Genetics of Dilated Cardiomyopathy

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

REVIEwS

Genetics of dilated cardiomyopathy:


practical implications for heart failure
management
Andrew N. Rosenbaum   1, Katherine E. Agre   2 and Naveen L. Pereira1,3,4*
Abstract | Given the global burden of heart failure, strategies to understand the underlying
cause or to provide prognostic information are critical to reducing the morbidity and mortality
associated with this highly prevalent disease. Cardiomyopathies often have a genetic cause,
and the field of heart failure genetics is progressing rapidly. Through a deliberate investigation,
evaluation for a familial component of cardiomyopathy can lead to increased identification of
pathogenic genetic variants. Much research has also been focused on identifying markers of risk
in patients with cardiomyopathy with the use of genetic testing. Advances in our understanding
of genetic variants have been slightly offset by an increased recognition of the heterogeneity of
disease expression. Greater breadth of genetic testing can increase the likelihood of identifying a
variant of uncertain significance, which is resolved only rarely by cellular functional validation and
segregation analysis. To increase the use of genetics in heart failure clinics, increased availability
of genetic counsellors and other providers with experience in genetics is necessary. Ultimately,
through ongoing research and increased clinical experience in cardiomyopathy genetics, an
improved understanding of the disease processes will facilitate better clinical decision-​making
about the therapies offered, exemplifying the implementation of precision medicine.

The global prevalence of heart failure is approximately discussion of the pragmatic use of genetic testing in
26 million patients, and the current estimates of the heart failure clinics for patients presenting with new-​
incidence and prevalence of heart failure in the USA are onset cardiomyopathy. We aim to provide clinicians with
nearly 1 million per year and >6 million patients, respec- a focused, evidence-​based guide to the use of genetic
tively1–3. Although some marginal declines in the inci- testing to facilitate the management of genetic cardio­
dence of heart failure can be attributed to a reduction in myopathies. An extensive overview of the molecular
risk factors for cardiovascular disease and improvements mechanisms and pathogenesis of DCM is not provided,
1
Department of
in management, the need to identify and treat heart nor is a comprehensive summary of all the genetic vari-
Cardiovascular Medicine, failure is growing, particularly given an overall 5-year ants that have been identified, but these areas have been
Mayo Clinic, Rochester, survival of only 50% after diagnosis of heart failure4. reviewed previously14–16.
MN, USA. The global economic burden related to heart failure is
2
Department of Clinical approximately US$120 billion3 and, in the USA, $30 bil- Background
Genomics, Mayo Clinic,
lion of spending per year is estimated to be attributable Defining idiopathic and familial DCM
Rochester, MN, USA.
to direct and indirect costs resulting from heart failure Although generally straightforward in clinical practice,
3
William J. von Liebig Center
for Transplantation and
care5. The large burden of this disease means that devel- the diagnostic criteria for cardiomyopathies are important
Clinical Regeneration, Mayo oping effective management strategies is paramount. to consider in the context of applying appropriate genetic
Clinic, Rochester, MN, USA. Dilated cardiomyopathy (DCM) is the second most considerations to the diagnostic work-​up. DCM is gener-
4
Department of Molecular common cause of heart failure (after coronary artery ally defined as a left ventricular ejection fraction (LVEF)
Pharmacology and disease), underlying approximately 36% of all heart fail- of <45%, with a left ventricular end-​diastolic dimension
Experimental Therapeutics, ure cases, and has an estimated prevalence of >0.4% in >112% of that predicted for age and body surface area17.
Mayo Clinic, Rochester,
MN, USA.
the general population6–8. Of these patients, 25–30% are The term ‘idiopathic’ can be applied only after exclusion
estimated to have an identifiable familial component (as of the vast array of potential causes, many of which require
*e-​mail: pereira.naveen@
mayo.edu defined below)7,9–13. simple, non-​invasive tests and, increasingly, the use of
https://doi.org/10.1038/ In this Review, we give a broad perspective on cardiac MRI, with an emphasis on the clinical history18.
s41569-019-0284-0 the genetic causes of DCM to provide a context for a Familial DCM is diagnosed when two or more family

Nature Reviews | Cardiology


Reviews

can have a higher or lower yield. For example, a genetic


Key points
cause of hypertrophic cardiomyopathy (HCM) can be
• The frontier of genetics is rapidly advancing and, increasingly, genetic testing in heart identified in ≥50% of patients on the basis of the pres-
failure clinics is associated with benefit for family screening and individual ence of various clinical factors7. As in HCM, identify-
prognostication. ing the genetic cause of DCM can provide substantial
• A focused evaluation of the clinical characteristics and inheritance pattern of heart clinical insight into family screening, management and
failure or sudden cardiac death, in addition to consultation with a genetic counsellor prognosis29,30. However, other cardiomyopathies, such as
when appropriate, can facilitate a successful genetic evaluation.
restrictive or noncompaction cardiomyopathy, are less
• Genetic testing is recommended in all patients with familial dilated cardiomyopathy well described and might have a lower degree of famil-
(DCM) to facilitate screening, whereas guideline recommendations for testing in
ial involvement and a variable rate of identification of
patients with sporadic DCM differ, but specific clinical features might increase the
the pathogenic variant, which might partly be caused
yield of testing.
by differences in the interpretation or diagnosis of the
• Genetic testing in DCM is currently associated with the identification of a culprit
phenotype31–34.
variant in approximately 15–25% of patients with sporadic DCM and approximately
20–40% of patients with familial DCM.
Genetic aetiology in heart failure
• The identification of a pathogenic variant might have important predictive and
therapeutic implications; however, expression of the phenotype might depend on A targeted, three-​generation family history is of the
environmental triggers. utmost importance when one is assessing whether car-
• All first-​degree relatives of patients with familial DCM should undergo clinical
diomyopathy is sporadic or familial. In this Review, we
screening; guidelines for screening of first-​degree relatives of patients with sporadic use the term ‘sporadic’ to refer to any proband without a
DCM differ. family history of phenotypic manifestation of the disease
(more precisely termed ‘simplex’)7. The family history
also provides data on the likelihood of identifying a
members meet the criteria for DCM or when the patient genetic cause and is critical to evaluating the possibility
of interest (proband) meets the criteria for DCM and a of a syndromic association35. Table 2 provides a list of
first-​degree relative has experienced sudden cardiac death some common syndromic causes of DCM, with clinical
aged <35 years19. However, these definitions are arbitrary phenotype–gene correlations. Ascertainment of a ped-
and often difficult to document conclusively. In general, igree with phenotypic traits allows strategies for testing
familial DCM should be suspected when family members and for the evaluation of relatives to be determined36,37.
of the proband have been reported to have heart failure or Important elements of a family history include the
premature sudden cardiac death without a well-​defined age of relatives, health status, cardiac screening status of
cause. In addition, if left ventricular dilatation without a first-​degree relatives and cause of death (when applicable),
reduction in LVEF is detected during screening of first-​ with appropriate subsequent questions, particularly in the
degree family members, a diagnosis of familial DCM setting of red flags (Box 1). Furthermore, ascertainment of
should also be considered. ancestral background and the presence of consanguinity
is important because these factors affect the frequency of
Frequency of genetic variants potentially disease-​causing recessive alleles. Reviewing
A variant in the genome that has a ≥90% likelihood of death certificates and autopsy reports can provide further
causing DCM is referred to as a pathogenic or likely path- and more accurate insights into the cause of death.
ogenic genetic variant20. In contrast to many Mendelian Suspicion of familial cardiomyopathy should be high
disorders, genetic associations present in familial cardio­ in scenarios in which a family has more than one
myopathies have variable penetrance, and manifes- affected individual and in which a family member has
tations might occasionally be observed only when experienced sudden cardiac death. However, potential
exposure to an additional insult occurs21,22. Furthermore, limitations to the pedigree approach must be acknowl-
estimates of a genetic cause of DCM are solely on the edged, which include a lack of family history data, lim-
basis of well-​characterized known pathogenic or likely ited family structure and incomplete family cardiac
pathogenic genetic variants that have been identified screening.
so far; many more culprit genetic variants are likely to Elucidating a heritable link between family members
exist23,24. Nevertheless, estimates of the frequency of of a patient with cardiomyopathy can be difficult because of
underlying pathogenic variants range from 15% to 25% in incomplete penetrance and variable expressivity (such as
unselected patients with DCM and from 20% to 40% asymptomatic ventricular dysfunction and/or dilatation,
in patients with familial DCM25–28. Table 1 details the manifestations due to conduction tissue disease, heart
ten genes that have been most commonly implicated failure and the particularly challenging occurrence of sud-
in DCM and their estimated frequency in non-​syndromic den cardiac death)13,38. Subclinical or early clinical disease
DCM. Figure 1 shows the location in a cardiomyocyte that might not be apparent from the family history could
of the proteins encoded by these ten genes. Of note, be present in up to 29% of affected families39.
the likelihood of identifying a culprit genetic vari-
ant (the ‘yield’) is higher in syndromic DCM than in Genetics in the management of DCM
non-​syndromic DCM, particularly in those patients Clinical guidelines
with coexistent muscular dystrophies,  conduction Clinical guidelines have assisted clinicians in determin-
defects or hearing loss13. ing when to use genetic testing to aid in the diagnosis and
Compared with genetic testing in patients with DCM, management of idiopathic DCM. Each guideline empha-
genetic testing in patients with other cardiomyopathies sizes a thorough family history and physical examination

