
and do not proliferate under physiological conditions. However, cardiac hypertension, diabetes mellitus, chronic obstructive pulmonary disease,
tissue exhibits plasticity that enables the heart to respond to environ- anemia and chronic kidney disease induces a systemic inflammatory
mental demands, and cells can grow, shrink, or die in reaction to various state and these systemic disorders are thought to increase the risk of de-
physiological or pathological stresses. Cardiac hypertrophy is classified veloping HFpEF. Structural alterations of HFpEF are characterized with
as physiological when associated with normal cardiac function or as cardiomyocyte hypertrophy and interstitial fibrosis, and functional
pathological when associated with cardiac dysfunction. change is consisted of incomplete relaxation of myocardial strips and in-
Normal physiological enlargement of the heart chiefly occurs creased cardiomyocyte stiffness [5–8]. Replacement fibrosis is usually
through hypertrophy of cardiomyocytes in response to growth of the thought not to develop in HFpEF because cell death does not predomi-
body or exercise, and the enlarged cardiomyocytes receive adequate nantly develop in HFpEF. Whether HFrEF and HFpEF are distinct patho-
nourishment due to corresponding expansion of the capillary network. logical conditions or rather represent a syndrome that exists across a
Structural or functional cardiac abnormalities do not occur in this set- spectrum is yet to be defined [9]. Heart failure, defined on clinical
ting, and physiological hypertrophy is generally not considered to be a terms, derives from numerous different disorders, however, under-
risk factor for heart failure. In contrast, pathological hypertrophy is asso- standing mechanisms of cardiac hypertrophy continues to be essential
ciated with production of high levels of neurohumoral mediators, he- for describing pathologies in this critical condition.
modynamic overload, injury and loss of cardiomyocytes. In the
pathological setting, cardiomyocyte growth exceeds the capacity of
3. Various types of cardiac hypertrophy
the capillaries to adequately supply nutrients and oxygen, leading to
cardiac hypoxia and remodeling in rodents [1,2]. Because cardiac hyper-
Heart responses to environmental conditions and able to grow or
trophy plays a central role in cardiac remodeling and is an independent
shrink. Heart increases in size and depending on the types, strength
risk factor for cardiac events, understanding this process is critically
and duration of stimuli, it results in physiological or pathological cardiac
important.
hypertrophy. Physiological hypertrophy is characterized with normal or
Cardiac dysfunction is associated with a complex spectrum of path-
enhanced contractile function coupled with normal architecture and or-
ophysiological changes, including capillary rarefaction, metabolic de-
ganization of cardiac structure [10]. Pathological hypertrophy associates
rangement, sarcomere disorganization, altered calcium handling,
with increased cardiomyocytes death and fibrotic remodeling, and it is
inflammation, cellular senescence, cell death and fibrosis. Due to such
characterized with reduced systolic and diastolic function that often
complexity, no simple therapeutic approach is adequate for the
progresses towards heart failure. Primary triggering events of cardiac
management of this condition. There is some overlap between the
hypertrophy are mechanical stress and neurohumoral stimulation, and
mechanisms of physiological and pathological cardiac hypertrophy,
these contribute for the modulation of various cellular responses includ-
since there is evidence for a central role of Akt signaling or
ing gene expression, protein synthesis, sarcomere assembly, cell metab-
mechanotransduction in physiological hypertrophy, while neurohor-
olism, leading to the development and progression of cardiac
monal factors or continuously overloaded biomechanical stress
hypertrophy [11–13]. Accumulating evidence indicates that pathologi-
induce multiple signaling pathways, including Akt, in pathological
cal and physiological hypertrophy differs in the signaling pathways
hypertrophy.
that drives these processes. Studies suggest that left ventricular hyper-
trophy provides short-term benefit and long-term harm, however,
mechanism regulating this transition from adaptive to maladaptive hy-
2. Classification of heart failure
pertrophy is yet to be defined [14].
Cardiac hypertrophy can also be classified into various variants de-
There are two broad types of heart failure, heart failure with reduced
pending on the geometries of the heart, whether it develops eccentric
(HFrEF) and heart failure with preserved (HFpEF) ejection fraction.
or concentric growth (Fig. 1). Eccentric hypertrophy develops with vol-
HFrEF develops through the accumulation of myocardial damage and
ume overload and non-pathological eccentric hypertrophy shows in-
progressive loss of cardiomyocytes, and is commonly caused by myo-
creased ventricular volume with a coordinated growth in wall and
cardial infarction, hypertensive heart disease, or cardiomyopathy. Oxi-
septal thickness. In this condition, cardiomyocytes grow both in length
dative stress present within the cardiomyocytes induces
and width. Pathological eccentric hypertrophy generally develops
cardiomyocytes death and replacement fibrosis [3]. Cardiomyocyte
with cardiac diseases such as myocardial infarction and dilated cardio-
loss promotes alterations within the extracellular matrix and contrib-
myopathy, and lead to ventricular dilatation with preferential lengthen-
utes to left ventricular (LV) dilatation and eccentric LV remodeling [4].
ing of cardiomyocytes. Concentric hypertrophy shows an increase in
I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262 247

Fig. 1. Geometry of the heart and signaling pathways contributing mainly for physiological cardiac hypertrophy. Cardiac hypertrophy can be classified as eccentric or concentric
hypertrophy based on the geometry of the heart. Mechanotransduction and Insulin/IGF-1/Akt signaling induce physiological hypertrophy, but excessive activation of these pathways
promotes pathological hypertrophy. Thyroid hormone induces physiological hypertrophy and also has the potential to ameliorate pathological hypertrophy. SIRT3 and SIRT6
suppresses Akt, thereby inhibit hypertrophic responses.

free wall and septal thickness associated with reduced left ventricular described as “physiological” hypertrophy. There is a linear relationship
dimension. Cardiomyocytes typically increase in thickness more than between the increase of body weight and cardiac weight. The increase of
in length [6,15], and this condition usually develops under pathological cardiac weight during growth is largely attributable to enlargement
conditions such as hypertension or valvular diseases, although in some of cardiomyocytes, and there is an almost 3-fold increase of cardiomyo-
settings, exercise such as wrestling is known to induce non-pathological cyte diameter in humans during development from infants to adults.
concentric hypertrophy [16]. Cardiac hypertrophy is generally thought Physiological hypertrophy differs from the pathological hypertrophy
to initially develop as an adaptive response to reduce wall stress thereby that occurs in patients with hypertension, valvular heart disease or
diminish oxygen demand according to the law of Laplace. This is a myocardial infarction. Negative cardiac remodeling including fibrosis
pioneering stress adaptation hypothesis demonstrating that ventricular does not predominantly develop in physiological hypertrophy, and
wall stress is proportional to both ventricular pressure and cavity radius this response is thought harmless and even beneficial in healthy indi-
and inversely proportional to ventricular thickness [17]. Mechanisms viduals. It is generally accepted that both cardiac size and function are
regulating the geometry of the heart is elegantly reviewed by Molkentin dependent on angiogenesis, and dysregulation of coordination between
et al. [18] Several complex mechanisms are reported to be involved in cardiomyocyte growth and angiogenesis in the heart has a causal role in
the molecular basis of physiological and pathological cardiac the shift from adaptive cardiac hypertrophy to heart failure [1,2,19].
hypertrophy. Physiological cardiac hypertrophy develops through finite signals
from growth hormones and mechanical forces. Within a physiological
range, growth hormones such as insulin, insulin-like growth factor1
4. Mechanism of physiological cardiac hypertrophy and thyroid hormone, and mechanical forces induce physiological cardi-
ac hypertrophy via the activation of several signalings such as PI3K
Enlargement of the heart in response to growth, exercise, and preg- (phosphoinositide 3-kinase), Akt, AMP-activated protein kinase and
nancy is associated with maintenance of normal cardiac function and is mTOR (mammalian target of rapamycin) pathways [11,20–24].
248 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

4.1. Mechanosensors shows high affinity for the IGF-1 receptor (IGF-1R) and relatively low af-
finity for the insulin receptor (IR). The circulating level of IGF-1 is in-
Cardiomyocytes is equipped with a machinery to sense and respond versely correlated with cardiovascular disease [35]. Thus, a low IGF-1
to mechanical load, and this system is described as level is associated with a high risk of coronary artery disease [36,37]. A
mechanotransduction. Mechanotransduction enables cardiomyocytes low IGF-1 level also predicts a worse prognosis in patients with myocar-
to convert mechanical stimuli into biochemical events through the dial infarction [38], and elderly persons with low IGF-1 levels have an el-
modulation of signaling molecules. Several protein complexes are re- evated risk of heart failure [39]. IGF-1 is generally thought to have a
ported to link to mechanotransduction and mechanotransmission at cardioprotective role. For example, IGF-1 was reported to protect
three key sites within the cardiac myocyte/cytoskeleton; the sarcomere, cardiomyocytes against oxidative stress and ameliorate cardiac dys-
the intercalated disc and the sarcolemma. Proteins acting as function after myocardial infarction [40,41]. In the heart, IGF-1 is pre-
mechanotransducers responding upon mechanical load are the initial dominantly produced and secreted by fibroblasts, and fibroblast
step in the process that drives physiological and pathological cardiac hy- derived IGF-1 promoted cardiomyocyte hypertrophy and contributed
pertrophy. Mechanotransduction and its downstream molecules func- to suppress cardiac dysfunction [42]. IGF-1 is involved in regulating
tion initially as adaptive responses, however, prolonged and abnormal cell proliferation, differentiation, metabolism, and survival, and its ex-
loading conditions promotes maladaptive cardiac remodeling, associat- pression is critically important for normal prenatal/postnatal growth
ed with altered physiological function and pathological cardiac hyper- and development, with its biological effects being predominantly medi-
trophy (Fig. 1). At the sarcomere, titin and muscle LIM protein (MLP) ated via the IGF-1R. This receptor is reported to have dual activities,
are the two molecules broadly studied to be involved in force transmis- with the IGF-1/IGF-1R/Akt/mammalian target of rapamycin (mTOR)
sion and sensing that would regulate cardiac hypertrophy [25,26]. Both and IGF-1/IGF-1R/ERK signaling pathways being described as canonical
proteins form a vast network with interacting proteins such as titin-Cap pathways, while IGF-1/Gi/phospholipase C (PLC)/inositol 1,4,5 triphos-
(TCAP), calsarcin-1, four-and-a half-LIM domain protein-1 (FHL1) and phate (IP3)/Ca+ signaling is considered to be a non-canonical pathway
muscle-specific ankyrin repeat protein (MARP) family members. In ad- and crosstalk occurs between these pathways [43]. The effects of in-
dition to these molecules existing at the sarcomere, several other mole- creased IGF-1/IGF-1R signaling have been studied in skeletal muscle
cules are located in the intercalated disc (such as N-cadherin) and and in the myocardium. IGF-1 induces skeletal muscle hypertrophy,
sarcolemma (such as Integrins, Vinculin, Talin, Caveolin-3 and Dystro- generally via Akt/mTOR signaling [44], or via the Akt-independent
glycan), and reported to modulate physiology in the heart including car- mTOR/p70S6K pathway in some settings [45]. In athletes, IGF-1 level
diac hypertrophy. The molecular mechanisms of mechanotransduction in cardiac tissue (but not endothelin (ET)-1 or angiotensin II (AngII))
in cardiac hypertrophy and failure, and their therapeutic potentials are was higher than in control subjects, and serum IGF-1 level was shown
elegantly reviewed by Sheikh et al. [27] As described in this review, to increase with exercise [46,47]. These results suggested the contribu-
mechanisms underlying mechanotransduction are yet to be defined tion of IGF-1 in promoting physiological cardiac hypertrophy upon ex-
and further description is beyond the scope of this review. ercise. Genetic deletion of IGF-1 leads to significant reduction of body
weight during development and usually results in embryonic lethality
4.2. Thyroid hormone/thyroid hormone receptor signaling or death from respiratory failure shortly after birth. IGF-1R null mice
show dramatic phenotypic changes, with their birth weight being only
Thyroid hormone exerts its biological effect by binding to thyroid 45% of that for healthy littermates and 100% mortality from respiratory
hormone receptors (TRα and TRβ), and thyroid hormone-TR signaling failure [48,49]. Forced expression of IGF-1 in the heart promotes cardiac
pathway is involved in regulating cardiac contractility, structure, and hypertrophy with cardiac dysfunction [50] or without cardiac dysfunc-
electrophysiology. Whether thyroid hormone promotes physiological tion [51] depending on the transgenes used, the effect of IGF-1 on
cardiac hypertrophy in adults is still controversial, however, its role in non-cardiomyocytes, or the observation period. A murine model of car-
postnatal hypertrophy is well accepted. After birth, thyroid hormone in- diomyocyte-specific overexpression of Igf1r mimics physiological, not
creases dramatically in children [23]. Thyroid hormone promotes the pathological, cardiac hypertrophy by promoting gene transcription
physiological growth of the neonatal cardiomyocytes, and this is medi- that is not predominantly related to pathological cardiac growth [22].
ated by the activation of p38MAPK and PI3K/Akt/mTOR signaling [24, Interestingly, Igf1r deletion does not lead to any differences in the base-
28]. Thyroid hormone optimizes cardiac geometry, which is known to line phenotype of adult cardiomyocytes. However, cardiac growth in re-
contribute for improving myocardial performance [29]. Continuous in- sponse to exercise was inhibited in one study, but not in another study,
jection of thyroid hormone promoted cardiac hypertrophy in rats and possibly due to different exercise protocols [21,52]. Consistent with the
this was not associated with fibrosis and even reduced collagen gene ex- findings by Ikeda et al., suppression of IGF-1 signaling did not affect the
pression [30,31]. In a rat LV pressure overload model, thyroid hormone hypertrophic response to chronic β-adrenergic stimulation [53]. The re-
promoted non-significant increase in LV dry weight, and suppressed sults obtained by modulation of IGF-1 signaling vary between studies
heart failure. Myocytes treated with thyroid hormone were and animal models, possibly because of its role in a broad spectrum of
hypertrophied, but this was associated with an increase in α-MHC and biological activities that include cardiac growth, aging, metabolism,
sarcoplasmic reticulum Ca2+ (SERCA) expression, similar to physiolog- cell proliferation, and cell survival. However, it seems that activation
ical hypertrophy [32]. In a rat pathological hypertrophy model with of IGF-1 signaling is beneficial within a certain finite physiological
myocardial infarction, thyroid hormone changed cardiac geometry range, contributing to the maintenance of cardiac homeostasis (Fig.1).
and improved systolic cardiac function [33]. Studies indicate that thy-
roid hormone can convert pathological to physiological cardiac hyper- 4.4. Insulin/insulin receptor (IR)/Akt signaling—as a pathway mediating
trophy and details are reviewed and discussed by Pantos et al [34]. physiological hypertrophy

4.3. Insulin-like growth factor-1/insulin-like growth factor-1 receptor/Akt Insulin is a hormone secreted by pancreatic β-cells that was discov-
signaling ered in 1921 and was initially identified as a critical regulator of the
blood glucose level. Over time, evidence has been obtained to demon-
Insulin-like growth factor 1(IGF-1) is structurally similar to insulin strate that insulin has a broad range of biological activities, and it is
and is mainly synthesized and secreted by the liver in response to stim- now known that insulin signaling coordinates glucose/lipid metabo-
ulation by growth hormone, although it is also produced by extrahepat- lism, protein synthesis, cell growth and differentiation, apoptosis, and
ic tissues and acts locally in a paracrine or autocrine fashion. IGF-1 has a senescence [54,55]. Insulin binds to the insulin receptor (IR), after
molecular weight of 7.6 kDa and shares 50% homology with insulin. It which insulin receptor tyrosine kinase phosphorylates insulin receptor
I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262 249

