Вы находитесь на странице: 1из 8

The Ne w E n g l a nd Jo u r n a l o f Me d ic i ne

Review Articles

Mechanisms of Disease the width of individual cardiac myocytes and there-


fore in concentric hypertrophy. In hypertrophic car-
diomyopathy, mutant contractile proteins lead to
F R A N K L I N H . E P S T E I N , M. D. , Editor myofibrillar disarray and secondary hypertrophy of
myocytes. In most forms of cardiac hypertrophy, there
is an increase in the expression of embryonic genes,
S IGNALING P ATHWAYS FOR C ARDIAC including the genes for natriuretic peptides and fetal
contractile proteins.3 The induction of the natriuret-
H YPERTROPHY AND F AILURE ic peptide genes is a feature of hypertrophy in all
mammalian species and is a prognostic indicator of
JOHN J. HUNTER, M.D., clinical severity. Recently, evidence of the loss of my-
AND KENNETH R. CHIEN, M.D., PH.D. ocytes as a result of programmed cell death (apop-
tosis) has also been reported in both experimental
and clinical cardiac hypertrophy (Fig. 1).

H
EART failure is a leading cause of mortality GENETIC METHODS OF STUDYING
in the United States. As a result of advances CARDIAC HYPERTROPHY AND FAILURE
in genetic technology, a molecular basis of
heart failure is emerging.1,2 This review highlights the Cardiac hypertrophy and failure are highly complex
ways in which these insights are leading to new ther- disorders that arise as a result of a combination of
apeutic targets in patients with acquired forms of genetic, physiologic, and environmental factors. The
heart failure. identification of mutations involving a single gene
that are responsible for inherited forms of hyper-
MORPHOLOGIC CLASSIFICATION trophic cardiomyopathy, dilated cardiomyopathy, and
OF CARDIAC HYPERTROPHY ventricular arrhythmogenesis (the long-QT syndrome)
Myocardial hypertrophy is an early milestone dur- has allowed us to pinpoint several of the initiating
ing the clinical course of heart failure and an impor- events that can lead to features of heart failure in hu-
tant risk factor for subsequent cardiac morbidity and mans.4,5 There is still a broad gap, however, between
mortality. In response to a variety of mechanical, he- identifying the defective gene and understanding how
modynamic, hormonal, and pathologic stimuli, the this defect leads to the cardiac abnormalities. In this
heart adapts to increased demands for cardiac work regard, in vitro assays of cardiac muscle cells and stud-
by increasing muscle mass through the initiation of ies of genetically engineered animals are beginning
a hypertrophic response. At the cellular level, cardiac to identify the points in cardiac growth signaling that
myocytes respond to biomechanical stress by initiat- cause these distinct forms of cardiac hypertrophy and
ing several different processes that lead to hypertro- failure.
phy (Fig. 1). The so-called physiologic hypertrophy Assays of Cardiac-Muscle Cells
that occurs in elite athletes is associated with pro-
portional increases in the length and width of car- The ability to culture primary cardiac myocytes
diac myocytes. By contrast, the assembly of contrac- has resulted in the availability of a well-characterized
tile-protein units in series characterizes the eccentric in vitro system in which to study the hypertrophic
hypertrophy that occurs in patients with dilated car- response. Although they are based on neonatal car-
diomyopathy, with a relatively greater increase in the diac-muscle cells, studies of these cultures have led
length than in the width of myocytes. During pres- to the identification of signaling pathways that acti-
sure overload, new contractile-protein units are as- vate cellular responses known to occur during hy-
sembled in parallel, resulting in a relative increase in pertrophy in vivo, including an increase in cell size,
an increase in the expression of embryonic genes, and
the accumulation and assembly of contractile pro-
teins.6 By altering the expression of specific genes in
cultured cardiac myocytes, peptide hormones, growth
From the University of California San Diego–Salk Institute Program in
Molecular Medicine, Department of Medicine and Center for Molecular
factors, and cytokines have been identified that can
Genetics, University of California San Diego School of Medicine, La Jolla, activate specific features of the hypertrophic response
Calif. Address reprint requests to Dr. Chien at the Department of Medi- (Table 1).7 Among the most extensively characterized
cine, 0613-C, University of California San Diego, 9500 Gilman Dr., La
Jolla, CA 92093, or at kchien@ucsd.edu. of these substances are endothelin and angiotensin
©1999, Massachusetts Medical Society. II, insulin-like growth factor I, and other growth

1276 · Octo b er 2 1 , 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at MIT LIBRARIES on October 10, 2013. For personal use only. No other uses without permission.
Copyright © 1999 Massachusetts Medical Society. All rights reserved.
M EC H A NIS MS OF D IS EASE

Apoptosis Physiologic hypertrophy

Growth7
stimuli

Normal muscle cell


Concentric hypertrophy
Increased expression7
of embryonic genes

Eccentric hypertrophy
Sarcomeric disorganization

Figure 1. Morphology of Ventricular Muscle Cells in Cardiac Hypertrophy and Failure.