www.nature.com/nrcardio
Reviews

Table 1 | Top ten genes in which dCM-​associated variants most commonly occur7,28,35
gene (protein) Protein Estimated age of onset of dCM associated Notes ref.
function prevalence in (years) disorders
dCM (%)
TTN (titin) Sarcomere, 12–25a Usually 40–59 Usually isolated More common in women than 54

structural DCM in men; variable prognosis but


family clustering
LMNA (lamin A/C) Nuclear 4–6 (up to 30 30–49; conduction Subset of Emery– Most conduction disease 46

membrane if conduction disease usually Dreifuss muscular cases symptomatic over time;
disease also prominent dystrophy ventricular arrhythmias common
present)
MYH7 (β-​myosin Sarcomere 4–10a Variable: adolescence Primarily HCM; Prognosis variable depending 64

heavy chain) to 49 also LVNC and on site of genetic variant;


RCM homogeneity within families
MYH6 (α-​myosin Sarcomere 4 Variable: adolescence Primarily HCM Prognosis variable depending 64

heavy chain) to 49 on site of genetic variant;


homogeneity within families
MYPN (myopalladin) Sarcomere, 3–4 Variable: adolescence Usually isolated Variable presentation with some 70

Z-​disc to 59 DCM early deaths due to heart failure


DSP (desmoplakin) Desmosome 3–4 40–49 ARVC Identified in multiple patients 79

with advanced DCM although


natural history uncertain;
arrhythmic burden seems to be
much lower than in ARVC
RBM20 (RNA-​binding RNA-​binding 2–5 Adolescence to 39; can Usually isolated Rapid progression: SCD and 84

protein 20) protein, present with SCD or DCM end-​stage heart failure are
spliceosome advanced heart failure common, with a high incidence
of ventricular arrhythmias
TNNT2 (cardiac Sarcomere 2–3 Variable: adolescence HCM, LVNC and Prognosis variable depending 64

muscle troponin T) to 49 RCM on site of genetic variant;


homogeneity within families
SCN5A (sodium Ion channel 2–3 Adolescence Brugada syndrome Early onset, commonly with atrial 87

channel protein type and LQTS fibrillation; not associated with


5, α subunit) risk of ventricular arrhythmias
TPM1 Sarcomere 0.5–1.0 Variable: adolescence HCM, LVNC and Prognosis variable depending 64

(α-​tropomyosin) to 49 RCM on site of genetic variant;


homogeneity within families
ARVC, arrhythmogenic right ventricular cardiomyopathy ; DCM, dilated cardiomyopathy ; HCM, hypertrophic cardiomyopathy ; LQTS, long QT syndrome; LVNC;
left ventricular noncompaction cardiomyopathy ; RCM, restrictive cardiomyopathy ; SCD, sudden cardiac death. aHigher end of the range if DCM is familial.

in patients with DCM and appropriate screening of that interval screening should occur for phenotypic
first-​degree family members of affected patients. manifestations, in addition to appropriate disease
Regarding screening for familial DCM, the 2013 counselling25,41. In the absence of a family history or an
ACC/AHA heart failure guidelines40, the 2009 Heart identifiable pathogenic genetic variant, the ACC/AHA
Failure Society of America (HFSA) guidelines29 (which guidelines recommend that clinical screening of fam-
were updated in 2018 in a joint publication with the ily members of patients with sporadic idiopathic DCM
American College of Medical Genetics (ACMG)25), should be left to the discretion of the family members’
the 2010 ESC guidelines for genetic testing and counsel- physicians and is not routinely recommended40. The
ling41 and the 2016 AHA scientific statement on DCM42 AHA scientific statement on DCM similarly indicates
recommend clinical screening of adult first-​degree rela­ that clinical screening of first-​degree relatives might be
tives of affected individuals, with electrocardiographic reasonable42. However, because of uncertainties about
and echocardiographic examination of left ventricular the distinction between familial and sporadic cardiomy-
function and size every 3–5 years. Additionally, accord- opathies, the ESC recommends routine clinical screen-
ing to a 2011 joint expert consensus statement from ing in all first-​degree relatives of patients with DCM if
the Heart Rhythm Society (HRS) and European Heart genetic information is unavailable41.
Rhythm Association (EHRA)43, if a pathogenic or likely Regarding genetic testing in familial DCM specifi-
pathogenic variant is present in a proband, cascade cally, the updated 2018 HFSA/ACMG guidelines and
screening should be conducted; this recommendation the 2010 ESC guidelines recommend testing of the most
is supported by the guidelines from the ACMG, ESC affected family member25,41. The 2013 ACC/AHA guide-
and AHA25,41,42. If a family member undergoes genetic lines and the 2016 AHA scientific statement indicate that
screening and a pathogenic variant is identified in the genetic testing can be considered together with genetic
absence of phenotypic manifestations, both the 2018 counselling, although specific recommendations about
HFSA guidelines and the 2010 ESC guidelines indicate whom to test are not given40,42.

