Вы находитесь на странице: 1из 11

REVIEWS

Molecular basis of physiological


heartgrowth: fundamental concepts
and new players
Marjorie Maillet1, Jop H.van Berlo1 and Jeffery D.Molkentin1,2
Abstract | The heart hypertrophies in response to developmental signals as well as increased
workload. Although adult-onset hypertrophy can ultimately lead to disease, cardiac
hypertrophy is not necessarily maladaptive and can even be beneficial. Progress has been
made in our understanding of the structural and molecular characteristics of physiological
cardiac hypertrophy, as well as of the endocrine effectors and associated signalling
pathways that regulate it. Physiological hypertrophy is initiated by finite signals, which
include growth hormones (such as thyroid hormone, insulin, insulin-like growth factor 1
and vascular endothelial growth factor) and mechanical forces that converge on a limited
number of intracellular signalling pathways (such as PI3K, AKT, AMP-activated protein
kinase and mTOR) to affect gene transcription, protein translation and metabolism.
Harnessing adaptive signalling mediators to reinvigorate the diseased heart could have
important medical ramifications.

Myocardium
The mammalian heart is a muscle, the fundamental response to pathological conditions such as hyper
From the Greek mys (muscle) function of which is to pump blood throughout the tension and myocardial infarction from coronary artery
and kardia (heart). It is the circulatory system to deliver oxygen and nutrients to disease, which eventually give rise to ventricular remod
thick middle muscular layer of organs and to transport carbon dioxide back to the elling and dilatation, fibrosis, and diminished cardiac
the heart that contracts.
lungs. In response to an increased workload, typically output 2,3. However, the myocardium can also undergo an
caused by pathological or physiological stimulation, the adaptive form of cardiac hypertrophy called physiologi
heart undergoes a growth process named hypertrophy, cal hypertrophy, which is fundamentally different from
which decreases ventricular wall stress and maintains or pathological hypertrophy because the heart does not
even augments pump function (BOX1). However, hyper develop disease and can even benefit from it6. A heart
trophy, from the Greek for increased growth, is a more undergoing physiological hypertrophy shows enhanced
complex phenomenon than this simple definition might vascular perfusion and metabolism, and the growth
suggest. Cardiomyocytes, which represent 85% of the process is initiated by molecular pathways specific to
heart mass, are the contracting cells of the heart, and physiological hypertrophy. This Review discusses the
they contain an arrayed series of basic contractile units unique structural, functional, metabolic and circulatory
1
Department of Pediatrics,
University of Cincinnati,
called sarcomeres13 (BOX1). Unlike other cell types that features of physiological hypertrophy, although we do
Cincinnati Childrens Hospital comprise the heart (that is, endothelial cells, fibroblasts, not attempt to cover the even more elaborate literature
Medical Center, Cincinnati, immune cells and progenitor cells), mammalian cardio related to pathological cardiac hypertrophy.
Ohio 45229, USA. myocytes become terminally differentiated shortly
2
Howard Hughes Medical
after birth and mostly lose their ability to proliferate, Characteristics of physiological hypertrophy
Institute, University of
Cincinnati, Cincinnati although a low level of cardiomyocyte turnover occurs Physiological cardiac hypertrophy drives the normal
Childrens Hospital Medical throughout life1,4,5. As a consequence, although cardiac growth of the heart from birth to early adulthood
Center, Cincinnati, mass can be increased by fibroblast proliferation, and (which is known as postnatal or maturational hyper
Ohio45229, USA. possibly also by progenitor cell activity and some trophy, hereafter referred to as postnatal hypertrophy),
Correspondence to J.D.M.
e-mail:
cardiomyocyte renewal, mass increase primarily occurs the growth of the maternal heart during pregnancy,
jeff.molkentin@cchmc.org through the hypertrophy of individual cardiomyocytes and the growth of the heart in well-conditioned
doi:10.1038/nrm3495 (BOX 1) . Cardiac hypertrophy commonly occurs in athletes as a result of extreme and/or repetitive exercise.

38 | JANUARY 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Cardiac hypertrophy geometries


The heart has the ability to increase its size and, depending on the stimulus, this results in physiological or pathological
hypertrophy. Hypertrophy itself decreases ventricular wall stress by increasing the thickness of this wall. It follows Laplaces
law, which says wall stress (or tension) is an inverse function of wall thickness (tension=(pressureradius)/(2wall
thickness)). Cardiac hypertrophy can be classed as either eccentric or concentric growth, based on the geometries of the
heart and individual cardiomyocytes (see the figure). Non-pathological eccentric hypertrophy is characterized by an
increase in ventricular volume with a coordinated growth in wall and septal thicknesses, where individual cardiomyocytes
grow in both length and width. However, eccentric hypertrophy under pathological conditions (myocardial infarction or
dilated cardiomyopathy) can lead to wall dilation with preferential lengthening of cardiomyocytes. Physiological
stimulation (such as pregnancy or endurance training) can also induce a less pronounced form of eccentric hypertrophy2,6.
Concentric hypertrophy is characterized by a reduction in left ventricular chamber dimension and an increase in free wall
and septal thicknesses, and individual cardiomyocytes typically increase in thickness more than in length (this results in a
decreased length/width ratio)2,6. Concentric hypertrophy usually arises owing to pathological conditions such as chronic
hypertension or valvular stenosis. Isometric exercise training, such as wrestling or weight-lifting, also induces a milder form
of concentric cardiac hypertrophy that is not known to be pathological1. The heart can go from a normal state to a state of
physiological hypertrophy and back, although pathological hypertrophy that produces heart failure may be less reversible.

Physiological Pathological

Normal adult heart


Neonatal heart Right ventricle Left ventricle

Diameter
Development

Sarcomere Thickness
Width
Exercise or Length Cardiomyopathy
pregnancy

Eccentric
Eccentric Concentric Concentric

Nature Reviews | Molecular Cell Biology


Valvularstenosis
Postnatal hypertrophy is induced by greater circula described in cross-country skiers in the late nineteenth
Also called heart valve disease.
Valvular stenosis occurs in tory demands of the growing organism and by high century, was later defined more definitively by electro
response to stiffening, circulatory levels of growth hormone and insulin-like cardiographic studies in the mid1950s in marathon
thickening, fusion or blockage growth factor 1 (IGF1) 7. During postnatal hyper runners11,12 (BOX1).
of one or more valves of the trophy, some of the cardiomyocytes become binucle Physiological hypertrophy is a mild form of growth,
heart. The heart comprises
four valves: the mitral, aortic,
ated, expand their contractile apparatus, remodel their typically characterized by a 1020% increase in heart
tricuspid and pulmonic valves. extracellular matrix as the heart chambers enlarge and weight normalized to body weight. Heart mass in
wall and septal thicknesses increase, and improve exci professional athletes assessed by echocardiography
Transverse-tubule system tation contraction coupling efficiency through more routinely demonstrated a slightly, albeit significantly,
Also called the Ttubule
elaborate calcium cycling between the transverse-tubule increased value compared with sedentary age-matched
system. A Ttubule is a deep
invagination of the sarcolemma system and sarcoplasmic reticulum8,9. In pregnancy- control individuals1315. In healthy women, pregnancy
(cardiomyocyte plasma induced physiological hypertrophy, cardiac growth is is associated with a transient 1020% increase in total
membrane) enriched in the greatest in the third trimester, as it is associated heart mass16. By contrast, in all mammals, postnatal
excitationcontraction coupling with the largest expansion in circulatory volume and hypertrophy typically results in a twofold or greater
molecules. Ttubule system
refers to the network of
cardiac output requirements for the mother given the increase in left ventricular mass between birth and
Ttubules within an adult rapid growth of the fetus10. Finally, exercise-induced adulthood as a result of an increase in the average
cardiomyocyte. cardiac hypertrophy, which was initially empirically diameter of cardiomyocytes17,18.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JANUARY 2013 | 39