Phenotypically distinct changes in the morphology of myocytes occur in response to various growth stimuli. The expression of
embryonic genes such as natriuretic peptides is increased in both eccentric and concentric hypertrophy, but not in physiologic hy-
pertrophy, in response to exercise. Myofibrillar disarray (sarcomeric disorganization) is typical of hypertrophic cardiomyopathies;
this disorganization is focal and is accompanied by more widespread increases in the cross-sectional area of myocytes.

TABLE 1. POTENTIAL THERAPEUTIC AND MOLECULAR TARGETS IN THE SIGNALING PATHWAYS INVOLVED
IN HEART FAILURE.*

GOAL TYPE OF DRUGS MOLECULAR TARGET

Inhibition of pathologic hyper- Antagonists of Gqa-dependent receptors Angiotensin II receptor


trophy Endothelin-1 receptor
? Novel receptors
Inhibitors of intracellular kinase Antagonists of ras, p38, and c-jun
cascades N-terminal kinase (JNK)
? Novel kinases
Promotion of physiologic hyper- Growth hormone Growth-hormone receptor
trophy Insulin-like growth factor I Insulin-like growth factor I receptor
Inhibition of neurohumoral over- Beta-blockers b1-Adrenergic receptor
stimulation
Enhancement of contractile and Relief of inhibition of sarcoplasmic retic- Phospholamban inhibitors
relaxation responses ulum calcium ATPase
Agents that counteract the desensitiza- Inhibitors of b-adrenergic–receptor kinase
tion of G protein–coupled receptor
kinases
Relief of energy deprivation Angiogenic growth factors Vascular endothelial-derived growth factor
Fibroblast growth factor 5
Agents involved in angiogenesis
? Others
Inhibition of pathways of apopto- Promoters of myocyte survival gp130 ligands (e.g., cardiotrophin 1)
sis of myocytes Neuregulin
Inhibitors of apoptosis Caspase inhibitors
Inhibitors of cytokines Tumor necrosis factor a, ? others

*A more complete description of each of these classes has been published.7

Vol ume 341 Numb e r 17 · 1277

The New England Journal of Medicine


Downloaded from nejm.org at MIT LIBRARIES on October 10, 2013. For personal use only. No other uses without permission.
Copyright © 1999 Massachusetts Medical Society. All rights reserved.
The Ne w E n g l a nd Jo u r n a l o f Me d ic i ne

Pressure overload

Interleukin-6 family of cytokines

gp130 LIF receptor


Cardiac myocyte

Hypertrophic signals. Apoptotic signals.


(ras, Gqa, p38b) (Gqa, p38a)

Organization of sarcomeres . Apoptosis


Increased expression of.
embryonic genes

Compensatory hypertrophy Heart failure

Figure 2. Pathways Involved in Hypertrophy, Apoptosis, and Survival of Myocytes during the Transition between Cardiac
Hypertrophy and Heart Failure in Response to Biomechanical Stress.
Biomechanical stress, such as chronic hypertension and pressure overload, activates multiple parallel and converging
signals for hypertrophy and apoptosis, which represent two distinct outcomes. At the same time, biomechanical stress
also leads to the induction of gp130-dependent ligands, such as cardiotrophin 1. This cytokine binds to its receptor,
which consists of gp130–LIF (leukemia inhibitory factor) receptor heterodimers, resulting in the activation of down-
stream gp130 pathways that block the actions of apoptotic pathways. In the absence of gp130, the response of cardiac
myocytes to biomechanical stress is shifted toward apoptosis, resulting in the loss of functional myocytes and the onset
of heart failure. Thus, the outcome of biomechanical stress is dependent on the balance between these two contradic-
tory signal-transduction pathways.

factors that activate either heteromeric (Gq) or low- Cardiac Hypertrophy and Failure
molecular-weight guanosine triphosphate (GTP)– in Genetically Altered Animals
binding protein (ras) signaling pathways, as well as Mice have a heart rate of over 500 beats per
cardiotrophin 1 and other members of the interleu- minute and an aorta that is 1 mm in diameter, but
kin-6 cytokine family that activate cellular responses they are a valid model for studying both pressure-
by means of the transmembrane signal transducer overload hypertrophy and heart failure, because of
gp130. A relatively distinct pattern of cardiac cellular the similarities of these disorders in mice and hu-
responses has been associated with each of these mans.12-14 The ability to engineer precise mutations
substances, implying that their actions are specific. in the heart, coupled with the ability to quantitate
To a certain extent, this specificity reflects the acti- the effects of these mutations on cardiac function
vation of different downstream intracellular kinase in vivo,15,16 has led to the recognition of a pre-
cascades that stimulate the appearance of specific fea- viously unsuspected set of signaling pathways and
tures of myocardial-cell hypertrophy (Fig. 2). Study molecules that stimulate specific aspects of cardiac
of these downstream signaling pathways has identi- growth. The effects of both the overexpression and
fied kinases that generate primarily hypertrophic, ap- the loss of individual cardiac genes in animals have
optotic, and anti-apoptotic signals,8-10 as well as ki- been studied, and we will describe examples of each.
nases that regulate the assembly of myofilaments (rho These models are useful not simply because they
kinase).11 In addition, nuclear signaling proteins have replicate human disease but also because they allow
been found that activate and suppress various cardiac the differentiation of the many different processes
genes during hypertrophy.7 that together cause such conditions as pressure-