Nature Reviews | Cardiology


Reviews

Sodium channel cells45,46. LMNA variants most commonly result in both


protein type 5 (SCN5A) DCM and conduction system disease, with the for-
Plasma membrane mer generally occurring in the fourth to fifth decade
Cytoplasm of life, eventually requiring pacing in most patients46.
Myosin heavy chain Cardiac muscle α-Tropomyosin Additionally, the risk of sudden cardiac death is high
Titin (TTN) (MYH6 and MYH7) troponin T (TNNT2) (TPM1) (up to 46%) in patients with LMNA variants compared
with controls (OR 3.7, 95% CI 2.3–5.9, P < 0.001), and
many patients require pacemaker implantation47.
The prevalence of LMNA variants as a cause of DCM
is not definitively known, but general estimates range
from 4% to 8%28,46,48. In a highly selected population
of tertiary referral patients with familial DCM, LMNA
Myopalladin Z-disc variants were identified in 30% of those presenting with
(MYPN) DCM and conduction system disease, in 88% of those
Nucleus Lamin A/C
(LMNA) presenting with DCM and a muscular dystrophy syn-
Desmoplakin drome and even in 25% of those presenting with isolated
(DSP) conduction system disease without DCM49.
RNA-binding
protein 20 (RBM20) Sarcomeric protein cardiomyopathies. Variants in
Desmosome Mitochondrion
genes that encode sarcomeric proteins, which are
involved in contractile force generation and cellular
anchoring, underlie a range of cardiomyopathies, from
Fig. 1 | Cellular locations of proteins associated with dCM. The graphic shows the hypertrophic remodelling to ventricular dilatation, and
approximate cellular locations of the ten proteins and their respective genes in which phenotypic variability is common50. Variants in some
dilated cardiomyopathy (DCM)-causing pathogenic variants most commonly occur genes encoding sarcomeric proteins that are associated
(see Table 1).
with DCM might exert their effect through impaired
responses to physiological stress51,52. The sarcomeric
The guidelines differ somewhat with respect to spo- protein of greatest attention in the context of DCM is
radic DCM (in the absence of a family history of the titin, encoded by TTN. Several types of variants in TTN
disease) largely because of the unknown diagnostic yield that cause truncation of the protein have been identi-
and clinical utility of genetic screening in this popu­ fied53. Variants in TTN might cause up to 25% of cases
lation. The ESC and ACC/AHA guidelines emphasize of familial DCM, inherited in an autosomal-​dominant
that, unless particular clinical features (a family history manner, and 12–18% of cases of sporadic DCM54,55.
of cardiac conduction disease, premature sudden cardiac Variants in TTN are not generally associated with car-
death or the recommendation of particular therapies) diac conduction defects or skeletal muscle disease53,54.
are present, routine genetic testing is not systematically The presentation of titin cardiomyopathy is generally
recommended40,41, whereas the HFSA/ACMG guidelines similar to that of idiopathic forms of DCM, with onset
recommend that genetic testing can facilitate cascade in the fifth to sixth decade of life55. Prognosis is also
screening and should be performed in all patients with similar, but patients with titin cardiomyopathy might
DCM25. The HRS/EHRA guidelines recommend testing have a better response to medical therapy than those
in patients with DCM and clinically significant cardiac with idiopathic DCM56.
conduction disease or a family history of premature, Putative pathogenic variants have also been identified
unexpected death43. in genes encoding proteins involved in the sarcomeric
As a result of publications describing a new pheno- contractile apparatus, namely (in order of prevalence)
typic paradigm of arrhythmogenic cardiomyopathy, MYH7 (encoding myosin 7, also known as β-myosin
emphasizing the substantial overlap between DCM, heavy chain), MYH6 (encoding myosin 6, also known
arrhythmogenic cardiomyopathy and heritable arrhyth- as α-​myosin heavy chain), TNNT2 (encoding cardiac
mia syndromes, the HRS has released an updated con- muscle troponin T), MYBPC3 (encoding cardiac-​type
sensus document44 to address comprehensively the myosin-​binding protein C), TNNI3 (encoding cardiac
unique electrophysiological considerations (including muscle troponin I), TPM1 (encoding α-​tropomyosin),
mechanisms, implantable cardioverter–defibrillator MYL2 (encoding myosin regulatory light chain 2), MYL3
therapy and management of dysrhythmia) as well as gen- (encoding myosin light chain 3) and ACTC1 (encoding
eral management of cardiomyopathies with arrhythmia α-​cardiac actin)50,57–60. More frequently identified in
as a prominent component. HCM, MYBPC3 variants constitute 30–40% of HCM
pathogenic variants, and MYH7 variants constitute
Genetic variants causing DCM 20–30% of HCM pathogenic variants57,61–63. However,
Laminopathies. Lamin A/C is a nuclear envelope protein variants in these genes have also been discovered to con-
encoded by LMNA that functions as a support element, tribute to DCM, with phenotypic heterogeneity driven
with a structure homologous to that of intermediate fil- by mutation locus64,65. The proportion of familial DCM
aments. The pathogenesis of LMNA-​associated DCM that results from variants in genes that encode contrac-
involves disruption of chromatin organization in divid- tile proteins might be as high as 10%, with substantial
ing cells and of signal transduction in non-​dividing phenotypic heterogeneity64.

www.nature.com/nrcardio
Reviews

Variants also occur in genes encoding proteins of the mechanical and signalling functions. Variants in genes
Z-​disc, which can similarly result in DCM phenotypes. encoding desmosomal proteins are traditionally associ-
DCM-​causing variants have been identified in ACTN2 ated with arrhythmogenic right ventricular cardiomyop­
(encoding α-​actinin 2), ANKRD1 (encoding ankyrin athy. However, left ventricular dysfunction can occur in a
repeat domain-​containing protein 1), CSRP3 (encoding DCM pattern independently of right ventricular involve-
muscle LIM protein), LDB3 (encoding LIM domain-​ ment and, therefore, the true pathogenesis of arrhythmo-
binding protein 3), MYOZ2 (encoding myozenin 2), genic right ventricular cardiomyopathy or DCM
TCAP (encoding telethonin) and VCL (encoding vin- secondary to variants in genes encoding desmoso­mal
culin)66–69. Variants in MYPN, encoding myopalladin, proteins is incompletely understood78.
another important Z-​disc-associated protein with struc- The major culprit variants for DCM seem to be
tural and signalling functions, were found to occur in located in the desmosomal genes DSP (encoding desmo-
European patients with DCM at a frequency of 3–4%70. plakin) and PKP2 (encoding plakophilin 2). Pathogenic
variants in these genes might be under-​recognized.
Myofibrillar cardiomyopathy. Beyond the sarcomere One report described a prevalence of 13% for patho-
unit, intermediate filaments maintain the structural genic variants in genes encoding desmosomal proteins
integrity of the myofibril, allow force generation and among unselected patients undergoing cardiac trans-
facilitate signalling. The main intermediate filament plantation for idiopathic DCM79, whereas other studies
protein in both cardiomyocytes and skeletal myocytes of unselected patients with isolated DCM have identified
is desmin, encoded by DES71. Variants in DES are gen- DSP as being the fourth most common gene to have
erally inherited in an autosomal-​dominant fashion and DCM-​causing variants28.
characteristically present with conduction defects caus-
ing syncopal events or even sudden cardiac death71,72. Tafazzin-​related cardiomyopathy. Variants in TAZ (also
DES variants have also been associated with restrictive known as G4.5), which encodes a variety of proteins
cardiomyopathy, with a characteristic presentation of involved in the regulation of cardiolipin, namely the taf-
concomitant atrioventricular block73. azzin proteins, can cause Barth syndrome (features of
Pathogenic variants in FLNC, encoding another which include DCM, short stature, lactic acidosis and neu-
structural protein, filamin C, were previously thought to tropenia) in infants80,81. However, other variants in TAZ
affect both skeletal and cardiac muscle, but research has can result in a less severe phenotype of DCM. Variants in
now confirmed a cardiac-​specific, DCM phenotype74. DTNA, encoding dystrobrevin-​α, might cause an X-​linked,
Pathogenic variants in FLNC are often highly pene- noncompaction cardiomyopathy phenotype80,82,83.
trant and associated with a high risk of sudden cardiac
death75,76. Guidelines give a class IIa recommendation for Other genetic variants associated with cardiomyopathies.
cardioverter–defibrillator implantation if a pathogenic Several other genetic variants have been associated with
FLNC variant is present and the LVEF is <45%44. DCM, with diverse pathogenic mechanisms. Variants in
Another cause of myofibrillar cardiomyopathy is var- RBM20, which encodes RNA-​binding protein 20, which is
iants in BAG3, which encodes an intracellular chaperone involved in providing mRNA post-​transcriptional modifi-
protein. In a severe phenotype, BAG3 variants result in cations in spliceosomes, were identified in 2009 as a cause
severe muscular dystrophy in children77. of DCM84. Rapid progression towards advanced heart fail-
ure is not uncommon by the fourth decade of life, and the
Desmosomal cardiomyopathy. Desmosomes are pro- occurrence of ventricular arrhythmias and sudden cardiac
teins involved in the structure and function of the death is high in patients with RBM20 variants84,85.
intercalated discs, which are important for transmis- Variants in SCN5A, encoding the α-​subunit of the
sion of electrical activity from cell to cell and also have cardiac sodium channel, are typically associated with
Brugada syndrome or long QT syndrome but might
also increase the propensity for heart failure86. The
Table 2 | dCM clinical phenotype–gene associations DCM phenotype of individuals with SCN5A variants can
distinguishing clinical features of dCM at Suspected variant gene include conduction disturbance and arrhythmias86. The
presentation or syndrome age of onset of cardiomyopathy associated with SCN5A
DCM with prominent conduction disease LMNA variants is early, in the second to third decade of life87.
DCM or restrictive cardiomyopathy with conduction DES or LMNA
disease or sudden cardiac death and skeletal myopathy Important syndromic DCMs
Mitochondrial myopathies. An extensive range of car-
Early-​onset, rapidly progressive DCM with ventricular RBM20
arrhythmias diomyopathies are associated with mitochondrial disor-
ders. Mitochondrial myopathies display two inheritance
Early-​onset DCM with history of muscular dystrophy DMD patterns: maternal transmission (owing to segregation
Early-​onset, rapidly progressive DCM with X-​linked DMD (X-​linked DCM) of mitochondria in dividing cells) and autosomal dom-
inheritance pattern, without muscular dystrophy inant (because some nuclear proteins also interact with
Ophthalmoplegia, conduction defects and DCM Kearns–Sayre syndrome mitochondria and can cause disease)88. Genetic testing for
(mitochondrial) mitochondrial myopathies relies on gene sequencing
Myoclonic epilepsy , septal hypertrophy and DCM MERRF syndrome of mitochondrial DNA for known pathogenic variants;
(mitochondrial) unlike with nuclear genes, phenotypic expression of vari-
DCM, dilated cardiomyopathy ; MERRF, myoclonus epilepsy with ragged red fibres. ants in mitochondrial genes is proportional to the number

