You are on page 1of 14

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

Review article

Mechanisms of Disease
Robert S. Schwartz, M.D., Editor

Inherited Cardiomyopathies
Hugh Watkins, M.D., Ph.D., Houman Ashrafian, B.M., B.Ch., D.Phil.,
and Charles Redwood, Ph.D.

I
nherited cardiomyopathies are a major cause of heart disease in From the Department of Cardiovascular
all age groups, often with an onset in adolescence or early adult life. Not only the Medicine (H.W., H.A., C.R.) and the Well-
come Trust Centre for Human Genetics
patients but also their families can be severely burdened by these illnesses. More (H.W.), University of Oxford, Oxford,
than 20 years ago, the first disease gene for hypertrophic cardiomyopathy was United Kingdom. Address reprint re-
identified.1,2 This finding led to the concept that hypertrophic cardiomyopathy is a quests to Dr. Watkins at the Department
of Cardiovascular Medicine, West Wing,
disease of the sarcomere.3 Similar advances in the elucidation of the genetic basis of John Radcliffe Hospital, Oxford OX3 9DU,
other forms of cardiomyopathy, as well as in other inherited cardiovascular diseases, United Kingdom, or at hugh.watkins@
soon followed. cardiov.ox.ac.uk.

The identification of disease genes in numerous inherited diseases has raised N Engl J Med 2011;364:1643-56.
expectations for new forms of treatment, but experience has shown that such novel Copyright 2011 Massachusetts Medical Society.

therapies rarely follow.4 For some inherited cardiomyopathies, however, there are
realistic prospects that molecular insights will soon lead to novel treatments. This
review focuses on recent findings regarding the mechanisms underlying cardio-
myopathies that will inform clinical practice and guide the search for therapeutic
targets.

Cl a ssific at ion of Inher i ted C a r diom yopathie s

The long-standing classification of inherited cardiomyopathies according to func-


tional and morphologic features is crude yet clinically useful. Despite considerable
heterogeneity within the categories of hypertrophic, dilated, restrictive, arrhythmo-
genic right ventricular, and other types of cardiomyopathies, these diagnostic clas-
sifications can predict major complications and delineate treatment options for each
group. Finer resolution of these categories is possible with the aid of molecular
genetics, which can identify clinically significant subtypes. Molecular insights, how-
ever, do not supersede the clinical classification,5,6 since different mutations within
the same gene can underlie different disorders (Fig. 1). Mutations that affect adjacent
amino acids in the -myosin heavy chain, for example, cause either hypertrophic
cardiomyopathy or dilated cardiomyopathy.7 All the inherited cardiomyopathies are
genetically heterogeneous; within each category there are multiple disease genes, and
many different mutations, each of which is uncommon. Nevertheless, technical ad-
vances now allow routine genetic testing of families.7 The degree of genetic hetero-
geneity varies among the cardiomyopathies and determines the extent to which a fi-
nal common pathway of pathogenesis can be identified for each condition.

H y per t rophic C a rdiom yopath y, a dise a se


of The S a rc omer e

Hypertrophic cardiomyopathy is an autosomal dominant disease characterized by


unexplained hypertrophy of the left ventricle (and sometimes of the right ventricle),

n engl j med 364;17 nejm.org april 28, 2011 1643


The New England Journal of Medicine
Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Hypertrophic Dilated
cardiomyopathy cardiomyopathy Arrhythmogenic right
ventricular cardiomyopathy
Mendelian genetic Genetic Genetic
Sarcomeric Sarcomeric, lamin A/C Desmosome proteins
AMPK Dystrophin complex, TMEM43
Sarcomeric
LAMP2, GLA Z-disk, and others
Mitochondrial
Acquired
Nonmendelian genetic Nutritional
Toxins and drugs
Myocarditis
Endocrine
Autoimmune
Peripartum

Restrictive Left ventricular


cardiomyopathy noncompaction

Figure 1. Clinical Categories of Inherited Cardiomyopathies and Their Genetic Basis.


The clinical entities hypertrophic cardiomyopathy and dilated cardiomyopathy share some disease genes with each
other, as well as with restrictive cardiomyopathy and left ventricular noncompaction, which are less common. Arrhyth-
mogenic right ventricular cardiomyopathy appears to be a genetically distinct category, although its clinical phenotype
cannot always be easily distinguished from that of dilated cardiomyopathy. AMPK denotes AMP-activated protein kinase,
GLA -galactosidase A, LAMP2 lysosomal-associated membrane protein 2, and TMEM43 transmembrane protein 43.
Classes of genes shown in red are the overwhelmingly predominant cause of disease within the respective categories.

often with predominant involvement of the inter- Analyses in vitro and in mouse models of car-
ventricular septum. Other hallmark features are diomyopathy have shown increased contractility
myocyte disarray and fibrosis (Fig. 2). Hypertro- of mutant myofilaments due to altered myosin ki-
phic cardiomyopathy was termed a disease of netics, increased thin-filament calcium sensitivity,
the sarcomere when the first three disease or changes in cMyBP-Cmediated regulation.11,12
genes to be identified were found to encode com- These perturbations trigger signaling pathways
ponents of the contractile apparatus of heart
muscle.3 Mutations in nine genes encoding sar- Figure 2 (facing page). Pathogenesis of Hypertrophic
comeric proteins have now been convincingly Cardiomyopathy.
shown to cause hypertrophic cardiomyopathy. In hypertrophic cardiomyopathy, mutations in sarcomeric
proteins generally increase myofilament activation and
Disease-causing mutations in any one of these
result in myocyte hypercontractility and excessive energy
genes are found in up to two thirds of patients use (Panel A). Alterations in myocardial energy status
with hypertrophic cardiomyopathy. Mutations in can also result from primary mutations affecting myo-
MYH7, encoding the -myosin heavy chain, and in cardial energy generation (e.g., mitochondrial transfer
MYBPC3, encoding cardiac myosin-binding protein RNA mutations). These mitochondrial defects and mu-
tations in the cardiac energy-sensing apparatus (e.g.,
C (cMyBP-C), are the most common, each ac-
AMP-activated protein kinase [AMPK]) recapitulate a hy-
counting for one fourth to one third of all cases of pertrophic cardiomyopathylike phenotype. Alterations
the disease; the remaining seven genes each ac- in myocardial energetics and in calcium handling com-
count for less than 1% to 5% of cases.8 The mu- bined with stimulation of signaling pathways (e.g., the
tations generally cause single amino acid substi- Janus-associated kinasesignal transducers and activa-
tors of transcription [JAK-STAT] signaling pathway) dimin-
tutions in proteins that become incorporated into
ish myocyte relaxation and promote myocyte growth,
the sarcomere. However, about half the reported with aberrant tissue architecture (i.e., myofibrillar disarray
MYBPC3 mutations are truncations; these, and and myocardial fibrosis) (Panel B, hematoxylin and eosin).
some MYBPC3 missense mutations, can result in In patients with hypertrophic cardiomyopathy, these
haploinsufficiency, a condition in which the changes often result in gross hypertrophy, with especially
prominent septal hypertrophy (arrow) as compared with
gene product of the wild-type allele cannot com-
the normal heart, as shown on the cardiac magnetic res-
pensate for the decreased product from the mutant onance images (Panel C).
allele.9,10

1644 n engl j med 364;17 nejm.org april 28, 2011

The New England Journal of Medicine


Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

A Cardiac myocyte Sarcolemma


Altered energy sensing
AMPK mutations
Decreased energy production AMPK
Mitochondrial mutations