substrate (IRS) and activates PI3K, leading to generation of suppression of mitochondrial fatty acid oxidation [72]. Androgens
phosphoinositide-3,4,5 triphosphate that mediates the phosphorylation such as testosterone promote cardiac hypertrophy and heart failure.
and activation of Akt. It is generally accepted that insulin signaling is Anti-androgen therapy was shown to suppress Akt-mTOR signaling
critically important for the normal physiological growth and develop- and attenuate cardiac hypertrophy and left ventricular dysfunction in
ment of cardiac tissue (Fig.1). Cardiac specific overexpression of Akt a model of left ventricular pressure overload [73]. In a murine pressure
was shown to enhance cardiac function with left ventricular hypertro- overload model, heterozygous depletion of Akt significantly attenuated
phy [56]. The Akt-mTOR signaling was increased in a murine treadmill cardiomyocyte hypertrophy and improved cardiac dysfunction [1]. Al-
exercise model [57]. PI3K-Akt signaling is reported to increase Ca2 + though it is widely accepted that Akt is the critical regulator coordinat-
handling and improve cardiac inotropism, and was mediated partially ing angiogenesis and physiological growth of the myocardium, there is
via the stabilization of L-type Ca2 + channel (LTCC) [58]. Preferential accumulating evidence that excessive Akt signaling has a pathological
translocation of Akt to the sarcoplasmic reticulum was shown to en- effect under some circumstances by promoting dysregulated cardiac
hance cardiomyocyte contractility without promoting cardiac hypertro- hypertrophy that results in tissue hypoxia and contributes to the pro-
phy by the phosphorylation of phospholamban [59]. Homozygous gression of heart failure.
deletion of Ir in cardiac tissue leads to a significant decrease of cardio-
myocyte size and heart weight, with persistence of a fetal gene expres- 4.6. Role of sirtuins in modulating Akt signaling
sion pattern and reduced cardiac function [20]. Depletion of Ir in the
heart induces mitochondrial dysfunction by reducing the expression Sirtuins (silent mating type information regulation 2 homologs)
of fatty acid oxidation genes and their regulators [60,61]. Systemic de- have recently received considerable attention because of their role in
pletion of Irs1 or Irs2 led to the postnatal general growth retardation aging. Calorie restriction is the best established method of increasing
[62,63]. Recently, an interesting paper by Ikeda et al has described the longevity in various models ranging from Caenorhabditis elegans to
role of IGF-1 and IR mediated signaling in regulating hypertrophic re- mice, and the levels of SIRTs 1, 3, and 6 are reported to be increased
sponse induced by exercise. Homozygous deletion of Igf1r (Igf1r−/−) by calorie restriction [74–77]. It is generally accepted that SIRT3 and
or Ir (Ir−/−) in cardiac tissue is associated with normal or reduced base- SIRT6 are involved in regulation of the aging process in mammals [78–
line cardiac growth, respectively, while the hypertrophic response to 81]. SIRT3 and SIRT6 were recently reported to inhibit cardiac hypertro-
exercise is normal in both cases. Igf1r−/− mice lacking one Ir allele in phy. SIRT3 null mice develop cardiac hypertrophy with cardiac dysfunc-
cardiomyocytes (Igf1r−/− Ir+/−) and Ir−/− mice lacking one Igf1r allele tion, while overexpression of SIRT3 in the heart attenuates agonist-
in cardiomyocytes (Igf1r+/− Ir−/−) show attenuation of cardiac hyper- induced cardiac hypertrophy [82]. Inhibition of SIRT3 expression or sup-
trophy in response to exercise. The phenotype of Igf1r+/− Ir−/− mice pression of SIRT3 activity is associated with an increase of reactive oxy-
is more severe, suggesting that the IR and its downstream molecules gen species (ROS) and activation of Akt signaling [82,83]. Moreover,
are more closely involved in physiological hypertrophy related to exer- specific overexpression of SIRT6 in the heart inhibits cardiac hypertro-
cise [52]. It remains unclear whether crosstalk occurs between the IR phy induced by agonists or pressure overload, and this anti-hypertro-
and IGF-1R during development of pathological cardiac hypertrophy. phic effect of SIRT6 is mediated via inhibition of IGF-1/Akt signaling
Both insulin/IR signaling and IGF-1/IGF-1R signaling share downstream [84] (Fig. 1). SIRT6 interacts with c-Jun, resulting in deacetylation of his-
effectors such as Akt, so further studies are needed to determine wheth- tone H3K9, and thus inhibits Akt expression at the transcriptional level.
er one signaling pathway is more beneficial than the other. The role of SIRT1 in cardiac hypertrophy is complex. Marked eleva-
tion of SIRT1 expression (12.5-fold) induces pathological hypertrophy
4.5. Akt is a key molecule for cardiac hypertrophy associated with cardiac dysfunction, whereas less marked induction
(7.5-fold) attenuates age-related cardiac hypertrophy [85]. In addition,
Akt is the best-characterized molecule in the downstream pathways haploinsufficiency of SIRT1 attenuates cardiac hypertrophy due to pres-
of IGF-1R and IR signaling. It is involved in diverse cellular processes, in- sure overload, while SIRT1 overexpression has a detrimental effect in
cluding the regulation of cell growth, survival, metabolism, and angio- pressure overload models [86]. SIRT1 deacetylates Akt and thus pro-
genesis. Akt promotes cardiac hypertrophy via modulation of several motes its binding to phosphoinositide-3,4,5 triphosphate and subse-
signaling pathways. Eukaryotic translation initiation factor 4E-binding quent activation. Both SIRT1 and Akt stimulate endothelial cell
protein 1(4E-BP1) inhibits the initiation of translation and is negatively proliferation, differentiation, and migration, a process known as
regulated by mTOR, while PI3K-Akt-mTOR signaling suppresses 4E-BP1 sprouting and branching. Neovascularization is suppressed in SIRT-1
and promotes compensatory cardiac hypertrophy. Specific depletion of null mice, while SIRT-1 deficient zebrafish display dysregulation of en-
mTOR in the heart inhibits the protective hypertrophic response and dothelial sprouting and develop malformations of the vascular network
promotes cardiac dysfunction during pressure overload [64]. Akt sup- [87]. Further studies are needed to elucidate the role of SIRT1 in coordi-
presses CCAAT/enhanced-binding protein (C/EBP)β and upregulates nating cardiac angiogenesis and hypertrophy.
Cbp/p300-interacting transactivator with ED-rich carboxy-terminal do-
main 4 (CITED4), which is negatively regulated by C/EBPβ. CITED4 in- 5. Mechanisms of pathological cardiac hypertrophy
creases the expression of GATA4 and promotes cardiac hypertrophy.
Systemic heterozygous C/EBPβ depletion promotes physiological cardi- Enlargement of the heart in response to hypertensive stress, myo-
ac hypertrophy, ameliorates systolic dysfunction, and increases the sur- cardial injury, or excessive neurohumoral activation is associated with
vival rate in a murine model of left ventricular pressure overload [65]. cardiac dysfunction and is described as “pathological hypertrophy”
Activation of Akt initially promotes growth of the heart while preserv- [88]. Cardiac function is initially maintained in pressure overload-in-
ing cardiac function [66–68], but constitutive activation of this signaling duced cardiac hypertrophy, and this is described as the adaptive
pathway leads to pathological hypertrophy and cardiac dysfunction phase. However, sustained pressure overload promotes the transition
[19]. Mice that are transgenic for PI3K, IGF-1, or IGF-1R and mice sub- from the adaptive to maladaptive phase, which is characterized by a re-
jected to exercise training show increased Akt expression (by 4-fold, duced ejection fraction and left ventricular dilatation. Ventricular wall
6-fold, 6-fold, and 1.5- to 2-fold, respectively), and cardiac dysfunction stress is proportional to both the left ventricular (LV) cavity radius
does not occur within this Akt expression range [22,69–71]. However, and LV pressure, while it is inversely proportional to LV wall thickness
forced expression of Akt in cardiac tissue for 6 weeks (a 15-fold increase [17,89]. For this reason, the initial stages of pressure overload hypertro-
compared with normal littermates) caused pathological hypertrophy phy are described as “compensatory”, and were thought to be beneficial.
and cardiac dysfunction [19], while enhancement of cardiac Akt signal- However, LV hypertrophy has been reported as an independent risk fac-
ing promoted pathological hypertrophy associated with progressive tor for adverse cardiovascular events, such as heart failure and
250 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

arrhythmia [90,91]. Furthermore, data from both clinical and basic stud- triphosphate (Ins(1,4,5) P3), which induces intracellular Ca2+ release
ies indicate that cardiac function can be maintained by therapy and activation of calcineurin/NFAT signaling [13,98]. Calcineurin is a
targeting suppression of LV hypertrophy in the setting of hemodynamic Ca2+-activated serine-threonine protein phosphatase that dephosphor-
stress [92–94]. These findings have led to rethinking of the role of cardi- ylates NFAT in the cytoplasm, inducing its nuclear translocation and the
ac hypertrophy under these conditions and it has been suggested that expression of genes involved in hypertrophy. Activation of calcineurin/
heart failure can be treated through modulation of cardiac hypertrophy NFAT signaling alone is sufficient to induce pathological cardiac hyper-
induced by stress [95,96]. trophy [99–101] (Fig. 2). In contrast, physiological cardiac hypertrophy
In the process of hypertrophic remodeling, it is widely accepted that induced by exercise or pregnancy is not associated with increased cal-
the nature of the stress determines the phenotype. Thus, intermittent cineurin/NFAT signaling [102]. Recently, transient receptor potential
pressure overload induces diastolic dysfunction and vascular rarefaction cation (TRPC) channels were reported to be involved in activation of cal-
before cardiac hypertrophy, unlike the changes in exercise models cineurin/NFAT signaling. TRPC channels regulate the movement of Ca2+
employing swimming or running [97]. Pathological cardiac hypertrophy and Na+ in specific microdomains, and expression of these channels
is usually associated with increased rates of myocytes death and fibrotic was reported to be increased along with activation in pathological hy-
remodeling, that would promote systolic and diastolic dysfunction, gen- pertrophy or heart failure [103–106]. TRPC1, TRPC3, and TRPC6 are
erally predisposing to the development of heart failure. The activation of the best known of several isoforms, and inhibition of the gene expres-
mechanotransduction and neurohormonal mechanisms are the initial sion of these isoforms attenuates cardiac hypertrophy induced by ago-
step that drives pathological hypertrophy preceding heart failure. Pro- nists and pressure overload [103,107].
teins acting as mechanotransducers are crucial for physiological hyper-
trophy and dysregulation of this system promotes pathological 5.2. β-adrenergic receptor signaling and Ca2+/calmodulin-dependent ki-
hypertrophy [11]. Adrenergic nervous system and renin-angiotensin nase II signaling
system are extensively studied neurohormonal mechanisms, initially
activated upon stress to increase contractility and survival in the early Patients with heart failure have high blood catecholamine levels and
phase, and become pathological in the chronic phase. The β-adrenergic increased adrenergic tone is correlated with a poor clinical outcome
receptor blockers, angiotensin-converting enzyme inhibitors and angio- [108]. High adrenergic activity is associated with cardiac hypertrophy,
tensin receptor blockers are well accepted to reduce left ventricular and the basal plasma norepinephrine concentration was shown to pre-
mass and contribute for favorable clinical outcome in patients with dict the extent of cardiac hypertrophy in hypertensive patients inde-
heart failure. Different signaling members are proposed to mediate car- pendently of systolic blood pressure and body mass index [109]. It
diac hypertrophy, and several molecules are reported to be involved in was initially considered that β-blockers were contraindicated for pa-
pathogenic signaling pathways and these are discussed next. tients with heart failure, but these drugs later became first-line therapy
for severe heart failure based on data from a number of large-scale clin-
5.1. Calcineurin-nuclear factor of activated T cells (NFAT) signaling ical trials [110,111]. Calmodulin-dependent kinase II (CaMKII) is a ser-
ine/threonine kinase that is regulated by the Ca2 +/calmodulin
Both physiological and pathological cardiac hypertrophy are associ- complex, ROS, and exchange proteins directly activated by cAMP
ated with elevation of cardiomyocyte Ca2+ levels. Ca2+ modulates the (EPAC). Catecholamines bind to adrenergic receptors, which are classi-
activity of various Ca2+-dependent signaling pathways, including cal- fied as G-protein-coupled receptors, and signaling via these receptors
cineurin/nuclear factor of activated T cells (NFAT) signaling and cal- activates CaMKII. Activation of the renin-angiotensin-aldosterone sys-
modulin-dependent kinase II signaling. Neurohumoral mediators such tem increases the angiotensin II level, and directly induces cardiac hy-
as catecholamines and angiotensin II bind to seven-transmembrane re- pertrophy via activation of CaMKII signaling.
ceptors that are coupled to G proteins. Gq signaling activates phospho- Studies performed in humans and rodents have shown that cardiac
lipase C (PLC), and this catalyses the synthesis of inositol 1,4,5- CaMKII activity is increased in heart failure. Gain of CaMKII function

Fig. 2. Signaling pathways for pathological cardiac hypertrophy. Neurohumoral factors, such as catecholamines or angiotensin II, upregulate calcineurin/NFAT and CaMKII/MEF-2 signaling
to induce pathological cardiac hypertrophy. IL-6/gp130/JAK/STAT signaling also promotes pathological hypertrophy. Nitric oxide and natriuretic peptide-coupled/cGMP/PKG signaling is
anti-hypertrophic, while PDE5 and PDE9A promote pathological cardiac hypertrophy by suppressing this signaling pathway.
I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262 251

leads to cardiac hypertrophy, while inhibition of CaMKII ameliorates of the MKK1/ERK1/2 pathway, and also protects against cell death [135],
myocardial hypertrophy and improves heart failure [112–115]. There while deletion of ERK (Erk1–/– and Erk2+/–) does not attenuate the hy-
are four isoforms of CaMKII encoded by different genes (α, β, γ and δ). pertrophic response to various stimuli [136]. These results indicate
Each isoform has a different tissue distribution and CaMKIIδ is the that ERK1/2 signaling is pro-hypertrophic, but is not essential for the de-
major isoform in the myocardium. CaMKIIδ null mice showed less velopment of cardiac hypertrophy. It was recently reported that the
prominent cardiac hypertrophy in response to pressure overload ERK1/2 signaling pathway coordinates eccentric and concentric growth
along with amelioration of systolic dysfunction and a higher survival of the heart. Mice with complete cardiac deletion of ERK1/2 (Erk1–/–
rate [116,117]. Activation of Gq/PLC/IP3 signaling by norepinephrine, MHC-Cre Erk2fl/fl) developed eccentric cardiac hypertrophy (lengthen-
phenylephrine, or endothelin-1 induces the growth of neonatal rat ven- ing) and systolic dysfunction in response to pressure overload or stimu-
tricular myocytes, along with elevated expression of fetal genes such as lation with angiotensin II/phenylephrine, while Mkk1 transgenic mice
those encoding atrial natriuretic peptide (ANP), brain natriuretic pep- showed concentric growth (wall thickening) of the heart [137]. Al-
tide (BNP), myosin light chain-2, or α and β-myosin heavy chain. Forced though p38MAPK signaling is generally accepted to be pathological, its
expression of CaMKII results in the same phenotypic changes as induc- role in promoting cardiac hypertrophy is puzzling and still controver-
tion of neurohumoral agonist/Gq signaling, while pharmacological inhi- sial. Activation of MKK6-p38MAPK signaling induces hypertrophy,
bition of CaMKII suppresses such negative remodeling [118–120]. whereas activation of MKK3 signaling promotes apoptosis in isolated
CaMKII induces cardiomyocyte growth via phosphorylation of class II neonatal cardiomyocytes. In contrast, studies in mouse models have in-
histone deacetylases (HDACs), especially HDAC4 and HDAC5, by pro- dicated that p38MAPK does not induce hypertrophy, but plays a role in
moting the export of these molecules from the nucleus, leading to dere- regulating apoptosis, fibrosis, and left ventricular dilatation [138].
pression of MEF-2-mediated gene expression and cardiac hypertrophy Forced activation of cardiac MKK3-p38MAPK signaling leads to dilated
[115,121–123]. In short, activation of CaMKII is sufficient to induce cardiomyopathy with early death, while activation of MKK6-p38MAPK
pathological cardiac hypertrophy (Fig. 2). signaling results in restrictive cardiomyopathy and early death. Howev-
er, left ventricular hypertrophy does not develop in either of these
5.3. cGMP/protein kinase G signaling models [139]. JNK mainly has an anti-hypertrophic effect and is report-
ed to repress cardiac hypertrophy through inhibition of calcineurin/
Nitric oxide (NO) and natriuretic peptide-coupled signaling is medi- NFAT signaling [140]. MKK4 is a positive regulator of JNK and a key me-
ated by a second messenger, cyclic guanosine monophosphate (cGMP). diator that prevents the transition from an adaptive response to mal-
cGMP is involved in regulating physiological processes in the myocardi- adaptive cardiac hypertrophy through modulation of calcineurin/NFAT
um, including cell growth and apoptosis, and activation of cGMP/pro- signaling [141].
tein kinase G (PKG) signaling is reported to inhibit cardiac MAPK phosphatase 1 (MKP-1) is a negative feedback inhibitor of
hypertrophy [124]. Phosphodiesterases (PDEs) specifically cleave the MAPK signaling, and constitutive expression of MKP-1 in the heart
3′,5′-cyclic phosphate moiety of cyclic adenosine monophosphate downregulates p38MAPK, JNK1/2, and ERK1/2, and also prevents induc-
(cAMP) and/or cGMP. At least 21 PDE genes have been cloned and clas- tion of hypertrophy by catecholamines or aortic banding [142]. Recent-
sified into 11 families. Among them, PDE-5, PDE-6, and PDE-9 specifical- ly, dual-specificity phosphatases 1 and 4 (Dusp1 and 4, also known as
ly act on cGMP; PDE-4, PDE-7, and PDE-8 are specific for cAMP; and MKP-1 and 4, respectively) were reported to have a cardioprotective
PDE-1, PDE-2, PDE-3, PDE-10, and PDE-11 interact with both cAMP role through inhibition of p38MAPK. Dusp1 and 4 null mice show eleva-
and cGMP [125]. Activity of both cGMP and PKG is reduced in patients tion of p38MAPK with no change of JNK or ERK1/2 levels, and have a low
with heart failure [126]. There is evidence that a PDE-5 inhibitor has survival rate associated with systolic dysfunction and cardiac dilatation
an anti-hypertrophic effect in the setting of pressure overload, which [143].
is associated with activation of PKG and its target molecule, regulator
of G protein signaling 2 (RGS2), and with suppression of TRPC6/calcine- 5.5. Insulin/insulin receptor (IR)/Akt signaling—as a pathway mediating
urin/NFAT signaling [127,128]. Recently, cGMP-selective PDE9A was de- pathological hypertrophy
tected in mammalian hearts and was reported to contribute to
suppression of cGMP signaling, thereby promoting cardiac hypertrophy It is widely accepted that insulin signaling pathway is essential in
and remodeling [129] (Fig. 2). promoting physiological hypertrophy, however, accumulating evidence
has indicated that insulin/insulin receptor (IR)/Akt signaling should be
5.4. Protein kinase C and mitogen-activated protein kinase signaling maintained within a physiological range to become beneficial. Animal
studies and large-scale clinical trials have not provided conclusive evi-
The α-adrenergic receptors (α-ARs) couple with Gq, and Gq-protein dence about whether insulin signaling is cardioprotective in adults.
kinase C (PKC) signaling and mitogen-activated protein kinase (MAPK) Complete suppression of insulin signaling is detrimental through inhibi-
signaling promote cardiac hypertrophy in mice, while suppression of tion of physiological hypertrophy, while overactivation of this signaling
PKC conversely inhibits GPCR-mediated cardiac hypertrophy [130,131]. pathway induces pathological cardiac hypertrophy and disturbs ho-
MAPK signaling has an important role in the regulation of cell prolif- meostasis. Chronic hyperinsulinemia induces pathological hypertrophy
eration, cell growth, and stress responses. It is induced in by promoting angiotensin II signaling [144]. Also, intensive glycemic
cardiomyocytes by small G proteins (Ras, Rac, Rho, etc.), G-protein- control with insulin did not reduce cardiovascular events in diabetic pa-
coupled receptors, and stress, and is followed by several levels of phos- tients, but instead was associated with an increase of events [145]. In a
phorylation-based amplification. There are three main branches of murine model of chronic pressure overload, cardiac insulin signaling
MAPK signaling, which are p38MAPK, c-Jun N-terminal kinases was markedly increased and mismatch developed between cardiomyo-
(JNKs), and extracellular signal-regulated kinase (ERK)1/2. The mito- cyte size and vascularity, inducing myocardial hypoxia, cardiomyocyte
gen-activated protein kinase kinase (MKK)1/2/ERK1/2/NFAT signaling, death, and cardiac dysfunction. Conversely, inhibition of cardiac insulin
MKK 4/7/JNKs/MEF2 signaling, and MKK3/6/p38MAPK/GATA4 signal- signaling by heterozygous Ir deletion in the heart or whole body hetero-
ing pathways are involved in modulation of cardiac hypertrophy. Nearly zygous Akt deletion significantly attenuated cardiomyocyte hypertro-
all of the MAPK signaling components are activated in end-stage heart phy and improved cardiac dysfunction due to pressure overload.
failure in humans and in animal models of pathological cardiac hyper- These findings suggest that sustained activation of cardiac insulin sig-
trophy [132–134]. It is generally considered that the ERK1/2 signaling naling causes myocardial hypoxia and systolic dysfunction when the
pathway is pro-hypertrophic. Forced expression of MKK1 induces phys- heart is subjected to pressure overload [1]. Kemi et al. have shown
iological cardiac hypertrophy with cardiomyocyte growth via activation that Akt-mTOR signaling is increased with physiological cardiac
252 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