Nature Reviews | Cardiology


Reviews

Box 1 | Family history red flags Becker muscular dystrophy can manifest with a
pattern similar to that of Duchenne muscular dystro-
Any of the following findings in the family history should prompt further investigation: phy, although cardiac involvement is not universal and,
• Sudden cardiac death when present, the age of onset of DCM tends to be later.
• Sudden infant death syndrome A more severe form of cardiomyopathy associated with
• Unexplained syncope the DMD gene is X-​linked DCM, which does not involve
• Seizures without objective ictal activity skeletal muscle and is associated with early onset (in the
• Heart transplantation in the absence of coronary artery disease second decade of life) and rapid progression93.
• Presence of a pacemaker and/or implantable cardioverter–defibrillator in a young
LMNA variants are associated with a number of
person (aged <60 years) autosomal-​dominant disorders of the skeletal muscle.
• Muscle weakness or myopathy
In Emery–Dreifuss muscular dystrophy and limb gir-
dle muscular dystrophy type 1B, progressive wasting of
• Drowning of an experienced swimmer
skeletal muscle occurs46, but the cardiac manifestations
• Motor vehicle accident in the absence of intoxication or vehicle malfunction
(such as DCM, atrioventricular block and ventricular
• Individual who died of a supposed heart attack at a young age, in the absence of arrhythmias) are similar to those in diseases associated
known coronary risk factors and with no autopsy performed
with LMNA variants in which a cardiac phenotype pre-
dominates94,95. Phenotypic cardiac manifestations in
of mutant mitochondria in the cell88. A definitive diag- Emery–Dreifuss muscular dystrophy and limb girdle
nosis is made only after pathology examination involv- muscular dystrophy type 1B are driven by the location
ing assays for mitochondrial quantity and function or by of the variant within the LMNA gene96. Emery–Dreifuss
genetic testing89. Together, mitochondrial DNA-​related muscular dystrophy can also be associated with an
diseases constitute approximately 3% of DCMs88. X-​linked inheritance pattern through variants in EMD,
Despite the variety in presentation, the more com- which encodes emerin, a nuclear laminar protein.
mon mitochondrial myopathy syndromes can have
characteristic cardiac features. For example, in Kearns– Types of genetic testing
Sayre syndrome, which often presents by adolescence Candidate genetic variant testing. Testing for specific
and is characterized by progressive ophthalmoplegia and variants in an asymptomatic individual is recommended
retinal degeneration, cardiac conduction abnormalities if a first-​degree relative has DCM that is associated with
(especially heart block) are present in most patients88–90. a pathogenic or likely pathogenic gene variant; this
In MELAS (mitochondrial encephalopathy, lactic aci- approach is referred to as predictive testing because the
dosis and stroke-​like episodes) syndrome, heart failure individual does not have symptoms or known structural
can develop but is characteristically accompanied by disease. In the presence of specific clinical findings, such
substantial and symmetrical left ventricular hypertro- as conduction tissue disease, advanced heart block in the
phy, which can present in childhood or adulthood88,90. setting of DCM, or the presence of concomitant skeletal
In MERRF (myoclonus epilepsy with ragged red fibres) muscle involvement in the form of an asymptomatic ele-
syndrome, which usually manifests during childhood, vation in creatine kinase level or a manifest neuromus-
substantial left ventricular dysfunction can occur90. cular disorder, the yield of genetic testing is high, and
targeted single-​gene testing or the use of a syndromic
Lysosomal storage disorders. Lysosomal storage disor- cardiomyopathy gene panel can be considered97.
ders include diseases resulting in the accumulation of
specific cellular material, with pathogenesis involving Cardiomyopathy gene panel testing. Various commer-
the heart. The presentation, manifestations and severity cially available gene panels are routinely used to iden-
of the disease processes differ greatly. The cardiac man- tify a genetic variant in cardiomyopathies. The number
ifestations of these lysosomal storage diseases have been and types of genes offered for sequencing differ between
reviewed previously91. panels. The genes have typically been found to be asso-
ciated with cardiomyopathies in previous functional
Neuromuscular syndromes. The archetype of a neuro- studies or by familial segregation analysis. A study eval-
muscular syndrome affecting the heart is Duchenne uating a large gene panel of 46 genes in 766 unselected
muscular dystrophy. Genetic variants in DMD, encoding patients with DCM indicated that up to 37% might have
dystrophin, are associated with an X-​linked, recessive an underlying culprit variant28. In smaller gene panels
pattern of inheritance. Like desmin, dystrophin con- of five to ten genes, the yield of testing was <10%, but
nects the sarcomeric proteins to the plasma membrane increased to >30% with a larger panel (46 genes), albeit
via the dystroglycan complex. In Duchenne muscular with a high rate of variants of uncertain significance
dystrophy, muscle contraction results in the loss of (VUSs; 60%)28. Figure 2 shows an algorithm to deter-
cell membrane integrity and eventually cell death and mine whether cardiomyopathy gene panel testing should
fibrofatty replacement92. Duchenne muscular dystrophy be considered.
cardiomyopathy typically manifests in the second dec-
ade of life and always involves a clear history of skeletal Whole-​exome sequencing. The clinical use of whole-​
myopathy92. Given that cardiac involvement is inevita- exome sequencing is increasing as the cost associated
ble if death from other causes is averted, screening for with this form of testing has decreased considerably
cardiomyopathy is essential and is performed yearly in over time, and information about the presence of var-
adult patients92. iants in unrelated genes can be obtained. Whole-​exome

www.nature.com/nrcardio
Reviews

sequencing can also facilitate the identification of in the proband. Third, whole-​exome sequencing gener-
novel variants, particularly in genes not known to be ally detects single-​nucleotide substitutions but can miss
involved in pathogenesis, primarily by trio testing (test- larger structural genetic changes, such as copy-​number
ing a proband and both parents to determine whether variants, long insertions or deletions, or repetitive DNA
the variant segregates with the disease)98. The rate of segments, which can result in changes in gene expres-
identification of a pathogenic variant in selected, pre- sion. Additionally, as indicated by the name, this type of
viously undiagnosed Mendelian disorders can be as testing does not comprehensively sequence non-​coding
high as 25%99. However, one study evaluating the utility regions (intergenic areas and introns); variation in these
of whole-​exome sequencing in HCM found that novel regions can be involved in disease pathogenesis through
variants not included in the standard gene panel were the regulation of gene expression19. Given the limitations
identified in 9% of patients100. in our knowledge of the functional importance of var-
The routine use of whole-​exome sequencing is asso- iants in intronic or intergenic regions, whole-​genome
ciated with substantial limitations101. Most importantly, sequencing is not routinely used in clinical practice.
the depth of coverage is limited to the genes of interest,
so the test results must be carefully interpreted because Counselling before genetic testing
several issues can arise, particularly the misattribution Pretest counselling should be provided to all patients
of a variant as being pathogenic. Second, the yield of with DCM, ideally by an experienced provider with
trio testing for the identification of novel variants can knowledge of DCM genetics, such as a genetic counsel-
be low if that pathogenic variant has occurred de novo lor, to facilitate patient understanding of the rationale

DCM

Known DCM-related genetic Exclude known causes of DCM


variant in family member?

High-strength recommendations Family history


• Disease-appropriate screening Yes No • Two or more family members
• Take a targeted family history with DCM?
• Genetic counselling referral • First-degree relative with sudden
• Appropriate management according cardiac death aged <50 years?
to disease process
Yes No
Familial Sporadic

Intermediate-strength recommendations Intermediate-strength recommendations


• HFSA/ESC: Consider testing the • HFSA: Consider testing all patients
most-affected family member with new-onset DCM
• ACC/AHA: Consider genetic testing • ESC: Consider testing for rare or
• Referral to an expert in cardiovascular specific cardiomyopathy, especially
genetics to help to guide counselling if there are therapeutic implications
• ACC/AHA: Consider testing if conduction
disease present or family history of
sudden cardiac death

Genetic testing

Pathogenic or likely Variant of uncertain Benign or likely No variant


pathogenic variant significance benign variant

Genetically Monitor for variant


proven DCM reclassification over time

High-strength recommendations Idiopathic DCMa


• Institute appropriate disease-management
strategies
• Initiate cascade screening of family
• Genetic counselling referral for changes
in variant reclassification over time

Fig. 2 | genetic testing in non-​syndromic dCM. Recommended algorithm for the consideration of genetic testing in
patients with non-​syndromic dilated cardiomyopathy (DCM). HFSA , Heart Failure Society of America. aFor idiopathic DCM,
first-​degree relatives continue to be at elevated risk; consider retesting in light of the discovery of additional culprit genes.