Ca2+
ATP Sarcoplasmic
reticulum
ADP Increased use of
energy by the sarcomere

Increased myofilament activation

Actin

Tropomyosin Troponin complex


-myosin
heavy
chain

Cardiac myosin- Regulatory and essential


binding protein C myosin light chains

Hypertrophic cardiomyopathy Normal myocardial tissue

Myocyte
disarray

COLOR FIGURE

Draft 10 4/12/11
Author Watkins
n engl j med 364;17 nejm.org april 28, 2011 Fig # 2
1645
Title
The New England Journal of Medicine
Downloaded from nejm.org on August 14, 2017. For personal use only. No otherME
uses without
Forti permission.
DE
Copyright 2011 Massachusetts Medical Society. All rights reserved.Longo
Artist Knoper
The n e w e ng l a n d j o u r na l of m e dic i n e

that induce cardiac hypertrophy and are likely to pha-actinin-2. Rare variants of TCAP (telethonin),25
contribute to the diastolic dysfunction in hyper- ANKRD1 (encoding cardiac ankyrin repeat protein,
trophic cardiomyopathy. The elevated sarcoplas- or CARP),26 JPH2 (junctophilin-2),27 and MYOZ2
mic calcium concentration during diastole, as (myozenin-2)28 have been described in candidate-
documented in mouse models of hypertrophic gene analyses and studies of small families, but
cardiomyopathy,13 is likely to promote signaling their role in the disease is unclear. All these genes
(e.g., by means of calcineurinnuclear factor of encode proteins that are not integral components
activated T cells [NFAT] and calcium-calmodu- of the contractile apparatus, which suggests the
lindependent protein kinase II)14; the changes involvement of additional mechanisms. These vari-
in calcium handling may also confer a predispo- ants potentially disrupt processes common to the
sition to arrhythmias.15 downstream consequences of myofilament mu-
At least two mechanisms explain how sarco- tations, such as mechanosensory signaling and
meric mutations alter calcium balance. First, calcium handling. Mutations in PRKAG2, which
mutations affecting the thin-filament regulatory encodes the 2 subunit of AMP-activated protein
proteins tropomyosin, troponin T, and troponin I kinase (AMPK), produce a phenocopy of hypertro-
all enhance calcium sensitivity by increasing the phic cardiomyopathy accompanied by the Wolff
affinity of troponin C for calcium16; mutations ParkinsonWhite syndrome and progressive heart
affecting myosin and cMyBP-C also increase this block.29 AMPK, an important energy sensor, inter-
affinity through the formation of additional cross- acts with multiple signaling cascades.30 Although
bridges between thick and thin filaments. Since glycogen accumulation probably contributes to
troponin is the principal dynamic calcium buffer myocyte hypertrophy, early activation of hyper-
in the sarcoplasm,17 the increased affinity should trophic signaling pathways also occurs in trans-
elevate calcium levels during diastole.18 Second, genic mice in which mutant PRKAG2 is overex-
sarcomeric mutations increase the energy re- pressed.31
quirements of myosin ATPase. Since the cross- Rodents have been used to test proposed
bridge cycle, which generates the contractile force therapeutic targets, in some cases leading to pilot
of the myocyte, accounts for about 70% of the studies in humans. The L-type calcium-channel
cardiomyocytes ATP consumption, contractile inhibitor diltiazem prevented dysregulation of cal-
inefficiency could compromise the energetics of cium in the sarcoplasmic reticulum and cardiac
the myocyte.19 The energy deficiency could reduce hypertrophy in mice with a myosin heavy-chain
the activity of other ATP-consuming processes mutation.32 A phase 2 trial of diltiazem in patients
such as ion pumps (in particular, the sarcoendo- in the preclinical hypertrophic phase of cardiomy-
plasmic reticulum Ca2+ ATPase [SERCA]), thereby opathy is in progress (ClinicalTrials.gov number,
reducing calcium uptake during diastole. There NCT00319982).
is evidence of increased tension-dependent ATP Therapies to improve cardiac energetics have
consumption (tension cost) in isolated myofibril also been tested. In a randomized trial of perhexi-
preparations20 and of compromised energetics in line in patients with nonobstructive hypertrophic
mouse models21 and in patients with cardiomy- cardiomyopathy and activity-limiting symptoms,
opathy, including mutation carriers before hyper- the partial inhibition of fatty acid oxidation, in the
trophy has developed.22 Moreover, other diseases context of the oxygen limitation due to microvas-
that limit myocardial energy production, includ- cular disease in hypertrophic cardiomyopathy,33
ing mitochondrial transfer RNA mutations, cause improved cardiac ATP levels and diastolic func-
a form of cardiac hypertrophy that resembles tion, reduced symptoms, and increased exercise
hypertrophic cardiomyopathy.19 capacity.34,35 Progressive interstitial cardiac fibro-
Other disease genes have been implicated in sis, resulting from nonmyocyte (e.g., fibroblast)-
hypertrophic cardiomyopathy, albeit sometimes mediated activation of transforming growth fac-
with less than robust evidence. Cosegregation in tor signaling, is a feature of hypertrophic
large families with members affected by hyper- cardiomyopathy.36-38 The finding that preemptive
trophic cardiomyopathy23,24 supports pathogenic angiotensin II type 1receptor inhibition pre-
roles for mutations in CSRP3, which encodes mus- vented myocardial fibrosis in a mouse model of
cle LIM protein, and in ACTN2, which encodes al- cardiomyopathy,38 as well as encouraging results