hypertrophy upon exercise, in contrast, they also showed that this sig- 6.1.2. Modification of DNA and histones
naling pathway is reduced with pathological hypertrophy in a murine Distinct families of DNA elements described as promoters and en-
LV-pressure overload model. This report indicates that Akt-mTOR sig- hancers regulate gene transcription. By modulating the structure of
naling may diverge physiological from pathological hypertrophy [57]. chromatin, epigenetic modifications regulate the access of promoters
Ras homology enriched in brain (Rheb) is the positive regulator for and enhancers to DNA and thus control gene expression. Acetylation
mTOR, and Rheb-mTOR signaling was shown to be activated in the of histones on lysine residues promotes the relaxation of chromatin,
heart upon metabolic stress, especially in a cardiac ischemia model, leading to transcriptional activation, while suppression of histone acet-
and promote myocardial damage via the suppression of autophagy ylation leads to condensation of chromatin and inhibition of gene ex-
[146]. Systemic insulin resistance is prevalent among patients with sys- pression. Active promoters are characterized by high levels of
tolic dysfunction and it has been reported to increase the future risk of monoacetylated lysine 9 and lysine 14 (H3K9ac and H3K14ac) and
heart failure [147–149]. Although several lines of evidence have sug- trimethylated (me3) lysine 4 (H3K4me3) on histone H3. Inactive pro-
gested a link between heart failure and systemic metabolic dysfunction, moters are characterized by methylated DNA, trimethylation of lysine
the underlying molecular mechanisms and clinical implications have 9 and lysine 27 on histone H3 (H3K9me3 and H3K27me3), and
been unclear. Recently, pressure overload was shown to induce adipose deacetylation of histones. Active enhancers are enriched in H3K27ac,
tissue inflammation by promoting lipolysis, leading to systemic meta- while lack of H3K27ac and a high level of H3K27me3 define an enhancer
bolic dysfunction. It was also demonstrated that suppression of adipose as “poised”.
tissue inflammation improved systemic insulin resistance Acetylation of histones is regulated by histone acetyltransferases
(hyperinsulinemia) and ameliorated cardiac dysfunction induced by (HATs) and histone deacetylases (HDACs). Overactivation of HATs,
chronic pressure overload, possibly via suppression of excessive cardiac such as CREB-binding protein (CBP) and p300, induces hypertrophy
insulin signaling [1,150]. These results indicate that insulin signaling and left ventricular remodeling in mice [159,160]. Hypertrophy of
also needs to be regulated within a certain physiological range to main- non-necrotic tissue in a myocardial infarction model also requires
tain homeostasis, with too much or too little signaling via the IR being p300 HAT activity [161], and phenylephrine-induced cardiac hypertro-
detrimental (Fig.1). phy is associated with high p300 activity. HDACs have an opposing
role to HATs by mediating the removal of acetyl groups. There are
three main classes of HDACs, which are class I (HDAC1, 2, 3, and 8),
6. Modifiers of cardiac hypertrophy class II (HDAC4, 5, 6, 7, 9, and 10), and class III. Class II HDACs have an
anti-hypertrophic effect [162]. In mice, depletion of HDAC5 or HDAC9
6.1. Endogenous factors was reported to increase sensitivity to stress signals and enhance cardi-
ac hypertrophy, owing to the role of these enzymes in silencing MEF2C
6.1.1. Autophagy [163]. Recently, an increased load on the heart was shown to induce nu-
Autophagy is an evolutionary conserved catabolic system involved clear export of HDAC4, demethylation of H3K9, and activation of ANP,
in degradation of unnecessary or dysfunctional cellular components with these responses not requiring an increase of histone acetylation
through lysosomal machinery, thus recycling amino acids for the syn- [164]. Unlike class II HDACs, the class I HDACs mediate hypertrophic re-
thesis of proteins that are essential for cell survival. Autophagy is acti- sponses. HDAC2 induces cardiac hypertrophy by repression of INPP5F,
vated by various stresses, including starvation, ischemia-reperfusion, the gene coding for phosphatidylinositide phosphatase SAC2, which is
infection, ROS, and hypoxia [151]. A basal level of autophagy is essential a negative regulator of the Akt/GSK-3β pathway [165]. Similar to class
for removal and renewal of dysfunctional mitochondria and for main- II HDACs, class III HDACs are associated with inhibition of cardiac hyper-
taining cellular homeostasis. The role of stress-induced autophagy has trophy and with improved cardiomyocyte survival. Modulation of
yet to be completely defined, although it is known to promote or atten- HDAC expression, including suppression of specific pro-hypertrophic
uate pathology in several disease models [152–154]. An increase of the HDACs, has the potential to become next-generation therapy for heart
autophagic response is protective during cardiac ischemia [155], but ac- failure.
tivation of autophagy is associated with deterioration of cardiac func- Methylation of cytosine on DNA induces the repression of transcrip-
tion in a pressure overload model [156] or an ischemia-reperfusion tion. Three DNA methyltransferases (DNMTs) have been identified in
model [155]. There is accumulating evidence that autophagy has a role mammals (DNMT1, 3A, 3B). The epigenetic profile undergoes alter-
in cardiac hypertrophy. Autophagy protein 5 (encoded by Atg5) is the ations in end-stage cardiomyopathy. Increased expression of certain
key molecule in formation of autophagic vesicles. In adult mice, cardi- genes in the failing heart is associated with promoter demethylation,
ac-specific deletion of Atg5 was reported to induce cardiac hypertrophy, but hypermethylation does not show a clear correlation with reduced
systolic dysfunction, and left ventricular dilatation [157]. Charged gene expression [166]. Significant hypomethylation at satellite regions
multivesicular body protein 2B (CHMP2B) regulates the fusion of has also been found in failing hearts, and is correlated with a 27-fold in-
autophagosomes and lysosomes, and therefore is essential for autopha- crease of the corresponding transcripts [167]. Accordingly, further stud-
gy. Atrogin-1 degrades CHMP2B and maintains it at a physiological level. ies are warranted to determine the role of alterations of DNA
Whole body deletion of atrogin-1 leads to a marked increase of methylation in the progression of heart failure.
CHMP2B, along with suppression of the autophagic response in cardiac
tissue and significant cardiac hypertrophy at the age of 16 months. 6.1.3. MicroRNA
While systolic cardiac dysfunction does not develop, there is significant MicroRNA (miRNA) is a class of small non-coding RNAs involved in
reduction of the lifespan, possibly due to diastolic dysfunction induced repression of translation or transcriptional degradation of target
by cardiac fibrosis [158]. Another study showed that heterozygous dele- mRNAs and Mirbase lists N 2000 known human miRNAs [168]. A single
tion of Becn1 (the gene coding for Beclin-1 that is involved in early miRNA may have tens to hundreds of target genes and there is accumu-
autophagosome formation) inhibits autophagy in cardiomyocytes and lating evidence that miRNAs have a role in cardiac development, hyper-
ameliorates pathological remodeling caused by pressure overload. In trophy, and failure. In mice with left ventricular pressure overload or
contrast, forced expression of Becn1 upregulates autophagic responses calcineurin overexpression, 11 miRNAs showed upregulation, and five
and promotes pathological cardiac remodeling [156]. These results indi- of them were also increased in the failing human heart [169]. Forced ex-
cate that autophagic responses are critically involved in maintaining ho- pression of these five miRNAs (miR-23a, miR-23b, miR-24, miR-195 and
meostasis in response to cardiac stress, with the pathological effects of miR-214Z) induced hypertrophy of primary cultured cardiomyocytes
inhibiting autophagy being dependent on the animal model, extent of [169,170]. In addition, overexpression of miR-212/132 in
induction, and timing of observation. cardiomyocytes induced pathological cardiac hypertrophy and heart
I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262 253

failure in mice. Conversely, cardiac hypertrophy and systolic dysfunc- in end-stage failure [189]. In rats with abdominal aortic constriction and
tion due to pressure overload were significantly ameliorated in miR- compensated cardiac hypertrophy, glycolysis increases without oxida-
212/132 null mice. FoxO3 is a transcriptional molecule involved in tion of glucose [190]. In rat LV-pressure overload model generated
anti-hypertrophic and pro-autophagic responses. It is located down- with clipping of ascending aorta, total lactate production per gram left
stream of (and is negatively regulated by) the insulin signaling path- ventricular weights was higher in hypertrophied hearts than non-
way, with activation of insulin signaling promoting translocation of hypertrophied controls when evaluated 11–12 weeks after the opera-
FoxO3 from the nucleus and inhibiting its activity. Both miR-212 and tion [191]. In another rat LV-pressure overload model due to aortic
miR-132 were reported to directly inhibit FoxO3 expression and induce arch constriction, it was reported that cardiac oxidation of glucose in-
pathological hypertrophy via upregulation of calcineurin/NFAT signal- creased initially during the phase of compensated hypertrophy, follow-
ing and suppression of autophagy [170], while cardiomyocyte-specific ed by a decrease after the onset of systolic dysfunction [180]. In mice
overexpression of miR-208a induced pathological hypertrophy associ- with LV-pressure overload, glucose oxidation, glycolysis and lactate ox-
ated with cardiac dysfunction [171]. Moreover, miR-22 is reported to idation were reduced 6 weeks after the operation [192]. Interestingly,
be increased in the failing human heart [172]. Stimulation by angioten- spontaneously hypertensive rats show higher glucose oxidation rates
sin II or phenylephrine increases miR-22 expression in cultured in the phase of hypertrophy before cardiac dysfunction occurs [193].
cardiomyocytes along with enlargement of cardiomyocytes, while sup- On the other hand, glucose oxidation is unchanged in rats with myocar-
pression of miR-22 ameliorates angiotensin II- or phenylephrine-in- dial infarction, despite the development of systolic dysfunction [194],
duced cardiomyocyte hypertrophy [173]. However, in vivo deletion of while patients with idiopathic dilated cardiomyopathy show increased
miR-22 leads to cardiac dilatation and contractile dysfunction associat- total glucose utilization [195]. These findings suggest that changes of
ed with reduction of SERCA2a pump activity [174]. Several other glucose utilization depend on the pathogenesis and stage of heart failure
miRNAs are also involved in promotion or suppression of cardiac hyper- [177].
trophy and the details were recently reviewed elsewhere [175,176]. Glucose is transported into the cytosol by glucose transporters
(GLUTs), with GLUT1 being the major transporter in the fetal heart
6.1.4. Metabolism and GLUT4 having this role in the adult heart. Modulating GLUT1 or
Heart weight and myocyte fiber size are reduced by starvation or se- GLUT4 expression was reported to regulate hypertrophy. Deletion of
vere weight loss. Heart size is increased in obesity without hypertension GLUT1 in cardiomyocytes did not alter cardiac hypertrophy, fibrosis,
or other cardiovascular and/or metabolic abnormalities, while modest or capillary rarefaction in response to pressure overload. Although
weight loss is associated with reduction of heart size without hemody- GLUT1 depletion reduced glycolysis and glucose oxidation by 50%,
namic changes in normotensive obese subjects. Thus, it is highly possi- with a reciprocal increase of FA oxidation, there was no exacerbation
ble that there are mechanisms which link heart size to the systemic of systolic dysfunction [196]. Forced expression of GLUT1 in cardiac tis-
nutritional status. sue attenuated pathological remodeling due to left ventricular pressure
The heart requires a huge amount of ATP to allow it to continuously overload, but promoted cardiac hypertrophy and failed to prevent sys-
pump blood. Under physiological conditions, more than 95% of the ATP tolic dysfunction [197]. Cardiomyocyte-specific deletion of GLUT4 in-
utilized in the heart is generated by oxidative phosphorylation, with duces cardiomyocyte hypertrophy with increased expression of ANP
most of the remainder coming from glycolysis (5%) and to a lesser ex- and BNP in the ventricles. Myocardial fibrosis does not develop, and
tent from the Krebs cycle. Between 60 and 70% of the ATP generated is both basal and isoproterenol-induced contraction is preserved,
used for cardiac contraction and the remaining 30–40% is consumed representing the compensated phase of cardiac hypertrophy [198].
by various ion pumps. Approximately 70−90% of cardiac ATP originates Under hypoxic conditions, oxidation of glucose is generally considered
from oxidation of fatty acids (FAs), while the remaining 10–30% is pro- to be a protective response that generates ATP, but switching from FA
duced by oxidation of glucose, lactate, and ketone bodies [177]. During to glucose was reported to activate mTOR signaling and promote cardiac
glycolysis, glucose is converted into pyruvate and this is subsequently hypertrophy [199].
converted to lactate or oxidized in the mitochondria. It is generally ac- It was recently reported that fructose metabolism is elevated in the
cepted that utilization of FAs is impaired in the failing heart. FA uptake failing heart and contributes to the progression of cardiac hypertrophy.
is reduced in models of heart failure induced by rapid pacing and a Hypoxia inducible factor-1α (HIf-1α) increases the expression of splice
high-salt diet [178,179], and FA transporter activity is reduced when factor 3b subunit 1 (SF3B1) and mediates alternative splicing of
systolic function is impaired [179–182]. In some models, enzymes in- ketohexokinase (KHK) pre-mRNA. Splice switching of KHK-A to KHK-
volved in FA oxidation and the FA oxidation rate are decreased in the C promotes fructolysis and induces cardiac hypertrophy by upregula-
early (compensated) phase of left ventricular hypertrophy [180,183], tion of lipid and protein biosynthesis. Depletion of SF3B1 suppressed
while this occurs during the late phase along with cardiac dysfunction cardiac hypertrophy in a murine model of left ventricular pressure over-
in other models [182]. It was thought that FA overload developed in load, and SF3B1 was markedly increased in patients with hypertrophic
the failing heart, promoting oxidative stress derived from toxic lipid de- cardiomyopathy and aortic stenosis [200]. These results suggest that in-
rivatives [184], but studies based on pharmacological inhibition of car- hibition of the SF3B1- KHK-C axis and modulation of fructolysis could
diac FA have challenged this concept. In mice fed a high fat diet, potentially be new targets for treating heart failure, but more investiga-
myocardial insulin resistance developed and promoted left ventricular tions are required to define the pathological link between metabolic
remodeling in response to pressure overload [185]. Another study dysfunction and cardiac hypertrophy.
showed reduction of cardiac hypertrophy and improvement of cardiac
function after maintaining Dahl salt-sensitive rats on a high fat diet 6.2. Exogenous factors
[186], while acute suppression of FA oxidation led to significant worsen-
ing of systolic function in patients with end-stage heart failure [187]. 6.2.1. Fibroblasts
Therefore, additional investigation of the role of FA metabolism in Fibroblasts account for 27% of all cardiac cells in mice and 64% in rats,
heart failure is required to elucidate the therapeutic potential of modu- while non-cardiomyocytes (mainly fibroblasts) account for 72% of total
lating FA metabolism. cells in humans [201,202]. Thus, fibroblasts are one of the chief cell
FA and glucose negatively regulate each other, so that use of one types in cardiac tissue, contributing to maintenance of homeostasis
substrate directly inhibits use of the other (the Randle cycle) [188]. In under physiological conditions and to tissue remodeling in response
the physiological state, the heart predominantly generates ATP from to stress. Cardiac fibroblasts express extracellular matrix (ECM) recep-
fatty acids, but progression of heart failure promotes utilization of glu- tors coupling mechanical stimuli to functional responses, and are critical
cose and the heart eventually becomes unable to utilize either substrate regulators of the composition and stiffness of the ECM and fibrotic
254 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

response [203,204]. Myocardial fibrosis and stiffness is an important 6.2.2. Endothelial cells
component of diastolic dysfunction. Mechanical stress activates cardiac Studies indicate that endothelial cells communicate with
fibroblasts and promotes the production and release of bioactive medi- cardiomyocytes by secreting autocrine and paracrine mediators. Cardiac
ators [205]. Cardiac fibroblasts originate from several sources, including endothelial cells secrete vasodilative and vasoconstrictive factors such
resident fibroblasts, bone marrow progenitors, and endothelial cells. as nitric oxide (NO) and endothelin-1 (ET-1). They also secrete physio-
Pressure overload induces rapid expansion of resident fibroblasts origi- logical active substances such as C-type natriuretic peptide (CNP),
nating from the epicardium and endocardium, rather than fibroblasts neuregulins (NRGs) and apelin. It is well known that angiogenesis per
derived from hematopoietic precursors [206]. The role of cardiac fibro- se is sufficient to promote cardiac hypertrophy possibly because an in-
blasts in cardiac hypertrophy is not yet clearly defined, partly due to crease in the supply of nutrients and oxygen would itself induce hyper-
the lack of fibroblast-specific genes. Several Cre systems (including trophic responses. Accumulating evidence indicates that some of the
Fsp1-Cre, Postn-Cre, Tcf21-Cre, Col1a1-Cre, and Col1a2-Cre) have been humoral mediators secreted by endothelial cells, as described above,
created to specifically deplete target genes in fibroblasts. Some of would also modulate this process. NO and CNP are secreted from cardiac
these systems were found to be nonspecific under physiological or path- endothelial cells and up-regulate cyclic GMP (cGMP)-cGMP dependent
ological conditions, however, accumulating evidence indicates that fi- protein kinase 1 (PKGI) signaling, contributing for the suppression of
broblasts communicate with cardiomyocytes by secreting humoral cardiac hypertrophy [3,220]. Atrial natriuretic peptide (ANP) and B-
mediators such as IGF-1, fibroblast growth factor-2 (FGF-2), type natriuretic peptide (BNP) are secreted from cardiomyocytes and
transforming growth factor-β1 (TGF-β1), interleukin-33(IL-33) in an also known to activate cGMP-PKGI signaling, thereby inhibit cardiac hy-
autocrine and paracrine manner. Krüppel-like factor 5 was recently re- pertrophy [129,220]. Neuregulins (NRGs) are part of the epidermal
ported to have a critical influence on cardiac function through regula- growth factor family known to be involved in broad biological process-
tion of fibroblasts. In a fibroblast Klf5 deletion model generated by es. Endothelial cells in adult heart are reported to produce and secrete
crossing Postn-Cre mice with Klf5flox/flox mice, cardiac hypertrophy NRG-1, which binds to erythroblastic leukemia viral oncogene homolog
and fibrosis due to pressure overload were both attenuated. Interesting- (ErbB) receptor. In neonatal rat cardiomyocytes, addition of NRG-1 in-
ly, Postn-Cre Klf5flox/flox mice subjected to severe pressure overload also duced cardiac hypertrophy, and this was suppressed by ErbB inhibition
showed amelioration of cardiac hypertrophy and fibrosis, despite signif- [221,222]. The role of NRG-1-ErbB signaling in pathological hypertro-
icant cardiac dysfunction along with increased mortality. IGF-1 expres- phy is yet to be defined. Apelin is reported to be secreted by endocardial
sion was transactivated by Klf5 in fibroblasts and this promoted and endothelial cells and bind to apelin receptor (also known as APJ re-
cardiomyocyte hypertrophy contributing for the suppression of cardiac ceptor) in cardiomyocytes. Injection of apelin did not induce cardiac hy-
dysfunction [42]. Fibroblast growth factor-2 (FGF-2) is predominantly pertrophy in rodents and apelin knockout model mice developed
expressed by cardiac fibroblasts and induces cardiac hypertrophy by cardiac hypertrophy upon LV pressure-overload. APJ is activated by me-
the activation of mitogen-activated protein kinase signaling [207,208]. chanical stretch and mediates hypertrophic response, thereby genetic
Hypertrophic response in cardiomyocytes is significantly attenuated in depletion of APJ suppressed load induced cardiac hypertrophy in a β-
FGF-2 null mice in a murine LV pressure overload model [209]. Cardiac arrestin dependent manner [223]. ET-1 is a molecule well known to in-
fibroblasts are also the main source for the production of connective tis- duce cardiac hypertrophy. Addition of ET1 into cardiomyocytes pro-
sue growth factor (CTGF). In cultured cardiomyocytes, CTGF induced motes hypertrophy and suppression of ET1 receptor attenuated
cardiomyocyte hypertrophy by activating Akt signaling [210]. Forced cardiac hypertrophy in a rat LV pressure overload model [224,225].
expression of CTGF was shown to promote age-dependent cardiac hy- ET1 is secreted from endothelial cells, cardiomyocytes and fibroblasts.
pertrophy and dilatation [211]. Recently, fibroblast-derived microRNA Interestingly, endothelial ET1 depletion led to a worsening of cardiac
21 (miR-21) was reported to promote cardiomyocyte hypertrophy, function with comparable cardiac hypertrophy upon LV pressure over-
while pharmacological inhibition of miR-21 ameliorated the hypertro- load [226], and further studies are needed to clearly define the cell spe-
phic response to angiotensin II [212]. TGF-β is secreted by cardiac fibro- cific role of ET1 in heart failure.
blasts, cardiomyocytes and endothelial cells. TGF-β/Smad3 signaling is
described as canonical pathway, contributing for the progression of car- 6.2.3. Immune cells
diac fibrosis. Neutralization of TGF-β in a rat LV pressure-overload In the physiological state, resident cardiac macrophages are pre-
model attenuated fibroblast activation and collagen mRNA expression, dominantly maintained through self-renewal and to a lesser extent by
but did not affect cardiac hypertrophy [213]. Another study showed recruitment of monocytes. Although pro-inflammatory cytokines such
that the inhibition of TGF-β reduces non-myocyte proliferation and at- as TNF-α are known to promote cardiac hypertrophy, the role of inflam-
tenuate cardiac hypertrophy in a murine hypertrophic cardiomyopathy matory cells in the process of hypertrophy is yet to be defined. Mono-
model [214]. TGF-β/TGF-β activated kinase 1(TAK1) signaling is de- cyte-derived macrophages increase in response to cardiac injury or
scribed as non-canonical pathway, and in addition to its contribution stress. Cardiac hypertrophy or fibrosis does not develop after infusion
for promoting fibrosis, it is reported to enhance cardiac hypertrophy. of angiotensin II for 2 days, but the macrophage population is already
TAK1 is activated upon LV pressure-overload and forced expression of expanding at this time, suggesting that resident and recruited immune
TAK1 induced cardiac hypertrophy and heart failure [215]. Conversely, cells are involved in the early response to stress that precedes cardiac
dominant negative form of TAK1 attenuated TGF-β induced cardiomyo- hypertrophy [227]. In hypertensive Ren-2 rats, depletion of macro-
cyte hypertrophy [216]. These results suggest that the suppression of phages by clodronate resulted in systolic dysfunction and cardiac dilata-
TGF-β/TAK1 signaling would become anti-fibrotic and anti-hypertro- tion, but these changes were not associated with cardiac hypertrophy
phic therapy in heart failure [203]. Fibroblasts are reported to secrete [228]. Infusion of angiotensin II ameliorated cardiac fibrosis in MCP-1
anti-hypertrophic factors. Adrenomedullin is secreted by cardiac fibro- KO mice, but cardiac hypertrophy was not improved [229]. Among the
blasts, cardiomyocytes and endothelial cells and suppress cardiomyo- various types of macrophages, it is widely accepted that M1 (classically
cyte hypertrophy [217]. Adrenomedullin loss of function murine activated) macrophages are inflammatory cells that produce pro-in-
model exhibit marked cardiac hypertrophy and fibrosis in response to flammatory cytokines, while M2 (alternatively activated) macrophages
aortic constriction [218]. IL-33 is predominantly produced in cardiac fi- are anti-inflammatory cells [230,231]. Several subsets of macrophages
broblasts upon mechanical stress and suppresses cardiac hypertrophy have been reported in the mouse heart. The CCR2+ CD11chi subset is
induced by AngII, phenylephrine or LV pressure overload [219]. Taken predominantly derived from circulating monocytes and is involved in
together, these results indicate that fibroblasts can directly regulate car- cardiac inflammation [227], but other cardiac macrophage subsets
diac hypertrophy and that modulation of fibroblast function could be may contribute to maintenance of homeostasis. Since several inflamma-
important for suppression of cardiac remodeling. tory cytokines induce cardiac hypertrophy, analyzing specific
I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262 255