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Importantly, cardiac function is preserved during unique type of growth response. The initiating stimuli
physiological cardiac hypertrophy. Measurements of for triggering physiological cardiac hypertrophy can be
systolic function, as assessed by echocardiography, are separated into stretch-sensitive mechanisms or biochem
similar in both professional athletes and sedentary con ical signals that include neuro-endocrine factors and
trol individuals19. Similarly, diastolic function remains hormones, and will be discussedbelow.
normal or is marginally enhanced in the athletes heart19.
In addition, physiological cardiac hypertrophy induced by Initiating signals: hormones and receptors
pregnancy or exercise is fully reversible. In healthy women The biochemical signals that underlie physiological
the hypertrophy of the left ventricle normalizes 8weeks hypertrophy are well characterized and include ligands
after parturition10. Moreover, echocardiographic studies for a network of tyrosine kinase receptors and nuclear
showed that posterior wall thickness and estimated left receptors. Whereas pathological cardiac hypertrophy is
ventricular mass regressed within a few weeks of decon mediated by the neuro-endocrine hormones endothelin1
ditioning in college- and Olympic-trained athletes20,21. and angiotensin II (BOX2), physiological hypertrophy
This collectively suggests that physiological hypertrophy appears to be initiated by different ligands that include
induced by exercise or pregnancy is likely to be harm thyroid hormone, insulin, growth hormone and IGF1
less, and perhaps even beneficial, in healthy individuals, (REF.2) (FIG.1). Cardiac concentrations of IGF1 but not
although pre-existing conditions such as inherited cardio endothelin 1 or angiotensin II were upregulated in pro
myopathies, arrhythmogenic channelopathies or angiogenic fessional athletes33. Vascular endothelial growth factors
imbalance can promote disease and even premature death (VEGFs) also affect physiological cardiac hypertrophy.
in athletes and mothers22,23. For example, postnatal heart growth was impaired in
There is also a net induction in angiogenesis during Vegfb-null mice34, and mice and rats overexpressing Vegfb
physiological hypertrophy. Exercise training enhances in the heart developed cardiac hypertrophy character
coronary blood flow capacity, coronary artery diameter ized by enlarged cardiomyocytes with preserved cardiac
and the capillary to cardiomyocyte ratio throughout the function, at least early in life35,36.
heart 2426. Finally, hypertrophy can affect the metabolic
substrate profile of the myocardium. Early in cardiac Thyroid hormone as a transcriptional activator of post-
development, glycolysis is the major source of energy natal growth. Although the influence of the thyroid
for proliferating cardiomyocytes. However, after birth, hormone triiodothyronine (T3) on adult physiological
mitochondrial capacity increases and fatty acid oxidation hypertrophy remains controversial, its role in postnatal
becomes the primary metabolic pathway for generating hypertrophy (developmental growth) is more accepted.
ATP in the heart27. Whereas pathological cardiac hyper Thyroid hormone levels increase dramatically in children
trophy is initially associated with a switch from an oxidative after birth37. In mice, circulating T3 levels transiently
to a glycolytic metabolic profile, exercise-induced hyper increase after birth, reaching a maximum during the
trophy relies on maintaining a proper balance of glycolytic second postnatal week (a 2,000fold increase compared
and fatty acid oxidation, as both are augmented28,29. to the level of T3 at birth) and returning to lower levels
An important molecular feature of physiological at the end of the third week of life38. T3 exerts a direct
Systolic function hypertrophy is the absence of induction of the molecular transcriptional effect on contractile and calcium handling
The performance of the left stress fetal gene programme that is classically associated proteins as the heart matures during postnatal develop
ventricle during systole, which with the development of pathological hypertrophy30. This ment. In rodents, T3 regulates the postnatal switch from
is the contraction of the heart. pathology-induced stress programme includes induction the transcription of Myh7 (which encodes -MHC) to the
The best index of left ventricle
systolic function is ejection
of mRNA for atrial natriuretic factor (ANF), brain natriu transcription of genes encoding -MHC 39. T3 sig
fraction, which is calculated retic peptide (BNP), skeletal actin and myosin heavy nals through two different thyroid hormone receptors
as the difference between chain (-MHC; also known as myosin heavy chain 7)1. (TR and TR); by acting as a dimer with the retinoic
end-diastolic and end-systolic Genes encoding calcium-handling proteins also remain acid receptor, these receptors function as transcription
left ventricle volume, divided
unchanged during physiological hypertrophy, whereas factors that can change gene expression40. In a similar
by the end-diastolic left
ventricle volume. many such genes are altered in pathological hypertrophy way to -MHC, T3 positively regulates expression of
or heart failure1. Moreover, physiological hypertrophy is sarcoplasmic/endoplasmic reticulum calcium ATPase2
Diastolic function not significantly associated with interstitial or replace (SERCA2) while inhibiting -MHC and phospholamban
The performance of the left ment fibrosis as observed in pathological hypertrophy 1. expression4043. T3 also activates the transcription of the
ventricle during diastole, which
is the relaxation of the heart
For example, levels of collagen I, which confers stiffness to 1 adrenergic receptor, sodium and potassium channels,
and the filling of the ventricle. the fibrotic matrix of the heart, remain unchanged in the cardiac troponin I (CTNI), ANF, sodium/calcium
left ventricles of rats subjected to endurance training 31,32. exchanger (NCX), TR1 and adenylyl cyclase (AC)
Arrhythmogenic Similarly, markers of myofibroblast activation that under typesV and VI40. Interestingly, cytosolic TR1 interacts
channelopathies
lie pathological remodelling of the heart, such as smooth with the regulatory p85 subunit of PI3K, which might
Genetic or acquired cardiac ion
channel diseases. Ion channels muscle -actin, are not detected in exercised rat hearts32. be a potential mechanism underlying protein synthesis
(sodium, potassium and Physiological hypertrophy at the organ level is trig regulation in the heart41.
calcium channels) control the gered at the cardiomyocyte level by a restricted number of
electrical activity of the heart. hypertrophic stimuli, the signals from which converge on Insulin regulates protein synthesis and gene expression.
Abnormal electrical activity
can lead to cardiac arrhythmias
a limited number of intracellular signal transduction path In the heart, insulin signalling directly affects glucose
(irregular cardiac rhythm) and ways that alter gene expression in the nucleus and increase transport, glycolysis, glucose oxidation, glycogen syn
sudden death. the rates of protein and RNA synthesis to coordinate this thesis, fatty acid oxidation and protein synthesis 44,45.

40 | JANUARY 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 2 | Pathological hypertrophy signalling pathways


Pathological hypertrophy is induced by disorders such as systemic or pulmonary hypertension, myocardial infarction,
coronary artery disease, genetic mutations in genes encoding sarcomeric proteins, diabetic and metabolic
cardiomyopathy, viral and bacterial myocarditis, valvular insufficiency and congenital heart defects3. Pathological
hypertrophy is associated with increased rates of myocyte death, fibrotic remodelling and decreased systolic and
diastolic function that often progresses towards heart failure. Angiotensin II, endothelin 1 and catecholamines bind to
seven-transmembrane receptors that are coupled to and activate heterotrimeric Gproteins. In particular,
Gq/11signalling activates phospholipase C (PLC), which catalyses the synthesis of inositol 1,4,5triphosphate
(Ins(1,4,5)P3) and DAG3. Ins(1,4,5)P3 production induces intracellular Ca2+ release to then activate calcium/calmodulin-
dependent protein kinase (CaMK) or calcineurin, which mediate
cardiomyocyte growth2. Calcineurin is a calcium-dependent protein
phosphatase that dephosphorylates nuclear factor of activated T cells AT-R or /-AR Catecholamine
(NFAT) transcription factors. NFAT forms complexes with the cofactors Endo-R Angiotensin Plasma
GATA4 or myocyte enhancer factor2 (MEF2) to transactivate the membrane
transcription of hypertrophic target genes that are typically
maladaptive, although some beneficial or cellular protective genes are Gq protein AC
also activated. Forexample, numerous genetic or pharmacologic gain-
G protein
and lossoffunction approaches have demonstrated the importance of
the calciumcalcineurinNFAT pathway in regulating the pathological MAPKKK
growth of the heart2 (FIG.2), although calcineurinNFAT signalling PKA
downstream of insulin-like growth factor 1 (IGF1) signalling could be Ca2+
adaptive62. Other examples of mediators of pathological cardiac MAPKK Calcineurin
hypertrophy are the p38 and cJun Nterminal kinase (JNK) branches of
the MAPK cascade, which phosphorylate and activate the GATA4
transcription factor. Sustained p38 or JNK activation in the heart leads JNK p38 NFAT
to cardiomyopathy and heart failure, whereas lossoffunction studies
demonstrate their function as negative regulators of cardiac
hypertrophy2. Catecholamines can also signal to adenylyl cyclase (AC)
to induce activation of protein kinase A (PKA), which then
phosphorylates an array of intracellular targets, leading to increased Nucleus
calcium release and enhanced contractility; the net effect of this
appears to be cardiomyopathic, as it results in increased myocyte
MEF2 NFAT
apoptosis and necrosis. /-AR,- or -adrenergic receptor; AT-R,
Maladaptive
angiotensin II receptor; Endo-R, endothelin 1 receptor; MAPKK, MAPK genes
MEF2 GATA4
kinase; MAPKKK, MAPK kinase kinase.