1646 n engl j med 364;17 nejm.org april 28, 2011

The New England Journal of Medicine


Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

from a small clinical study, supports further in- defective transmission of force, affect stretch-
vestigation of this approach.39 sensing mechanisms involving titin, or both.51,52
The arginine-14 deletion in phospholamban (a
membrane protein of muscle cells that regulates
Dil ated C a r diom yopath y,
a fina l c om mon pheno t y pe SERCA) causes excessive inhibition of the calci-
w i th di v er se c ause s um pump and thus reduces calcium reuptake
during diastole.53 The pathogenic effects of other
The main features of dilated cardiomyopathy are mutations (e.g., those in LMNA, encoding the
left ventricular dilatation, systolic dysfunction, lamin A and C nuclear envelope proteins)54 are
myocyte death, and myocardial fibrosis (Fig. 3). less clear. Nevertheless, the diverse changes in
Analysis of asymptomatic relatives of affected cardiomyocyte structure and function result in
patients indicates that familial disease accounts autophagy, a pathway of protein and organelle
for one third to one half of cases.40,41 More than degradation, and ultimately apoptosis.55,56
40 disease genes have been identified; the most The molecular complexity of dilated cardiomy-
common mode of inheritance is autosomal dom- opathy suggests only a limited scope for specific
inant transmission, although autosomal reces- disease-modifying therapies. Broad-based ap-
sive and X-linked forms have been described.42,43 proaches, perhaps involving regenerative medi-
Dilated cardiomyopathy is sometimes inherited cine, may be needed.
with other phenotypes, both cardiac (e.g., con-
duction disorder)44 and noncardiac (e.g., sensori-
A r r h y thmo genic R igh t
neural hearing loss).45 Unlike hypertrophic car- V en t r icul a r C a r diom yopath y,
diomyopathy, dilated cardiomyopathy is caused a dise a se of the de smosome
by mutations in genes that encode components
of a wide variety of cellular compartments and The main feature of arrhythmogenic right ven-
pathways, including the nuclear envelope, con- tricular cardiomyopathy (ARVC) is fibrofatty re-
tractile apparatus, the force transduction appa- placement of the myocardium, mainly in the right
ratus (e.g., Z-disk and costamere), gene tran- ventricle but also in the left ventricle.57 This
scription and splicing machinery, and calcium change results in the predominant clinical feature
handling (Fig. 3).36,42,43 of susceptibility to ventricular arrhythmias. The
Given the diversity of affected cellular pro- disease is familial, and typically autosomal domi-
cesses, multiple proximal factors probably con- nant, in about half the cases. Mutations in five
tribute to contractile dysfunction of cardiomyo- genes that encode desmosomal proteins (desmo-
cytes before cell death and fibrotic repair occur. plakin, plakoglobin, plakophilin 2, desmoglein 2,
In dilated cardiomyopathy, mutations in the and desmocollin 2) have been found in ARVC and
genes encoding contractile proteins result in in two related autosomal recessive disorders, Naxos
functional changes that are the opposite of the disease (ARVC accompanied by woolly hair and
changes caused by mutations in the same con- palmoplantar keratoderma) and the Carvajal syn-
tractile genes that cause hypertrophic cardiomy- drome (which has a similar dermatologic pheno-
opathy. Mutations in the -myosin heavy chain type but in which left ventricular involvement is
gene depress motor function in dilated cardio- predominant) (Fig. 4). The majority of causative
myopathy,46,47 and mutations in genes for thin- mutations are insertions or deletions or nonsense
filament regulatory proteins reduce the calcium mutations that result in premature truncation of
sensitivity of contractile regulation and the af- the encoded proteins. Two other, nondesmosomal
finity of troponin for calcium16,48; hence, these genes have been implicated in ARVC: one for
mutations depress the generation of force. Several transforming growth factor 3 (TGF-3) and the
disease genes encode components of the Z-disk other for transmembrane protein 43 (TMEM43).59,60
(e.g., Cypher/ZASP),49 the structure at the bound- The existence of further mapped loci indicates
ary of each sarcomere, or the costamere (e.g., that additional disease genes remain to be dis-
-sarcoglycan), the structural complex that links covered in ARVC.61
the contractile apparatus to the sarcolemma and Desmosomes mediate intercellular attachments
extracellular matrix.50 These mutations may cause and anchor cytoplasmic domains of membrane

n engl j med 364;17 nejm.org april 28, 2011 1647


The New England Journal of Medicine
Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A
Dystroglycan
complex
Sarcoglycan
Reduced sarcolemmal complex
Ca2+
integrity 3 Na+ T tubule
Caveolin Ca2+

Sarcolemma ATP

3 Na+ 2 K+ Ca2+

Decreased energy Ryanodine


production Syntrophins receptor
Dystrophin and dystrobrevin
Aberrant Ca2+
Mitochondrion Disruption of force Ca2+ handling
transmission Ca2+

SERCA and
Actin-associated phospho- Sarcoplasmic
ADP ATP cytoskeleton Ca2+ lamban reticulum

Decreased myofilament
Myofibril activation

Abnormalities of the
nuclear envelope

Nucleus
Lamin
Nuclear pore A and C
Aberrant Aberrant
Emerin transcription splicing

B C

Cardiac muscle

Fibrosis

COLOR FIGURE

proteins to the intermediate desmin filaments of cycle. Damage to the cell surfaces,
Draft 5
causing4/12/11
cell de-
cardiomyocytes. Mutant desmosomes may there- tachment and cell death,Author
probably 62 Ex-
Watkinsensues.
fore compromise cell-to-cell adhesion at interca- perimental data indicating that mutant desmo-
Fig # 3
Title
lated disks, lessening the ability of myocytes to somes also cause remodeling of gap junctions63
ME
withstand mechanical forces during the cardiac explain how electrocardiographic
DE
Forti
Longo
changes and
Artist Knoper
AUTHOR PLEASE NOTE:
Figure has been redrawn and type has been reset
Please check carefully

Issue date 4/28/11


1648 n engl j med 364;17 nejm.org april 28, 2011

The New England Journal of Medicine


Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

tracellular lipid perturbations and may contrib-


Figure 3 (facing page). Pathogenesis of Dilated
Cardiomyopathy. ute to the fibrofatty change (Fig. 4).66
Dilated cardiomyopathy is the end phenotype of diverse Thus, although existing therapy for end-stage
mutations in heterogeneous pathways ranging from ARVC includes conventional therapy for heart
components of the membrane-scaffolding apparatus failure, genetic insights predict that the restitu-
(e.g., sarcoglycan and dystrophinopathies), sarcomeric tion of Wnt/catenin myocardial signaling and
proteins (which exhibit reduced myofilament activation,
modification of lipid-metabolism pathways (e.g.,
unlike the case for hypertrophic cardiomyopathy), nuclear
envelope proteins (e.g., lamin A and C), calcium-handling by PPARG modifiers) may represent more tar-
proteins (e.g., phospholamban [PLN]), transcription co- geted, disease-modifying therapies.
factors (e.g., eyes absent homolog 4 [EYA4]), and RNA
splicing (e.g., ribonucleic acidbinding protein [RBM20]),
to the cell energy-generating machinery (Panel A). L e ssons L e a r ned from
Although these mutations are highly diverse in terms of Molecul ar Genetic Family Studies
the pathways affected, their common features are im-
paired contraction, insurmountable cellular compromise The diversity of the cardiomyopathies results from
with consequent cell death and fibrotic repair (Panel B, genetic, allelic, epigenetic, and environmental het-
hematoxylin and eosin), and ultimately, gross cardiac erogeneity, all of which contribute to the pheno-
thinning and dilatation. Such biventricular dilatation and
type (Fig. 5). Here we summarize how studies of
wall thinning relative to the cavity dimension (arrows)
can be detected on cardiac magnetic resonance imaging cardiomyopathies improve our understanding of
(Panel C). simple monogenic conditions and their poly-
genic counterparts.

ventricular arrhythmias can develop before the loss Incomplete and Age-Related Penetrance
of myocytes and dysfunction of the right ventricle As in most other autosomal dominant disorders,
become apparent (the concealed phase of disease). inherited cardiomyopathies show marked pheno-
However, this mechanical defect does not typic variability, even within families. Penetrance
explain the right ventricular predominance and the proportion of mutation carriers with clin-
the prominence of inflammation and fibrofatty ically detectable disease increases with age
change. Desmosomal proteins also modify the but remains less than 100%. In most persons
Wnt/catenin signaling pathway, which is im- with hypertrophic cardiomyopathy, the hypertro-
portant for myogenesis in the heart. Increased phy is manifested in adolescence, whereas the
nuclear translocation of plakoglobin, which is age at onset in patients with sarcomeric dilated
caused by the reduced plakoglobin-sequestering cardiomyopathy is bimodal (with peaks during
capacity of mutant desmosomes, appears to sup- childhood and mid-adulthood).67 The disease is
press Wnt signaling of cardiac progenitor cells.64 gradually progressive in patients with dilated car-
Redistribution of plakoglobin is a central feature diomyopathy due to LMNA.68 It is uncommon to
of ARVC and could serve as a diagnostic test for find numerous persons with clinically apparent
the disease in postmortem tissue and, conceiv- ARVC in a single pedigree, indicating a low level
ably, in endomyocardial-biopsy specimens.58 The of penetrance.
predilection for involvement of the right ventri-
cle in ARVC probably depends on properties of Variable Expressivity
cardiac progenitor cells in the second heart Early reports of each of the cardiomyopathies de-
field, the embryonic source of the right ventricle. scribed patients with severe forms of the disease.
These primitive right-ventricle precursor cells Subsequent studies, however, have shown that
are prone to differentiate into adipocytes (be- most affected persons have mild, sometimes atyp-
cause of reduced transcription mediated by T-cell ical disease; as a result, the number of cases in a
factor/lymphoid enhancer factor [Tcf/Lef ]), ren- given family, and thus the proportion of familial
dering them more susceptible to the reduced cases, is greater than originally suspected. Only
Wnt signaling.65 Adipogenic transcription fac- a minority of patients with hypertrophic cardio-
tors, such as peroxisome proliferatoractivated myopathy have the classic feature of outflow ob-
receptor gamma (PPARG) (which is known to struction at rest, and up to half the cases of idio-
drive TMEM43 expression), may also mediate in- pathic dilated cardiomyopathy are familial.40,41 It

n engl j med 364;17 nejm.org april 28, 2011 1649


The New England Journal of Medicine
Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A Sarcolemma Outer
dense
Plakophilin Desmoglein plaque Intermediate
filaments