macrophage subsets could help to elucidate their roles in cardiac hyper- Studies indicate that intercellular communications mediated by pro
trophy. In addition, the roles of other immune cells, such as T lympho- or anti-inflammatory cytokines secreted from cardiomyocytes and non-
cytes, mast cells, and neutrophils, in several models of cardiac cardiomyocytes, modulate cardiac hypertrophy and pathologies in
hypertrophy have been reviewed by Ryan et al. [232]. heart failure [232,256,257] (Fig. 3A, B).
As detailed above, various mechanisms contribute to the develop-
ment or suppression of pathological cardiac hypertrophy. Further stud-
6.3. Endogenous and exogenous factors ies are needed to elucidate these mechanisms in sufficient detail to
allow development of new therapies for severe heart failure that target
6.3.1. Cytokines the suppression of cardiac hypertrophy.
There is increasing evidence that inflammation has a central role in
heart failure. In patients with dilated cardiomyopathy, ischemic heart 7. Angiogenic response in physiological versus pathological cardiac
disease, and myocarditis, cardiac tissue is infiltrated by pro-inflammato- hypertrophy
ry cells such as macrophages and the levels of inflammatory cytokines
are elevated [233]. Studies using several rodent models to investigate Coordination of cardiac growth and angiogenesis appears to be crit-
the role of inflammation in heart failure have indicated that an in- ically important in separating physiological or adaptive hypertrophy
creased inflammatory response generally has an unfavorable impact from pathological cardiac hypertrophy. It has long been observed that
on cardiac homeostasis [234–237]. Levels of pro-inflammatory cyto- capillary rarefaction occurs in cardiac dysfunction, whereas the number
kines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-6, and of capillaries is maintained or increased in hearts with physiological hy-
IL-1β, have been reported to show a correlation with the severity of pertrophy [258–260]. Vascular endothelial growth factor (VEGF) is a
heart failure [238]. Cardiomyocyte-specific gain of function for TNF-α major angiogenic molecule with a pivotal role in vessel formation, de-
induces cardiac hypertrophy and fibrosis, resulting in dilated cardiomy- velopment, and maintenance of homeostasis in several key organs
opathy with cardiac dysfunction [239]. Conversely, systemic TNF-α de- [261–263]. Hypertrophic stimuli initially upregulate VEGF expression
letion ameliorates cardiac hypertrophy and remodeling in a pressure in the heart, allowing cardiac growth and angiogenesis to be coordinat-
overload model [240]. Interestingly, TNF-α was shown to have a ed in the adaptive phase of hypertrophy. Shiojima et al. provided the
cardioprotective role in desmin-deficient mice through ectopic expres- first evidence that disruption of coordination between cardiac hypertro-
sion of keratins K8 and K18 [241]. Induction of IL-6 by angiotensin II was phy and angiogenesis promotes the transition to heart failure [19]. VEGF
reported to have a causal role in the development of cardiac hypertro- deficiency has been reported to occur during pressure overload, which
phy. Overexpression of both IL-6 and the IL-6 receptor in mice induces would lead to a decrease of the capillary density in the heart and pro-
concentric cardiac hypertrophy, fibrosis, and diastolic dysfunction mote the transition from compensatory hypertrophy to cardiac failure
[242,243], while deletion of IL-6 ameliorates angiotensin II-induced car- [264]. Hypoxia inducible factor-1α (HIF-1α) and GATA4 are widely
diac hypertrophy, inflammation, and fibrosis [244–246]. IL-6 binds to studied regulators of VEGF, with Hif-1α being the major positive regu-
the IL-6 receptor and activates receptor subunit glycoprotein-130 lator of VEGF under hypoxic conditions. During the acute phase of left
(gp130). The IL-6/gp130/Janus kinase (JAK)/signal transducer and acti- ventricular pressure overload, Hif-1α and VEGF maintain pace with
vator of transcription (STAT) pathway promotes cardiac hypertrophy myocardial hypertrophy, while suppression of Hif-1α has a causal role
and cardiac dysfunction [247] (Fig. 2). Although the results of these in the decline of VEGF expression and evidence of cardiac hypoxia dur-
studies suggested a pathological role of IL-6 in cardiac remodeling, dele- ing the late phase of pressure overload [2]. Recently, the concept that
tion of IL-6 had no effect on cardiac hypertrophy or fibrosis in a pressure cardiac hypoxia develops in non-ischemic heart failure is challenged
overload model. However, deletion of IL-6st, the gene coding for gp130, by a study analyzing dilated cardiomyopathy (DCM) patients. Dass et
resulted in dilated cardiomyopathy with impaired cardiac function and al. have performed cardiac magnetic resonance imaging and concluded
increased mortality [248]. Thus, further investigation is required to elu- that although microvascular dysfunction develops with DCM, cardiac
cidate the role of IL-6 in cardiac hypertrophy under physiological and hypoxia does not develop in this condition. This study suggested the
pathological conditions. It was reported that Il1b-deficient mice with dissociation between microvascular dysfunction and oxygenation in
pressure overload show reduced cardiac hypertrophy and improved DCM patients at least in a rest condition [265]. Further studies are need-
cardiac function, indicating that signaling via IL-1β promotes patholog- ed to test whether cardiac hypoxia develops in non-ischemic heart fail-
ical hypertrophy [249]. Treatment with IL-1β induces cardiac dysfunc- ure patients.
tion in mice, while a clinical study showed that blockade of IL-1β The zinc finger transcription factor GATA4 is abundantly expressed
signaling increases oxygen consumption and improves exercise toler- from the embryonic period to adulthood and regulates cardiac-specific
ance in patients with systolic heart failure [250]. These findings suggest gene expression. GATA4 promotes angiogenesis by binding to the pro-
that further clinical trials are warranted to determine whether inhibi- moter region of VEGF, thereby regulating transcription of this gene.
tion of IL-1β has a favorable effect on heart failure. Forced expression of GATA4 in adult murine cardiomyocytes enhances
Nuclear factor kappa-light-chain-enhancer of activated B cells (NF- angiogenesis, while myocyte-specific deletion of GATA4 leads to a de-
κB) is one of the critical regulators of inflammation and is known to in- crease of capillary density [266]. Activation of GATA4 is predominantly
crease the production of pro-inflammatory cytokines such as TNF-α. Re- independent of hypoxia and is induced by various molecules involved
cently, NF-κB was reported to mediate angiotensin II and endothelin-1 in cardiac hypertrophy, such as ERK, p38MAPK, and Akt. Either ERK or
induced cardiomyocyte hypertrophy [251], but data obtained from p38MAPK phosphorylates GATA4, leading to up-regulation of its tran-
transgenic mice are conflicting. In a murine pressure overload model, scriptional activity [267,268]. Interestingly, Hif-1α and GATA4 seem to
deletion of p65 NF-κB in cardiomyocytes reduced cardiac function regulate the VEGF promoter by acting on distinct regulatory elements
along with decreased vascularity and increased fibrosis [252], but an- [2,266]. There is evidence to suggest that Akt is one of the critical regu-
other study in a pressure overload model showed that NF-κB/NFAT sig- lators of angiogenic responses and cell growth. In a hind limb ischemia
naling would promote cardiac hypertrophy and ventricular remodeling model, Akt1 mediates VEGF production and promotes angiogenesis
[253]. [269], and Akt also promotes homing of endothelial cells to ischemic re-
Interleukin 10 (IL-10) is an anti-inflammatory cytokine, and recently gions in vivo [270]. Besides Akt/GATA4 signaling, Akt influences angio-
it was shown to suppress LV-pressure overload induced cardiac hyper- genesis by promoting the production of nitric oxide (NO) [271–273]. It
trophy by the inhibition of NF-κB signaling [254]. Interestingly, in the has been proposed that an increase of angiogenesis per se is sufficient
LV-pressure overload model, T-cell accumulation was shown to contrib- to induce cardiac hypertrophy via an NO-dependent or independent
ute for an increase in IL-10 expression in cardiac tissue [255]. mechanisms. Forced expression of PR39, a peptide regulator of
256 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

Fig. 3. Modifiers of cardiac hypertrophy. A; Endogenous factors contributing for the induction or suppression of cardiac hypertrophy. Dysregulation in autophagy, DNA and histone
modification, micro RNA and metabolism in cardiomyocytes promote cardiac hypertrophy. TNF-α secreted from cardiomyocytes induces cardiac hypertrophy, in contrast, atrial
natriuretic peptide (ANP), brain natriuretic peptide (BNP) and adrenomedullin inhibit this response in an autocrine manner. B; Exogenous factors contributing for the induction or
suppression of cardiac hypertrophy. Endothelial cells secrete anti-hypertrophic mediators such as nitric oxide (NO), C-type natriuretic peptide (CNP), apelin and adrenomedullin. They
also secrete a pro-hypertrophic factor such as neuregulin-1 (NRG-1). Fibroblasts secrete predominantly pro-hypertrophic molecules such as transforming growth factor β (TGF-β),
insulin like growth factor-1 (IGF-1), fibroblast growth factor-2 (FGF-2), connective tissue growth factor (CTGF) and microRNA21 (miR-21). They secrete anti-hypertrophic molecules
like adrenomedullin and interleukin-33 (IL-33). Immune cells also contribute for pro or anti-hypertrophic responses, and these are mediated by TNF-α or interleukin-10 (IL-10)
respectively.

angiogenesis, or placental growth factor induced cardiac hypertrophy in and hormonal changes. Peripartum cardiomyopathy (PPCM) is an un-
the absence of hypertrophic stimulations partially via NO mediated sig- common complication of pregnancy defined as development of heart
naling [274,275]. In a murine model of cardiac tissue-specific, tetracy- failure with a reduced ejection fraction (b45%) in the last month of
cline-regulated gain of function for Akt (15-fold increase compared pregnancy or within 5 months of delivery in a patient who has no
with normal littermates), short–term induction of Akt (2 weeks) in- other cause of heart failure [276]. The pathogenesis of PPCM is un-
duced physiological hypertrophy together with induction of VEGF, but known, but imbalance of cardiac angiogenesis and reduced vascular
forced expression of this gene for 6 weeks led to pathological hypertro- density was recently shown to be associated with its development
phy, cardiac dysfunction, and reduced VEGF expression associated with [277].
myocardial capillary rarefaction [19]. These results suggest that the ef- Heart failure with reduced ejection fraction (HFrEF) develops due to
fect of Akt on the heart depends on the duration and extent of its activa- the accumulation of myocardial damage and is commonly caused by
tion, and that sustained Akt signaling can be detrimental by promoting myocardial infarction, hypertensive heart disease, or cardiomyopathy.
pathological hypertrophy. A marked decrease of myocardial capillary density has been found in pa-
Accumulating evidence indicates that capillary rarefaction has a tients with idiopathic dilated cardiomyopathy who develop congestive
pathological role in several disorders. During normal pregnancy, physi- heart failure, suggesting that capillary rarefaction has a causative role
ological cardiac hypertrophy occurs due to expansion of blood volume in the progression of this disease [278]. Interestingly, mechanical
I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262 257