1278 · Oc to b er 2 1 , 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at MIT LIBRARIES on October 10, 2013. For personal use only. No other uses without permission.
Copyright © 1999 Massachusetts Medical Society. All rights reserved.
MEC H A NIS MS OF D IS EASE

overload hypertrophy and congestive heart failure in (endothelin-126,27), angiotensin II,28,29 interleukin-6–
humans. related cytokines (cardiotrophin 130,31), and growth
In the tissue-restricted approach to overexpression, factors that activate receptor tyrosine kinases (insu-
the regulatory region from a cardiac-specific gene is lin-like growth factor I32,33).
fused to a candidate gene of interest and used to One of the first genetically defined models of con-
produce transgenic mice that express the candidate centric ventricular hypertrophy resulted from cardiac-
gene specifically in cardiac-muscle cells because of directed overexpression of the a1b-adrenergic recep-
the ability of the regulatory sequences to restrict ex- tor.34 This confirmed previous work in cultured
pression to the heart.17 The availability of well-char- cardiac myocytes demonstrating that a-adrenergic
acterized regulatory regions of cardiac-specific genes stimulation induced a hypertrophic response. a-Adre-
has allowed the expression of candidate signaling mol- nergic receptors share common intracellular signal-
ecules in the heart and even in the ventricles alone. ing pathways with other hypertrophic growth fac-
For example, transgenic mice have been produced tors, including angiotensin II and endothelin-1. In
that express an active mutant of ras, a protein that each of these pathways, signaling that results in hy-
mediates many growth-related responses in cardiac pertrophy proceeds by means of the Gqa subunit
myocytes as well as in cancer cells.18 Since the regu- of heteromeric G protein, which was found to be
latory sequences of a ventricle-specific gene control both necessary and sufficient to cause hypertrophy
the expression of this active form of ras, the heart is in cultured cardiac-muscle cells.35 Subsequently, over-
the only tissue in the animals in which the ras path- expression of Gqa itself was found to induce both
ways are activated.19,20 High levels of expression of a hypertrophic and an apoptotic response.36,37 Fur-
ras result in hypertrophic cardiomyopathy, including thermore, a protein inhibitor of Gqa, whose expres-
massive cardiac hypertrophy, heart failure, and sud- sion was also targeted to the heart by transgenic
den death, but not the dilatation of any heart cham- techniques, had no effect on cardiac structure or func-
ber (unpublished data). Increased concentrations of tion in unstressed mice, but it prevented hypertro-
ras messenger RNA were recently described in en- phy when pressure overload was induced by con-
domyocardial-biopsy specimens from humans with stricting the ascending aorta.38 Taken together, these
familial hypertrophic cardiomyopathy.21 results suggest that Gqa-dependent pathways have a
It is possible to disrupt, or “target,” a gene of inter- critical role in the development of myocardial hy-
est in a mouse by replacing it with a mutated sequence pertrophy (Fig. 2).
early in embryogenesis.22 Mice that are heterozygous The activation of cell-surface receptors and their
for the mutated allele can be mated to produce ho- immediate signaling targets, such as ras and Gqa, by
mozygous mice that do not have a functional copy cardiac growth factors is the first step in initiating the
of the targeted gene. For example, mice with dele- growth of myocytes (Fig. 2). Increases in intracellu-
tions of a muscle-restricted cytoskeletal protein have lar calcium concentrations in response to these growth
features of dilated cardiomyopathy,23 a finding that factors may also activate calmodulin-dependent path-
supports a causative role for disrupted cytoskeleton ways.39-41 According to in vitro and in vivo results,
components in the pathogenesis of cardiomyopathy. the primary downstream effectors are the mitogen-
Since many structural and signaling components of activated protein kinases, including c-jun N-terminal
cardiac myocytes are common to other tissues, gene kinase and p38.7,9,10,31 These kinases are particularly
targeting may be lethal; the animals may die from important switches in the pathways between apop-
defects in other tissues before the role of the gene tosis and adaptive hypertrophy. For example, in mice
in the heart can be studied. This difficulty can now p38 mitogen-activated protein kinases are strongly
be avoided by techniques to engineer heart-specific activated by pressure overload, and upstream kinases
gene deletions.24,25 that specifically activate p38 cause the growth of cul-
tured myocytes. However, the activation of p38 is
PRESSURE OVERLOAD AND CONCENTRIC also accompanied by an increase in the rate of apop-
HYPERTROPHY tosis.9 The two isoforms of p38, a and b, have oppo-
The extent of ventricular hypertrophy in patients site effects on apoptosis when stimulated by upstream
is a powerful predictor of adverse events. According- activators: p38a increases apoptosis, whereas p38b
ly, identifying the signals that mediate the pathways inhibits it (Fig. 2).9
from mechanical stress to downstream cellular events
has been a major area of interest. Both myocytes and CHAMBER DILATATION AND DILATED
nonmyocytes are direct biomechanical sensors of he- CARDIOMYOPATHY
modynamic load. Growth signals are generated by Dilated cardiomyopathy represents a final common
the release of growth factors and cytokines, which pathway of the myocardium in response to many dif-
lead to a regionally localized response. The factors ferent pathologic conditions. This has led to the ob-
that have been implicated in this response include vious conclusion that there are common pathways to
peptides that stimulate G protein–coupled receptors cardiac dilatation and failure. Local myocardial inju-