Desmoplakin
Desmocollin
Nucleus
Nuclear plakoglobin
translocation

Plakoglobin

Altered Wnt/
catenin
signaling

Intracellular
space Impaired cell adhesion and
Gap junction intercellular communication

B No ARVC ARVC

N-cadherin

Plakoglobin

C D

Fat

Fibrosis

Muscle

COLOR FIGURE

Draft 4 4/11/11
1650 n engl j med 364;17 nejm.org april 28, 2011 Author Watkins
Fig # 4
The New England Journal of Medicine Title
Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
ME Forti
Copyright 2011 Massachusetts Medical Society. All rights reserved. DE Longo
Artist Knoper
mechanisms of disease

Figure 4 (facing page). Pathogenesis of Arrhythmogenic


missense mutations in the same genes encoding
Right Ventricular Cardiomyopathy. sarcomeric proteins. Since these diseases arise
Arrhythmogenic right ventricular cardiomyopathy from mutations with opposing biophysical conse-
(ARVC) results from perturbation of one of three quences,48 a variant breeds true within each
groups of desmosomal proteins: transmembrane pro- family; there has been no reliable documentation
teins (e.g., the desmosomal cadherins, desmoglein and of families in which a single sarcomere mutation
desmocollin), proteins anchored directly to intermedi-
ate filaments (e.g., desmoplakin), and the armadillo
causes hypertrophic cardiomyopathy in some mem-
family of proteins (e.g., plakoglobin and plakophilin), bers and dilated cardiomyopathy in others. How-
which bind the desmosomal cadherins to desmoplakin ever, other aspects of the cardiomyopathy pheno-
(Panel A). In addition to the disruption of desmosomal type can vary within families, indicating the
mechanical function, which can lead to the death of absence of a precise relationship between the mu-
myocytes under physical stress, the suppression of
canonical Wnt/catenin signaling by nuclear plako-
tation and its biophysical consequences. For ex-
globin translocation appears to promote adipogenesis ample, apical hypertrophic cardiomyopathy most-
in mesodermal precursors. Panel B shows immuno- ly occurs in families affected primarily by typical
fluorescence images of left ventricular myocardium hypertrophic cardiomyopathy; in only a minority
from patients with ARVC and controls without ARVC. of cases does apical hypertrophic cardiomyopathy
Although both patients with ARVC and those without
show a strong junctional signal for N-cadherin, a non-
have a consistent relationship with a specific mu-
desmosomal adhesion molecule, plakoglobin, is mark- tation (e.g., Glu101Lys in the alpha cardiac actin
edly reduced in patients with ARVC whether or not the gene [ACTC1]).73 Similarly, familial restrictive car-
section shows typical pathological changes of fibrofatty diomyopathy is part of the spectrum of sarcomeric
replacement (Panel B). These changes explain the pro- hypertrophic cardiomyopathy, with a loose rela-
gressive fibrofatty replacement of ventricular myocardi-
um (Panel C, hematoxylin and eosin), with progressive
tionship between certain mutations and this vari-
gross effects on ventricular morphology and function, ant of the phenotype.74 Left ventricular noncom-
classically, but not exclusively, with right ventricular pre- paction is characterized by myocardium with a
dominance (arrows), as shown on cardiac magnetic spongy appearance. The disease may reflect a fail-
resonance imaging (Panel D). (Panel B reprinted from ure of normal development and sometimes occurs
Asimaki et al.58 with the permission of the publisher.)
together with cardiac and extracardiac develop-
mental defects, but progressive dysfunction in
has also become apparent that ARVC often goes adults indicates that left ventricular noncom-
unrecognized and is more common than was paction is a newly recognized aspect of cardiac
first thought.69 Left ventricular noncompaction, remodeling. In some families, the phenotype is
initially considered a rare disorder associated consistently manifested (the genetic basis of
with very high rates of cardioembolism and heart such families is unknown), but cases of non-
failure,70 is now considered to be substantially compaction do occur in families with otherwise
more common and less severe than was previ- typical hypertrophic or dilated cardiomyopathy
ously believed.71 Incomplete penetrance requires attributable to sarcomeric mutations.49,75
diagnostic criteria of less than the usual strin-
gency for first-degree relatives, in whom the pri- Phenocopies
or risk is generally 50%; clinicians caring for The term phenocopy refers to apparently similar
families at risk now use complex diagnostic algo-disorders with different causes. Distinctions among
rithms to interpret minor abnormalities.69 The such conditions can be clinically important, be-
corollary is that, in the general population, pa-cause disorders with similar cardiac morphology
tients with subtle features of inherited cardiomy-
can have different inheritance patterns, natural
opathies are difficult to recognize. Thus, popula-
histories, or responses to therapy. Certain autoso-
tion screening is generally ineffective; instead,mal dominant cardiomyopathies (those caused by
cascade screening (sequential identification of PRKAG2 mutations) and X-linked cardiomyopa-
related family members, increasingly guided by thies (Fabrys disease and Danons disease) share
genetic testing) is key.72 clinical features with sarcomeric hypertrophic
cardiomyopathy yet are distinct disorders.29,76,77
Genetic Heterogeneity and Allelic Disorders Such phenocopies may also inform our under-
Hypertrophic cardiomyopathy and dilated cardio- standing of disease mechanisms. Although hyper-
myopathy can be allelic, each caused by specific trophy due to PRKAG2 mutations is often attrib-

n engl j med 364;17 nejm.org april 28, 2011 1651


The New England Journal of Medicine
Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Modifier Genes Environmental Effects


In the locus HCM
Common or rare Age
in cis or in trans Athletic training
Additional coding variants Hypertension
Noncoding variants that Coronary disease
alter level of expression DCM
Outside the locus Age
Common polymorphisms Pregnancy
or rare mutations affecting Cytotoxic drugs
same system or down- Viral infection
stream pathways Coronary disease

Causal
Phenotype
Mutation

Post-translational and Epigenetic


Effects
DNA modification (e.g., methylation)
Histone modification (e.g., acetylation)
Translational control (e.g., microRNA)
Post-translational modifications

Figure 5. Complexities of the GenotypePhenotype Relationship in Inherited Cardiomyopathies.