Nature Reviews | Molecular Cell Biology

Insulin binds to the tyrosine kinase insulin receptor synthesized and secreted by the liver in response tosys
(IR), the activation of which leads to the recruitment temic growth hormone (GH) and then targeted to the
and phosphorylation of the adaptor proteins insulin tissues in which it mediates growth. However, IGF1
receptor substrate 1 (IRS1) and IRS2, which activate can also be produced by target tissues, where it directly
the PI3KAKT signalling pathway (see below). Mice acts in an autocrine and paracrine manner 52. For exam
null for Irs1 or Irs2 displayed a general postn atal ple, cardiac IGF1 and serum GH and IGF1 levels were
growth retardation phenotype 46,47. Heart-specific found to be transiently increased following exercise33,53.
deletion of the Inrs (insulin receptor) gene confirmed Systemic and local IGF1 signalling were also shown
the role of insulin in postnatal heart growth, as hearts to be essential for postnatal growth in general54. IGF1
from these mice were smaller and individual cardio binds the IR and the IGF1 receptor (IGF1R), a trans
myocyte volumes were reduced48. Moreover, heart- membrane tyrosine kinase receptor that activates the
specific Inrs-knockout mice showed exacerbated PI3KAKTphosphoinositide-dependent protein
hypertrophy with pathological stimulation, which was kinase 1 (PDK1)glycogen synthase kinase3 (GSK3)
associated with mitochondrial dysfunction, suggesting pathway (see below). Igf1null mice or Igf1rnull mice
that in the absence of this physiological growth path display a growth retardation phenotype and perinatal
way hearts were more prone to pathological hyper lethality, confirming the centrality of this ligand in
trophy 4951. Altogether, these studies indicate that mediating developmental growth55.
insulin signalling serves as a fundamental baseline Results obtained in genetically modified mice gen
regulator of physiological growth of the heart, and the erally confirm the role of IGF1 in regulating physio
heart is not only smaller in its absence but is also more logical cardiac hypertrophy. Initial results obtained
Myocarditis prone topathology. in mice with MHC-driven overexpression of IGF1 in
Inflammation of the heart the heart showed a physiological benefit, but this
caused by a viral or bacterial IGF1 as an initiator of physiological growth signalling. improvement was likely to have occurred through
infection or an autoimmune
disease. Myocarditis
The best-described and perhaps most crucial signal postnatally related cardiomyocyte hyperplasia56. Later,
sometimes induces eccentric ling pathway regulating physiological cardiac hyper separate transgenic mice overexpressing IGF1 under
hypertrophy and heart failure. trophy is that initiated by IGF1. This ligand is mostly the control of the skeletal actin promoter developed

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JANUARY 2013 | 41

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Initiating signals: mechanosensors


VEGFB Signal IGF1 or insulin Mechanotransduction is the fundamental process that
T3 Time Plasma permits the conversion of mechanical forces into bio
VEGFR1 IGF1R or IR membrane
chemical signals. In cardiomyocytes, extracellular,
PI3K PTEN IRS1/2
AMP/ATP intracellular or intercellular mechanical forces (changes
PDK1 in pressure, volume, stiffness, and so on) are sensed
TSC1/2 LKB1 TAK1
by specialized transducing proteins within the plasma
RHEB membrane, by focal adhesion complexes or internally
AMPK within the cardiac Z line that contains mechanosensing
AKT mTORC1 proteins (FIG.2). These sensing systems then activate
signalling pathways that initiate physiological cardiac
MEK1 GSK3 4EBP1 CaMKK
Metabolism hypertrophy (FIG.1).
Transient receptor potential (TRP) channels, which
ERK1/2 eIF2B eIF4E S6K were originally identified in chicken skeletal myocytes61
ATP
and later identified in essentially all tissues and cell types,
Protein synthesis can function as stretch-sensitive signalling mediators
that permeate calcium and other cations. This stretch-
Energy production
activated calcium current could then mediate growth
Transcription signalling in the heart through downstream effectors such
T3
as calcineurinnuclear factor of activated T cells (NFAT)
RXR TR FOXO
C/EBP Adaptive genes signalling62. The TRP canonical (TRPC) subfamily is com
CITED4 SRF PGC1 prised of TRPC3, TRPC6 and TRPC7 (which are activated
by diacylglycerol (DAG) that is generated by G protein-
Figure 1 | Physiological hypertrophy signalling pathways. Physiological hypertrophy is coupled receptor signalling) as well as TRPC1, TRPC4
an adaptive form of cardiac hypertrophy. The figure Nature Reviews
depicts | Molecular
central signalling Cell Biology
pathways of
and TRPC5 (which can be activated by DAG and deple
physiological hypertrophy discussed in the text. Physiological hypertrophy is initiated by
intermittent signals of triiodothyronine (T3), vascular endothelial growth factor B (VEGFB),
tion of intracellular calcium stores)61. TRPC1, TRPC4,
insulin and insulin-like growth factor 1 (IGF1), as illustrated by the oscillating curve. TRPC5 and TRPC6 are also known to be activated by
Thegrowth hormones activate membrane-localized tyrosine kinase receptors (VEGF stretching, and might thus participate in stretch sensing
receptor 1 (VEGFR1), IGF1 receptor (IGF1R) or insulin receptor (IR)) and nuclear receptors and downstream signalling to induce hypertrophic heart
(thyroid hormone receptor (TR)), which trigger intracellular signalling pathways specific growth. Indeed, TRPC1 and TRPC6 have been shown to
to physiological hypertrophy. These signalling pathways regulate the transcription of be directly activated by stretching, and both have been
adaptive genes, protein synthesis, metabolism and energy production. The growth signals implicated in regulating cardiac hypertrophy 6366.
centre on common signalling branches controlled by ERK1/2, PI3K, AKT and mTOR Integrins can also mediate mechanotransduction
complex 1 (mTORC1), whereas AMP-activated protein kinase (AMPK) governs metabolic through their attachment to the extracellular matrix.
adaptive reprogramming. 4EBP1, eukaryotic translation initiation factor 4Ebinding
Integrins are plasma membrane-spanning hetero
protein 1; C/EBP, CCAAT/enhancer binding protein-; CaMKK, calcium/calmodulin-
dependent protein kinase kinase-; CITED4, CBP/p300 interacting transactivator 4;
dimers comprised of and subunits that bind extra
eIF2B, eukaryotic translation initiation factor 2B; FOXO, forkhead box O; GSK3, cellular matrix components such as fibronectin, collagen
glycogen synthase kinase3; IRS1/2, insulin receptor substrate 1 or 2; LKB1, liver or laminin. The cytoplasmic tail of the subunit can
kinaseB1; PDK1, phosphoinositide-dependent protein kinase 1; PGC1, peroxisome transmit information on the basis of the composition
proliferator-activated receptor- co-activator 1; RHEB, RAS homologue enriched in brain; and stretching of the extracellular matrix through sig
RXR, retinoic acid receptor; S6K, S6 kinase; SRF, serum response factor; TAK1, transforming nalling proteins that reside within the focal adhesion
growth factor -activated kinase 1; TSC1/2, tuberous sclerosis complex 1 or 2. complex inside the cell. These integrin-associated com
plexes are composed of non-receptor tyrosine kinases,
such as focal adhesion kinase (FAK) or integrin-linked
a physiological form of cardiac hypertrophy, although kinase (ILK), which recruit signalling proteins such as
the prolongation of this signal well into adulthood the RHO GTPases, PI3K and protein kinase C (PKC).
resulted in pathological cardiac hypertrophy character Cardiac-specific ablation of Itgb1 (which encodes the
ized by increased fibrosis and decreased cardiac 1subunit) or a global deletion of Itgb3 (which encodes
function 57. By comparison, hearts overexpressing the 3 subunit) worsened hypertrophic disease induced
IGF1R (driven by MHC) displayed cardiac hyper by pressure overload67,68. However, although integrins
trophy characterized by an increase in cardiomyocyte certainly mediate stretch response signalling in fibro
volume, absence of histopathology and increased blasts and other cells, it remains unclear whether they
systolic function at 3 and 10months of age58. However, function in an analogous manner in cardiomyocytes.
Ig fr1 deletion in adult cardiomyocytes using the The cardiac Z line, which is the site of thin fila
MHC-Cre transgene, which induced recombination ment anchoring, has also been implicated in mechano
few days after birth, did not affect the baseline growth transduction and stretch sensing through a large array of
phenotype of the heart, although adult mice with this structural proteins, such as muscle LIM protein (MLP;
deletion were resistant to exercise-induced hyper also known as CSRP3), telethonin, myopalladin, palla
trophy 59,60. These results clearly indicate thecentrality din, cypher (also known as LDB3), actinin-associated
of IGF1 and its receptor in signalling the physiological LIM protein (ALP; also known as PDLIM3), car
growth of theheart. diac ankyrin repeat protein (CARP; also known as

42 | JANUARY 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Integrin

ECM Mechanical forces Ca2+

TRPCs
Sarcolemma RHO GTPases
PI3K
ILK FAK
Sarcoplasmic reticulum
ERK1/2 PKC

Obs
Actin lament
-actinin Myomesin ALP
MLP
NEB
Myosin lament Cypher
Tele
Titin

Z line Sarcomere Z line Z line

Hypertrophic response

Nucleus
Figure 2 | Stretch-mechanosensing in the initiation of physiological hypertrophy. Mechanotransduction
Nature Reviews | Molecularconverts
Cell Biology
mechanical forces into biochemical signals. Activation of transient receptor potential canonical (TRPC) channels by
stretch leads to calcium influx and the activation of hypertrophic signalling pathways. Changes in the extracellular matrix
(ECM) are sensed by integrins that signal through RHO GTPases, integrin-linked kinase (ILK), focal adhesion kinase (FAK),
protein kinase C (PKC) and the pro-hypertrophic PI3K and ERK1/2. Stretch is also sensed within the sarcomeres at the
Zline by proteins such as muscle LIM protein (MLP), actinin-associated LIM protein (ALP), nebulette (NEB),
cypher,telethonin (Tele), obscurin (Obs) and the giant protein titin that senses both strain and stretch.