Nature Reviews | Cardiology


Reviews

for testing, increase empowerment and improve com- by more than two levels from the classification obtained
munication and dynamics within families. Given that by an evidence-​based algorithm applied by a group of
patients often communicate the results and implications clinicians104. Similar rates of interlaboratory variability
of genetic testing to at-​risk family members, their under- have been observed in other studies105,106. As a result of
standing of this information is imperative. Additionally, this heterogeneity in interpretation, an attempt to cen-
although the cost of genetic testing is decreasing, many tralize variant classification has been initiated, initially
patients face financial concerns especially for testing using MYH7 as a prototype because of the prevalence of
that is considered elective and does not directly affect pathogenic variants in this gene107.
medical care. Therefore, understanding the financial
implication of genetic testing for patients is important, VUS: determining significance. Often, genetic testing is
particularly because insurance coverage is not uniform. inconclusive owing to the absence of a variant or the pres-
However, a genetic counsellor is not trained to provide ence of a benign variant or one or more VUSs. Adherence
financial advice about testing. to the criteria outlined by the ACMG for determining
Patients must also understand issues relating to the pathogenesis of a variant is important20,107. In some
the privacy of their genetic testing results and the scenarios, the genetic significance of a VUS can be deter-
potential implications for insurability for themselves mined by performing a segregation analysis in family
and their family members. The Genetic Information members. Many genetic laboratories offer free testing
Nondiscrimination Act (GINA) is a federal law in the of family members if a VUS is found that meets certain
USA that prohibits the use of genetic information for criteria. However, this approach should be interpreted
discrimination by employers and health insurers. cautiously because segregation analysis does not always
However, this law has limitations because it does not determine causality, and penetrance can vary108. In vitro
apply to life, long-​term care or disability insurance. functional characterization of a VUS in induced pluri-
Exceptions also exist for some employers and forms of potent stem cell-​derived cardiomyocytes has also been
health insurance (such as for government employees and useful in determining its pathogenicity109.
active members of the military). Patients and families
might have difficulty obtaining life and health insurance, Cascade screening. The identification of a pathogenic or
even when seeking care for a pre-​existing condition102. likely pathogenic variant in a DCM-​susceptibility gene
Because private insurance has a smaller role in Europe, is an indication for family cascade screening. Testing
few countries have formally regulated the use of genetic of asymptomatic family members guides the necessity
testing for risk assessment, but several European coun- for echocardiographic screening and is a cost-​effective
tries provide protection against the use of genetic testing approach. If an at-​risk family member tests negative for
information for determining life insurance premiums or a previously identified pathogenic or likely pathogenic
eligibility. Most countries in Asia do not have similar variant in the context of a negative baseline cardiac
regulatory protection103. evaluation, ongoing cardiac evaluation is not usually
Pretest counselling provides a patient with the best warranted, depending on the strength of the association
opportunity to evaluate a wide range of genetic testing between the variant and the disease25. Family members
options, which hinges on risk assessment. As described who test positive for a previously identified pathogenic
above, genetic testing can range from site-​specific or or likely pathogenic variant should receive tailored
gene-​specific testing to large disease-​specific panels to cardiac screening and preventative measures. Cascade
whole-​exome sequencing. The implications and possible screening in first-​degree relatives can also help to deter-
results of these tests differ widely, and setting appropriate mine the need to perform genetic screening and cardiac
expectations for patients before they undergo a genetic evaluations in distant relatives.
test is important, especially with whole-​exome sequenc-
ing, which can result in secondary genetic findings that Genetic risk factors and prognosis
have other health implications. Additionally, many syn- Markers of genetic susceptibility. Genome-​wide asso-
dromic causes of DCM exist, so obtaining a syndromic ciation studies (GWAS) have sought to discover novel
gene panel is imperative if a patient is suspected of hav- genetic markers to predict the development of DCM;
ing a syndromic form of DCM. Fortunately, comprehen- however, these markers have not been implemented in
sive cardiomyopathy gene panels usually include genes clinical practice. The single-​nucleotide polymorphisms
that have been implicated in syndromic causes of DCM. (SNPs) identified in these studies suggest that common
genetic variation can confer increased risk at a popu-
Interpreting genetic test reports lation level110–113. However, associations are challenging
ACMG guidelines. The ACMG published a set of because attribution of causality can be particularly diffi-
guidelines in 2015 for the standardized interpretation cult in intronic regions, and mechanisms must be sought
of genetic variants. These guidelines have helped to for variation in novel loci114,115.
provide consistent terminology between laboratories The use of GWAS has also yielded potential insights
and clinicians in the classification of variants as being into differential risk and disparate outcomes according
pathogenic, likely pathogenic, a VUS, likely benign or to ethnicity116. Unique genetic risk markers in African
benign20. However, despite the objectivity of the criteria, American individuals have been discovered that are
differences in interpretation persist between laborato- associated not only with left ventricular remodelling
ries and clinicians, with one study indicating that the but also with incident heart failure events, and these
laboratory classifications of 18% of 688 variants differed risk markers are not present in similar European