The effect of the particular disease gene,Watkins
AUTHOR: and specific mutation, on the cardiomyopathy
RETAKE: 1st phenotype is modified ex-
2nd can vary greatly even among
tensively by genetic, epigenetic, and environmental factors. Accordingly, the phenotype
FIGURE: 5 of 5 3rd
relatives with the same mutation. Most such modifying factors remain unknown,
Revised
although examples that illustrate
the categories listed are emerging. DCM denotes
ARTIST: ts dilated cardiomyopathy, and HCM hypertrophic cardiomyopathy.
SIZE
6 col
TYPE: Line Combo 4-C H/T 33p9
AUTHOR, PLEASE NOTE:
uted to glycogen accumulation, Figure has been redrawn and type has been reset.
the increase
Pleasein
checktions between the disease gene and the pheno-
carefully.
cardiac mass cannot be explained by a simple type are currently of limited usefulness for man-
JOB: 363xx
bulk effect; instead, the glycogen probably initi- aging the ISSUE: care xx-xx-10
of individual patients; some
ates signaling mechanisms involved in sarcomer- quantitative differences exist, but there is sub-
ic hypertrophic cardiomyopathy.31,78 stantial overlap between disease-gene groups,
and exceptions are common.81-83 Allelic hetero-
GenotypePhenotype Correlations geneity further complicates attempts to correlate
In certain circumstances, knowledge of the gene genotype with phenotype, since the rarity of in-
underlying the cardiomyopathy will alter patient dividual mutations usually means that sufficient
care. One example is phenocopies of hypertro- clinical data are unavailable. Long-term efforts
phic cardiomyopathy with different inheritance will be needed to accumulate reliable evidence on
patterns and natural histories. Another example genotypephenotype correlations. Data based on
is the susceptibility to conduction disease of pa- results from proband series are particularly vul-
tients with dilated cardiomyopathy due to LMNA nerable to ascertainment bias.84
mutations; when this is sufficient to warrant Additional complexities include the presence
pacemaker insertion, use of an implantable car- of two or more variants, as either compound or
dioverterdefibrillator should be considered.79,80 double heterozygosity.85-87 The proportion of
However, for most cardiomyopathies, correla- genotyped persons with more than one variant

1652 n engl j med 364;17 nejm.org april 28, 2011

The New England Journal of Medicine


Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

is higher in diseases with low penetrance no- related phenotypes, suggesting that they may in
tably, arrhythmogenic right ventricular cardio- fact be silent polymorphisms.93,94
myopathy.88 The presence of multiple variants
complicates genetic testing in families (since it F u t ur e Prospec t s
may be difficult to determine whether a sec-
ond variant is itself sufficient to cause disease) The incomplete penetrance that complicates ge-
and confounds genotypephenotype correlations netic evaluation of families with a cardiomyopa-
if only one allele is analyzed. thy paradoxically raises hopes that the develop-
ment of novel disease-modifying therapies may
Nonmendelian Variants and Modifier Effects be achievable. The underlying mutations cause
The widespread detailed sequencing of the genes subtle cellular perturbations11 that are tolerated
implicated in the cardiomyopathies should cul- by all mutation carriers for a period and in
minate in identification of a spectrum of vari- many cases, throughout life which suggests
ants, ranging from alleles that clearly cause dis- that compensatory mechanisms exist. The tran-
ease through variants of uncertain significance sition to overt disease can be abrupt in both hy-
to silent polymorphisms. A well-validated exam- pertrophic and dilated cardiomyopathies,95,96
ple of a common susceptibility variant is an in- suggesting a tipping point that triggers decom-
tronic deletion in MYBPC3, which causes a partial pensation. Novel therapies may need only to sub-
splicing defect (as opposed to the complete de- tly shift cellular variables to sustain the compen-
fect in typical autosomal dominant hypertrophic sated state, particularly if therapy begins in
cardiomyopathy) and confers susceptibility to asymptomatic mutation carriers identified by cas-
various cardiomyopathies in people whose fami- cade screening in families. Hypertrophic cardio-
lies come from the Indian subcontinent.89 It is myopathy may be the most tractable cardiomy-
likely that in a proportion of all cardiomyopa- opathy, since specific therapeutic targets have
thies, inheritance has a nonmendelian pattern, been identified downstream of the perturbation
in which alleles with only modest effects con- in contractile regulation, hypertrophied cardio-
verge; the likelihood of familial disease in these myocytes can undergo remodeling, and the dis-
cases is low, and the disease may be milder. Pa- ease does not depend on myocyte death. ARVC
tients with hypertrophic cardiomyopathy who do also appears to have a final common pathway
not have a family history of the disease are less with sufficient specificity to be targeted, partic-
likely to carry pathogenic sarcomeric mutations90 ularly if aberrant Wnt/catenin signaling rath-
and usually have a relatively mild phenotype.91 er than a mechanical defect is central to the dis-
Validation of variants, or modifier genes, with an order. The multiple primary defects underlying
intermediate effect is difficult because they can- dilated cardiomyopathy appear to be the most
not be tested by means of cosegregation. Com- difficult to target. Here, and also in other car-
mon variants can be evaluated with the use of diomyopathies, avoidance of environmental pre-
tests for association in large studies,89 but a sta- cipitants that could trigger decompensation96-98
tistical demonstration of an increased mutation could be important.
load is needed for rare variants, which requires Supported by the British Heart Foundation Centre of Excel-
sequencing of both case patients and controls.92 lence at Oxford, Wellcome Trust, European Commission Frame-
work Programme Grant (241577), and Oxford Partners National
The fact that variants occur in case patients but not Institute for Health Research Comprehensive Biomedical Re-
in controls is not adequate to prove a pathogenic search Centre.
role because control subjects often bear similarly Dr. Watkins reports being listed as a patent holder on patents
held by Harvard University for methods for detecting disease-
rare but different variants. This limitation is a associated mutations in hypertrophic cardiomyopathy; and Dr.
problem with many recent candidate-gene stud- Ashrafian reports holding a European method-of-use patent for
ies in cardiomyopathy. Some patients who have perhexiline in systolic heart failure and having patents pending
for its use in diastolic heart failure and hypertrophic cardiomy-
hypertrophic cardiomyopathy without a sarcomere opathy and for its use in systolic heart failure in countries out-
mutation may not have an inherited disease at all side Europe.
the variants in these cases will be chance We thank Dr. Mary Sheppard, Royal Brompton Hospital, Lon-
don, for the histologic images and Dr. Theodoros Karamitsos
findings. In keeping with this point, identical and Professor Stefan Neubauer, University of Oxford, Oxford,
variants are sometimes reported as causes of un- United Kingdom, for the cardiac magnetic resonance images.

n engl j med 364;17 nejm.org april 28, 2011 1653


The New England Journal of Medicine
Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