ANKRD1), ankyrin, nebulette and obscurin (FIG.2). PI3KPTEN as a nodal signalling integrator. In response
Deficiencies or mutations in some of these proteins to insulin and IGF1 signal reception at the sarcolemma,
lead to cardiomyopathy 69,70. Titin is another interest signals converge on a common effector, PI3K. PI3Ks
ing candidate for a role in mechanotransduction, as it are heterodimeric lipid kinases localized at the plasma
spans from Z line to Z line (where it interacts with other membrane, where they catalyse the formation of phos
sarcomeric proteins), provides direct structural support phatidylinositol3,4,5trisphosphate (PtdIns(3,4,5)P3).
by regulating the distention of sarcomeres and binds Once synthesized, PtdIns(3,4,5)P3 recruits cytosolic
well over 20 proteins with roles in stretchspring sensing70 effectors through their pleckstrin homology (PH)
(FIG.2). Moreover, mutations in titin in humans also domains. PtdIns(3,4,5)P3 is inactivated by lipid phos
cause dilated cardiomyopathy 71. Thus, there are multi phatases such as PTEN, which acts as an endogenous
ple systems in place that could regulate physiological PI3K inhibitor 45. PI3Ks are subdivided into three classes
hypertrophy through a direct load-sensing mechanism based on sequence homologies of the catalytic domains
within the cardiomyocyteitself. and substrate specificity. Among them, class Ia PI3Ks
(PI3K, PI3K and PI3K) are especially important for
Mediators of physiological hypertrophy physiological hypertrophy because they are activated
Many of the growth-inducing hormones, such as T3, by the IR and IGF1R, and integrins. Class Ia PI3Ks are
insulin or IGF1, and the stretch-sensitive signalling path heterodimers composed of a regulatory subunit (p85
ways, which are possibly initiated by integrins, TRPCs or or p85, or their truncated splice variants p50 or p55,
sarcomeric Zline proteins, converge on a finite number respectively) and a catalytic subunit (p110, p110 or
Stretchspring sensing of intracellular signalling pathways. This enables them p110)45. Overexpression of a constitutively active p110
Translation of changes in the
to directly transduce compensated or physiological in the mouse heart under the control of the MHC
cardiomyocyte extracellular
environment (stretch) and the growth signals into changes in gene expression, protein promoter produced physiological growth of the heart 72.
sarcomeres elasticity (spring) turnover, protein degradation and RNA processing to Conversely, dominant negative p110 overexpression
into biochemical hypertrophic directly mediate the growth characteristics of the heart in the heart induced a non-pathological atrophy and
signals. (FIG.1). In addition, select microRNAs have been identi repressed growth induced by IGF1R overexpression
Sarcolemma
fied that are likely to directly regulate the physiological and exercise training 58,72,73. These results were later con
Specialized plasma membrane growth response of the myocardium. This area and the firmed by gene targeting. Muscle-specific deletion of
of a myocyte. relevant references are discussed in BOX3. PI3K regulatory subunit 1 (Pik3r1; which encodes p85)

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JANUARY 2013 | 43

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 3 | MicroRNAs and physiological cardiac hypertrophy


membrane-targeted myrAKT1 in the mouse heart for
2weeks induced a reversible physiological hypertrophy,
MicroRNAs (miRNAs) are highly conserved, small, non-coding RNAs of 1825 whereas sustained AKT1 expression for 6weeks caused
nucleotides that generally downregulate gene or protein expression post- heart failure87. By comparison, nuclear targeting of
transcriptionally. With respect to adaptive growth regulation, the muscle-specific AKT1 did not affect heart growth but did induce hyper
miRNAs miR1 and miR133 are downregulated in adult animal models of
plasia88,89. Mechanistically, AKT1 directly promotes
physiological cardiac hypertrophy116. Hypertensive rats undergoing exercise training
show reduced muscle levels of miR16 and miR21 (which target vascular endothelial
protein translation, in part by inhibiting GSK3 activ
growth factor (VEGF) and BCL2, respectively) but normal levels of miR126 (which ity, which negatively regulates eukaryotic translation ini
targets PI3K)117. However, other studies suggest that miRNAs might be important tiation factor 2B (eIF2B)45,90,91 (FIG.1). Consistent with
regulators of postnatal heart growth. For example, miR195, a member of the miR15 these observations, overexpression of GSK3 in the adult
family, participates in the postnatal mitotic arrest of cardiomyocytes by heart blocked postnatal hypertrophy before ultimately
downregulating cell cycle genes118. Transgenic mice overexpressing miR195 under inducing heart failure92. As a final mechanism, AKT was
the control of the myosin heavy chain (MHC) promoter had hypoplastic hearts shown to inhibit the activity of forkhead box proteinO3
associated with downregulation of cell cycle genes that included the checkpoint (FOXO3), which results in reduced general protein
kinase 1 (REF.118). Similarly, the overexpression of miRNAs targeting thyroid hormone turnover and catabolism mediated by the ubiquitin
signalling, such as miR208a, negatively regulates MHC expression through thyroid
ligases atrogin 1 (also known as FBXO32) and muscle-
hormone receptor-associated protein 1 (THRAP1; also known as MED13)119,120.
Finally,miR27a silences the thyroid hormone 1 receptor, thereby negatively
specific RING finger protein 1 (MURF1; also known as
regulating MHC gene expression121. Thereare undoubtedly other miRNAs that will TRIM63), and hence favours the net protein accumula
be uncovered as regulators of physiological hypertrophy. tion needed for hypertrophy 93. Altogether these results
indicate that short-term AKT activation promotes an
adaptive cellular growth programme, whereas sustained
in a Pik3r2 (which encodes p85)-null background or AKT signalling leads to pathological hypertrophy and
cardiac-specific deletion of Pik3ca (which encodes heart failure. Thus, the duration of a signal or its fre
p110) confirmed that class Ia PI3Ks are essential for quency is likely to be crucial in determining the overall
physiological growth of the heart 74,75. Finally, cardiac- beneficial versus detrimental effect on theheart.
restricted Pten deletion promoted heart growth at the
organ and cellular levels76. Thus, PI3K and PTEN are mTOR increases mRNA translation in physiological
central signalling integrators of the physiological growth hypertrophy. mTOR is part of two distinct serine/
response. threonine kinase complexes, mTOR complex 1
(mTORC1) and mTORC2, the latter of which is not
AKT directly promotes protein translation. AKT is a sensitive to rapamycin. Both complexes regulate cell
serine/threonine protein kinase, the activity of which growth and survival, and have also been implicated in
is primarily regulated by PtdIns(3,4,5)P3mediated controlling adaptive growth of the heart. mTORC1 con
membrane recruitment and by PDK1 activity. PDK1 tains mTOR, regulatory associated protein of mTOR
is a kinase that translocates to the membrane follow (RAPTOR) and G protein subunit-like (GL; also
ing PtdIns(3,4,5)P3 synthesis, where it activates AKT, known as LST8). mTORC1 is activated following AKT-
atypical PKCs and the ribosomal p70 S6 kinase (S6K). mediated inhibition of tuberous sclerosis complex 1
PI3Kdependent PDK1 activation leads to direct AKT (TSC1) and TSC2, the latter of which functions as a
activation through phosphorylation of this kinase by GTPase-activating protein for RAS homologue enriched
PDK1 at Thr308 (REFS45,77). Heart mass and individ in brain (RHEB), which then directly activates mTORC1
ual cardiomyocyte volumes were significantly reduced (REF.45) (FIG.1). mTORC1 can also be activated by amino
in mice null for Pdk1 specifically in the heart, which acids and inhibited by AMP-activated protein kinase
suggests a role for this kinase during postnatal develop (AMPK). Once activated, mTORC1 promotes ribosomal
ment 78. There are three genes encoding AKT Akt1, protein production through direct regulation of S6Ks
Akt2 and Akt3 although Akt1 and Akt2 are the main and by inhibiting eIF4Ebinding protein 1 (4EBP1),
cardiac isoforms45,79. Global loss of Akt2 in gene-targeted which then allows unrestrained cap-dependent trans
mice produced insulin resistance with no other discerni lation by eIF4E45. mTORC2 may also control adaptive
ble phenotype80. However, Akt1null mice displayed gen growth through its ability to directly phosphorylate
eral growth impairment and, remarkably, were refractory and activate AKT, which then secondarily leads to the
to physiological cardiac hypertrophy in response to activation of mTORC1.
swimming training 8183. Similarly, overexpression of a mTOR signalling has been explored in the heart. For
dominant negative AKT1 mutant (AKT1K179M) in the example, pharmacologic inhibition of mTORC1 with
heart prevented hypertrophic growth84. Cardiac over rapamycin reverses cardiac hypertrophy induced by
expression of a constitutively active AKT1 mutant AKT overexpression84,87. Similarly, deletion of Tsc1 in the
(either AKT1T308D/S473D or AKT1E40) or a membrane- heart induced early neonatal cardiac hypertrophy, lead
localized AKT1 mutant (myrAKT1) initially promoted ing to heart failure that could be delayed by rapamycin
hypertrophy with physiological characteristics in young treatment 94. RHEB overexpression invitro induced
animals, although prolongation of such a signal was ulti a 4EBP1dependent hypertrophy that was probably
mately pathological8486. Indeed, the magnitude of AKT1 adaptive, given the lack of fetal gene re-expression95.
activation and its cellular location are crucial determi However, overexpression of a dominant negative mTOR
nants of its cellular effects. Conditional expression of the in the heart did not inhibit the hypertrophic response