Vol ume 341 Numb e r 17 · 1279

The New England Journal of Medicine


Downloaded from nejm.org at MIT LIBRARIES on October 10, 2013. For personal use only. No other uses without permission.
Copyright © 1999 Massachusetts Medical Society. All rights reserved.
The Ne w E n g l a nd Jo u r n a l o f Me d ic i ne

ry can cause progressive and sometimes deleterious toskeleton and the extracellular matrix in the patho-
dilatation and thinning of the ventricular wall. genesis of cardiomyopathy. Moreover, a molecular
In about 25 percent of patients with idiopathic di- defect involved in familial dilated cardiomyopathy in
lated cardiomyopathy, the disorder is familial and ge- humans has been mapped to the cytoskeletal region
netic, and it is likely to be genetic in some nonfamil- of the cardiac actin gene.43 One of the first examples
ial cases as well.4 Indeed, the first example of familial of a genetic link between the cytoskeleton and dilat-
dilated cardiomyopathy for which the genetic basis ed cardiomyopathy was provided by studies of mice
was defined was Duchenne’s muscular dystrophy. In that have a deficiency in a muscle-specific LIM (lin-1,
this and related muscular dystrophies, the molecular ISL-1, and mec-3) domain protein23 and have many
defect is in the dystrophin–dystroglycan–laminin features of the dilated cardiomyopathy that occurs in
transmembrane complex that connects the actin cy- humans. This cytoskeletal protein may be a compo-
toskeleton of the muscle cells to structural proteins nent of a biomechanical sensor pathway that trans-
that are synthesized by fibroblasts surrounding the duces hemodynamic force into specific signaling re-
myocytes (Fig. 3). In these dystrophies, there is an sponses. Disruption of other cytoskeletal proteins,
impairment of the normal linkage by which force such as desmin, plakoglobin, and N-cadherin, results
generated by individual myocytes is translated into in cardiac dilatation and impaired cardiac function
work done by the muscle tissue as a unit, and exces- during fetal development or after birth. In summary,
sive stresses on individual myocytes cannot be spread increased biomechanical stress on cardiac myocytes,
across that muscle. either through genetic abnormalities or through ex-
The recent demonstration that the molecular de- cessive stress on the chamber wall due to myocyte loss
fect in Syrian hamsters with cardiomyopathy lies in or severe hemodynamic loading, generates a persist-
the d-sarcoglycan component of this complex42 fur- ent signal for ventricular growth and hypertrophy.2,44
ther implicates the linkage between the myocyte cy- By contrast, mutations in sarcomeric proteins cause

Extracellular matrix Laminin-2

Sarcoglycans a
Dystroglycans
d g a b
b

a-Actinin
Syntrophins
MLP Z-disk
Cytoskeletal7
? Dystrophin
a-actin7
7
Desmin7
7

Figure 3. Primary Structural Components of the Linkage between the Cytoskeleton and the Extracellular Matrix, Includ-
ing Actin, the Dystrophin–Glycoprotein Complex, and Laminin-2 (Merosin).
Genetic defects in these components lead to dilated cardiomyopathy, with or without associated skeletal myopathy.
This complex is physically associated with the Z-disk of cardiac myocytes, the Z-disk components desmin (associated
with dilated cardiomyopathy in humans and mice) and a-actinin, and a muscle-specific cytoskeletal protein (MLP) (as-
sociated with dilated cardiomyopathy in mice).23 The question mark indicates an unknown factor.