unloading by use of a left ventricular assist device (LVAD) is reported to Japan Heart Foundation Research Grant, The Senri Life Science Founda-
increase the myocardial microvascular density in patients with HFrEF, tion, SENSHIN Medical Research Foundation, ONO Medical Research
although it accompanied by cardiac fibrosis [279]. Foundation, Tsukada Grant for Niigata University Medical Research,
Approximately half of all heart failure patients are considered to The Nakajima Foundation, SUZUKEN memorial foundation, HOKUTO
have heart failure with a preserved ejection fraction (HFpEF), and this Corporation, Inamori Foundation, Mochida Memorial Foundation for
type of heart failure is predominant among elderly persons [280]. Medical & Pharmaceutical Research, Banyu Foundation Research
Heart failure is common in the elderly population since its prevalence Grant, Grant for Basic Science Research Projects from The Sumitomo
shows a dramatic increase with aging (0.3/0.2% among men/women Foundation (to I.S.), and by a grant from Bourbon (to T.M. and I.S).
aged 20–39 years, 1.9/1.4% at 40–59 years, and 14.7/12.8% at
≥80 years) [281]. A recent study showed that cardiac hypertrophy, epi-
cardial coronary artery disease (CAD), coronary microvascular rarefac- References
tion, and myocardial fibrosis have a higher prevalence in HFpEF
[1] I. Shimizu, T. Minamino, H. Toko, S. Okada, H. Ikeda, N. Yasuda, et al., Excessive car-
patients than age-matched controls. Interestingly, the heart weight, car- diac insulin signaling exacerbates systolic dysfunction induced by pressure over-
diac fibrosis, and microvascular density were similar in HFpEF patients load in rodents, J. Clin. Invest. 120 (2010) 1506–1514.
with or without CAD, while capillary rarefaction was correlated with [2] M. Sano, T. Minamino, H. Toko, H. Miyauchi, M. Orimo, Y. Qin, et al., p53-induced
inhibition of Hif-1 causes cardiac dysfunction during pressure overload, Nature
myocardial fibrosis, indicating that cardiac endothelial cell remodeling 446 (2007) 444–448.
has a causal role in the onset and progression of HFpEF [3,282]. There [3] W.J. Paulus, C. Tschope, A novel paradigm for heart failure with preserved ejection
is evidence from several sources that age-related heart failure can de- fraction: comorbidities drive myocardial dysfunction and remodeling through cor-
onary microvascular endothelial inflammation, J. Am. Coll. Cardiol. 62 (2013)
velop in the absence of established risk factors such as hypertension,
263–271.
obesity, diabetes, or atherosclerosis [283–285]. Aging is a normal pro- [4] J.S. Janicki, G.L. Brower, J.D. Gardner, A.L. Chancey, J.A. Stewart Jr., The dynamic in-
cess, but since the incidence and prevalence of heart failure increases teraction between matrix metalloproteinase activity and adverse myocardial re-
modeling, Heart Fail. Rev. 9 (2004) 33–42.
with age, aging seems to predispose for pathological cardiac hypertro-
[5] L. van Heerebeek, A. Borbely, H.W. Niessen, J.G. Bronzwaer, J. van der Velden, G.J.
phy. In a recent study, exposure to the circulation of young mice caused Stienen, et al., Myocardial structure and function differ in systolic and diastolic
regression of cardiac hypertrophy in aged mice. This study showed that heart failure, Circulation 113 (2006) 1966–1973.
circulating growth differentiation factor 11 (GDF11) was responsible for [6] D.E. Selby, B.M. Palmer, M.M. LeWinter, M. Meyer, Tachycardia-induced diastolic
dysfunction and resting tone in myocardium from patients with a normal ejection
regression of age-related cardiac hypertrophy, and that the GDF11 level fraction, J. Am. Coll. Cardiol. 58 (2011) 147–154.
declines with aging [286]. Another group showed that pathological car- [7] M. Kasner, D. Westermann, B. Lopez, R. Gaub, F. Escher, U. Kuhl, et al., Diastolic tis-
diac hypertrophy does not develop in mice aged over 24 months old, sue Doppler indexes correlate with the degree of collagen expression and cross-
linking in heart failure and normal ejection fraction, J. Am. Coll. Cardiol. 57
and in this setting, GDF11 had no effect on cardiac structure or function. (2011) 977–985.
In this paper, in-vitro studies showed that GDF11 promote cardiomyo- [8] A. Borbely, J. van der Velden, Z. Papp, J.G. Bronzwaer, I. Edes, G.J. Stienen, et al.,
cyte hypertrophy [287]. GDF11 was recently reported to improve vascu- Cardiomyocyte stiffness in diastolic heart failure, Circulation 111 (2005)
774–781.
lature and enhance neurogenesis in brain [288]. The role of GDF11 in the [9] J.S. Burchfield, M. Xie, J.A. Hill, Pathological ventricular remodeling: mechanisms:
maintenance of cardiac homeostasis is yet to be defined, and further part 1 of 2, Circulation 128 (2013) 388–400.
studies are needed to elucidate other potential molecular mechanisms [10] K.L. Weeks, J.R. McMullen, The athlete's heart vs. the failing heart: can signaling ex-
plain the two distinct outcomes? Physiology 26 (2011) 97–105.
providing a link between aging and cardiac hypertrophy.
[11] R.C. Lyon, F. Zanella, J.H. Omens, F. Sheikh, Mechanotransduction in cardiac hyper-
trophy and failure, Circ. Res. 116 (2015) 1462–1476.
8. Conclusions [12] G.S. Francis, K.M. McDonald, J.N. Cohn, Neurohumoral activation in preclinical
heart failure. Remodeling and the potential for intervention, Circulation 87
(1993) IV90–IV96.
Heart failure develops as the result of multiple factors affecting car- [13] M. Maillet, J.H. van Berlo, J.D. Molkentin, Molecular basis of physiological heart
diac function in a complex manner, and this challenging condition does growth: fundamental concepts and new players, Nat. Rev. Mol. Cell Biol. 14
not respond to simple one-dimensional treatment. Cardiac hypertrophy (2013) 38–48.
[14] G.G. Schiattarella, J.A. Hill, Is inhibition of hypertrophy a good therapeutic strategy
is one of the most critical components of cardiac remodeling. Over the in ventricular pressure overload? Circulation 131 (2015) 1435–1447.
last decade, our understanding of cardiac hypertrophy has made signif- [15] J. Heineke, J.D. Molkentin, Regulation of cardiac hypertrophy by intracellular sig-
icant progress and several important signaling molecules and modifiers nalling pathways, Nat. Rev. Mol. Cell Biol. 7 (2006) 589–600.
[16] B.C. Bernardo, K.L. Weeks, L. Pretorius, J.R. McMullen, Molecular distinction be-
involved in hypertrophy have been identified. Continuing investigation tween physiological and pathological cardiac hypertrophy: experimental findings
of this field should further improve our understanding of the processes and therapeutic strategies, Pharmacol. Ther. 128 (2010) 191–227.
involved in hypertrophy and may lead to the establishment of next- [17] W. Grossman, D. Jones, L.P. McLaurin, Wall stress and patterns of hypertrophy in
the human left ventricle, J. Clin. Invest. 56 (1975) 56–64.
generation therapies for heart failure. [18] M. Maillet, J.H. van Berlo, J.D. Molkentin, Molecular basis of physiological heart
growth: fundamental concepts and new players, Nat. Rev. Mol. Cell Biol. 14
Disclosure (2013) 38–48.
[19] I. Shiojima, K. Sato, Y. Izumiya, S. Schiekofer, M. Ito, R. Liao, et al., Disruption of co-
ordinated cardiac hypertrophy and angiogenesis contributes to the transition to
I.S. and T.M. disclose no conflict of interests. heart failure, J. Clin. Invest. 115 (2005) 2108–2118.
[20] D.D. Belke, S. Betuing, M.J. Tuttle, C. Graveleau, M.E. Young, M. Pham, et al., Insulin
signaling coordinately regulates cardiac size, metabolism, and contractile protein
Acknowledgements
isoform expression, J. Clin. Invest. 109 (2002) 629–639.
[21] J. Kim, A.R. Wende, S. Sena, H.A. Theobald, J. Soto, C. Sloan, et al., Insulin-like
This work was supported by a Grant-in-Aid for Scientific Research, a growth factor I receptor signaling is required for exercise-induced cardiac hyper-
Grant-in-Aid for Scientific Research on Innovative Areas (Stem Cell trophy, Mol. Endocrinol. 22 (2008) 2531–2543.
[22] J.R. McMullen, T. Shioi, W.Y. Huang, L. Zhang, O. Tarnavski, E. Bisping, et al., The in-
Aging and Disease), and a Grant-in-Aid for Exploratory Research from sulin-like growth factor 1 receptor induces physiological heart growth via the
the Ministry of Education, Culture, Sports, Science and Technology phosphoinositide 3-kinase(p110alpha) pathway, J. Biol. Chem. 279 (2004)
(MEXT) of Japan and grants from the Ono Medical Research Foundation, 4782–4793.
[23] P. Stubbe, J. Gatz, P. Heidemann, A. Muhlen, R. Hesch, Thyroxine-binding globulin,
the Japan Diabetes Foundation, the Takeda Science Foundation, the triiodothyronine, thyroxine and thyrotropin in newborn infants and children, Hor-
Takeda Medical Research Foundation, and the Terumo Foundation (to mone and metabolic research = Hormon- und Stoffwechselforschung = Hor-
T.M.) as well as by a Grants-in-Aid for Young Scientists (Start-up) mones et metabolisme 10 (1978) 58–61.
[24] K. Kinugawa, M.Y. Jeong, M.R. Bristow, C.S. Long, Thyroid hormone induces cardiac
(JSPS KAKENHI Grant Number 26893080), and grants from the Uehara myocyte hypertrophy in a thyroid hormone receptor alpha1-specific manner that
Memorial Foundation, Takeda Science Foundation, Kowa Life Science requires TAK1 and p38 mitogen-activated protein kinase, Mol. Endocrinol. 19
Foundation, Manpei Suzuki Diabetes Foundation, Kanae Foundation, (2005) 1618–1628.
258 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

[25] B.R. Anderson, H.L. Granzier, Titin-based tension in the cardiac sarcomere: molec- [53] S. Moellendorf, C. Kessels, L. Peiseler, A. Raupach, C. Jacoby, N. Vogt, et al., IGF-IR
ular origin and physiological adaptations, Prog. Biophys. Mol. Biol. 110 (2012) signaling attenuates the age-related decline of diastolic cardiac function, Am. J.
204–217. Physiol. Endocrinol. Metab. 303 (2012) E213–E222.
[26] R. Knoll, M. Hoshijima, H.M. Hoffman, V. Person, I. Lorenzen-Schmidt, M.L. Bang, [54] A.R. Saltiel, C.R. Kahn, Insulin signalling and the regulation of glucose and lipid me-
et al., The cardiac mechanical stretch sensor machinery involves a Z disc complex tabolism, Nature 414 (2001) 799–806.
that is defective in a subset of human dilated cardiomyopathy, Cell 111 (2002) [55] M. Tatar, A. Bartke, A. Antebi, The endocrine regulation of aging by insulin-like sig-
943–955. nals, Science 299 (2003) 1346–1351.
[27] R.C. Lyon, F. Zanella, J.H. Omens, F. Sheikh, Mechanotransduction in cardiac hyper- [56] Y.K. Kim, S.J. Kim, A. Yatani, Y.H. Huang, G. Castelli, D.E. Vatner, et al., Mechanism of
trophy and failure, Circ. Res. 116 (2015) 1462–1476. enhanced cardiac function in mice with hypertrophy induced by overexpressed
[28] A. Kenessey, K. Ojamaa, Thyroid hormone stimulates protein synthesis in the car- Akt, J. Biol. Chem. 278 (2003) 47622–47628.
diomyocyte by activating the Akt-mTOR and p70S6K pathways, J. Biol. Chem. [57] O.J. Kemi, M. Ceci, U. Wisloff, S. Grimaldi, P. Gallo, G.L. Smith, et al., Activation or in-
281 (2006) 20666–20672. activation of cardiac Akt/mTOR signaling diverges physiological from pathological
[29] C. Pantos, I. Mourouzis, K. Markakis, N. Tsagoulis, M. Panagiotou, D.V. Cokkinos, hypertrophy, J. Cell. Physiol. 214 (2008) 316–321.
Long-term thyroid hormone administration reshapes left ventricular chamber [58] D. Catalucci, D.H. Zhang, J. DeSantiago, F. Aimond, G. Barbara, J. Chemin, et al., Akt
and improves cardiac function after myocardial infarction in rats, Basic Res. regulates L-type Ca2+ channel activity by modulating Ca-v alpha 1 protein stabil-
Cardiol. 103 (2008) 308–318. ity, J. Cell Biol. 184 (2009) 923–933.
[30] B. Ziegelhoffer-Mihalovicova, W. Briest, H.A. Baba, B. Rassler, H.G. Zimmer, The ex- [59] D. Catalucci, M.V.G. Latronico, M. Ceci, F. Rusconi, H.S. Young, P. Gallo, et al., At in-
pression of mRNA of cytokines and of extracellular matrix proteins in triiodothyro- creases sarcoplasmic reticulum Ca2+ cycling by direct phosphorylation of phos-
nine-treated rat hearts, Mol. Cell. Biochem. 247 (2003) 61–68. pholamban at Thr(17), J. Biol. Chem. 284 (2009) 28180–28187.
[31] J.L. Yao, M. Eghbali, Decreased collagen gene-expression and absence of fibrosis in [60] S. Boudina, H. Bugger, S. Sena, B.T. O'Neill, V.G. Zaha, O. Ilkun, et al., Contribution of
thyroid hormone-induced myocardial hypertrophy — response of cardiac fibro- impaired myocardial insulin signaling to mitochondrial dysfunction and oxidative
blasts to thyroid-hormone invitro, Circ. Res. 71 (1992) 831–839. stress in the heart, Circulation 119 (2009) 1272–1283.
[32] K.C. Chang, V.M. Figueredo, J.H. Schreur, K. Kariya, M.W. Weiner, P.C. Simpson, [61] S. Sena, P. Hu, D. Zhang, X. Wang, B. Wayment, C. Olsen, et al., Impaired insulin sig-
et al., Thyroid hormone improves function and Ca2+ handling in pressure over- naling accelerates cardiac mitochondrial dysfunction after myocardial infarction, J.
load hypertrophy. Association with increased sarcoplasmic reticulum Ca2+- Mol. Cell. Cardiol. 46 (2009) 910–918.
ATPase and alpha-myosin heavy chain in rat hearts, J. Clin. Invest. 100 (1997) [62] E. Araki, M.A. Lipes, M.E. Patti, J.C. Bruning, B. Haag 3rd, R.S. Johnson, et al., Alterna-
1742–1749. tive pathway of insulin signalling in mice with targeted disruption of the IRS-1
[33] C. Pantos, I. Mourouzis, N. Tsagoulis, K. Markakis, G. Galanopoulos, N. Roukounakis, gene, Nature 372 (1994) 186–190.
et al., Thyroid hormone at supra-physiological dose optimizes cardiac geometry [63] D.J. Burks, J. Font de Mora, M. Schubert, D.J. Withers, M.G. Myers, H.H. Towery,
and improves cardiac function in rats with old myocardial infarction, J. Physiol. et al., IRS-2 pathways integrate female reproduction and energy homeostasis, Na-
Pharmacol. 60 (2009) 49–56. ture 407 (2000) 377–382.
[34] C. Pantos, I. Mourouzis, D.V. Cokkinos, New insights into the role of thyroid hor- [64] D. Zhang, R. Contu, M.V. Latronico, J. Zhang, R. Rizzi, D. Catalucci, et al., MTORC1
mone in cardiac remodeling: time to reconsider? Heart Fail. Rev. 16 (2011) 79–96. regulates cardiac function and myocyte survival through 4E-BP1 inhibition in
[35] Z. Ungvari, A. Csiszar, The emerging role of IGF-1 deficiency in cardiovascular mice, J. Clin. Invest. 120 (2010) 2805–2816.
aging: recent advances, J. Gerontol. A Biol. Sci. Med. Sci. 67 (2012) 599–610. [65] P. Bostrom, N. Mann, J. Wu, P.A. Quintero, E.R. Plovie, D. Panakova, et al., C/EBPbeta
[36] P. Spallarossa, C. Brunelli, F. Minuto, D. Caruso, M. Battistini, S. Caponnetto, et al., controls exercise-induced cardiac growth and protects against pathological cardiac
Insulin-like growth factor-I and angiographically documented coronary artery dis- remodeling, Cell 143 (2010) 1072–1083.
ease, Am. J. Cardiol. 77 (1996) 200–202. [66] G. Condorelli, A. Drusco, G. Stassi, A. Bellacosa, R. Roncarati, G. Iaccarino, et al., Akt
[37] A. Juul, T. Scheike, M. Davidsen, J. Gyllenborg, T. Jorgensen, Low serum insulin-like induces enhanced myocardial contractility and cell size in vivo in transgenic mice,
growth factor I is associated with increased risk of ischemic heart disease: a popu- Proc. Natl. Acad. Sci. U. S. A. 99 (2002) 12333–12338.
lation-based case-control study, Circulation 106 (2002) 939–944. [67] T. Matsui, L. Li, J.C. Wu, S.A. Cook, T. Nagoshi, M.H. Picard, et al., Phenotypic spec-
[38] E. Conti, F. Andreotti, A. Sciahbasi, P. Riccardi, G. Marra, E. Menini, et al., Markedly trum caused by transgenic overexpression of activated Akt in the heart, J. Biol.
reduced insulin-like growth factor-1 in the acute phase of myocardial infarction, J. Chem. 277 (2002) 22896–22901.
Am. Coll. Cardiol. 38 (2001) 26–32. [68] T. Shioi, J.R. McMullen, P.M. Kang, P.S. Douglas, T. Obata, T.F. Franke, et al., Akt/pro-
[39] R.S. Vasan, L.M. Sullivan, R.B. D'Agostino, R. Roubenoff, T. Harris, D.B. Sawyer, et al., tein kinase B promotes organ growth in transgenic mice, Mol. Cell. Biol. 22 (2002)
Serum insulin-like growth factor I and risk for heart failure in elderly individuals 2799–2809.
without a previous myocardial infarction: the Framingham heart study, Ann. In- [69] K. Yamashita, J. Kajstura, D.J. Discher, B.J. Wasserlauf, N.H. Bishopric, P. Anversa,
tern. Med. 139 (2003) 642–648. et al., Reperfusion-activated Akt kinase prevents apoptosis in transgenic mouse
[40] M. Vinciguerra, M.P. Santini, W.C. Claycomb, A.G. Ladurner, N. Rosenthal, Local IGF- hearts overexpressing insulin-like growth factor-1, Circ. Res. 88 (2001) 609–614.
1 isoform protects cardiomyocytes from hypertrophic and oxidative stresses via [70] J.R. McMullen, T. Shioi, L. Zhang, O. Tarnavski, M.C. Sherwood, P.M. Kang, et al.,
SirT1 activity, Aging (Albany NY) 2 (2010) 43–62. Phosphoinositide 3-kinase(p110alpha) plays a critical role for the induction of
[41] M. Vinciguerra, M.P. Santini, C. Martinez, V. Pazienza, W.C. Claycomb, A. Giuliani, physiological, but not pathological, cardiac hypertrophy, Proc. Natl. Acad. Sci. U.
et al., mIGF-1/JNK1/SirT1 signaling confers protection against oxidative stress in S. A. 100 (2003) 12355–12360.
the heart, Aging Cell 11 (2012) 139–149. [71] T. Shioi, P.M. Kang, P.S. Douglas, J. Hampe, C.M. Yballe, J. Lawitts, et al., The con-
[42] N. Takeda, I. Manabe, Y. Uchino, K. Eguchi, S. Matsumoto, S. Nishimura, et al., Car- served phosphoinositide 3-kinase pathway determines heart size in mice, EMBO
diac fibroblasts are essential for the adaptive response of the murine heart to pres- J. 19 (2000) 2537–2548.
sure overload, J. Clin. Invest. 120 (2010) 254–265. [72] A.R. Wende, B.T. O'Neill, H. Bugger, C. Riehle, J. Tuinei, J. Buchanan, et al., Enhanced
[43] R. Troncoso, C. Ibarra, J.M. Vicencio, E. Jaimovich, S. Lavandero, New insights into cardiac Akt/protein kinase B signaling contributes to pathological cardiac hypertro-
IGF-1 signaling in the heart, Trends Endocrinol. Metab. 25 (2014) 128–137. phy in part by impairing mitochondrial function via transcriptional repression of
[44] D.J. Glass, Skeletal muscle hypertrophy and atrophy signaling pathways, Int. J. mitochondrion-targeted nuclear genes, Mol. Cell. Biol. 35 (2015) 831–846.
Biochem. Cell Biol. 37 (2005) 1974–1984. [73] C. Zwadlo, E. Schmidtmann, M. Szaroszyk, B. Kattih, N. Froese, H. Hinz, et al.,
[45] Y.H. Song, M. Godard, Y. Li, S.R. Richmond, N. Rosenthal, P. Delafontaine, Insulin- Antiandrogenic therapy with finasteride attenuates cardiac hypertrophy and left
like growth factor I-mediated skeletal muscle hypertrophy is characterized by in- ventricular dysfunction, Circulation 131 (2015) 1071–1081.
creased mTOR-p70S6K signaling without increased Akt phosphorylation, J. [74] A.S. Hebert, K.E. Dittenhafer-Reed, W. Yu, D.J. Bailey, E.S. Selen, M.D. Boersma, et al.,
Investig. Med. 53 (2005) 135–142. Calorie restriction and SIRT3 trigger global reprogramming of the mitochondrial
[46] E.T. Poehlman, C.J. Rosen, K.C. Copeland, The influence of endurance training on in- protein acetylome, Mol. Cell 49 (2013) 186–199.
sulin-like growth-factor-I in older individuals, Metabolism 43 (1994) 1401–1405. [75] Y. Kanfi, R. Shalman, V. Peshti, S.N. Pilosof, Y.M. Gozlan, K.J. Pearson, et al.,
[47] G.G.N. Serneri, M. Boddi, P.A. Modesti, I. Cecioni, M. Coppo, L. Padeletti, et al., In- Regulation of SIRT6 protein levels by nutrient availability, FEBS Lett. 582
creased cardiac sympathetic activity and insulin-like growth factor-I formation (2008) 543–548.
are associated with physiological hypertrophy in athletes, Circ. Res. 89 (2001) [76] X. Qiu, K. Brown, M.D. Hirschey, E. Verdin, D. Chen, Calorie restriction reduces ox-
977–982. idative stress by SIRT3-mediated SOD2 activation, Cell Metab. 12 (2010) 662–667.
[48] J.P. Liu, J. Baker, A.S. Perkins, E.J. Robertson, A. Efstratiadis, Mice carrying null mu- [77] C. Canto, J. Auwerx, Caloric restriction, SIRT1 and longevity, Trends Endocrinol.
tations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF re- Metab. 20 (2009) 325–331.
ceptor (Igf1r), Cell 75 (1993) 59–72. [78] D. Bellizzi, G. Rose, P. Cavalcante, G. Covello, S. Dato, F. De Rango, et al., A novel
[49] L. Powell-Braxton, P. Hollingshead, C. Warburton, M. Dowd, S. Pitts-Meek, D. VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated
Dalton, et al., IGF-I is required for normal embryonic growth in mice, Genes Dev. with survival at oldest ages, Genomics 85 (2005) 258–263.
7 (1993) 2609–2617. [79] Y. Kanfi, S. Naiman, G. Amir, V. Peshti, G. Zinman, L. Nahum, et al., The sirtuin SIRT6
[50] M.C. Delaughter, G.E. Taffet, M.L. Fiorotto, M.L. Entman, R.J. Schwartz, Local insulin- regulates lifespan in male mice, Nature 483 (2012) 218–221.
like growth factor I expression induces physiologic, then pathologic, cardiac hyper- [80] L. Fontana, L. Partridge, V.D. Longo, Extending healthy life span—from yeast to
trophy in transgenic mice, FASEB J. 13 (1999) 1923–1929. humans, Science 328 (2010) 321–326.
[51] K. Reiss, W. Cheng, A. Ferber, J. Kajstura, P. Li, B. Li, et al., Overexpression of insulin- [81] J.A. Baur, D. Chen, E.N. Chini, K. Chua, H.Y. Cohen, R. de Cabo, et al., Dietary
like growth factor-1 in the heart is coupled with myocyte proliferation in transgen- restriction: standing up for sirtuins, Science 329 (2010) 1012–1013 author
ic mice, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 8630–8635. reply 3–4.
[52] H. Ikeda, I. Shiojima, Y. Ozasa, M. Yoshida, M. Holzenberger, C.R. Kahn, et al., Inter- [82] N.R. Sundaresan, M. Gupta, G. Kim, S.B. Rajamohan, A. Isbatan, M.P. Gupta, Sirt3
action of myocardial insulin receptor and IGF receptor signaling in exercise-in- blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent anti-
duced cardiac hypertrophy, J. Mol. Cell. Cardiol. 47 (2009) 664–675. oxidant defense mechanisms in mice, J. Clin. Invest. 119 (2009) 2758–2771.
I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262 259