44 | JANUARY 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

to swimming exercise in mice96. Moreover, cardiac- with ageing 105. These lines of evidence suggest that
specific deletion of Mtor or Rptor resulted in heart fail ERK1/2 signalling provides a crucial component to the
ure without an initial phase of hypertrophy 97,98. Just as adaptive hypertrophic response that is protective, and it
perplexing, deletion or overexpression of S6K had no certainly protects cardiomyocytes from death following
effect in various models of physiological hypertrophy ischaemic injury 106.
in the mouse99. Taken together, these results underscore It thus appears that physiological hypertrophy is
the complexity associated with mTOR signalling in the triggered by a restricted number of intracellular sig
heart, such that pharmacologic inhibition with rapa nalling pathways centred around the nodal mediators
mycin can blunt physiological hypertrophy, but dele PI3KPTEN, AKT and ERK1/2, which regulate the tran
tion of mTOR or other pathway regulators gives rise to scription of a specific set of genes (for example, C/EBP
cardiac dilatation and pathology. These findings suggest controls an exercise-specific transcriptome) and increase
a crucial role for mTOR in basic heart homeostasis, as protein translation throughmTOR.
well as in adaptive hypertrophy.
AMPK in metabolic reprogramming
C/EBP regulates physiological hypertrophy. CCAAT/ The coordination of cardiac growth with metabo
enhancer binding protein (C/EBP) is a transcription lism is essential for an adaptive response. AMPK is
factor that controls cellular proliferation in many cell described as the nodal point energy sensor in all cells
types through changes in gene expression100. C/EBP that coordinates increases in metabolic output with
was identified as a factor specifically downregulated by nutrient utilization (FIG.1). AMPK is a heterotrimeric
exercise training in a differential screen for physiologi protein kinase composed of three subunits: the catalytic
cal and pathological cardiac hypertrophy effectors101. subunit, the subunit that links the and subu
Cardiomyocytes in which C/EBP was knocked down nits and binds glycogen, and the subunit that binds
by siRNA or mice in which Cebpb was deleted hetero AMP, ADP or ATP in a mutually exclusive manner 107.
zygously showed changes in gene expression profiles AMP binding to AMPK leads to its activation through
that mimic those seen in response to exercise. Cebpb- conformational changes that allow phosphorylation of
heterozygous mice displayed greater cardiomyocyte the subunit by the upstream kinases liver kinase B1
proliferation at baseline, as if they had been exercised, (LKB1; also known as STK11), calcium/calmodulin-
and were protected from pathological stress stimulation. dependent protein kinase kinase (CaMKK; also
Mechanistically, C/EBP functions downstream of known as CaMKK2) andtransforming growth factor
AKT1 to inhibit CBP/p300interacting transactivator4 activated kinase 1 (TAK1; also known as MAP3K7).
(CITED4)-induced proliferation. C/EBP is also pro Falling energy levels lead to AMPK activation by AMP
posed to compete with serum response factor (SRF) to that favours ATP production by stimulating fatty acid
regulate a transcriptome specific to exercise training 101. oxidation, glucose uptake and glycolysis, as well as by
This study shows that C/EBP is another important reducing ATP-dependent processes such as transcrip
regulator of an adaptive or physiological hypertrophy tion and protein synthesis107. AMPK is therefore crucial
response: downregulation of C/EBP permits the to cardiac homeostasis, and most studies suggest that
expression of beneficial or protective genes and allows long-term inhibition of AMPK exacerbates pathological
the expression of genes that enhance cardiomyocyte pro hypertrophy, leading to heart failure, whereas intermit
liferation and hence cell number in the heart. Thus, the tent AMPK activation could be cardioprotective. For
inhibition of C/EBP activity could be used to possibly example, mouse hearts null for Adipoq (an adipose
revitalize the damaged or cardiomyopathicheart. tissue-secreted hormone), in which AMPK activity
was decreased, were sensitized to pressure overload-
ERK1/2 in physiological hypertrophy. ERK1 and ERK2 induced hypertrophy 108. Mice null for Prkaa2, which
(ERK1/2; also known as MAPK3 and MAPK1, respec encodes AMPK2, developed an exacerbated pathologi
tively) are activated in the heart or cultured cardiomyo cal response to hypertrophic stimuli109,110. Interestingly,
cytes by a wide array of stress stimuli, some of which, deletion of Stk11 (which encodes LKB1) in the heart
such as growth factors and stretching, are consistent led to reduced ventricular cardiomyocyte size in young
with physiological growth. Indeed, overexpression of animals, suggesting a positive role of AMPK in permit
an activated MEK1 mutant protein in the heart, driven ting proper postnatal cardiac growth111,112. However,
by the MHC promoter, induced constitutive ERK1/2 loss of AMPK activity in older animals induced hyper
signalling; this produced a stable form of concentric car trophy that was normalized by a constitutively active
diac hypertrophy without fibrosis and with a significant AMPK mutant or with rapamycin treatment 112. With
enhancement in cardiac function that was associated respect to AMPK activation, treatment with the AMPK
with protection from ischaemiareperfusion injury to activator metformin improved cardiac function fol
the heart 102,103. Consistent with these observations, inhi lowing ischaemiareperfusion injury in mice through
bition of ERK1/2by overexpression of their dedicated a mechanism involving activation of endothelial nitric
phosphatase, dual specificity phosphatase 6 (DUSP6), oxide synthase (eNOS) and the mitochondrial tran
resulted in more cardiac dilation on stress stimulation104. scription factor peroxisome proliferator-activated
Similar results were observed in mice with combined receptor- co-activator 1 (PGC1)113. Collectively,
disruption of Mapk3 and Mapk1 in the heart: such mice these studies underscore the importance of metabolic
showed cardiac failure with stress stimulation, or even reprogramming during the hypertrophic response to

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JANUARY 2013 | 45

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

ensure proper adaptation during physiological heart were unable to grow in width (cross-sectional area)
growth. Thus, enhancing AMPK activity with specific and instead defaulted to a molecular programme of
pharmacologic agents could be an attractive therapeutic myocyte lengthening that led to heart failure105. With
avenue to explore for treating pathological conditions respect to medical relevance, the collective work in the
of theheart. field suggests that the activation of adaptive or protective
factors, even if they induce cardiac hypertrophy, could
Conclusions be a useful strategy in patients with heart failure. For
The concept that cardiac hypertrophy is deleterious example, selective induction of AMPK activity should
largely pertains to insults that are known to result in loss be protective and help to restore energy balance to the
of cardiovascular homeostasis or proper gene function. cardiomyocyte to support adaptive hypertrophy. Several
However, in the absence of a pre-existing condition, the pharmaceutical companies have programmes devoted
heart can clearly grow to a limited extent in response to to the development and application of selective AMPK
physiological stimuli without an increase in morbidity activators with just such a goal (cellular protection and
or mortality. Moreover, the heart can increase in size supportive adaptive growth). Moreover, intermittent or
by two- to threefold solely as a result of cellular hyper controlled activation of AKT, PI3K, ERK1/2 or mTOR
trophy during postnatal development. Physiological could be protective in supporting cardiac growth and
hypertrophy could therefore be considered as a way cardiomyocyte viability to prevent a further decline in
to prevent cardiac dysfunction and failure. Indeed, functional performance. However, a temporal compo
exercise conditioning by itself reversed or delayed the nent to such regulation might also be crucial. Indeed,
onset of disease and extended lifespan in mouse models physiological hypertrophy induced by exercise, and
of desmin-related cardiomyopathy or hypertrophic even by pregnancy, induces transient increases in car
cardiomyopathy 114,115. Studies in animal models largely diac load that are more phasic compared with sustained
support the selective hypothesis that failure of the heart overload signals associated with pathological signalling
to mount a compensated hypertrophy response through (BOX2). For example, it may be desirable to activate PI3K,
select nodal regulators of adaptive hypertrophic signal AKT, ERK1/2 or AMPK for only a few hours each day to
ling ultimately leads to pathology and potentially heart induce adaptive signals associated with exercise, which
failure. Moreover, one might even postulate that loss of might selectively augment myocardial growth and rein
adaptive hypertrophic ability and the underlying homeo vigorate metabolic functioning, while at the same time
static hypertrophic pathways that support cardiomyocyte antagonizing ongoing cell death. Thus, the challenge
cross-sectional area growth is a molecular event that moving forward will be to properly harness the molecu
directly promotes dilation and possibly heart failure. For lar effectors that program beneficial or physiological
example, deletion of Mapk1 and Mapk3 from the mouse cardiac growth in a controlled manner in an attempt to
heart produced a phenotype in which cardiomyocytes reinvigorate the failingheart.