1280 · Oc to b er 2 1 , 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at MIT LIBRARIES on October 10, 2013. For personal use only. No other uses without permission.
Copyright © 1999 Massachusetts Medical Society. All rights reserved.
MECH A NIS MS OF D IS EASE

hypertrophic cardiomyopathy but do not affect ven- effects (Fig. 2).9 Cell death may occur when the ap-
tricular systolic function. optotic forces exceed a certain threshold. The acti-
vation of apoptotic signals during the hypertrophic
Apoptosis of Cardiac Myocytes response of myocytes may explain the risk of death
Apoptosis is a mechanism by which cells can be associated with ventricular hypertrophy in humans.
eliminated without an inflammatory response. Evi- In support of this concept, mice with a loss-of-func-
dence of an increased rate of apoptosis has been de- tion mutation in the cytokine receptor gp130 of the
tected in failing hearts at the time of transplantation ventricular chamber have normal cardiac structure but
in humans, as well as in hearts from animals with ex- have massive cardiac apoptosis accompanied by rap-
perimentally induced hypertrophy and cardiomyop- idly progressive dilated cardiomyopathy when sub-
athy. Unlike necrosis, apoptosis leaves little or no jected to pressure overload.25 These studies indicate
histologic trace of the lost cells. Accordingly, docu- that the inhibition of apoptosis by gp130-dependent
menting its occurrence and estimating the extent of pathways in myocytes has a critical role in the tran-
the loss of myocytes as a result of apoptosis have sition between compensatory hypertrophy and overt
been problematic; therefore, the importance of ap- heart failure and suggest that the balance between ap-
optosis in the transition from compensatory hyper- optotic and hypertrophic pathways determines wheth-
trophy to heart failure has been unclear. er chamber dilatation will occur (Fig. 2).25
At the cellular level, there is normally a balance be-
tween apoptotic and anti-apoptotic signals, and cell Cardiac Function and Contractility
death occurs in response to a persistent shift in this The b-adrenergic–receptor pathway is a critical
balance. The cytokine tumor necrosis factor a, act- point of control for cardiac contractility in both nor-
ing through its receptor, activates both apoptotic mal and failing hearts (Fig. 4). The primary func-
and anti-apoptotic signals, with a tendency toward tional disturbance in dilated cardiomyopathy is im-
promoting apoptosis. Similarly, p21 ras induces both paired contractility, yet when contractility is decreased
apoptotic c-jun N-terminal kinase and anti-apoptotic in mice by overexpression of the calcium-regulatory
1-phosphatidylinositol 3-kinase signals. Among mito- protein phospholamban, the mass and volume of the
gen-activated protein kinases, the extracellular signal– cardiac chamber are no different from those in nor-
regulated kinases tend to be anti-apoptotic, c-jun mal mice.46 Moreover, when contractility is decreased,
N-terminal kinase promotes apoptosis,8 and as men- as in mice with mutations in the myosin heavy chain,
tioned, the a and b isoforms of p38 have opposing the result is hypertrophic cardiomyopathy, without

b-Adrenergic receptor

GSa Adenylyl cyclase


Sarcoplasmic.
reticulum

Cyclic AMP. PKA P

Phospho-7
b-Adrenergic–. lamban
receptor kinase
Calcium
Calcium pump

Figure 4. Regulation of the Contractile Function of Myocytes.


The contractile function of myocytes is regulated by changes in calcium flux into and out of the sarcoplasmic reticulum.
Activation of b-adrenergic receptors leads to increased uptake of calcium into the sarcoplasmic reticulum by the calcium
pump; the phosphorylation (P) of phospholamban by cyclic AMP-dependent protein kinase (PKA) removes its tonic in-
hibition of the calcium pump. b-Adrenergic receptors are desensitized in both heart failure and maladaptive hypertro-
phy; a substantial component in this desensitization is up-regulation of the b-adrenergic–receptor kinase (bARK). A de-
ficiency of phospholamban has recently been shown to halt the progression of heart failure and dilated cardiomyopathy
in a genetically based animal model.45

Vol ume 341 Numb e r 17 · 1281

The New England Journal of Medicine


Downloaded from nejm.org at MIT LIBRARIES on October 10, 2013. For personal use only. No other uses without permission.
Copyright © 1999 Massachusetts Medical Society. All rights reserved.
The Ne w E n g l a nd Jo u r n a l o f Me d ic i ne