[83] V.B. Pillai, N.R. Sundaresan, G. Kim, M. Gupta, S.B. Rajamohan, J.B. Pillai, et al., Exog- [113] U. Kirchhefer, W. Schmitz, H. Scholz, J. Neumann, Activity of cAMP-dependent pro-
enous NAD blocks cardiac hypertrophic response via activation of the SIRT3-LKB1- tein kinase and Ca2+/calmodulin-dependent protein kinase in failing and
AMP-activated kinase pathway, J. Biol. Chem. 285 (2010) 3133–3144. nonfailing human hearts, Cardiovasc. Res. 42 (1999) 254–261.
[84] N.R. Sundaresan, P. Vasudevan, L. Zhong, G. Kim, S. Samant, V. Parekh, et al., The [114] X. Ai, J.W. Curran, T.R. Shannon, D.M. Bers, S.M. Pogwizd, Ca2+/calmodulin-depen-
sirtuin SIRT6 blocks IGF-Akt signaling and development of cardiac hypertrophy dent protein kinase modulates cardiac ryanodine receptor phosphorylation and
by targeting c-Jun, Nat. Med. 18 (2012) 1643–1650. sarcoplasmic reticulum Ca2+ leak in heart failure, Circ. Res. 97 (2005) 1314–1322.
[85] R.R. Alcendor, S. Gao, P. Zhai, D. Zablocki, E. Holle, X. Yu, et al., Sirt1 regulates aging [115] J. Bossuyt, K. Helmstadter, X. Wu, H. Clements-Jewery, R.S. Haworth, M. Avkiran,
and resistance to oxidative stress in the heart, Circ. Res. 100 (2007) 1512–1521. et al., Ca2+/calmodulin-dependent protein kinase IIdelta and protein kinase D
[86] S. Oka, R. Alcendor, P. Zhai, J.Y. Park, D. Shao, J. Cho, et al., PPARalpha-Sirt1 complex overexpression reinforce the histone deacetylase 5 redistribution in heart failure,
mediates cardiac hypertrophy and failure through suppression of the ERR tran- Circ. Res. 102 (2008) 695–702.
scriptional pathway, Cell Metab. 14 (2011) 598–611. [116] H. Ling, T. Zhang, L. Pereira, C.K. Means, H. Cheng, Y. Gu, et al., Requirement for
[87] M. Potente, L. Ghaeni, D. Baldessari, R. Mostoslavsky, L. Rossig, F. Dequiedt, et al., Ca2+/calmodulin-dependent kinase II in the transition from pressure overload-
SIRT1 controls endothelial angiogenic functions during vascular growth, Genes induced cardiac hypertrophy to heart failure in mice, J. Clin. Invest. 119 (2009)
Dev. 21 (2007) 2644–2658. 1230–1240.
[88] J.A. Hill, E.N. Olson, Cardiac plasticity, N. Engl. J. Med. 358 (2008) 1370–1380. [117] J. Backs, T. Backs, S. Neef, M.M. Kreusser, L.H. Lehmann, D.M. Patrick, et al., The
[89] H. Sandler, H.T. Dodge, Left ventricular tension and stress in man, Circ. Res. 13 delta isoform of CaM kinase II is required for pathological cardiac hypertrophy
(1963) 91–104. and remodeling after pressure overload, Proc. Natl. Acad. Sci. U. S. A. 106 (2009)
[90] D. Levy, R.J. Garrison, D.D. Savage, W.B. Kannel, W.P. Castelli, Prognostic implica- 2342–2347.
tions of echocardiographically determined left ventricular mass in the Framing- [118] W. Zhu, Y. Zou, I. Shiojima, S. Kudoh, R. Aikawa, D. Hayashi, et al., Ca2+/calmodu-
ham heart study, N. Engl. J. Med. 322 (1990) 1561–1566. lin-dependent kinase II and calcineurin play critical roles in endothelin-1-induced
[91] M.J. Koren, R.B. Devereux, P.N. Casale, D.D. Savage, J.H. Laragh, Relation of left ven- cardiomyocyte hypertrophy, J. Biol. Chem. 275 (2000) 15239–15245.
tricular mass and geometry to morbidity and mortality in uncomplicated essential [119] M.T. Ramirez, X.L. Zhao, H. Schulman, J.H. Brown, The nuclear deltaB isoform of
hypertension, Ann. Intern. Med. 114 (1991) 345–352. Ca2+/calmodulin-dependent protein kinase II regulates atrial natriuretic factor
[92] Y. Zou, Y. Hiroi, H. Uozumi, E. Takimoto, H. Toko, W. Zhu, et al., Calcineurin plays a gene expression in ventricular myocytes, J. Biol. Chem. 272 (1997) 31203–31208.
critical role in the development of pressure overload-induced cardiac hypertrophy, [120] C.A. Sei, C.E. Irons, A.B. Sprenkle, P.M. McDonough, J.H. Brown, C.C. Glembotski, The
Circulation 104 (2001) 97–101. alpha-adrenergic stimulation of atrial natriuretic factor expression in cardiac
[93] S. Ichihara, T. Senbonmatsu, E. Price Jr., T. Ichiki, F.A. Gaffney, T. Inagami, Angioten- myocytes requires calcium influx, protein kinase C, and calmodulin-regulated
sin II type 2 receptor is essential for left ventricular hypertrophy and cardiac fibro- pathways, J. Biol. Chem. 266 (1991) 15910–15916.
sis in chronic angiotensin II-induced hypertension, Circulation 104 (2001) [121] J. Backs, K. Song, S. Bezprozvannaya, S. Chang, E.N. Olson, CaM kinase II selectively
346–351. signals to histone deacetylase 4 during cardiomyocyte hypertrophy, J. Clin. Invest.
[94] T. Senbonmatsu, S. Ichihara, E. Price Jr., F.A. Gaffney, T. Inagami, Evidence for angio- 116 (2006) 1853–1864.
tensin II type 2 receptor-mediated cardiac myocyte enlargement during in vivo [122] C.L. Zhang, T.A. McKinsey, S. Chang, C.L. Antos, J.A. Hill, E.N. Olson, Class II histone
pressure overload, J. Clin. Invest. 106 (2000) R25–R29. deacetylases act as signal-responsive repressors of cardiac hypertrophy, Cell 110
[95] J.A. Hill, M. Karimi, W. Kutschke, R.L. Davisson, K. Zimmerman, Z. Wang, et al., Car- (2002) 479–488.
diac hypertrophy is not a required compensatory response to short-term pressure [123] R. Passier, H. Zeng, N. Frey, F.J. Naya, R.L. Nicol, T.A. McKinsey, et al., CaM kinase sig-
overload, Circulation 101 (2000) 2863–2869. naling induces cardiac hypertrophy and activates the MEF2 transcription factor in
[96] J.H. van Berlo, M. Maillet, J.D. Molkentin, Signaling effectors underlying pathologic vivo, J. Clin. Invest. 105 (2000) 1395–1406.
growth and remodeling of the heart, J. Clin. Invest. 123 (2013) 37–45. [124] E. Takimoto, H.C. Champion, M. Li, D. Belardi, S. Ren, E.R. Rodriguez, et al., Chronic
[97] C. Perrino, S.V. Naga Prasad, L. Mao, T. Noma, Z. Yan, H.S. Kim, et al., Intermittent inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hy-
pressure overload triggers hypertrophy-independent cardiac dysfunction and vas- pertrophy, Nat. Med. 11 (2005) 214–222.
cular rarefaction, J. Clin. Invest. 116 (2006) 1547–1560. [125] R.C. Kukreja, F.N. Salloum, A. Das, Cyclic guanosine monophosphate signaling and
[98] J.D. Molkentin, J.R. Lu, C.L. Antos, B. Markham, J. Richardson, J. Robbins, et al., A cal- phosphodiesterase-5 inhibitors in cardioprotection, J. Am. Coll. Cardiol. 59 (2012)
cineurin-dependent transcriptional pathway for cardiac hypertrophy, Cell 93 1921–1927.
(1998) 215–228. [126] L. van Heerebeek, N. Hamdani, I. Falcao-Pires, A.F. Leite-Moreira, M.P. Begieneman,
[99] B.J. Wilkins, J.D. Molkentin, Calcium-calcineurin signaling in the regulation of car- J.G. Bronzwaer, et al., Low myocardial protein kinase G activity in heart failure with
diac hypertrophy, Biochem. Biophys. Res. Commun. 322 (2004) 1178–1191. preserved ejection fraction, Circulation 126 (2012) 830–839.
[100] J.D. Molkentin, J.R. Lu, C.L. Antos, B. Markham, J. Richardson, J. Robbins, et al., A cal- [127] E. Takimoto, N. Koitabashi, S. Hsu, E.A. Ketner, M. Zhang, T. Nagayama, et al., Reg-
cineurin-dependent transcriptional pathway for cardiac hypertrophy, Cell 93 ulator of G protein signaling 2 mediates cardiac compensation to pressure overload
(1998) 215–228. and antihypertrophic effects of PDE5 inhibition in mice, J. Clin. Invest. 119 (2009)
[101] S.R. Houser, J.D. Molkentin, Does contractile Ca2 + control calcineurin-NFAT 408–420.
signaling and pathological hypertrophy in cardiac myocytes? Sci. Signal. 1 [128] N. Koitabashi, T. Aiba, G.G. Hesketh, J. Rowell, M. Zhang, E. Takimoto, et al., Cyclic
(2008), pe31. GMP/PKG-dependent inhibition of TRPC6 channel activity and expression nega-
[102] B.J. Wilkins, Y.S. Dai, Bueno OF, S.A. Parsons, J. Xu, D.M. Plank, et al., Calcineurin/ tively regulates cardiomyocyte NFAT activation novel mechanism of cardiac stress
NFAT coupling participates in pathological, but not physiological, cardiac hypertro- modulation by PDE5 inhibition, J. Mol. Cell. Cardiol. 48 (2009) 713–724.
phy, Circ. Res. 94 (2004) 110–118. [129] D.I. Lee, G. Zhu, T. Sasaki, G.S. Cho, N. Hamdani, R. Holewinski, et al., Phosphodies-
[103] M. Seth, Z.S. Zhang, L. Mao, V. Graham, J. Burch, J. Stiber, et al., TRPC1 channels are terase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease,
critical for hypertrophic signaling in the heart, Circ. Res. 105 (2009) 1023–1030. Nature 519 (2015) 472–476.
[104] K. Kuwahara, Y. Wang, J. McAnally, J.A. Richardson, R. Bassel-Duby, J.A. Hill, et al., [130] T. Arimoto, Y. Takeishi, H. Takahashi, T. Shishido, T. Niizeki, Y. Koyama, et al., Car-
TRPC6 fulfills a calcineurin signaling circuit during pathologic cardiac remodeling, diac-specific overexpression of diacylglycerol kinase zeta prevents Gq protein-
J. Clin. Invest. 116 (2006) 3114–3126. coupled receptor agonist-induced cardiac hypertrophy in transgenic mice, Circula-
[105] T. Niizeki, Y. Takeishi, T. Kitahara, T. Arimoto, M. Ishino, O. Bilim, et al., Diacylglyc- tion 113 (2006) 60–66.
erol kinase-epsilon restores cardiac dysfunction under chronic pressure overload: [131] Y. Takeishi, P. Ping, R. Bolli, D.L. Kirkpatrick, B.D. Hoit, R.A. Walsh, Transgenic over-
a new specific regulator of Galpha(q) signaling cascade, Am. J. Physiol. Heart expression of constitutively active protein kinase C epsilon causes concentric car-
Circ. Physiol. 295 (2008) H245–H255. diac hypertrophy, Circ. Res. 86 (2000) 1218–1223.
[106] T. Ohba, H. Watanabe, M. Murakami, Y. Takahashi, K. Iino, S. Kuromitsu, et al., Up- [132] J.S. Gutkind, S. Offermanns, A new G(q)-initiated MAPK signaling pathway in the
regulation of TRPC1 in the development of cardiac hypertrophy, J. Mol. Cell. heart, Dev. Cell 16 (2009) 163–164.
Cardiol. 42 (2007) 498–507. [133] S. Haq, G. Choukroun, H. Lim, K.M. Tymitz, F. del Monte, J. Gwathmey, et al., Differ-
[107] X. Wu, P. Eder, B. Chang, J.D. Molkentin, TRPC channels are necessary mediators of ential activation of signal transduction pathways in human hearts with hypertro-
pathologic cardiac hypertrophy, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) phy versus advanced heart failure, Circulation 103 (2001) 670–677.
7000–7005. [134] K. Toischer, A.G. Rokita, B. Unsold, W. Zhu, G. Kararigas, S. Sossalla, et al., Differen-
[108] J.N. Cohn, Plasma norepinephrine and mortality, Clin. Cardiol. 18 (1995) I9–12. tial cardiac remodeling in preload versus afterload, Circulation 122 (2010)
[109] A.H. Strand, H. Gudmundsdottir, I. Os, G. Smith, A.S. Westheim, R. Bjornerheim, 993–1003.
et al., Arterial plasma noradrenaline predicts left ventricular mass independently [135] Bueno OF, L.J. De Windt, K.M. Tymitz, S.A. Witt, T.R. Kimball, R. Klevitsky, et al., The
of blood pressure and body build in men who develop hypertension over MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in
20 years, J. Hypertens. 24 (2006) 905–913. transgenic mice, EMBO J. 19 (2000) 6341–6350.
[110] M. Packer, A.J. Coats, M.B. Fowler, H.A. Katus, H. Krum, P. Mohacsi, et al., Effect of [136] N.H. Purcell, B.J. Wilkins, A. York, M.K. Saba-El-Leil, S. Meloche, J. Robbins, et al., Ge-
carvedilol on survival in severe chronic heart failure, N. Engl. J. Med. 344 (2001) netic inhibition of cardiac ERK1/2 promotes stress-induced apoptosis and heart
1651–1658. failure but has no effect on hypertrophy in vivo, Proc. Natl. Acad. Sci. U. S. A. 104
[111] P.A. Poole-Wilson, K. Swedberg, J.G. Cleland, A. Di Lenarda, P. Hanrath, M. Komajda, (2007) 14074–14079.
et al., Comparison of carvedilol and metoprolol on clinical outcomes in patients [137] I. Kehat, J. Davis, M. Tiburcy, F. Accornero, M.K. Saba-El-Leil, M. Maillet, et al., Extra-
with chronic heart failure in the Carvedilol Or Metoprolol European Trial cellular signal-regulated kinases 1 and 2 regulate the balance between eccentric
(COMET): randomised controlled trial, Lancet 362 (2003) 7–13. and concentric cardiac growth, Circ. Res. 108 (2011) 176–183.
[112] B. Hoch, R. Meyer, R. Hetzer, E.G. Krause, P. Karczewski, Identification and ex- [138] K. Nishida, O. Yamaguchi, S. Hirotani, S. Hikoso, Y. Higuchi, T. Watanabe, et al.,
pression of delta-isoforms of the multifunctional Ca2 +/calmodulin-depen- p38alpha mitogen-activated protein kinase plays a critical role in cardiomyocyte
dent protein kinase in failing and nonfailing human myocardium, Circ. Res. survival but not in cardiac hypertrophic growth in response to pressure overload,
84 (1999) 713–721. Mol. Cell. Biol. 24 (2004) 10611–10620.
260 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