1. Bernardo,B.C., Weeks,K.L., Pretorius,L. & 12. Henschen, S. Skilanglauf and skiwettlauf: ein 22. Sarquella-Brugada, G. et al. Genetics of sudden
McMullen,J.R. Molecular distinction between medizinische sportstudie. Mitt. Med. Klin. Uppsala 2, cardiac death in children and young athletes.
physiological and pathological cardiac hypertrophy: 1518 (1899). Cardiol.Young 24, 115 (2012).
experimental findings and therapeutic strategies. 13. Nishimura,T., Yamada,Y. & Kawai,C. 23. Patten,I.S. etal. Cardiac angiogenic imbalance leads
Pharmacol. Ther. 128, 191227 (2010). Echocardiographic evaluation of long-term effects of to peripartum cardiomyopathy. Nature 485,
2. Heineke,J. & Molkentin,J.D. Regulation of cardiac exercise on left ventricular hypertrophy and function in 333338 (2012).
hypertrophy by intracellular signalling pathways. professional bicyclists. Circulation 61, 832840 24. Heiss,H.W. etal. Studies on the regulation of
Nature Rev. Mol. Cell Biol. 7, 589600 (2006). (1980). myocardial blood flow in man. I.: Training effects on
3. Van Berlo,J.H., Maillet,M. & Molkentin,J.D. 14. Sugishita,Y., Koseki,S., Matsuda,M., Yamaguchi,T. & blood flow and metabolism of the healthy heart at rest
Signaling effectors underlying pathologic growth and Ito,I. Myocardial mechanics of athletic hearts in and during standardized heavy exercise. Bas. Res.
remodeling of the heart. J.Clin. Invest. (in the press). comparison with diseased hearts. Am. Heart J. 105, Cardiol. 71, 658675 (1976).
4. Bergmann, O. et al. Evidence for cardiomyocyte 273280 (1983). 25. Pelliccia,A. etal. Coronary arteries in physiological
renewal in humans. Science 324, 98102 (2009). 15. Dickhuth,H.H., Reindell,H., Lehmann,M. & Keul,J. hypertrophy: echocardiographic evidence of increased
5. Kajstura, J. et al. Cardiomyogenesis in the aging and [Capacity for regression of the athletic heart]. proximal size in elite athletes. Int. J.Sports Med. 11,
failing human heart. Circulation 126, 18691881 Z.Kardiol. 74 (Suppl. 7), 135143 (1985). 120126 (1990).
(2012). 16. Schannwell,C.M. etal. Left ventricular hypertrophy 26. Laughlin,M.H., Bowles,D.K. & Duncker,D.J.
6. Dorn, G. W. The fuzzy logic of physiological cardiac and diastolic dysfunction in healthy pregnant women. Thecoronary circulation in exercise training. Am.
hypertrophy. Hypertension 49, 962970 (2007). Cardiology 97, 7378 (2002). J.Physiol. Heart Circ. Physiol. 302, H10H23
7. Porrello,E.R., Widdop,R.E. & Delbridge,L.M. Early 17. Janz,K.F., Dawson,J.D. & Mahoney,L.T. Predicting (2012).
origins of cardiac hypertrophy: does cardiomyocyte heart growth during puberty: The Muscatine Study. 27. Lopaschuk,G.D. & Jaswal,J.S. Energy metabolic
attrition programme for pathological catchup growth Pediatrics 105, e63 (2000). phenotype of the cardiomyocyte during development,
of the heart? Clin. Exp. Pharmacol. Physiol. 35, 18. Hew,K.W. & Keller,K.A. Postnatal anatomical and differentiation, and postnatal maturation.
13581364 (2008). functional development of the heart: a species J.Cardiovasc. Pharmacol. 56, 130140 (2010).
8. Hopkins,S.F.Jr, McCutcheon,E.P. & Wekstein,D.R. comparison. Birth Defects Res. B.Dev. Reprod. 28. Gertz,E.W., Wisneski,J.A., Stanley,W.C. &
Postnatal changes in rat ventricular function. Circ. Res. Toxicol. 68, 309320 (2003). Neese,R.A. Myocardial substrate utilization during
32, 685691 (1973). 19. Pluim,B.M., Zwinderman,A.H., van der Laarse,A. & exercise in humans. Dual carbon-labeled carbohydrate
9. Clubb,F.J.Jr & Bishop,S.P. Formation of binucleated van der Wall,E.E. The athletes heart. A meta-analysis isotope experiments. J.Clin. Invest. 82, 20172025
myocardial cells in the neonatal rat. An index for of cardiac structure and function. Circulation 101, (1988).
growth hypertrophy. Lab. Invest. 50, 571577 336344 (2000). 29. Abel,E.D. & Doenst,T. Mitochondrial adaptations to
(1984). 20. Ehsani,A.A., Hagberg,J.M. & Hickson,R.C. physiological versus pathological cardiac hypertrophy.
10. Eghbali,M., Wang,Y., Toro,L. & Stefani,E. Heart Rapid changes in left ventricular dimensions and Cardiovasc. Res. 90, 234242 (2011).
hypertrophy during pregnancy: a better functioning mass in response to physical conditioning and Indepth review of the mitochondrial adaptations to
heart? Trends Cardiovasc. Med. 16, 285291 deconditioning. Am. J.Cardiol. 42, 5256 physiological or pathological cardiac hypertrophic
(2006). (1978). signals. Describes how the distinct cardiac
11. Winsor,T. & Beckner,G. Hypertrophy of the heart; 21. Maron,B.J., Pelliccia,A., Spataro,A. & Granata,M. metabolic profiles associated with physiological
electrocardiographic distinction between physiologic Reduction in left ventricular wall thickness after and pathological hypertrophy are initiated by
and pathologic enlargement. Calif. Med. 82, deconditioning in highly trained Olympic athletes. specific signalling pathways: PI3K, AMPK and
151158 (1955). Br.Heart J. 69, 125128 (1993). PGC1.