chamber dilatation.47 These observations, as well as the identification of targets whose actions could be
clinical and experimental studies of b-adrenergic– interrupted, thereby halting or perhaps reversing clin-
blocking drugs in patients with heart failure, suggest ical deterioration.
that impaired contractility in certain forms of dilated
cardiomyopathy may be a secondary phenomenon, REFERENCES
perhaps resulting from alterations in energy metab- 1. Chien KR, Grace AA. Principles of cardiovascular molecular and cellu-
olism or intracellular calcium handling. lar biology. In: Braunwald E, ed. Heart disease: a textbook of cardiovascu-
Animals have been developed that have increased lar medicine. 5th ed. Vol. 2. Philadelphia: W.B. Saunders, 1997:1626-49.
2. Chien KR. Stress pathways and heart failure. Cell 1999;98:555-8.
ventricular contractile function as a primary feature. 3. Chien KR, Zhu H, Knowlton KU, et al. Transcriptional regulation dur-
These include mice that overexpress b1- and b 2-adre- ing cardiac growth and development. Annu Rev Physiol 1993;55:77-95.
4. Seidman CE, Seidman JG. Molecular genetics of inherited cardiomyop-
nergic receptors48,49 or the Gsa protein to which it is athies. In: Chien KR, ed. Molecular basis of cardiovascular disease: a com-
coupled50; mice that overexpress a peptide inhibitor panion to Braunwald’s Heart Disease. Philadelphia: W.B. Saunders, 1999:
of the b-adrenergic–receptor kinase, the principal 251-63.
5. Curran ME, Sanguinetti MC, Keating MT. Molecular basis of inherited
desensitizer of b-adrenergic receptors51; and mice in cardiac arrhythmias. In: Chien KR, ed. Molecular basis of cardiovascular
which the phospholamban gene has been disrupted.52 disease: a companion to Braunwald’s Heart Disease. Philadelphia: W.B.
The increased risk of death among patients with heart Saunders, 1999:302-12.
6. Chien KR, Knowlton KU, Zhu H, Chien S. Regulation of cardiac gene
failure that is associated with chronic stimulation of expression during myocardial growth and hypertrophy: molecular studies
b-adrenergic agonists can be replicated in mice with of an adaptive physiologic response. FASEB J 1991;5:3037-46.
7. Hunter JJ, Grace AA, Chien KR. Molecular and cellular biology of car-
dilated cardiomyopathy due to a cytoskeletal gene diac hypertrophy and failure. In: Chien KR, ed. Molecular basis of heart
mutation.23 The offspring of genetic crosses between disease: a companion to Braunwald’s Heart Disease. Philadelphia: W.B.
these mice 23 and those overexpressing b 2-adrenergic Saunders, 1999:211-50.
8. Xia Z, Dickens M, Raingeaud J, Davis RJ, Greenberg ME. Opposing
receptors49 have a very high mortality rate.53 How- effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 1995;
ever, a genetic cross between the cardiomyopathic 270:1326-31.
mice 23 and those overexpressing the peptide inhibi- 9. Wang Y, Huang S, Sah VP, et al. Cardiac muscle cell hypertrophy and
apoptosis induced by distinct members of the p38 mitogen-activated pro-
tor of the b-adrenergic–receptor kinase results in a tein kinase family. J Biol Chem 1998;273:2161-8.
mouse with decreased chamber dimensions and im- 10. Wang Y, Su B, Sah VP, Brown JH, Han J, Chien KR. Cardiac hyper-
trophy induced by mitogen-activated protein kinase kinase 7, a specific ac-
proved contractile function.53 This suggests that the tivator for c-Jun NH2-terminal kinase in ventricular muscle cells. J Biol
deleterious effect of long-term exposure to inotropic Chem 1998;273:5423-6.
drugs, such as phosphodiesterase inhibitors, in pa- 11. Sah VP, Hoshijima M, Chien KR, Brown JH. Rho is required for Gal-
phaq and alpha1-adrenergic receptor signaling in cardiomyocytes: dissoci-
tients with heart failure may not be due to changes ation of Ras and Rho pathways. J Biol Chem 1996;271:31185-90.
in contractility alone.53 The different effects of over- 12. Chien KR. Genes and physiology: molecular physiology in genetically
expression of the b 2-adrenergic receptor and overex- engineered animals. J Clin Invest 1996;97:901-9.
13. Rockman HA, Ross RS, Harris AN, et al. Segregation of atrial-specific
pression of the inhibitor of the b-adrenergic–recep- and inducible expression of an atrial natriuretic factor transgene in an
tor kinase might also reflect differing downstream in vivo murine model of cardiac hypertrophy. Proc Natl Acad Sci U S A
1991;88:8277-81. [Erratum, Proc Natl Acad Sci U S A 1991;88:9907.]
effects on cardiac relaxation,54 or pathologic effects 14. Rockman HA, Ono S, Ross RS, et al. Molecular and physiological al-
of chronic b-adrenergic overstimulation48 as compared terations in murine ventricular dysfunction. Proc Natl Acad Sci U S A
with those resulting from relief of desensitization.53 1994;91:2694-8.
15. Lin MC, Rockman HA, Chien KR. Heart and lung disease in engi-
In this regard, phospholamban negatively regulates neered mice: technological miniaturization combined with the power of
the uptake of calcium by the sarcoplasmic reticulum molecular genetics makes the mouse a model animal for understanding hu-
(Fig. 4), and a deficiency of phospholamban can halt man cardiovascular and pulmonary disease. Nat Med 1995;1:749-51.
16. Christensen G, Wang Y, Chien KR. Physiological assessment of com-
progression of dilated cardiomyopathy and heart fail- plex cardiac phenotypes in genetically engineered mice. Am J Physiol 1997;
ure.54 b-Adrenergic pathways lead to the phospho- 272:H2513-H2524.
17. Hunter JJ, Zhu H, Lee KJ, Kubalak S, Chien KR. Targeting gene ex-
rylation of phospholamban, which reduces its activ- pression to specific cardiovascular cell types in transgenic mice. Hyperten-
ity and increases ATPase activity in the sarcoplasmic sion 1993;22:608-17.
reticulum. 18. Thorburn A, Thorburn J, Chen SY, et al. HRas-dependent pathways
can activate morphological and genetic markers of cardiac muscle cell hy-
pertrophy. J Biol Chem 1993;268:2244-9. [Erratum, J Biol Chem 1993;
CONCLUSIONS 268:16082.]
The decrease in cardiac performance in the failing 19. Hunter JJ, Tanaka N, Rockman HA, Ross J Jr, Chien KR. Ventricular
expression of a MLC-2v-ras fusion gene induces cardiac hypertrophy and
heart may be a consequence of alterations in specific selective diastolic dysfunction in transgenic mice. J Biol Chem 1995;270:
signaling molecules and their downstream pathways 23173-8.
in individual myocytes (Table 1). By analogy to car- 20. Gottshall KR, Hunter JJ, Tanaka N, et al. Ras-dependent pathways in-
duce obstructive hypertrophy in echo-selected transgenic mice. Proc Natl
cinogenesis, heart failure may be viewed as a pro- Acad Sci U S A 1997;94:4710-5.
gressive, multistep process involving physiologic and 21. Kai H, Muraishi A, Sugiu Y, et al. Expression of proto-oncogenes and
gene mutation of sarcomeric proteins in patients with hypertrophic cardio-
molecular initiators, promoters, suppressors, and ef- myopathy. Circ Res 1998;83:594-601.
fectors of the chronic course to heart-muscle failure.7 22. Young SG, Lusis AJ, Hammer RE. Genetically modified animal mod-
Further unraveling of the signals that cause specific els in cardiovascular research. In: Chien KR, ed. Molecular basis of cardio-
vascular disease: a companion to Braunwald’s Heart Disease. Philadelphia:
features of heart failure, coupled with the growing W.B. Saunders, 1999:37-85.
human genome data base, should ultimately lead to 23. Arber S, Hunter JJ, Ross J Jr, et al. MLP-deficient mice exhibit a dis-