References

1. Jarcho JA, McKenna W, Pare JA, et al. SG, et al. Familial hypertrophic cardiomy- Cardiac ankyrin repeat protein gene
Mapping a gene for familial hypertrophic opathy-linked mutant troponin T causes (ANKRD1) mutations in hypertrophic
cardiomyopathy to chromosome 14q1. stress-induced ventricular tachycardia and cardiomyopathy. J Am Coll Cardiol 2009;
N Engl J Med 1989;321:1372-8. Ca2+-dependent action potential remod- 54:334-42.
2. Geisterfer-Lowrance AA, Kass S, Tani- eling. Circ Res 2003;92:428-36. 27. Landstrom AP, Weisleder N, Batalden
gawa G, et al. A molecular basis for famil- 14. Bers DM, Guo T. Calcium signaling in KB, et al. Mutations in JPH2-encoded
ial hypertrophic cardiomyopathy: a beta cardiac ventricular myocytes. Ann N Y junctophilin-2 associated with hypertro-
cardiac myosin heavy chain gene mis- Acad Sci 2005;1047:86-98. phic cardiomyopathy in humans. J Mol
sense mutation. Cell 1990;62:999-1006. 15. Huke S, Knollmann BC. Increased Cell Cardiol 2007;42:1026-35.
3. Thierfelder L, Watkins H, MacRae C, myofilament Ca2+-sensitivity and ar- 28. Osio A, Tan L, Chen SN, et al.
et al. Alpha-tropomyosin and cardiac tro- rhythmia susceptibility. J Mol Cell Cardiol Myozenin 2 is a novel gene for human
ponin T mutations cause familial hyper- 2010;48:824-33. hypertrophic cardiomyopathy. Circ Res
trophic cardiomyopathy: a disease of the 16. Robinson P, Griffiths PJ, Watkins H, 2007;100:766-8.
sarcomere. Cell 1994;77:701-12. Redwood CS. Dilated and hypertrophic 29. Blair E, Redwood C, Ashrafian H, et
4. Dietz HC. New therapeutic approach- cardiomyopathy mutations in troponin al. Mutations in the gamma(2) subunit of
es to mendelian disorders. N Engl J Med and alpha-tropomyosin have opposing ef- AMP-activated protein kinase cause fa-
2010;363:852-63. fects on the calcium affinity of cardiac milial hypertrophic cardiomyopathy: evi-
5. Maron BJ, Towbin JA, Thiene G, et al. thin filaments. Circ Res 2007;101:1266- dence for the central role of energy com-
Contemporary definitions and classifica- 73. promise in disease pathogenesis. Hum
tion of the cardiomyopathies: an Ameri- 17. Smith GA, Dixon HB, Kirschenlohr Mol Genet 2001;10:1215-20.
can Heart Association Scientific State- HL, Grace AA, Metcalfe JC, Vandenberg 30. Kim AS, Miller EJ, Young LH. AMP-
ment from the Council on Clinical JI. Ca2+ buffering in the heart: Ca2+ activated protein kinase: a core signalling
Cardiology, Heart Failure and Transplan- binding to and activation of cardiac myo- pathway in the heart. Acta Physiol (Oxf)
tation Committee; Quality of Care and fibrils. Biochem J 2000;346:393-402. 2009;196:37-53.
Outcomes Research and Functional Ge- 18. Kataoka A, Hemmer C, Chase PB. 31. Banerjee SK, McGaffin KR, Huang
nomics and Translational Biology Inter- Computational simulation of hypertro- XN, Ahmad F. Activation of cardiac hy-
disciplinary Working Groups; and Coun- phic cardiomyopathy mutations in tropo- pertrophic signaling pathways in a trans-
cil on Epidemiology and Prevention. nin I: influence of increased myofilament genic mouse with the human PRKAG2
Circulation 2006;113:1807-16. calcium sensitivity on isometric force, Thr400Asn mutation. Biochim Biophys
6. Elliott P, Andersson B, Arbustini E, et ATPase and [Ca2+]i. J Biomech 2007;40: Acta 2010;1802:284-91.
al. Classification of the cardiomyopa- 2044-52. 32. Semsarian C, Ahmad I, Giewat M, et
thies: a position statement from the Euro- 19. Ashrafian H, Redwood C, Blair E, al. The L-type calcium channel inhibitor
pean Society of Cardiology Working Watkins H. Hypertrophic cardiomyopa- diltiazem prevents cardiomyopathy in a
Group on Myocardial and Pericardial Dis- thy: a paradigm for myocardial energy mouse model. J Clin Invest 2002;109:1013-
eases. Eur Heart J 2008;29:270-6. depletion. Trends Genet 2003;19:263-8. 20.
7. Kamisago M, Sharma SD, DePalma 20. Belus A, Piroddi N, Scellini B, et al. 33. Petersen SE, Jerosch-Herold M, Hud-
SR, et al. Mutations in sarcomere protein The familial hypertrophic cardiomyopa- smith LE, et al. Evidence for microvascular
genes as a cause of dilated cardiomyopa- thy-associated myosin mutation R403Q dysfunction in hypertrophic cardiomyop-
thy. N Engl J Med 2000;343:1688-96. accelerates tension generation and relaxa- athy: new insights from multiparametric
8. Richard P, Charron P, Carrier L, et al. tion of human cardiac myofibrils. J Physiol magnetic resonance imaging. Circulation
Hypertrophic cardiomyopathy: distribu- 2008;586:3639-44. 2007;115:2418-25.
tion of disease genes, spectrum of muta- 21. Spindler M, Saupe KW, Christe ME, et 34. Ashrafian H, Frenneaux MP, Opie LH.
tions, and implications for a molecular al. Diastolic dysfunction and altered ener- Metabolic mechanisms in heart failure.
diagnosis strategy. Circulation 2003;107: getics in the alphaMHC403/+ mouse Circulation 2007;116:434-48.
2227-32. [Erratum, Circulation 2004;109: model of familial hypertrophic cardiomy- 35. Abozguia K, Elliott P, McKenna WJ, et
3258.] opathy. J Clin Invest 1998;101:1775-83. al. Metabolic modulator perhexiline cor-
9. Marston S, Copeland O, Jacques A, et 22. Crilley JG, Boehm EA, Blair E, et al. rects energy deficiency and improves ex-
al. Evidence from human myectomy sam- Hypertrophic cardiomyopathy due to sar- ercise capacity in symptomatic hypertro-
ples that MYBPC3 mutations cause hyper- comeric gene mutations is characterized phic cardiomyopathy. Circulation 2010;
trophic cardiomyopathy through haploin- by impaired energy metabolism irrespec- 122:1562-9.
sufficiency. Circ Res 2009;105:219-22. tive of the degree of hypertrophy. J Am 36. OHanlon R, Grasso A, Roughton M
10. van Dijk SJ, Dooijes D, dos Remedios Coll Cardiol 2003;41:1776-82. et al. Prognostic significance of myocar-
C, et al. Cardiac myosin-binding protein C 23. Geier C, Gehmlich K, Ehler E, et al. dial fibrosis in hypertrophic cardiomyop-
mutations and hypertrophic cardiomyop- Beyond the sarcomere: CSRP3 mutations athy. J Am Coll Cardiol 2010;56:867-74.
athy: haploinsufficiency, deranged phos- cause hypertrophic cardiomyopathy. Hum 37. Ho CY, Lpez B, Coelho-Filho OR, et
phorylation, and cardiomyocyte dysfunc- Mol Genet 2008;17:2753-65. al. Myocardial fibrosis as an early mani-
tion. Circulation 2009;119:1473-83. 24. Chiu C, Bagnall RD, Ingles J, et al. festation of hypertrophic cardiomyopa-
11. Redwood CS, Moolman-Smook JC, Mutations in alpha-actinin-2 cause hyper- thy. N Engl J Med 2010;363:552-63.
Watkins H. Properties of mutant contrac- trophic cardiomyopathy: a genome-wide 38. Teekakirikul P, Eminaga S, Toka O, et
tile proteins that cause hypertrophic car- analysis. J Am Coll Cardiol 2010;55:1127- al. Cardiac fibrosis in mice with hypertro-
diomyopathy. Cardiovasc Res 1999;44:20- 35. phic cardiomyopathy is mediated by non-
36. 25. Hayashi T, Arimura T, Itoh-Satoh M, myocyte proliferation and requires Tgf-
12. Seidman JG, Seidman C. The genetic et al. Tcap gene mutations in hypertro- beta. J Clin Invest 2010;120:3520-9.
basis for cardiomyopathy: from mutation phic cardiomyopathy and dilated cardio- 39. Penicka M, Gregor P, Kerekes R, et al.
identification to mechanistic paradigms. myopathy. J Am Coll Cardiol 2004;44:2192- The effects of candesartan on left ven-
Cell 2001;104:557-67. 201. tricular hypertrophy and function in non-
13. Knollmann BC, Kirchhof P, Sirenko 26. Arimura T, Bos JM, Sato A, et al. obstructive hypertrophic cardiomyopa-