46 | JANUARY 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

30. Wilkins,B.J. etal. Calcineurin/NFAT coupling 51. Boudina,S. etal. Contribution of impaired myocardial 73. McMullen,J.R. etal. Phosphoinositide
participates in pathological, but not physiological, insulin signaling to mitochondrial dysfunction and 3kinase(p110) plays a critical role for the induction
cardiac hypertrophy. Circ. Res. 94, 110118 (2004). oxidative stress in the heart. Circulation 119, of physiological, but not pathological, cardiac
31. Burgess,M.L. etal. Exercise- and hypertension- 12721283 (2009). hypertrophy. Proc. Natl Acad. Sci. USA 100,
induced collagen changes are related to left ventricular 52. Efstratiadis,A. Genetics of mouse growth. Int. J.Dev. 1235512360 (2003).
function in rat hearts. Am. J.Physiol. 270, Biol. 42, 955976 (1998). This study, along with reference 72, showed that
H151H159 (1996). 53. Sutton,J. & Lazarus,L.A. Growth hormone in PI3K regulates the physiological growth of the heart
32. Jin,H. etal. Effects of exercise training on cardiac exercise: comparison of physiological and in gain and loss of function studies. PI3K activity
function, gene expression, and apoptosis in rats. pharmacological stimuli. J.Appl. Physiol. 41, controls exercise-induced physiological cardiac
Am.J.Physiol. Heart Circ. Physiol. 279, 523527 (1976). hypertrophy but not pathological hypertrophy.
H2994H3002 (2000). 54. Baker,J., Liu,J.P., Robertson,E.J. & Efstratiadis,A. 74. Luo,J. etal. Class IA phosphoinositide 3kinase
33. Neri Serneri,G.G. etal. Increased cardiac Role of insulin-like growth factors in embryonic and regulates heart size and physiological cardiac
sympathetic activity and insulin-like growth factorI postnatal growth. Cell 75, 7382 (1993). hypertrophy. Mol. Cell. Biol. 25, 94919502 (2005).
formation are associated with physiological 55. Liu,J.P., Baker,J., Perkins,A.S., Robertson,E.J. & 75. Lu,Z. etal. Loss of cardiac phosphoinositide 3kinase
hypertrophy in athletes. Circ. Res. 89, 977982 Efstratiadis,A. Mice carrying null mutations of the p110 results in contractile dysfunction. Circulation
(2001). genes encoding insulin-like growth factor I (Igf1) and 120, 318325 (2009).
34. Bellomo,D. etal. Mice lacking the vascular type 1 IGF receptor (Igf1r). Cell 75, 5972 (1993). 76. Crackower,M.A. etal. Regulation of myocardial
endothelial growth factorB gene (Vegfb) have smaller References 54 and 55 show that IGF1 signalling is contractility and cell size by distinct PI3KPTEN
hearts, dysfunctional coronary vasculature, and essential for postnatal growth. signaling pathways. Cell 110, 737749 (2002).
impaired recovery from cardiac ischemia. Circ. Res. 56. Reiss,K. etal. Overexpression of insulin-like growth This study showed that PTEN deletion in
86, e29e35 (2000). factor1 in the heart is coupled with myocyte cardiomyocytes promotes heart growth at the
35. Karpanen,T. etal. Overexpression of vascular proliferation in transgenic mice. Proc. Natl Acad. Sci. organ and cellular level. Overexpression of
endothelial growth factorB in mouse heart alters USA 93, 86308635 (1996). dominant negative p110 downstream of PTEN
cardiac lipid metabolism and induces myocardial 57. Delaughter,M.C., Taffet,G.E., Fiorotto,M.L., normalizes the phenotype.
hypertrophy. Circ. Res. 103, 10181026 (2008). Entman,M.L. & Schwartz,R.J. Local insulin-like 77. McManus,E.J. etal. The in vivo role of PtdIns(3,4,5)P3
36. Bry,M. etal. Vascular endothelial growth factorB acts growth factor I expression induces physiologic, then binding to PDK1 PH domain defined by knockin
as a coronary growth factor in transgenic rats without pathologic, cardiac hypertrophy in transgenic mice. mutation. EMBO J. 23, 20712082 (2004).
inducing angiogenesis, vascular leak, or inflammation. FASEB J. 13, 19231929 (1999). 78. Mora,A. etal. Deficiency of PDK1 in cardiac muscle
Circulation 122, 17251733 (2010). 58. McMullen,J.R. etal. The insulin-like growth factor 1 results in heart failure and increased sensitivity to
37. Stubbe,P., Gatz,J., Heidemann,P., Muhlen,A. & receptor induces physiological heart growth via the hypoxia. EMBO J. 22, 46664676 (2003).
Hesch,R. Thyroxine-binding globulin, triiodothyronine, phosphoinositide 3kinase(p110) pathway. J.Biol. 79. Oudit,G.Y. etal. The role of phosphoinositide3
thyroxine and thyrotropin in newborn infants and Chem. 279, 47824793 (2004). kinase and PTEN in cardiovascular physiology and
children. Horm. Metab. Res. 10, 5861 (1978). 59. Kim,J. etal. Insulin-like growth factor I receptor disease. J.Mol. Cell. Cardiol. 37, 449471 (2004).
38. Hadj-Sahraoui,N., Seugnet,I., Ghorbel,M.T. & signaling is required for exercise-induced cardiac 80. Cho,H. etal. Insulin resistance and a diabetes
Demeneix,B. Hypothyroidism prolongs mitotic activity hypertrophy. Mol. Endocrinol. 22, 25312543 (2008). mellitus-like syndrome in mice lacking the protein
in the post-natal mouse brain. Neurosci. Lett. 280, In this study, IGF1R targeted deletion in adult kinase Akt2 (PKB). Science 292, 17281731
7982 (2000). cardiomyocytes did not result in any baseline (2001).
39. Morkin,E. Regulation of myosin heavy chain genes in hypertrophic phenotype in young mice. 81. Chen,W.S. etal. Growth retardation and increased
the heart. Circulation 87, 14511460 (1993). IGF1Rtargeted hearts were resistant to exercise apoptosis in mice with homozygous disruption of the
40. Arsanjani,R., McCarren,M., Bahl,J.J. & Goldman,S. induced hypertrophy. Akt1 gene. Genes Dev. 15, 22032208 (2001).
Translational potential of thyroid hormone and its 60. Moellendorf,S. etal. IGFIR signaling attenuates the 82. Cho,H., Thorvaldsen,J.L., Chu,Q., Feng,F. &
analogs. J.Mol. Cell. Cardiol. 51, 506511 (2011). age-related decline of diastolic cardiac function. Birnbaum,M.J. Akt1/PKB is required for normal
41. Kenessey,A. & Ojamaa,K. Thyroid hormone Am.J.Physiol. Endocrinol. Metab. 303, e213e222 growth but dispensable for maintenance of glucose
stimulates protein synthesis in the cardiomyocyte by (2012). homeostasis in mice. J.Biol. Chem. 276,
activating the Akt-mTOR and p70S6K pathways. 61. Patel,A. etal. Canonical TRP channels and 3834938352 (2001).
J.Biol. Chem. 281, 2066620672 (2006). mechanotransduction: from physiology to disease 83. DeBosch,B. etal. Akt1 is required for physiological
42. Kenessey,A., Sullivan,E.A. & Ojamaa,K. Nuclear states. Pflugers Arch. 460, 571581 (2010). cardiac growth. Circulation 113, 20972104 (2006).
localization of protein kinase C induces thyroid 62. Musar,A., McCullagh,K.J., Naya,F.J., Olson,E.N. 84. Shioi,T. etal. Akt/protein kinase B promotes organ
hormone receptor1 expression in the cardiomyocyte. & Rosenthal,N. IGF1 induces skeletal myocyte growth in transgenic mice. Mol. Cell. Biol. 22,
Am. J.Physiol. Heart Circ. Physiol. 290, H381H389 hypertrophy through calcineurin in association with 27992809 (2002).
(2006). GATA2 and NFATc1. Nature 400, 581585 (1999). 85. Matsui,T. etal. Phenotypic spectrum caused by
43. Belakavadi,M., Saunders,J., Weisleder,N., 63. Maroto,R. etal. TRPC1 forms the stretch-activated transgenic overexpression of activated Akt in the
Raghava,P.S. & Fondell,J.D. Repression of cardiac cation channel in vertebrate cells. Nature Cell Biol. 7, heart. J.Biol. Chem. 277, 2289622901 (2002).
phospholamban gene expression is mediated by 179185 (2005). 86. Condorelli,G. etal. Akt induces enhanced myocardial
thyroid hormone receptor1 and involves targeted 64. Spassova,M.A., Hewavitharana,T., Xu,W., contractility and cell size in vivo in transgenic mice.
covalent histone modifications. Endocrinology 151, Soboloff,J. & Gill,D.L. A common mechanism Proc. Natl Acad. Sci. USA 99, 1233312338 (2002).
29462956 (2010). underlies stretch activation and receptor activation of 87. Shiojima,I. etal. Disruption of coordinated cardiac
44. Brownsey,R.W., Boone,A.N. & Allard,M.F. TRPC6 channels. Proc. Natl Acad. Sci. USA 103, hypertrophy and angiogenesis contributes to the
Actions of insulin on the mammalian heart: 1658616591 (2006). transition to heart failure. J.Clin. Invest. 115,
metabolism, pathology and biochemical mechanisms. 65. Seth,M. etal. TRPC1 channels are critical for 21082118 (2005).
Cardiovasc. Res. 34, 324 (1997). hypertrophic signaling in the heart. Circ. Res. 105, Showed that conditional expression of AKT1 in the
45. Shiojima,I. & Walsh,K. Regulation of cardiac growth 10231030 (2009). heart for 2weeks induces a reversible physiological
and coronary angiogenesis by the Akt/PKB signaling 66. Wu,X., Eder,P., Chang,B. & Molkentin,J.D. hypertrophy while sustained AKT1 expression for
pathway. Genes Dev. 20, 33473365 (2006). TRPCchannels are necessary mediators of pathologic 6weeks causes heart failure.
Comprehensive review on the role of PI3KAKT cardiac hypertrophy. Proc. Natl Acad. Sci. USA 107, 88. Shiraishi,I. etal. Nuclear targeting of Akt enhances
signalling pathways in regulating physiological 70007005 (2010). kinase activity and survival of cardiomyocytes.
cardiac hypertrophy, cardiac contractile function This study, along with reference 65, showed that Circ.Res. 94, 884891 (2004).
and coronary angiogenesis. TRPC channels are necessary mediators of cardiac 89. Rota,M. etal. Nuclear targeting of Akt enhances
46. Araki,E. etal. Alternative pathway of insulin signalling hypertrophy. ventricular function and myocyte contractility.
in mice with targeted disruption of the IRS1 gene. 67. Shai,S.Y. etal. Cardiac myocyte-specific excision of Circ.Res. 97, 13321341 (2005).
Nature 372, 186190 (1994). the 1 integrin gene results in myocardial fibrosis and 90. Haq,S. etal. Glycogen synthase kinase3 is a
47. Burks,D.J. etal. IRS2 pathways integrate female cardiac failure. Circ. Res. 90, 458464 (2002). negative regulator of cardiomyocyte hypertrophy.
reproduction and energy homeostasis. Nature 407, 68. Johnston,R.K. etal. 3 integrin-mediated J.Cell. Biol. 151, 117130 (2000).
377382 (2000). ubiquitination activates survival signaling during 91. Antos,C.L. etal. Activated glycogen synthase3
48. Belke,D.D. etal. Insulin signaling coordinately myocardial hypertrophy. FASEB J. 23, 27592771 suppresses cardiac hypertrophy in vivo. Proc. Natl
regulates cardiac size, metabolism, and contractile (2009). Acad. Sci. USA 99, 907912 (2002).
protein isoform expression. J.Clin. Invest. 109, 69. Cox,L., Umans,L., Cornelis,F., Huylebroeck,D. & 92. Michael,A. etal. Glycogen synthase kinase3
629639 (2002). Zwijsen,A. A broken heart: a stretch too far: an regulates growth, calcium homeostasis, and diastolic
Shows that insulin signalling controls postnatal overview of mouse models with mutations in stretch- function in the heart. J.Biol. Chem. 279,
cardiac growth. Cardiomyocyte-specific IR knockout sensor components. Int. J.Cardiol. 131, 3344 2138321393 (2004).
was shown to result in cardiomyocytes with a (2008). 93. Skurk,C. etal. The FOXO3a transcription factor
reduced volume, and postnatal contractile and 70. Linke,W.A. Sense and stretchability: the role of titin regulates cardiac myocyte size downstream of AKT
metabolic switches were altered. and titin-associated proteins in myocardial stress- signaling. J.Biol. Chem. 280, 2081420823 (2005).
49. Hu,P. etal. Minimally invasive aortic banding in mice: sensing and mechanical dysfunction. Cardiovasc. Res. 94. Malhowski,A.J. etal. Smooth muscle protein-
effects of altered cardiomyocyte insulin signaling 77, 637648 (2008). 22mediated deletion of Tsc1 results in cardiac
during pressure overload. Am. J.Physiol. Heart Circ. 71. Herman,D.S. etal. Truncations of titin causing dilated hypertrophy that is mTORC1mediated and reversed
Physiol. 285, H1261H1269 (2003). cardiomyopathy. N.Engl. J.Med. 366, 619628 by rapamycin. Hum. Mol. Genet. 20, 12901305
50. Sena,S. etal. Impaired insulin signaling accelerates (2012). (2011).
cardiac mitochondrial dysfunction after myocardial 72. Shioi,T. etal. The conserved phosphoinositide Showed that Tsc1 deletion results in lethal
infarction. J.Mol. Cell. Cardiol. 46, 910918 3kinase pathway determines heart size in mice. developmental and postnatal cardiac hypertrophy
(2009). EMBO J. 19, 25372548 (2000). that is reversed by rapamycin treatment.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JANUARY 2013 | 47