1282 · Octo b er 2 1 , 19 9 9

The New England Journal of Medicine


Downloaded from nejm.org at MIT LIBRARIES on October 10, 2013. For personal use only. No other uses without permission.
Copyright © 1999 Massachusetts Medical Society. All rights reserved.
MEC H A NIS MS OF D IS EASE

ruption of cardiac cytoarchitectural organization, dilated cardiomyopathy, 38. Akhter SA, Luttrell LM, Rockman HA, Iaccarino G, Lefkowitz RJ,
and heart failure. Cell 1997;88:393-403. Koch WJ. Targeting the receptor-Gq interface to inhibit in vivo pressure
24. Chen J, Kubalak SW, Chien KR. Ventricular muscle-restricted target- overload myocardial hypertrophy. Science 1998;280:574-7.
ing of the RXRalpha gene reveals a non-cell-autonomous requirement in 39. Gruver CL, DeMayo F, Goldstein MA, Means AR. Targeted develop-
cardiac chamber morphogenesis. Development 1998;125:1943-9. mental overexpression of calmodulin induces proliferative and hypertrophic
25. Hirota H, Chen J, Betz UAK, et al. Loss of a gp130 cardiac muscle growth of cardiomyocytes in transgenic mice. Endocrinology 1993;133:
cell survival pathway is a critical event in the onset of heart failure during 376-88.
biomechanical stress. Cell 1999;97:189-98. 40. Molkentin JD, Lu JR, Antos CL, et al. A calcineurin-dependent tran-
26. Shubeita HE, McDonough PM, Harris AN, et al. Endothelin induc- scriptional pathway for cardiac hypertrophy. Cell 1998;93:215-28.
tion of inositol phospholipid hydrolysis, sarcomere assembly, and cardiac 41. Mende U, Kagen A, Cohen A, Aramburu J, Schoen FJ, Neer EJ. Tran-
gene expression in ventricular myocytes: a paracrine mechanism for myo- sient cardiac expression of constitutively active Galphaq leads to hypertro-
cardial cell hypertrophy. J Biol Chem 1990;265:20555-62. phy and dilated cardiomyopathy by calcineurin-dependent and independ-
27. Yamazaki T, Komuro I, Kudoh S, et al. Endothelin-1 is involved in me- ent pathways. Proc Natl Acad Sci U S A 1998;95:13893-8.
chanical stress-induced cardiomyocyte hypertrophy. J Biol Chem 1996; 42. Nigro V, Okazaki Y, Belsito A, et al. Identification of the Syrian ham-
271:3221-8. ster cardiomyopathy gene. Hum Mol Genet 1997;6:601-7.
28. Sadoshima J, Izumo S. Molecular characterization of angiotensin 43. Olson TM, Michels VV, Thibodeau SN, Tai YS, Keating MT. Actin
II-induced hypertrophy of cardiac myocytes and hyperplasia of cardiac fi- mutations in dilated cardiomyopathy, a heritable form of heart failure. Sci-
broblasts: critical role of the AT1 receptor subtype. Circ Res 1993;73:413- ence 1998;280:750-2.
23. 44. Chen J, Chien KR. Complexity in simplicity: monogenic disorders and
29. Idem. The heterotrimeric G q protein-coupled angiotensin II receptor complex cardiomyopathies. J Clin Invest 1999;103:1483-5.
activates p21 ras via the tyrosine kinase-Shc-Grb2-Sos pathway in cardiac 45. Minamisawa S, Hoshijima M, Ward CA, et al. Genetic complementa-
myocytes. EMBO J 1996;15:775-87. tion identifies phospholamban–sarcoplasmic reticulum Ca++ ATPase inter-
30. Wollert KC, Taga T, Saito M, et al. Cardiotrophin-1 activates a distinct action as a critical Ca++ cycling defect that drives the progression of dilated
form of cardiac muscle cell hypertrophy: assembly of sarcomeric units in cardiomyopathy. Cell (in press).
series VIA gp130/leukemia inhibitory factor receptor-dependent pathways. 46. Kadambi VJ, Ponniah S, Harrer JM, et al. Cardiac-specific overexpres-