[139] P. Liao, D. Georgakopoulos, A. Kovacs, M. Zheng, D. Lerner, H. Pu, et al., The in vivo [167] S. Haider, L. Cordeddu, E. Robinson, M. Movassagh, L. Siggens, A. Vujic, et al., The
role of p38 MAP kinases in cardiac remodeling and restrictive cardiomyopathy, landscape of DNA repeat elements in human heart failure, Genome Biol. 13
Proc. Natl. Acad. Sci. U. S. A. 98 (2001) 12283–12288. (2012) R90.
[140] Q. Liang, Bueno OF, B.J. Wilkins, C.Y. Kuan, Y. Xia, J.D. Molkentin, c-Jun N-terminal [168] A. Kozomara, S. Griffiths-Jones, miRBase: integrating microRNA annotation and
kinases (JNK) antagonize cardiac growth through cross-talk with calcineurin-NFAT deep-sequencing data, Nucleic Acids Res. 39 (2011) D152–D157.
signaling, EMBO J. 22 (2003) 5079–5089. [169] E. van Rooij, L.B. Sutherland, N. Liu, A.H. Williams, J. McAnally, R.D. Gerard,
[141] W. Liu, M. Zi, J. Jin, S. Prehar, D. Oceandy, T.E. Kimura, et al., Cardiac-specific dele- et al., A signature pattern of stress-responsive microRNAs that can evoke
tion of mkk4 reveals its role in pathological hypertrophic remodeling but not in cardiac hypertrophy and heart failure, Proc. Natl. Acad. Sci. U. S. A. 103
physiological cardiac growth, Circ. Res. 104 (2009) 905–914. (2006) 18255–18260.
[142] Bueno OF, L.J. De Windt, H.W. Lim, K.M. Tymitz, S.A. Witt, T.R. Kimball, et al., The [170] A. Ucar, S.K. Gupta, J. Fiedler, E. Erikci, M. Kardasinski, S. Batkai, et al., The miRNA-
dual-specificity phosphatase MKP-1 limits the cardiac hypertrophic response in 212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy,
vitro and in vivo, Circ. Res. 88 (2001) 88–96. Nat. Commun. 3 (2012) 1078.
[143] M. Auger-Messier, F. Accornero, S.A. Goonasekera, Bueno OF, J.N. Lorenz, J.H. van [171] T.E. Callis, K. Pandya, H.Y. Seok, R.H. Tang, M. Tatsuguchi, Z.P. Huang, et al.,
Berlo, et al., Unrestrained p38 MAPK activation in Dusp1/4 double-null mice in- MicroRNA-208a is a regulator of cardiac hypertrophy and conduction in mice, J.
duces cardiomyopathy, Circ. Res. 112 (2013) 48–56. Clin. Invest. 119 (2009) 2772–2786.
[144] A.M. Samuelsson, E. Bollano, R. Mobini, B.M. Larsson, E. Omerovic, M. Fu, et al., [172] S.J. Matkovich, D.J. Van Booven, K.A. Youker, G. Torre-Amione, A. Diwan, W.H.
Hyperinsulinemia: effect on cardiac mass/function, angiotensin II receptor expres- Eschenbacher, et al., Reciprocal regulation of myocardial microRNAs and messen-
sion, and insulin signaling pathways, Am. J. Physiol. Heart Circ. Physiol. 291 (2006) ger RNA in human cardiomyopathy and reversal of the microRNA signature by bio-
H787–H796. mechanical support, Circulation 119 (2009) 1263–1271.
[145] H.C. Gerstein, M.E. Miller, R.P. Byington, D.C. Goff Jr., J.T. Bigger, J.B. Buse, et al., Ef- [173] X.D. Xu, X.W. Song, Q. Li, G.K. Wang, Q. Jing, Y.W. Qin, Attenuation of microRNA-22
fects of intensive glucose lowering in type 2 diabetes, N. Engl. J. Med. 358 (2008) derepressed PTEN to effectively protect rat cardiomyocytes from hypertrophy, J.
2545–2559. Cell. Physiol. 227 (2012) 1391–1398.
[146] S. Sciarretta, P.Y. Zhai, D. Shao, Y. Maejima, J. Robbins, M. Volpe, et al., Rheb is a crit- [174] P. Gurha, C. Abreu-Goodger, T. Wang, M.O. Ramirez, A.L. Drumond, S. van Dongen,
ical regulator of autophagy during myocardial ischemia pathophysiological impli- et al., Targeted deletion of microRNA-22 promotes stress-induced cardiac dilation
cations in obesity and metabolic syndrome, Circulation 125 (2012) (1134-U166). and contractile dysfunction, Circulation 125 (2012) 2751–2761.
[147] H. Ashrafian, M.P. Frenneaux, L.H. Opie, Metabolic mechanisms in heart failure, Cir- [175] R. Kumarswamy, T. Thum, Non-coding RNAs in cardiac remodeling and heart fail-
culation 116 (2007) 434–448. ure, Circ. Res. 113 (2013) 676–689.
[148] E. Ingelsson, J. Sundstrom, J. Arnlov, B. Zethelius, L. Lind, Insulin resistance and risk [176] Y.F. Melman, R. Shah, S. Das, MicroRNAs in heart failure: is the picture becoming
of congestive heart failure, J. Am. Med. Assoc. 294 (2005) 334–341. less miRky? Circ Heart Fail. 7 (2014) 203–214.
[149] R.M. Witteles, W.H. Tang, A.H. Jamali, J.W. Chu, G.M. Reaven, M.B. Fowler, Insulin [177] T. Doenst, T.D. Nguyen, E.D. Abel, Cardiac metabolism in heart failure: implications
resistance in idiopathic dilated cardiomyopathy: a possible etiologic link, J. Am. beyond ATP production, Circ. Res. 113 (2013) 709–724.
Coll. Cardiol. 44 (2004) 78–81. [178] J.C. Osorio, W.C. Stanley, A. Linke, M. Castellari, Q.N. Diep, A.R. Panchal, et al.,
[150] I. Shimizu, Y. Yoshida, T. Katsuno, K. Tateno, S. Okada, J. Moriya, et al., p53-induced Impaired myocardial fatty acid oxidation and reduced protein expression of
adipose tissue inflammation is critically involved in the development of insulin re- retinoid X receptor-alpha in pacing-induced heart failure, Circulation 106
sistance in heart failure, Cell Metab. 15 (2012) 51–64. (2002) 606–612.
[151] A.M. Choi, S.W. Ryter, B. Levine, Autophagy in human health and disease, N. Engl. J. [179] T. Kato, S. Niizuma, Y. Inuzuka, T. Kawashima, J. Okuda, Y. Tamaki, et al., Analysis of
Med. 368 (2013) 651–662. metabolic remodeling in compensated left ventricular hypertrophy and heart fail-
[152] L. Yang, P. Li, S. Fu, E.S. Calay, G.S. Hotamisligil, Defective hepatic autophagy in obe- ure, Circ. Heart Fail. 3 (2010) 420–430.
sity promotes ER stress and causes insulin resistance, Cell Metab. 11 (2010) [180] T. Doenst, G. Pytel, A. Schrepper, P. Amorim, G. Farber, Y. Shingu, et al., Decreased
467–478. rates of substrate oxidation ex vivo predict the onset of heart failure and contrac-
[153] R. Singh, Y. Xiang, Y. Wang, K. Baikati, A.M. Cuervo, Y.K. Luu, et al., Autophagy reg- tile dysfunction in rats with pressure overload, Cardiovasc. Res. 86 (2010)
ulates adipose mass and differentiation in mice, J. Clin. Invest. 119 (2009) 461–470.
3329–3339. [181] N. Rosenblatt-Velin, C. Montessuit, I. Papageorgiou, J. Terrand, R. Lerch,
[154] Y. Zhang, S. Goldman, R. Baerga, Y. Zhao, M. Komatsu, S. Jin, Adipose-specific dele- Postinfarction heart failure in rats is associated with upregulation of GLUT-1 and
tion of autophagy-related gene 7 (atg7) in mice reveals a role in adipogenesis, downregulation of genes of fatty acid metabolism, Cardiovasc. Res. 52 (2001)
Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 19860–19865. 407–416.
[155] Y. Matsui, H. Takagi, X. Qu, M. Abdellatif, H. Sakoda, T. Asano, et al., Distinct roles of [182] L.C. Heather, M.A. Cole, C.A. Lygate, R.D. Evans, D.J. Stuckey, A.J. Murray, et al., Fatty
autophagy in the heart during ischemia and reperfusion: roles of AMP-activated acid transporter levels and palmitate oxidation rate correlate with ejection fraction
protein kinase and Beclin 1 in mediating autophagy, Circ. Res. 100 (2007) in the infarcted rat heart, Cardiovasc. Res. 72 (2006) 430–437.
914–922. [183] M.N. Sack, T.A. Rader, S. Park, J. Bastin, S.A. McCune, D.P. Kelly, Fatty acid oxidation
[156] H. Zhu, P. Tannous, J.L. Johnstone, Y. Kong, J.M. Shelton, J.A. Richardson, et al., Car- enzyme gene expression is downregulated in the failing heart, Circulation 94
diac autophagy is a maladaptive response to hemodynamic stress, J. Clin. Invest. (1996) 2837–2842.
117 (2007) 1782–1793. [184] L.H. Opie, J. Knuuti, The adrenergic-fatty acid load in heart failure, J. Am. Coll.
[157] A. Nakai, O. Yamaguchi, T. Takeda, Y. Higuchi, S. Hikoso, M. Taniike, et al., The role Cardiol. 54 (2009) 1637–1646.
of autophagy in cardiomyocytes in the basal state and in response to hemodynam- [185] M.J. Raher, H.B. Thibault, E.S. Buys, D. Kuruppu, N. Shimizu, A.L. Brownell, et al., A
ic stress, Nat. Med. 13 (2007) 619–624. short duration of high-fat diet induces insulin resistance and predisposes to ad-
[158] T. Zaglia, G. Milan, A. Ruhs, M. Franzoso, E. Bertaggia, N. Pianca, et al., Atrogin-1 de- verse left ventricular remodeling after pressure overload, Am. J. Physiol. Heart
ficiency promotes cardiomyopathy and premature death via impaired autophagy, Circ. Physiol. 295 (2008) H2495–H2502.
J. Clin. Invest. 124 (2014) 2410–2424. [186] I.C. Okere, D.J. Chess, T.A. McElfresh, J. Johnson, J. Rennison, P. Ernsberger, et al.,
[159] R.J. Gusterson, E. Jazrawi, I.M. Adcock, D.S. Latchman, The transcriptional co-activa- High-fat diet prevents cardiac hypertrophy and improves contractile function in
tors CREB-binding protein (CBP) and p300 play a critical role in cardiac hypertro- the hypertensive dahl salt-sensitive rat, Clin. Exp. Pharmacol. Physiol. 32 (2005)
phy that is dependent on their histone acetyltransferase activity, J. Biol. Chem. 278 825–831.
(2003) 6838–6847. [187] H. Tuunanen, E. Engblom, A. Naum, K. Nagren, B. Hesse, K.E. Airaksinen, et al., Free
[160] T. Yanazume, K. Hasegawa, T. Morimoto, T. Kawamura, H. Wada, A. Matsumori, fatty acid depletion acutely decreases cardiac work and efficiency in
et al., Cardiac p300 is involved in myocyte growth with decompensated heart fail- cardiomyopathic heart failure, Circulation 114 (2006) 2130–2137.
ure, Mol. Cell. Biol. 23 (2003) 3593–3606. [188] P.J. Randle, Regulatory interactions between lipids and carbohydrates: the glucose
[161] S. Miyamoto, T. Kawamura, T. Morimoto, K. Ono, H. Wada, Y. Kawase, et al., Histone fatty acid cycle after 35 years, Diabetes Metab. Rev. 14 (1998) 263–283.
acetyltransferase activity of p300 is required for the promotion of left ventricular [189] M. Jullig, A.J. Hickey, C.C. Chai, G.L. Skea, M.J. Middleditch, S. Costa, et al., Is the fail-
remodeling after myocardial infarction in adult mice in vivo, Circulation 113 ing heart out of fuel or a worn engine running rich? A study of mitochondria in old
(2006) 679–690. spontaneously hypertensive rats, Proteomics 8 (2008) 2556–2572.
[162] J. Lu, T.A. McKinsey, R.L. Nicol, E.N. Olson, Signal-dependent activation of the MEF2 [190] H. Degens, K.F. de Brouwer, A.J. Gilde, M. Lindhout, P.H. Willemsen, B.J. Janssen,
transcription factor by dissociation from histone deacetylases, Proc. Natl. Acad. Sci. et al., Cardiac fatty acid metabolism is preserved in the compensated hypertrophic
U. S. A. 97 (2000) 4070–4075. rat heart, Basic Res. Cardiol. 101 (2006) 17–26.
[163] S. Chang, T.A. McKinsey, C.L. Zhang, J.A. Richardson, J.A. Hill, E.N. Olson, Histone [191] F.R. Eberli, C.S. Apstein, S. Ngoy, B.H. Lorell, Exacerbation of left-ventricular ische-
deacetylases 5 and 9 govern responsiveness of the heart to a subset of stress sig- mic diastolic dysfunction by pressure-overload hypertrophy — modification by
nals and play redundant roles in heart development, Mol. Cell. Biol. 24 (2004) specific-inhibition of cardiac angiotensin converting enzyme, Circ. Res. 70 (1992)
8467–8476. 931–943.
[164] M. Hohl, M. Wagner, J.C. Reil, S.A. Muller, M. Tauchnitz, A.M. Zimmer, et al., HDAC4 [192] P. Zhabyeyev, M. Gandhi, J. Mori, R. Basu, Z. Kassiri, A. Clanachan, et al., Pressure-
controls histone methylation in response to elevated cardiac load, J. Clin. Invest. overload-induced heart failure induces a selective reduction in glucose oxidation
123 (2013) 1359–1370. at physiological afterload, Cardiovasc. Res. 97 (2013) 676–685.
[165] C.M. Trivedi, Y. Luo, Z. Yin, M. Zhang, W. Zhu, T. Wang, et al., Hdac2 regulates the [193] M.E. Christe, R.L. Rodgers, Altered glucose and fatty acid oxidation in hearts of the
cardiac hypertrophic response by modulating Gsk3 beta activity, Nat. Med. 13 spontaneously hypertensive rat, J. Mol. Cell. Cardiol. 26 (1994) 1371–1375.
(2007) 324–331. [194] P.A. Amorim, T.D. Nguyen, Y. Shingu, M. Schwarzer, F.W. Mohr, A. Schrepper, et al.,
[166] M. Movassagh, M.K. Choy, D.A. Knowles, L. Cordeddu, S. Haider, T. Down, et al., Dis- Myocardial infarction in rats causes partial impairment in insulin response associ-
tinct epigenomic features in end-stage failing human hearts, Circulation 124 ated with reduced fatty acid oxidation and mitochondrial gene expression, J.
(2011) 2411–2422. Thorac. Cardiovasc. Surg. 140 (2010) 1160–1167.
I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262 261