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

95. Wang,Y. etal. Rheb activates protein synthesis and 103. Lips,D.J. etal. MEK1ERK2 signaling pathway 114. Maloyan, A. et al. Exercise reverses preamyloid
growth in adult rat ventricular cardiomyocytes. protects myocardium from ischemic injury in vivo. oligomer and prolongs survival in B-crystallin-based
J.Mol. Cell. Cardiol. 45, 812820 (2008). Circulation 109, 19381941 (2004). desmin-related cardiomyopathy. Proc. Natl Acad. Sci.
96. Shen,W.H. etal. Cardiac restricted overexpression of 104. Purcell,N.H. etal. Genetic inhibition of cardiac ERK1/2 USA 104, 59956000 (2007).
kinase-dead mammalian target of rapamycin (mTOR) promotes stress-induced apoptosis and heart failure 115. Konhilas,J.P. etal. Exercise can prevent and reverse
mutant impairs the mTOR-mediated signaling and but has no effect on hypertrophy in vivo. Proc.Natl the severity of hypertrophic cardiomyopathy. Circ. Res.
cardiac function. J.Biol. Chem. 283, 1384213849 Acad. Sci. USA 104, 1407414079 (2007). 98, 540548 (2006).
(2008). 105. Kehat,I. etal. Extracellular signal-regulated kinases 1 116. Care,A. etal. MicroRNA133 controls cardiac
97. Zhang,D. etal. MTORC1 regulates cardiac function and 2 regulate the balance between eccentric and hypertrophy. Nature Med. 13, 613618 (2007).
and myocyte survival through 4EBP1 inhibition in concentric cardiac growth. Circ. Res. 108, 176183 117. Fernandes, T. et al. Exercise training prevents the
mice. J.Clin. Invest. 120, 28052816 (2010). (2011). microvascular rarefaction in hypertension balancing
98. Shende,P. etal. Cardiac raptor ablation impairs 106. Kehat,I. & Molkentin,J.D. Extracellular signal- angiogenic and apoptotic factors: role of
adaptive hypertrophy, alters metabolic gene regulated kinase 1/2 (ERK1/2) signaling in cardiac microRNAs16, -21, and -126. Hypertension 59,
expression, and causes heart failure in mice. hypertrophy. Ann. NY Acad. Sci. 1188, 96102 513520 (2012).
Circulation 123, 10731082 (2011). (2010). 118. Porrello,E.R. etal. MiR15 family regulates postnatal
This study, along with reference 97, shows that 107. Horman,S., Beauloye,C., Vanoverschelde,J.L. & mitotic arrest of cardiomyocytes. Circ. Res. 109,
cardiac-specific mTOR or RAPTOR ablation results Bertrand,L. AMP-activated protein kinase in the 670679 (2011).
in heart failure without an initial phase of control of cardiac metabolism and remodeling. 119. van Rooij,E. etal. Control of stress-dependent cardiac
hypertrophy. Curr.Heart Fail. Rep. 9, 164173 (2012). growth and gene expression by a microRNA. Science
99. McMullen,J.R. etal. Deletion of ribosomal S6 108. Shibata,R. etal. Adiponectin-mediated modulation of 316, 575579 (2007).
kinases does not attenuate pathological, physiological, hypertrophic signals in the heart. Nature Med. 10, 120. Callis,T.E. etal. MicroRNA208a is a regulator of
or insulin-like growth factor 1 receptor- 13841389 (2004). cardiac hypertrophy and conduction in mice. J.Clin.
phosphoinositide 3kinase-induced cardiac 109. Zarrinpashneh,E. etal. AMPK2 counteracts the Invest. 119, 27722786 (2009).
hypertrophy. Mol. Cell. Biol. 24, 62316240 (2004). development of cardiac hypertrophy induced by 121. Nishi,H. etal. MicroRNA27a regulates cardiac
100. Tsukada,J., Yoshida,Y., Kominato,Y. & Auron,P.E. isoproterenol. Biochem. Biophys. Res. Commun. 376, myosin heavy chain gene expression by targeting
The CCAAT/enhancer (C/EBP) family of basic-leucine 677681 (2008). thyroid hormone receptor 1 in neonatal rat
zipper (bZIP) transcription factors is a multifaceted 110. Zhang,P. etal. AMP activated protein kinase2 ventricular myocytes. Mol. Cell. Biol. 31, 744755
highly-regulated system for gene regulation. deficiency exacerbates pressure-overload-induced left (2011).
Cytokine54, 619 (2011). ventricular hypertrophy and dysfunction in mice.
101. Bostrom,P. etal. C/EBP controls exercise-induced Hypertension 52, 918924 (2008). Acknowledgements
cardiac growth and protects against pathological 111. Sakamoto,K. etal. Deficiency of LKB1 in heart This work was supported by grants from the US National
cardiac remodeling. Cell 143, 10721083 (2010). prevents ischemia-mediated activation of AMPK2 but Institutes of Health (J.D.M., J.H.v.B. and M.M.), and the
Shows that C/EBP is specifically downregulated by not AMPK1. Am. J.Physiol. Endocrinol. Metab. 290, Howard Hughes Medical Institute (J.D.M).
exercise training. C/EBP downregulation increases e780e788 (2006).
cardiomyocyte proliferation and leads to the 112. Ikeda,Y. etal. Cardiac-specific deletion of LKB1 leads Competing interests statement
transcription of genes specific to exercise. to hypertrophy and dysfunction. J.Biol. Chem. 284, The authors declare no competing financial interests.
102. Bueno,O.F. etal. The MEK1ERK1/2 signaling 3583935849 (2009).
pathway promotes compensated cardiac hypertrophy 113. Gundewar,S. etal. Activation of AMP-activated
in transgenic mice. EMBO J. 19, 63416350 (2000). protein kinase by metformin improves left ventricular FURTHER INFORMATION
Demonstrates that activation of MEK1ERK1/2 function and survival in heart failure. Circ. Res. 104, Jeffery D.Molkentins homepage:
signalling in the mouse heart induces a 403411 (2009). http://www.cincinnatichildrens.org/molkentinlab
non-pathological form of compensated cardiac Shows that metformin exerts its cardioprotective ALL LINKS ARE ACTIVE IN THE ONLINE PDF
hypertrophy. effects through AMPK activation.

48 | JANUARY 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved

Вам также может понравиться