1654 n engl j med 364;17 nejm.org april 28, 2011

The New England Journal of Medicine


Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

thy: a pilot, randomized study. J Mol results in lethal, hereditary cardiomyopa- pathogenesis of arrhythmogenic right
Diagn 2009;11:35-41. thy. Proc Natl Acad Sci U S A 2006;103: ventricular cardiomyopathy. Cardiovasc
40. Baig MK, Goldman JH, Caforio AL, 1388-93. Res 2009;84:83-90.
Coonar AS, Keeling PJ, McKenna WJ. Fa- 54. Malhotra R, Mason PK. Lamin A/C 67. Lakdawala NK, Dellefave L, Redwood
milial dilated cardiomyopathy: cardiac deficiency as a cause of familial dilated CS, et al. Familial dilated cardiomyopathy
abnormalities are common in asymptom- cardiomyopathy. Curr Opin Cardiol 2009; caused by an alpha-tropomyosin muta-
atic relatives and may represent early dis- 24:203-8. tion: the distinctive natural history of sar-
ease. J Am Coll Cardiol 1998;31:195-201. 55. Mudd JO, Kass DA. Tackling heart comeric dilated cardiomyopathy. J Am
41. DeWitt MM, MacLeod HM, Soliven B, failure in the twenty-first century. Nature Coll Cardiol 2010;55:320-9.
McNally EM. Phospholamban R14 dele- 2008;451:919-28. 68. Fatkin D, MacRae C, Sasaki T, et al.
tion results in late-onset, mild, hereditary 56. Foo RS, Mani K, Kitsis RN. Death be- Missense mutations in the rod domain of
dilated cardiomyopathy. J Am Coll Cardi- gets failure in the heart. J Clin Invest the lamin A/C gene as causes of dilated
ol 2006;48:1396-8. 2005;115:565-71. cardiomyopathy and conduction-system
42. Jefferies JL, Towbin JA. Dilated car- 57. Sen-Chowdhry S, Morgan RD, Cham- disease. N Engl J Med 1999;341:1715-24.
diomyopathy. Lancet 2010;375:752-62. bers JC, McKenna WJ. Arrhythmogenic 69. Marcus FI, McKenna WJ, Sherrill D,
43. Dellefave L, McNally EM. The genetics cardiomyopathy: etiology, diagnosis, and et al. Diagnosis of arrhythmogenic right
of dilated cardiomyopathy. Curr Opin treatment. Annu Rev Med 2010;61:233-53. ventricular cardiomyopathy/dysplasia: pro-
Cardiol 2010 February 24 (Epub ahead of 58. Asimaki A, Tandri H, Huang H, et al. posed modification of the task force cri-
print). A new diagnostic test for arrhythmogenic teria. Circulation 2010;121:1533-41.
44. Wolf CM, Wang L, Alcalai R, et al. La- right ventricular cardiomyopathy. N Engl 70. Oechslin EN, Attenhofer Jost CH, Ro-
min A/C haploinsufficiency causes dilat- J Med 2009;360:1075-84. jas JR, Kaufmann PA, Jenni R et al. Long-
ed cardiomyopathy and apoptosis-trig- 59. Beffagna G, Occhi G, Nava A, et al. term follow-up of 34 adults with isolated
gered cardiac conduction system disease. Regulatory mutations in transforming left ventricular noncompaction: a distinct
J Mol Cell Cardiol 2008;44:293-303. growth factor-beta3 gene cause arrhyth- cardiomyopathy with poor prognosis.
45. Schnberger J, Wang L, Shin JT, et al. mogenic right ventricular cardiomyopa- J Am Coll Cardiol 2000;36:493-500.
Mutation in the transcriptional coactiva- thy type 1. Cardiovasc Res 2005;65:366- 71. Lofiego C, Biagini E, Pasquale F, et al.
tor EYA4 causes dilated cardiomyopathy 73. Wide spectrum of presentation and vari-
and sensorineural hearing loss. Nat Genet 60. Merner ND, Hodgkinson KA, Hay- able outcomes of isolated left ventricular
2005;37:418-22. wood AF, et al. Arrhythmogenic right ven- non-compaction. Heart 2007;93:65-71.
46. Schmitt JP, Debold EP, Ahmad F, et al. tricular cardiomyopathy type 5 is a fully 72. Wordsworth S, Leal J, Blair E, et al.
Cardiac myosin missense mutations penetrant, lethal arrhythmic disorder DNA testing for hypertrophic cardiomy-
cause dilated cardiomyopathy in mouse caused by a missense mutation in the opathy: a cost-effectiveness model. Eur
models and depress molecular motor TMEM43 gene. Am J Hum Genet 2008; Heart J 2010;31:926-35.
function. Proc Natl Acad Sci U S A 2006; 82:809-21. 73. Arad M, Penas-Lado M, Monserrat L,
103:14525-30. 61. Matolweni LO, Bardien S, Rebello G, et al. Gene mutations in apical hypertro-
47. Debold EP, Schmitt JP, Patlak JB, et al. et al. Arrhythmogenic right ventricular phic cardiomyopathy. Circulation 2005;
Hypertrophic and dilated cardiomyopathy cardiomyopathy type 6 (ARVC6): support 112:2805-11.
mutations differentially affect the molec- for the locus assignment, narrowing of 74. Kubo T, Gimeno JR, Bahl A, et al.
ular force generation of mouse alpha- the critical region and mutation screen- Prevalence, clinical significance, and ge-
cardiac myosin in the laser trap assay. Am ing of three candidate genes. BMC Med netic basis of hypertrophic cardiomyopa-
J Physiol Heart Circ Physiol 2007;293: Genet 2006;7:29. thy with restrictive phenotype. J Am Coll
H284-H291. 62. Kirchhof P, Fabritz L, Zwiener M, et Cardiol 2007;49:2419-26.
48. Mirza M, Marston S, Willott R, et al. al. Age- and training-dependent develop- 75. Klaassen S, Probst S, Oechslin E, et al.
Dilated cardiomyopathy mutations in ment of arrhythmogenic right ventricular Mutations in sarcomere protein genes in
three thin filament regulatory proteins cardiomyopathy in heterozygous plako- left ventricular noncompaction. Circula-
result in a common functional pheno- globin-deficient mice. Circulation 2006; tion 2008;117:2893-901.
type. J Biol Chem 2005;280:28498-506. 114:1799-806. 76. Maron BJ, Roberts WC, Arad M, et al.
49. Vatta M, Mohapatra B, Jimenez S, et 63. Kaplan SR, Gard JJ, Protonotarios N, Clinical outcome and phenotypic expres-
al. Mutations in Cypher/ZASP in patients et al. Remodeling of myocyte gap junc- sion in LAMP2 cardiomyopathy. JAMA
with dilated cardiomyopathy and left ven- tions in arrhythmogenic right ventricular 2009;301:1253-9.
tricular non-compaction. J Am Coll Car- cardiomyopathy due to a deletion in plak- 77. Monserrat L, Gimeno-Blanes JR,
diol 2003;42:2014-27. oglobin (Naxos disease). Heart Rhythm Marin F, et al. Prevalence of Fabry disease
50. Tsubata S, Bowles KR, Vatta M, et al. 2004;1:3-11. in a cohort of 508 unrelated patients with
Mutations in the human delta-sarcogly- 64. Garcia-Gras E, Lombardi R, Giocon- hypertrophic cardiomyopathy. J Am Coll
can gene in familial and sporadic dilated do MJ, et al. Suppression of canonical Cardiol 2007;50:2399-403.
cardiomyopathy. J Clin Invest 2000;106: Wnt/beta-catenin signaling by nuclear 78. Watkins H, Ashrafian H, McKenna
655-62. plakoglobin recapitulates phenotype of WJ. The genetics of hypertrophic cardio-
51. Bowles NE, Bowles KR, Towbin JA. arrhythmogenic right ventricular cardio- myopathy: Teare redux. Heart 2008;94:
The final common pathway hypothesis myopathy. J Clin Invest 2006;116:2012-21. 1264-8.
and inherited cardiovascular disease: the 65. Lombardi R, Dong J, Rodriguez G, et 79. Pasotti M, Klersy C, Pilotto A, et al.
role of cytoskeletal proteins in dilated al. Genetic fate mapping identifies sec- Long-term outcome and risk stratifica-
cardiomyopathy. Herz 2000;25:168-75. ond heart field progenitor cells as a tion in dilated cardiolaminopathies. J Am
52. Miller MK, Granzier H, Ehler E, Grego- source of adipocytes in arrhythmogenic Coll Cardiol 2008;52:1250-60.
rio CC. The sensitive giant: the role of titin- right ventricular cardiomyopathy. Circ 80. Meune C, Van Berlo JH, Anselme F,
based stretch sensing complexes in the Res 2009;104:1076-84. Bonne G, Pinto YM, Duboc D. Primary
heart. Trends Cell Biol 2004;14:119-26. 66. Djouadi F, Lecarpentier Y, Hbert JL, prevention of sudden death in patients
53. Haghighi K, Kolokathis F, Gramolini Charron P, Bastin J, Coirault C. A poten- with lamin A/C gene mutations. N Engl J
AO, et al. A mutation in the human phos- tial link between peroxisome proliferator- Med 2006;354:209-10.
pholamban gene, deleting arginine 14, activated receptor signalling and the 81. Watkins H, McKenna WJ, Thierfelder