[195] V.G. Davila-Roman, G. Vedala, P. Herrero, L. de las Fuentes, J.G. Rogers, D.P. Kelly, [224] H. Ito, Y. Hirata, M. Hiroe, M. Tsujino, S. Adachi, T. Takamoto, et al., Endothelin-1 in-
et al., Altered myocardial fatty acid and glucose metabolism in idiopathic dilated duces hypertrophy with enhanced expression of muscle-specific genes in cultured
cardiomyopathy, J. Am. Coll. Cardiol. 40 (2002) 271–277. neonatal rat cardiomyocytes, Circ. Res. 69 (1991) 209–215.
[196] R.O. Pereira, A.R. Wende, C. Olsen, J. Soto, T. Rawlings, Y. Zhu, et al., GLUT1 deficien- [225] H. Ito, M. Hiroe, Y. Hirata, H. Fujisaki, S. Adachi, H. Akimoto, et al., Endothelin ETA
cy in cardiomyocytes does not accelerate the transition from compensated hyper- receptor antagonist blocks cardiac hypertrophy provoked by hemodynamic over-
trophy to heart failure, J. Mol. Cell. Cardiol. 72 (2014) 95–103. load, Circulation 89 (1994) 2198–2203.
[197] R.O. Pereira, A.R. Wende, C. Olsen, J. Soto, T. Rawlings, Y. Zhu, et al., Inducible over- [226] S. Heiden, N. Vignon-Zellweger, S. Masuda, K. Yagi, K. Nakayama, M.
expression of GLUT1 prevents mitochondrial dysfunction and attenuates structural Yanagisawa, et al., Vascular endothelium derived endothelin-1 is required
remodeling in pressure overload but does not prevent left ventricular dysfunction, for normal heart function after chronic pressure overload in mice, PLoS ONE
J. Am. Heart Assoc. 2 (2013), e000301. 9 (2014), e88730.
[198] E.D. Abel, H.C. Kaulbach, R. Tian, J.C. Hopkins, J. Duffy, T. Doetschman, et al., Cardiac [227] S. Epelman, K.J. Lavine, A.E. Beaudin, D.K. Sojka, J.A. Carrero, B. Calderon, et al., Em-
hypertrophy with preserved contractile function after selective deletion of GLUT4 bryonic and adult-derived resident cardiac macrophages are maintained through
from the heart, J. Clin. Invest. 104 (1999) 1703–1714. distinct mechanisms at steady state and during inflammation, Immunity 40
[199] J.C. Schisler, T.J. Grevengoed, F. Pascual, D.E. Cooper, J.M. Ellis, D.S. Paul, et al., Car- (2013) 91–104.
diac energy dependence on glucose increases metabolites related to glutathione [228] H.R. Zandbergen, U.C. Sharma, S. Gupta, J.W. Verjans, S. van den Borne, S. Pokharel,
and activates metabolic genes controlled by mechanistic target of rapamycin, J. et al., Macrophage depletion in hypertensive rats accelerates development of car-
Am. Heart Assoc. 4 (2015). diomyopathy, J. Cardiovasc. Pharmacol. Ther. 14 (2009) 68–75.
[200] P. Mirtschink, J. Krishnan, F. Grimm, A. Sarre, M. Horl, M. Kayikci, et al., HIF-driven [229] S.B. Haudek, J. Cheng, J. Du, Y. Wang, J. Hermosillo-Rodriguez, J. Trial, et al., Mono-
SF3B1 induces KHK-C to enforce fructolysis and heart disease, Nature 522 (2015) cytic fibroblast precursors mediate fibrosis in angiotensin-II-induced cardiac hy-
444–449. pertrophy, J. Mol. Cell. Cardiol. 49 (2010) 499–507.
[201] I. Banerjee, J.W. Fuseler, R.L. Price, T.K. Borg, T.A. Baudino, Determination of cell [230] J.I. Odegaard, A. Chawla, Pleiotropic actions of insulin resistance and inflammation
types and numbers during cardiac development in the neonatal and adult rat in metabolic homeostasis, Science 339 (2013) 172–177.
and mouse, Am. J. Physiol. Heart Circ. Physiol. 293 (2007) H1883–H1891. [231] C.N. Lumeng, J.L. Bodzin, A.R. Saltiel, Obesity induces a phenotypic switch in adi-
[202] H.W. Vliegen, A. van der Laarse, C.J. Cornelisse, F. Eulderink, Myocardial pose tissue macrophage polarization, J. Clin. Invest. 117 (2007) 175–184.
changes in pressure overload-induced left ventricular hypertrophy. A study [232] R.A. Frieler, R.M. Mortensen, Immune cell and other noncardiomyocyte regulation
on tissue composition, polyploidization and multinucleation, Eur. Heart J. 12 of cardiac hypertrophy and remodeling, Circulation 131 (2015) 1019–1030.
(1991) 488–494. [233] B. Devaux, D. Scholz, A. Hirche, W.P. Klovekorn, J. Schaper, Upregulation of cell ad-
[203] A. Leask, Getting to the heart of the matter new insights into cardiac fibrosis, Circ. hesion molecules and the presence of low grade inflammation in human chronic
Res. 116 (2015) 1269–1276. heart failure, Eur. Heart J. 18 (1997) 470–479.
[204] M. Gregor, S. Osmanagic-Myers, G. Burgstaller, M. Wolfram, I. Fischer, G. Walko, [234] Y. Kayama, T. Minamino, H. Toko, M. Sakamoto, I. Shimizu, H. Takahashi, et al., Car-
et al., Mechanosensing through focal adhesion-anchored intermediate filaments, diac 12/15 lipoxygenase-induced inflammation is involved in heart failure, J. Exp.
FASEB J. 28 (2014) 715–729. Med. 206 (2009) 1565–1574.
[205] D. MacKenna, S.R. Summerour, F.J. Villarreal, Role of mechanical factors in modu- [235] N.G. Frangogiannis, C.W. Smith, M.L. Entman, The inflammatory response in myo-
lating cardiac fibroblast function and extracellular matrix synthesis, Cardiovasc. cardial infarction, Cardiovasc. Res. 53 (2002) 31–47.
Res. 46 (2000) 257–263. [236] T. Nagai, T. Anzai, H. Kaneko, Y. Mano, A. Anzai, Y. Maekawa, et al., C-reactive pro-
[206] T. Moore-Morris, N. Guimaraes-Camboa, I. Banerjee, A.C. Zambon, T. Kisseleva, A. tein overexpression exacerbates pressure overload-induced cardiac remodeling
Velayoudon, et al., Resident fibroblast lineages mediate pressure overload-induced through enhanced inflammatory response, Hypertension 57 (2011) 208–215.
cardiac fibrosis, J. Clin. Invest. 124 (2014) 2921–2934. [237] J.D. Schilling, H.M. Machkovech, A.H. Kim, R. Schwendener, J.E. Schaffer, Macro-
[207] M.A. Bogoyevitch, P.E. Glennon, M.B. Andersson, A. Lazou, C.J. Marshall, P.H. phages modulate cardiac function in lipotoxic cardiomyopathy, Am. J. Physiol.
Sugden, Acidic fibroblast growth factor or endothelin-1 stimulate the MAP kinase Heart Circ. Physiol. 303 (2012) H1366–H1373.
cascade in cardiac myocytes, Biochem. Soc. Trans. 21 (1993) 358S. [238] S. Rosenkranz, M. Flesch, K. Amann, C. Haeuseler, H. Kilter, U. Seeland, et al., Alter-
[208] C. Pellieux, A. Foletti, G. Peduto, J.F. Aubert, J. Nussberger, F. Beermann, et al., Dilat- ations of beta-adrenergic signaling and cardiac hypertrophy in transgenic mice
ed cardiomyopathy and impaired cardiac hypertrophic response to angiotensin II overexpressing TGF-beta(1), Am. J. Physiol. Heart Circ. Physiol. 283 (2002)
in mice lacking FGF-2, J. Clin. Invest. 108 (2001) 1843–1851. H1253–H1262.
[209] J.E. Schultz, S.A. Witt, M.L. Nieman, P.J. Reiser, S.J. Engle, M. Zhou, et al., Fibroblast [239] T. Kubota, C.F. McTiernan, C.S. Frye, S.E. Slawson, B.H. Lemster, A.P. Koretsky, et al.,
growth factor-2 mediates pressure-induced hypertrophic response, J. Clin. Invest. Dilated cardiomyopathy in transgenic mice with cardiac-specific overexpression of
104 (1999) 709–719. tumor necrosis factor-alpha, Circ. Res. 81 (1997) 627–635.
[210] N. Hayata, Y. Fujio, Y. Yamamoto, T. Iwakura, M. Obana, M. Takai, et al., Connective [240] M. Sun, M. Chen, F. Dawood, U. Zurawska, J.Y. Li, T. Parker, et al., Tumor necrosis
tissue growth factor induces cardiac hypertrophy through Akt signaling, Biochem. factor-alpha mediates cardiac remodeling and ventricular dysfunction after pres-
Biophys. Res. Commun. 370 (2008) 274–278. sure overload state, Circulation 115 (2007) 1398–1407.
[211] A.N. Panek, M.G. Posch, N. Alenina, S.K. Ghadge, B. Erdmann, E. Popova, et al., Con- [241] S. Papathanasiou, S. Rickelt, M.E. Soriano, T.G. Schips, H.J. Maier, C.H. Davos, et al.,
nective tissue growth factor overexpression in cardiomyocytes promotes cardiac Tumor necrosis factor-alpha confers cardioprotection through ectopic expression
hypertrophy and protection against pressure overload, PLoS ONE 4 (2009). of keratins K8 and K18, Nat. Med. 21 (2015) 1076–1084.
[212] C. Bang, S. Batkai, S. Dangwal, S.K. Gupta, A. Foinquinos, A. Holzmann, et al., Cardiac [242] H. Hirota, K. Yoshida, T. Kishimoto, T. Taga, Continuous activation of gp130, a sig-
fibroblast-derived microRNA passenger strand-enriched exosomes mediate car- nal-transducing receptor component for interleukin 6-related cytokines, causes
diomyocyte hypertrophy, J. Clin. Invest. 124 (2014) 2136–2146. myocardial hypertrophy in mice, Proc. Natl. Acad. Sci. U. S. A. 92 (1995)
[213] F. Kuwahara, H. Kai, K. Tokuda, M. Kai, A. Takeshita, K. Egashira, et al., Transforming 4862–4866.
growth factor-beta function blocking prevents myocardial fibrosis and diastolic [243] G.C. Melendez, J.L. McLarty, S.P. Levick, Y. Du, J.S. Janicki, G.L. Brower, Interleukin 6
dysfunction in pressure-overloaded rats, Circulation 106 (2002) 130–135. mediates myocardial fibrosis, concentric hypertrophy, and diastolic dysfunction in
[214] P. Teekakirikul, S. Eminaga, O. Toka, R. Alcalai, L.B. Wang, H. Wakimoto, et al., rats, Hypertension 56 (2010) 225–231.
Cardiac fibrosis in mice with hypertrophic cardiomyopathy is mediated by [244] B. Coles, C.A. Fielding, S. Rose-John, J. Scheller, S.A. Jones, V.B. O'Donnell, Classic in-
non-myocyte proliferation and requires Tgf-beta, J. Clin. Invest. 120 (2010) terleukin-6 receptor signaling and interleukin-6 trans-signaling differentially con-
3520–3529. trol angiotensin II-dependent hypertension, cardiac signal transducer and activator
[215] D. Zhang, V. Gaussin, G.E. Taffet, N.S. Belaguli, M. Yamada, R.J. Schwartz, et al., TAK1 of transcription-3 activation, and vascular hypertrophy in vivo, Am. J. Pathol. 171
is activated in the myocardium after pressure overload and is sufficient to provoke (2007) 315–325.
heart failure in transgenic mice, Nat. Med. 6 (2000) 556–563. [245] F. Ma, Y. Li, L. Jia, Y. Han, J. Cheng, H. Li, et al., Macrophage-stimulated cardiac fibro-
[216] K. Ono, T. Ohtomo, J. Ninomiya-Tsuji, M. Tsuchiya, A dominant negative TAK1 in- blast production of IL-6 is essential for TGF beta/Smad activation and cardiac fibro-
hibits cellular fibrotic responses induced by TGF-beta, Biochem. Biophys. Res sis induced by angiotensin II, PLoS ONE 7 (2012), e35144.
Commun. 307 (2003) 332–337. [246] H. Meier, J. Bullinger, G. Marx, A. Deten, L.C. Horn, B. Rassler, et al., Crucial role of
[217] T. Tsuruda, J. Kato, K. Kitamura, M. Kawamoto, K. Kuwasako, T. Imamura, et al., An interleukin-6 in the development of norepinephrine-induced left ventricular re-
autocrine or a paracrine role of adrenomedullin in modulating cardiac fibroblast modeling in mice, Cell. Physiol. Biochem. 23 (2009) 327–334.
growth, Cardiovasc. Res. 43 (1999) 958–967. [247] A.M. Terrell, P.R. Crisostomo, G.M. Wairiuko, M. Wang, E.D. Morrell, D.R. Meldrum,
[218] P. Niu, T. Shindo, H. Iwata, S. Iimuro, N. Takeda, Y.L. Zhang, et al., Protective effects Jak/STAT/SOCS signaling circuits and associated cytokine-mediated inflammation
of endogenous adrenomedullin on cardiac hypertrophy, fibrosis, and renal dam- and hypertrophy in the heart, Shock 26 (2006) 226–234.
age, Circulation 109 (2004) 1789–1794. [248] H. Hirota, J. Chen, U.A. Betz, K. Rajewsky, Y. Gu, J. Ross Jr., et al., Loss of a gp130 car-
[219] S. Sanada, D. Hakuno, L.J. Higgins, E.R. Schreiter, A.N.J. McKenzie, R.T. Lee, IL-33 and diac muscle cell survival pathway is a critical event in the onset of heart failure
ST2 comprise a critical biomechanically induced and card ioprotective signaling during biomechanical stress, Cell 97 (1999) 189–198.
system, J. Clin. Invest. 117 (2007) 1538–1549. [249] S. Honsho, S. Nishikawa, K. Amano, K. Zen, Y. Adachi, E. Kishita, et al., Pressure-me-
[220] M. Kuhn, Cardiology: a big-hearted molecule, Nature 519 (2015) 416–417. diated hypertrophy and mechanical stretch induces IL-1 release and subsequent
[221] K. Lemmens, K. Doggen, G.W. De Keulenaer, Role of neuregulin-1/ErbB signaling in IGF-1 generation to maintain compensative hypertrophy by affecting Akt and
cardiovascular physiology and disease: implications for therapy of heart failure, JNK pathways, Circ. Res. 105 (2009) 1149–1158.
Circulation 116 (2007) 954–960. [250] B.W. Van Tassell, R.A. Arena, S. Toldo, E. Mezzaroma, T. Azam, I.M. Seropian, et al.,
[222] E.M. Parodi, B. Kuhn, Signalling between microvascular endothelium and Enhanced interleukin-1 activity contributes to exercise intolerance in patients with
cardiomyocytes through neuregulin, Cardiovasc. Res. 102 (2014) 194–204. systolic heart failure, PLoS ONE 7 (2012), e33438.
[223] M.C. Scimia, C. Hurtado, S. Ray, S. Metzler, K. Wei, J. Wang, et al., APJ acts as a dual [251] V. Rajapurohitam, A. Kilic, S. Javadov, M. Karmazyn, Role of NF-kappaB and p38
receptor in cardiac hypertrophy, Nature 488 (2012) 394–398. MAPK activation in mediating angiotensin II and endothelin-1-induced
262 I. Shimizu, T. Minamino / Journal of Molecular and Cellular Cardiology 97 (2016) 245–262

stimulation in leptin production and cardiomyocyte hypertrophy, Mol. Cell. [270] J. Hur, C.H. Yoon, C.S. Lee, T.Y. Kim, I.Y. Oh, K.W. Park, et al., Akt is a key modulator
Biochem. 366 (2012) 287–297. of endothelial progenitor cell trafficking in ischemic muscle, Stem Cells 25 (2007)
[252] H. Javan, A.M. Szucsik, L. Li, C.L. Schaaf, M.E. Salama, C.H. Selzman, Cardiomyocyte 1769–1778.
p65 nuclear factor-kappaB is necessary for compensatory adaptation to pressure [271] D. Fulton, J.P. Gratton, T.J. McCabe, J. Fontana, Y. Fujio, K. Walsh, et al., Regulation of
overload, Circ. Heart Fail. 8 (2015) 109–118. endothelium-derived nitric oxide production by the protein kinase Akt, Nature 399
[253] Q. Liu, Y. Chen, M. Auger-Messier, J.D. Molkentin, Interaction between NFkappaB (1999) 597–601.
and NFAT coordinates cardiac hypertrophy and pathological remodeling, Circ. [272] S. Dimmeler, I. Fleming, B. Fisslthaler, C. Hermann, R. Busse, A.M. Zeiher, Activation
Res. 110 (2012) 1077–1086. of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation,
[254] S.K. Verma, P. Krishnamurthy, D. Barefield, N. Singh, R. Gupta, E. Lambers, et al., In- Nature 399 (1999) 601–605.
terleukin-10 treatment attenuates pressure overload-induced hypertrophic re- [273] Y. Rikitake, K. Hirata, S. Kawashima, M. Ozaki, T. Takahashi, W. Ogawa, et al., In-
modeling and improves heart function via signal transducers and activators of volvement of endothelial nitric oxide in sphingosine-1-phosphate-induced angio-
transcription 3-dependent inhibition of nuclear factor-kappa B, Circulation 126 genesis, Arterioscler. Thromb. Vasc. Biol. 22 (2002) 108–114.
(2012) (418-+). [274] D. Tirziu, E. Chorianopoulos, K.L. Moodie, R.T. Palac, Z.W. Zhuang, M. Tjwa, et al.,
[255] F.M.Y. Yang, A.P. Dong, P. Mueller, J. Caicedo, A.M. Sutton, J. Odetunde, et al., Coro- Myocardial hypertrophy in the absence of external stimuli is induced by angiogen-
nary artery remodeling in a model of left ventricular pressure overload is influ- esis in mice, J. Clin. Invest. 117 (2007) 3188–3197.
enced by platelets and inflammatory cells, PLoS ONE 7 (2012). [275] I.M. Jaba, Z.W. Zhuang, N. Li, Y. Jiang, K.A. Martin, A.J. Sinusas, et al., NO triggers
[256] A. Ghigo, I. Franco, F. Morello, E. Hirsch, Myocyte signalling in leucocyte recruit- RGS4 degradation to coordinate angiogenesis and cardiomyocyte growth, J. Clin.
ment to the heart, Cardiovasc. Res. 102 (2014) 270–280. Invest. 123 (2013) 1718–1731.
[257] T. Kamo, H. Akazawa, I. Komuro, Cardiac nonmyocytes in the hub of cardiac hyper- [276] M.M. Givertz, Cardiology patient page: peripartum cardiomyopathy, Circulation
trophy, Circ. Res. 117 (2015) 89–98. 127 (2013) e622–e626.
[258] O. Hudlicka, M. Brown, S. Egginton, Angiogenesis in skeletal and cardiac muscle, [277] I.S. Patten, S. Rana, S. Shahul, G.C. Rowe, C. Jang, L. Liu, et al., Cardiac angiogenic im-
Physiol. Rev. 72 (1992) 369–417. balance leads to peripartum cardiomyopathy, Nature 485 (2012) 333–338.
[259] M.L. Marcus, S. Koyanagi, D.G. Harrison, D.B. Doty, L.F. Hiratzka, C.L. Eastham, Ab- [278] E.P. Tsagalou, M. Anastasiou-Nana, E. Agapitos, A. Gika, S.G. Drakos, J.V. Terrovitis,
normalities in the coronary circulation that occur as a consequence of cardiac hy- et al., Depressed coronary flow reserve is associated with decreased myocardial
pertrophy, Am. J. Med. 75 (1983) 62–66. capillary density in patients with heart failure due to idiopathic dilated cardiomy-
[260] R.J. Tomanek, Response of the coronary vasculature to myocardial hypertrophy, J. opathy, J. Am. Coll. Cardiol. 52 (2008) 1391–1398.
Am. Coll. Cardiol. 15 (1990) 528–533. [279] S.G. Drakos, A.G. Kfoury, E.H. Hammond, B.B. Reid, M.P. Revelo, B.Y. Rasmusson,
[261] I. Shimizu, T. Aprahamian, R. Kikuchi, A. Shimizu, K.N. Papanicolaou, S. et al., Impact of mechanical unloading on microvasculature and associated central
MacLauchlan, et al., Vascular rarefaction mediates whitening of brown fat in obe- remodeling features of the failing human heart, J. Am. Coll. Cardiol. 56 (2010)
sity, J. Clin. Invest. 124 (2014) 2099–2112. 382–391.
[262] Y. Kitamoto, H. Tokunaga, K. Tomita, Vascular endothelial growth factor is an es- [280] B.A. Borlaug, The pathophysiology of heart failure with preserved ejection fraction,
sential molecule for mouse kidney development: glomerulogenesis and Nat. Rev. Cardiol. 11 (2014) 507–515.
nephrogenesis, J. Clin. Invest. 99 (1997) 2351–2357. [281] D. Lloyd-Jones, R.J. Adams, T.M. Brown, M. Carnethon, S. Dai, G. De Simone, et al.,
[263] N. Ferrara, Vascular endothelial growth factor: basic science and clinical progress, Heart disease and stroke statistics–2010 update: a report from the American
Endocr. Rev. 25 (2004) 581–611. Heart Association, Circulation 121 (2010) e46–e215.
[264] Y. Izumiya, I. Shiojima, K. Sato, D.B. Sawyer, W.S. Colucci, K. Walsh, Vascular endo- [282] S.F. Mohammed, S. Hussain, S.A. Mirzoyev, W.D. Edwards, J.J. Maleszewski, M.M.
thelial growth factor blockade promotes the transition from compensatory cardiac Redfield, Coronary microvascular rarefaction and myocardial fibrosis in heart fail-
hypertrophy to failure in response to pressure overload, Hypertension 47 (2006) ure with preserved ejection fraction, Circulation 131 (2015) 550–559.
887–893. [283] E.G. Lakatta, Cardiovascular regulatory mechanisms in advanced age, Physiol. Rev.
[265] S. Dass, C.J. Holloway, L.E. Cochlin, O.J. Rider, M. Mahmod, M. Robson, et al., No ev- 73 (1993) 413–467.
idence of myocardial oxygen deprivation in nonischemic heart failure, Circ. Heart [284] H. Shih, B. Lee, R.J. Lee, A.J. Boyle, The aging heart and post-infarction left ventric-
Fail. 8 (2015) 1088–1093. ular remodeling, J. Am. Coll. Cardiol. 57 (2011) 9–17.
[266] J. Heineke, M. Auger-Messier, J. Xu, T. Oka, M.A. Sargent, A. York, et al., Cardiomyo- [285] K. Prasun, R.T. Das, V. Panagia, N.S. Dhalla, β-adrenergic signal transduction path-
cyte GATA4 functions as a stress-responsive regulator of angiogenesis in the mu- way in developing and aging hearts, Heart Fail. Rev. 23-41 (1997).
rine heart, J. Clin. Invest. 117 (2007) 3198–3210. [286] F.S. Loffredo, M.L. Steinhauser, S.M. Jay, J. Gannon, J.R. Pancoast, P. Yalamanchi,
[267] N.S. Belaguli, J.L. Sepulveda, V. Nigam, F. Charron, M. Nemer, R.J. Schwartz, Cardiac et al., Growth differentiation factor 11 is a circulating factor that reverses age-relat-
tissue enriched factors serum response factor and GATA-4 are mutual coregulators, ed cardiac hypertrophy, Cell 153 (2013) 828–839.
Mol. Cell. Biol. 20 (2000) 7550–7558. [287] S.C. Smith, X.X. Zhang, X.Y. Zhang, P. Gross, T. Starosta, S. Mohsin, et al., GDF11 does
[268] Q. Liang, R.J. Wiese, Bueno OF, Y.S. Dai, B.E. Markham, J.D. Molkentin, The transcrip- not rescue aging-related pathological hypertrophy, Circ. Res. 117 (2015) 926–932.
tion factor GATA4 is activated by extracellular signal-regulated kinase 1- and 2- [288] L. Katsimpardi, N.K. Litterman, P.A. Schein, C.M. Miller, F.S. Loffredo, G.R.
mediated phosphorylation of serine 105 in cardiomyocytes, Mol. Cell. Biol. 21 Wojtkiewicz, et al., Vascular and neurogenic rejuvenation of the aging mouse
(2001) 7460–7469. brain by young systemic factors, Science 344 (2014) 630–634.
[269] E. Ackah, J. Yu, S. Zoellner, Y. Iwakiri, C. Skurk, R. Shibata, et al., Akt1/protein kinase
Balpha is critical for ischemic and VEGF-mediated angiogenesis, J. Clin. Invest. 115
(2005) 2119–2127.

You might also like