www.nature.com/nrcardio
Reviews

cohorts117,118. Studies that implicate common genetic var- least one D allele of the ACE gene was associated with
iation as risk loci need to be replicated in large, diverse lower survival, especially if the patients were not receiv-
and well-​characterized populations and functionally val- ing β-​blocker therapy131. However, in another trial in
idated before being used in the clinic in the form of risk which patients were stratified according to the presence
scores for disease prediction. of previously identified pathogenic variants in AGTR1,
no differences in long-​term mortality were identified132.
Use of genetics in prognosis. In addition to predicting the Although not associated with a large effect size, multiple
risk of developing DCM, considerable interest exists in SNPs have been identified that increase the risk of ACE
using genetic markers to determine prognosis in DCM inhibitor-​induced angio-​oedema133. Routine testing for
and to help with risk stratification. For example, patients SNPs associated with differential responses to neuro-
with pathogenic LMNA variants might be at increased hormonal blockade is not routinely performed in the
risk of ventricular arrhythmias, irrespective of the degree clinical setting owing to a lack of clinical validation and
to which LVEF is reduced, meriting the implantation of utility, but continued research to individualize medical
a cardioverter–defibrillator for the primary prevention therapy holds promise to improve outcomes134.
of sudden cardiac death119,120. SNPs can also be inform-
ative for risk stratification. For example, in a large-​scale Gene–environment interaction. The interaction between
GWAS, one SNP on chromosome band 5q22 was asso- environmental factors and the underlying genetic make-​
ciated with a 36% increased risk of death in patients with up of an individual is critically important. Analogous
heart failure121. Other GWAS have identified an associa- to a multiple-​hit hypothesis in cancer, environmental
tion between SNPs and an increased risk of heart failure factors and triggers might affect the phenotypic expres-
and cardiovascular events122,123. However, the routine sion of DCM in a patient with an underlying genetic
use of genetic testing for prognostication in arrhythmic predisposition135. For example, peripartum cardiomy­
DCM requires further data44. opathy has been associated not only with a familial pre-
disposition but might be caused by the combination of
Heart failure medical therapy and pharmacogenetics. an underlying pathogenic variant and the physiological
Given the ubiquitous use of neurohormonal blockade stress of pregnancy136,137. Similarly, genetic predisposi-
therapy in heart failure, with excellent data to support its tion might affect the cardiotoxicity of anthracyclines and
use for cardiac reverse remodelling, functional improve- other antineoplastic agents138. Indeed, when carefully
ment and increased survival, substantial interest lies in evaluated, environmental triggers might be associated
identifying which patients will benefit more or less from with a generally worse prognosis if an underlying genetic
these therapies. Research has provided some insight predisposition to the disease exists139.
into the relative efficacy of neurohormonal blockade in
patients with heart failure stratified by genetic variants, Uncertainties and future directions
contributing to the application of pharmacogenetics. Several barriers exist to the use of genetics in the care of
Some differences in individual response to β-​blockade patients with DCM. The foremost problem limiting our
are likely to be caused by genetic polymorphisms within identification of culprit genes is the frequency of VUSs,
the population in the gene encoding the β-​adrenergic which require functional studies or segregation analy-
receptor. Indeed, polymorphisms in ADRB1 and in GRK5, ses for interpretation28. Some genetic variants might be
encoding the downstream G-​protein-coupled recep- necessary but not sufficient for the phenotypic manifes-
tor kinase 5, influence responsiveness to β-​blockade21. tation of DCM and might require other so far unidenti-
Other data have shown that amino acid substitutions fied genetic or environmental factors to manifest overt
in the β-​adrenergic receptor, such as Arg389Gly, might disease135. Additionally, as described above, most of the
affect responsiveness to β-​blockade, possibly mediated isolated culprit genes that have been identified are auto-
through decreased activation of the renin–angiotensin– somal dominant or X-​linked, but many more genetic
aldosterone system124–126. However, this polymorphism variants might be inherited in an autosomal-​recessive
has not been associated with a change in morbidity or pattern, which can be more difficult to identify in familial
mortality in substudies of the WOSCOPS trial127 or the DCM and so are potentially under-​recognized23.
MERIT-​HF trial128. Broad interest exists in understanding the possible
Similarly, genes encoding angiotensin II and its asso- role of epigenomics and other ‘omics’ in chronic dis-
ciated receptors, which are involved in the pathogenesis ease and has only lately been evaluated in the context
of heart failure and are a target of antagonism in rou- of cardiomyopathy. Changes in DNA methylation have
tine guideline-​directed medical therapy, are associated been shown to alter the expression of genes involved
with polymorphisms that confer a differential response in energy metabolism, with an effect on cardiomyo-
to therapy. Data suggest that having a homozygous DD cyte function140. An integrated omics approach might
genotype for the ACE gene, which raises angiotensin II improve our understanding of disease pathogenesis, and
levels, combined with a polymorphism in AGTR1 the ‘omic’ patterns developed using artificial intelligence
(encoding the type 1 angiotensin II receptor) might con- in large populations could potentially be used for precision
fer higher levels of activation of the renin–angiotensin– diagnosis, prognosis and treatment of heart failure.
aldosterone system and worse prognosis, despite treat- The limited access to genetic counsellors and geneti-
ment with angiotensin-​c onverting enzyme (ACE) cists has restricted the use of genetics in the routine care
inhibitors129,130. In a prospective, observational study of of patients with DCM. One method to improve access is
patients with congestive heart failure, the presence of at to use videoconferencing for interviews (telemedicine),

Nature Reviews | Cardiology


Reviews

in this context termed ‘telegenetics’, which is supported genetic markers for the risk of DCM have been identi-
by most genetic counsellors141,142. However, new and fied. The routine use of expanded gene panels in DCM
innovative methods to educate and inform patients and increases the diagnostic yield, albeit with an increased
physicians need to be developed to improve access to risk of identifying VUSs. High-​throughput, in vitro
genetic testing that could complement genetic counsel- cellular functional validation and segregation analysis
ling by licensed providers. Additionally, on a practical have helped to interpret VUSs. Implementation of rou-
level, poor patient literacy and ineffective education or tine genetic testing for DCM in heart failure clinics is a
communication between physicians and patients about challenge, with limited availability of genetic counsellors.
the results and implications of genetic testing can hinder The heart failure community needs to make progress in
the evaluation or create challenges for interpretation. the identification and validation of genetic markers of
DCM risk, disease progression and response to therapy,
Conclusions which will eventually fulfil the promise of precision
Substantial progress has been made in the field of car- medicine in heart failure, with the hope of improved
diovascular genetics and in our understanding of the outcomes for patients and their families.
genetic basis of DCM. Increasing numbers of culprit rare
genetic variants have been validated, and new common Published online xx xx xxxx