n engl j med 364;17 nejm.org april 28, 2011 1655


The New England Journal of Medicine
Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.
mechanisms of disease

L, et al. Mutations in the genes for cardiac trophic cardiomyopathy associated with candidate gene for total anomalous pul-
troponin T and alpha-tropomyosin in hy- triple sarcomere protein gene mutations. monary venous return. Hum Mutat 2008;
pertrophic cardiomyopathy. N Engl J Med J Am Coll Cardiol 2010;55:1444-53. 29:468-74.
1995;332:1058-64. 88. Xu T, Yang Z, Vatta M, et al. Com- 94. Duboscq-Bidot L, Charron P, Ruppert
82. Niimura H, Bachinski LL, Sangwatan- pound and digenic heterozygosity con- V, et al. Mutations in the ANKRD1 gene
aroj S, et al. Mutations in the gene for tributes to arrhythmogenic right ventricu- encoding CARP are responsible for hu-
cardiac myosin-binding protein C and late- lar cardiomyopathy. J Am Coll Cardiol man dilated cardiomyopathy. Eur Heart J
onset familial hypertrophic cardiomy- 2010;55:587-97. 2009;30:2128-36.
opathy. N Engl J Med 1998;338:1248-57. 89. Dhandapany PS, Sadayappan S, Xue Y, 95. Maron BJ, Niimura H, Casey SA, et al.
83. Van Driest SL, Ellsworth EG, Ommen et al. A common MYBPC3 (cardiac myosin Development of left ventricular hypertro-
SR, Tajik AJ, Gersh BJ, Ackerman MJ. binding protein C) variant associated with phy in adults in hypertrophic cardiomy-
Prevalence and spectrum of thin filament cardiomyopathies in South Asia. Nat Gen- opathy caused by cardiac myosin-binding
mutations in an outpatient referral popu- et 2009;41:187-91. protein C gene mutations. J Am Coll Car-
lation with hypertrophic cardiomyopathy. 90. Andersen PS, Havndrup O, Hougs L, diol 2001;38:315-21.
Circulation 2003;108:445-51. et al. Diagnostic yield, interpretation, and 96. van Spaendonck-Zwarts KY, van Tint-
84. Blair E, Redwood C, Watkins H. As- clinical utility of mutation screening of elen JP, van Veldhuisen DJ, et al. Peripar-
certainment strategies and genotype: sarcomere encoding genes in Danish hy- tum cardiomyopathy as a part of familial
phenotype correlations in hypertrophic pertrophic cardiomyopathy patients and dilated cardiomyopathy. Circulation 2010;
cardiomyopathy. Circulation 2003;108(4): relatives. Hum Mutat 2009;30:363-70. 121:2169-75.
e24-e25. 91. Olivotto I, Girolami F, Ackerman MJ, 97. Noutsias M, Fechner H, de Jonge H, et
85. Blair E, Price SJ, Baty CJ, Ostman- et al. Myofilament protein gene mutation al. Human coxsackie-adenovirus receptor
Smith I, Watkins H. Mutations in cis can screening and outcome of patients with is colocalized with integrins alpha(v)
confound genotype-phenotype correla- hypertrophic cardiomyopathy. Mayo Clin beta(3) and alpha(v)beta(5) on the cardio-
tions in hypertrophic cardiomyopathy. Proc 2008;83:630-8. myocyte sarcolemma and upregulated in
J Med Genet 2001;38:385-8. 92. Matkovich SJ, Van Booven DJ, Hindes A, dilated cardiomyopathy: implications for
86. Ingles J, Doolan A, Chiu C, Seidman J, et al. Cardiac signaling genes exhibit un- cardiotropic viral infections. Circulation
Seidman C, Semsarian C. Compound and expected sequence diversity in sporadic 2001;104:275-80.
double mutations in patients with hyper- cardiomyopathy, revealing HSPB7 poly- 98. Xiong D, Lee GH, Badorff C, et al.
trophic cardiomyopathy: implications for morphisms associated with disease. J Clin Dystrophin deficiency markedly increases
genetic testing and counselling. J Med Invest 2010;120:280-9. enterovirus-induced cardiomyopathy: a
Genet 2005;42(10):e59. 93. Cinquetti R, Badi I, Campione M, et genetic predisposition to viral heart dis-
87. Girolami F, Ho CY, Semsarian C, et al. al. Transcriptional deregulation and a ease. Nat Med 2002;8:872-7.
Clinical features and outcome of hyper- missense mutation define ANKRD1 as a Copyright 2011 Massachusetts Medical Society.

IMAGES IN CLINICAL MEDICINE


The Journal welcomes consideration of new submissions for Images in Clinical
Medicine. Instructions for authors and procedures for submissions can be found
on the Journals Web site at NEJM.org. At the discretion of the editor, images that
are accepted for publication may appear in the print version of the Journal,
the electronic version, or both.

1656 n engl j med 364;17 nejm.org april 28, 2011

The New England Journal of Medicine


Downloaded from nejm.org on August 14, 2017. For personal use only. No other uses without permission.
Copyright 2011 Massachusetts Medical Society. All rights reserved.

You might also like