J Biol Chem 1996;271:9535-45. sion of phospholamban alters calcium kinetics and resultant cardiomyocyte
31. Sheng Z, Knowlton K, Chen J, Hoshijima M, Brown JH, Chien KR. mechanics in transgenic mice. J Clin Invest 1996;97:533-9.
Cardiotrophin 1 (CT-1) inhibition of cardiac myocyte apoptosis via a mi- 47. Geisterfer-Lowrance AA, Christe M, Conner DA, et al. A mouse
togen-activated protein kinase-dependent pathway: divergence from down- model of familial hypertrophic cardiomyopathy. Science 1996;272:731-4.
stream CT-1 signals for myocardial cell hypertrophy. J Biol Chem 1997; 48. Engelhardt S, Hein L, Wiesmann F, Lohse MJ. Progressive hypertro-
272:5783-91. phy and heart failure in beta1-adrenergic receptor transgenic mice. Proc
32. Ito H, Hiroe M, Hirata Y, et al. Insulin-like growth factor-I induces Natl Acad Sci U S A 1999;96:7059-64.
hypertrophy with enhanced expression of muscle specific genes in cultured 49. Milano CA, Allen LF, Rockman HA, et al. Enhanced myocardial func-
rat cardiomyocytes. Circulation 1993;87:1715-21. tion in transgenic mice overexpressing the beta 2-adrenergic receptor. Sci-
33. Duerr RL, Huang S, Miraliakbar HR, Clark R, Chien KR, Ross J Jr. ence 1994;264:582-6.
Insulin-like growth factor-1 enhances ventricular hypertrophy and function 50. Iwase M, Bishop SP, Uechi M, et al. Adverse effects of chronic endog-
during the onset of experimental cardiac failure. J Clin Invest 1995;95: enous sympathetic drive induced by cardiac GS alpha overexpression. Circ
619-27. Res 1996;78:517-24.
34. Milano CA, Dolber PC, Rockman HA, et al. Myocardial expression 51. Koch WJ, Rockman HA, Samama P, et al. Cardiac function in mice
of a constitutively active alpha 1B-adrenergic receptor in transgenic mice overexpressing the beta-adrenergic receptor kinase or a beta ARK inhibitor.
induces cardiac hypertrophy. Proc Natl Acad Sci U S A 1994;91:10109-13. Science 1995;268:1350-3.
35. LaMorte VJ, Thorburn J, Absher D, et al. Gq- and ras-dependent 52. Luo W, Grupp IL, Harrer J, et al. Targeted ablation of the phospho-
pathways mediate hypertrophy of neonatal rat ventricular myocytes follow- lamban gene is associated with markedly enhanced myocardial contractility
ing alpha 1-adrenergic stimulation. J Biol Chem 1994;269:13490-6. and loss of beta-agonist stimulation. Circ Res 1994;75:401-9.
36. D’Angelo DD, Sakata Y, Lorenz JN, et al. Transgenic Galphaq over- 53. Rockman HA, Chien KR, Choi DJ, et al. Expression of a beta-adre-
expression induces cardiac contractile failure in mice. Proc Natl Acad Sci nergic receptor kinase 1 inhibitor prevents the development of myocardial
U S A 1997;94:8121-6. failure in gene-targeted mice. Proc Natl Acad Sci U S A 1998;95:7000-5.
37. Adams JW, Sakata Y, Davis MG, et al. Enhanced Galphaq signaling: a 54. Xiao RP, Avdonin P, Zhou YY, et al. Coupling of beta2-adrenoceptor
common pathway mediates cardiac hypertrophy and apoptotic heart failure. to Gi proteins and its physiological relevance in murine cardiac myocytes.
Proc Natl Acad Sci U S A 1998;95:10140-5. Circ Res 1999;84:43-52.

Vol ume 341 Numb e r 17 · 1283

The New England Journal of Medicine


Downloaded from nejm.org at MIT LIBRARIES on October 10, 2013. For personal use only. No other uses without permission.
Copyright © 1999 Massachusetts Medical Society. All rights reserved.

Вам также может понравиться