1. Mozaffarian, D. et al. Heart disease and stroke 20. Richards, S. et al. Standards and guidelines for the 39. Baig, M. K. et al. Familial dilated cardiomyopathy:
statistics — 2016 update a report from the American interpretation of sequence variants: a joint consensus cardiac abnormalities are common in asymptomatic
Heart Association. Circulation 133, e38–e360 recommendation of the American College of Medical relative and may represent early disease. J. Am. Coll.
(2016). Genetics and Genomics and the Association for Cardiol. 31, 195–201 (1998).
2. Bui, A. L., Horwich, T. B. & Fonarow, G. C. Epidemiology Molecular Pathology. Am. Coll. Med. Genet. Genomics 40. Yancy, C. W. et al. 2013 ACCF/AHA guideline for
and risk profile of heart failure. Nat. Rev. Cardiol. 8, 17, 405–424 (2015). the management of heart failure: a report of the
30–41 (2011). 21. Cresci, S. et al. Clinical and genetic modifiers of long-​ American College of Cardiology Foundation/American
3. Ponikowski, P. et al. Heart failure: preventing disease term survival in heart failure. J. Am. Coll. Cardiol. 54, Heart Association task force on practice guidelines.
and death worldwide. ESC Heart Fail. 1, 4–25 (2014). 432–444 (2009). J. Am. Coll. Cardiol. 62, e147–e239 (2013).
4. Gerber, Y. et al. A contemporary appraisal of the heart 22. Liew, C.-C. & Dzau, V. J. Molecular genetics and 41. Charron, P. et al. Genetic counselling and testing
failure epidemic in Olmsted County, Minnesota, 2000 genomics of heart failure. Nat. Rev. Genet. 5, in cardiomyopathies: a position statement of the
to 2010. JAMA Intern. Med. 175, 996–1004 (2015). 811–825 (2004). European Society of Cardiology Working Group on
5. Heidenreich, P. A. et al. Forecasting the impact of 23. Harakalova, M. et al. A systematic analysis of genetic Myocardial and Pericardial Diseases. Eur. Heart J. 31,
heart failure in the United States a policy statement dilated cardiomyopathy reveals numerous 2715–2728 (2010).
from the American Heart Association. Circ. Heart Fail. ubiquitously expressed and muscle-​specific genes. 42. Bozkurt, B. et al. Current diagnostic and treatment
6, 606–619 (2013). Eur. J. Heart Fail. 17, 484–493 (2015). strategies for specific dilated cardiomyopathies:
6. Redfield, M. M. et al. Burden of systolic and diastolic 24. Tayal, U., Prasad, S. & Cook, S. A. Genetics and a scientific statement from the American Heart
ventricular dysfunction in the community. JAMA 289, genomics of dilated cardiomyopathy and systolic heart Association. Circulation 134, e579–e646 (2016).
194 (2003). failure. Genome Med. 9, 1–14 (2017). 43. Ackerman, M. J. et al. HRS/EHRA expert consensus
7. Hershberger, R. E., Hedges, D. J. & Morales, A. 25. Hershberger, R. E. et al. Genetic evaluation of statement on the state of genetic testing for the
Dilated cardiomyopathy: the complexity of a diverse cardiomyopathy: a clinical practice resource of the channelopathies and cardiomyopathies: this
genetic architecture. Nat. Rev. Cardiol. 10, 531–547 American College of Medical Genetics and Genomics document was developed as a partnership between
(2013). (ACMG). Genet. Med. 20, 899–909 (2018). the Heart Rhythm Society (HRS) and the European
8. Baldasseroni, S. et al. Left bundle-​branch block is 26. Lakdawala, N. K. et al. Genetic testing for dilated Heart Rhythm Association (EHRA). Heart Rhythm 8,
associated with increased 1-year sudden and total cardiomyopathy in clinical practice. J. Card. Fail. 18, 1308–1339 (2011).
mortality rate in 5517 outpatients with congestive 296–303 (2012). 44. Towbin, J. A. et al. 2019 HRS expert consensus
heart failure: a report from the Italian Network on 27. Ganesh, S. K. et al. Genetics and genomics for the statement on evaluation, risk stratification, and
Congestive Heart Failure. Am. Heart J. 143, 398–405 prevention and treatment of cardiovascular disease: management of arrhythmogenic cardiomyopathy.
(2002). update. a scientific statement from the American Heart Rhythm https://doi.org/10.1016/j.
9. Cahill, T. J., Ashrafian, H. & Watkins, H. Genetic Heart Association. Circulation 128, 2813–2851 hrthm.2019.05.007 (2019).
cardiomyopathies causing heart failure. Circ. Res. 113, (2013). 45. Sylvius, N. et al. In vivo and in vitro examination of the
660–675 (2013). 28. Pugh, T. J. et al. The landscape of genetic variation in functional significances of novel lamin gene mutations
10. Burkett, E. L. & Hershberger, R. E. Clinical and genetic dilated cardiomyopathy as surveyed by clinical DNA in heart failure patients. J. Med. Genet. 42, 639–647
issues in familial dilated cardiomyopathy. J. Am. Coll. sequencing. Genet. Med. 16, 601–608 (2014). (2005).
Cardiol. 57, 1641–1649 (2011). 29. Hershberger, R. E. et al. Genetic evaluation of 46. Fatkin, D. et al. Missense mutations in the rod
11. Jefferies, J. L. & Towbin, J. A. Dilated cardiomyopathy. cardiomyopathy—a Heart Failure Society of America domain of the lamin A/C gene as causes of dilated
Lancet 375, 752–762 (2010). practice guideline. J. Card. Fail. 24, 281–302 (2009). cardiomyopathy and conduction-​system disease.
12. Michels, V. et al. The frequency of familial dilated 30. Adams, D. R. & Eng, C. M. Next-​generation N. Engl. J. Med. 341, 1715–1724 (1999).
cardiomyopathy in a series of patients with idiopathic sequencing to diagnose suspected genetic disorders. 47. van Berlo, J. H. et al. Meta-​analysis of clinical
dilated cardiomyopathy. N. Engl. J. Med. 326, 77–82 N. Engl. J. Med. 379, 1353–1362 (2018). characteristics of 299 carriers of LMNA gene
(1992). 31. Kushwaha, S. S., Fallon, J. T. & Fuster, V. Restrictive mutations: do lamin A/C mutations portend a high risk
13. Grünig, E. et al. Frequency and phenotypes of familial cardiomyopathy. N. Engl. J. Med. 336, 267–276 of sudden death? J. Mol. Med. 83, 79–83 (2004).
dilated cardiomyopathy. J. Am. Coll. Cardiol. 31, (1997). 48. Parks, S. B. et al. Lamin A/C mutation analysis in a
186–194 (1998). 32. Finsterer, J. Cardiogenetics, neurogenetics, and cohort of 324 unrelated patients with idiopathic or
14. McNally, E. M. & Mestroni, L. Dilated cardiomyopathy: pathogenetics of left ventricular hypertrabeculation/ familial dilated cardiomyopathy. Am. Heart J. 156,
genetic determinants and mechanisms. Circ. Res. 121, noncompaction. Pediatr. Cardiol. 30, 659–681 161–169 (2008).
731–748 (2017). (2009). 49. van Tintelen, J. P. et al. High yield of LMNA mutations
15. Rosenbaum, A. N. & Pereira, N. L. Updates on the 33. Towbin, J. A., Lorts, A. & Jefferies, J. L. Left ventricular in patients with dilated cardiomyopathy and/or
genetic paradigm in heart failure. Curr. Treat. Options non-​compaction cardiomyopathy. Lancet 386, conduction disease referred to cardiogenetics
Cardiovasc. Med. 21, 37 (2019). 813–825 (2015). outpatient clinics. Am. Heart J. 154, 1130–1139
16. Briasoulis, A., Asleh, R. & Pereira, N. in Encyclopedia 34. Gallego-​Delgado, M. et al. Idiopathic restrictive (2007).
of Cardiovascular Research and Medicine 1st edn cardiomyopathy is primarily a genetic disease. 50. Morita, H., Seidman, J. G. & Seidman, C. E. Genetic
(eds Sawyer, D. & Vasan, R.) 368–379 (Elsevier, J. Am. Coll. Cardiol. 67, 3021–3023 (2016). causes of human heart failure. J. Clin. Invest. 115,
2018). 35. Haas, J. et al. Atlas of the clinical genetics of human 518–526 (2005).
17. Elliott, P. Cardiomyopathy: diagnosis and management dilated cardiomyopathy. Eur. Heart J. 36, 1123–1135 51. Vikhorev, P. G. et al. Abnormal contractility in
of dilated cardiomyopathy. Heart 84, 106–106 (2015). human heart myofibrils from patients with dilated
(2000). 36. Bennett, R. L. The Practical Guide to the Genetic cardiomyopathy due to mutations in TTN and
18. Japp, A. G., Gulati, A., Cook, S. A., Cowie, M. R. & Family History 2nd edn (Wiley, 2010). contractile protein genes. Sci. Rep. 7, 14829 (2017).
Prasad, S. K. The diagnosis and evaluation of dilated 37. Arbustini, E. et al. The MOGE(S) classification 52. Gramlich, M. et al. Stress-​induced dilated cardio­
cardiomyopathy. J. Am. Coll. Cardiol. 67, 2996–3010 for a phenotype-​genotype nomenclature of myopathy in a knock-​in mouse model mimicking
(2016). cardiomyopathy: endorsed by the world heart human titin-​based disease. J. Mol. Cell. Cardiol. 47,
19. Sweet, M. E., Taylor, M. R. & Mestroni, L. Diagnosis, federation. Glob. Heart 8, 355–382 (2013). 352–358 (2009).
prevalence, and screening of familial dilated 38. Mcnally, E. M., Golbus, J. R. & Puckelwartz, M. J. 53. Gerull, B. et al. Mutations of TTN, encoding the
cardiomyopathy. Expert. Opin. Orphan Drugs 3, Genetic mutations and mechanisms in dilated giant muscle filament titin, cause familial dilated
869–876 (2015). cardiomyopathy. J. Clin. Invest. 123, 19–26 (2013). cardiomyopathy. Nat. Genet. 30, 201–204 (2002).

www.nature.com/nrcardio
Reviews

54. Herman, D. S. et al. Truncations of titin causing dilated a clinicopathological study. Heart 97, 1744–1752 laboratories: a single-​center experience. Genet. Med.
cardiomyopathy. N. Engl. J. Med. 366, 619–628 (2011). 20, 369–373 (2018).
(2012). 80. Ichida, F. et al. Novel gene mutations in patients with 105. Amendola, L. M. et al. Performance of ACMG-​AMP
55. Tayal, U. et al. Phenotype and clinical outcomes of titin left ventricular noncompaction or barth syndrome. variant-​interpretation guidelines among nine
cardiomyopathy. J. Am. Coll. Cardiol. 70, 2264–2274 Circulation 103, 1256–1263 (2001). laboratories in the clinical sequencing exploratory
(2017). 81. Captur, G. & Nihoyannopoulos, P. Left ventricular research consortium. Am. J. Hum. Genet. 99, 247
56. Jansweijer, J. A. et al. Truncating titin mutations are non-​compaction: genetic heterogeneity, diagnosis (2016).
associated with a mild and treatable form of dilated and clinical course. Int. J. Cardiol. 140, 145–153 106. Harrison, S. M. et al. Clinical laboratories collaborate
cardiomyopathy. Eur. J. Heart Fail. 19, 512–521 (2010). to resolve differences in variant interpretations
(2017). 82. D’Adamo, P. et al. The X-​linked gene G4.5 is responsible submitted to ClinVar. Genet. Med. 19, 1096–1104
57. Veselka, J., Anavekar, N. S. & Charron, P. Hypertrophic for different infantile dilated cardiomyopathies. Am. J. (2017).
obstructive cardiomyopathy. Lancet 389, 1253–1267 Hum. Genet. 61, 862–867 (1997). 107. Kelly, M. A. et al. Adaptation and validation of the
(2017). 83. Bione, S. et al. A novel X-​linked gene, G4.5. is ACMG/AMP variant classification framework for
58. Mogensen, J. et al. Frequency and clinical expression responsible for Barth syndrome. Nat. Genet. 12, MYH7-associated inherited cardiomyopathies:
of cardiac troponin I mutations in 748 consecutive 385–389 (1996). recommendations by ClinGen’s Inherited
families with hypertrophic cardiomyopathy. J. Am. 84. Brauch, K. M. et al. Mutations in ribonucleic acid Cardiomyopathy Expert Panel. Genet. Med. 20,
Coll. Cardiol. 44, 2315–2325 (2004). binding protein gene cause familial dilated 351–359 (2018).
59. Moolman, J. C. et al. Sudden death due to troponin T cardiomyopathy. J. Am. Coll. Cardiol. 54, 930–941 108. Hoffman-​Andrews, L. The known unknown: the
mutations. J. Am. Coll. Cardiol. 29, 549–555 (1997). (2009). challenges of genetic variants of uncertain significance
60. Watkins, H. et al. Mutations in the genes for cardiac 85. van den Hoogenhof, M. M. G. et al. RBM20 mutations in clinical practice. J. Law Biosci. 4, 648–657 (2017).
troponin t and α-​tropomyosin in hypertrophic induce an arrhythmogenic dilated cardiomyopathy 109. Lv, W. et al. Functional annotation of TNNT2 variants
cardiomyopathy. N. Engl. J. Med. 332, 1058–1064 related to disturbed calcium handling. Circulation of uncertain significance with genome-​edited
(1995). 138, 1330–1342 (2018). cardiomyocytes. Circulation 138, 2852–2854 (2018).
61. Fujino, N. et al. A novel mutation Lys273Glu in the 86. Olson, T. M. et al. Sodium channel mutations and 110. Stark, K. et al. Genetic association study identifies
cardiac troponin T gene shows high degree of susceptibility to heart failure and atrial fibrillation. HSPB7 as a risk gene for idiopathic dilated
penetrance and transition from hypertrophic to dilated JAMA 293, 447–454 (2005). cardiomyopathy. PLOS Genet. 6, e1001167 (2010).
cardiomyopathy. Am. J. Cardiol. 89, 29–33 (2002). 87. McNair, W. P. et al. SCN5A mutation associated 111. Villard, E. et al. A genome-​wide association study
62. Kokado, H. et al. Clinical features of hypertrophic with dilated cardiomyopathy, conduction disorder, identifies two loci associated with heart failure due to
cardiomyopathy caused by a Lys183 deletion and arrhythmia. Circulation 110, 2163–2167 dilated cardiomyopathy. Eur. Heart J. 32, 1065–1076
mutation in the cardiac troponin I gene. Circulation (2004). (2011).
102, 663–669 (2000). 88. Brega, A., Narula, J. & Arbustini, E. Functional, 112. Cappola, T. P. et al. Common variants in HSPB7 and
63. Regitz-​Zagrosek, V., Erdmann, J., Wellnhofer, E., Raible, J. structural, and genetic mitochondrial abnormalities FRMD4B associated with advanced heart failure.
& Fleck, E. Novel mutation in the α-​tropomyosin gene in myocardial diseases. J. Nucl. Cardiol. 8, 89–97 Circ. Cardiovasc. Genet. 3, 147–154 (2010).
and transition from hypertrophic to hypocontractile (2001). 113. Meder, B. et al. A genome-​wide association study
dilated cardiomyopathy. Circulation 102, e112–e116 89. Zaragoza, M. V., Brandon, M. C., Diegoli, M., Arbustini, E. identifies 6p21 as novel risk locus for dilated
(2000). & Wallace, D. C. Mitochondrial cardiomyopathies: how cardiomyopathy. Eur. Heart J. 35, 1069–1077
64. Kamisago, M. et al. Mutations in sarcomere protein to identify candidate pathogenic mutations by (2014).
genes as a cause of dilated cardiomyopathy. N. Engl. mitochondrial DNA sequencing, MITOMASTER and 114. Cappola, T. P. et al. Loss-​of-function DNA sequence
J. Med. 343, 1688–1696 (2000). phylogeny. Eur. J. Hum. Genet. 19, 200–207 (2011). variant in the CLCNKA chloride channel implicates the
65. Daehmlow, S. et al. Novel mutations in sarcomeric 90. Anan, R. et al. Cardiac involvement in mitochondrial cardio-​renal axis in interindividual heart failure risk
protein genes in dilated cardiomyopathy. Biochem. diseases: a study on 17 patients with documented variation. Proc. Natl Acad. Sci. USA 108, 2456–2461
Biophys. Res. Commun. 298, 116–120 (2002). mitochondrial DNA defects. Circulation 91, 955–961 (2011).
66. Bos, J. M. & Ackerman, M. J. Z-​disc genes in (1995). 115. Franaszczyk, M. et al. The BAG3 gene variants in
hypertrophic cardiomyopathy: stretching the 91. Nair, V., Belanger, E. C. & Veinot, J. P. Lysosomal Polish patients with dilated cardiomyopathy: four
cardiomyopathies? J. Am. Coll. Cardiol. 55, storage disorders affecting the heart: a review. novel mutations and a genotype-​phenotype
1136–1138 (2010). Cardiovasc. Pathol. 39, 12–24 (2019). correlation. J. Transl Med. 12, 192 (2014).
67. Knöll, R. et al. The cardiac mechanical stretch sensor 92. Kamdar, F. & Garry, D. J. Dystrophin-​deficient 116. Parsa, A. et al. Hypertrophy-​associated polymorphisms
machinery involves a Z disc complex that is defective cardiomyopathy. J. Am. Coll. Cardiol. 67, 2533–2546 ascertained in a founder cohort applied to heart failure
in a subset of human dilated cardiomyopathy. Cell (2016). risk and mortality. Clin. Transl Sci. 4, 17–23 (2011).
111, 943–955 (2002). 93. Towbin, J. A. et al. X-​linked dilated cardiomyopathy - 117. Fox, E. R. et al. Genome-​wide association study of
68. Olson, T. M. et al. Metavinculin mutations alter actin molecular-​genetic evidence of linkage to the Duchenne cardiac structure and systolic function in African
interaction in dilated cardiomyopathy. Circulation muscular-​dystrophy (dystrophin) gene at the Xp21 Americans: the Candidate Gene Association Resource
105, 431–437 (2002). locus. Circulation 87, 1854–1865 (1993). (CARe) study. Circ. Cardiovasc. Genet. 6, 37–46
69. Villard, E. et al. Mutation screening in dilated 94. Mounkes, L. C., Burke, B. & Stewart, C. L. The A-​type (2013).
cardiomyopathy: prominent role of the beta myosin lamins: nuclear structural proteins as a focus for 118. Smith, N. L. et al. Association of genome-​wide
heavy chain gene. Eur. Heart J. 26, 794–803 (2005). muscular dystrophy and cardiovascular diseases. variation with the risk of incident heart failure in
70. Duboscq-​Bidot, L. et al. Mutations in the Z-​band Trends Cardiovasc. Med. 11, 280–285 (2001). adults of European and African ancestry: a prospective
protein myopalladin gene and idiopathic dilated 95. Bonne, G. et al. Mutations in the gene encoding lamin meta-​analysis from the Cohorts for Heart and Aging
cardiomyopathy. Cardiovasc. Res. 77, 118–125 A/C cause autosomal dominant Emery-​Dreifuss Research in Genomic Epidemiology (CHARGE)
(2008). muscular dystrophy. Nat. Genet. 21, 285–288 (1999). Consortium. Circ. Cardiovasc. Genet. 3, 256–266
71. Dalakas, M. et al. Desmin myopathy, a skeletal 96. Nigro, V. & Savarese, M. Genetic basis of limb-​girdle (2010).
myopathy with cardiomyopathy caused by mutations muscular dystrophies: the 2014 update. Acta Myol. 119. Meune, C. et al. Primary prevention of sudden death
in the desmin gene. N. Engl. J. Med. 342, 770–780 33, 1–12 (2014). in patients with lamin A/C gene mutations. N. Engl. J.
(2000). 97. Mestroni, L. et al. Guidelines for the study of familial Med. 354, 209–210 (2006).
72. Li, D. et al. Desmin mutation responsible for idiopathic dilated cardiomyopathies. Eur. Heart J. 20, 93–102 120. Bécane, H. M. et al. High incidence of sudden death
dilated cardiomyopathy. Circulation 100, 461–464 (1999). with conduction system and myocardial disease due to
(1999). 98. Xue, Y., Ankala, A., Wilcox, W. R. & Hegde, M. R. lamins A and C gene mutation. Pacing Clin.
73. Arbustini, E. et al. Desmin accumulation restrictive Solving the molecular diagnostic testing conundrum Electrophysiol. 23, 1661–1666 (2000).
cardiomyopathy and atrioventricular block associated for Mendelian disorders in the era of next-​generation 121. Smith, J. G. et al. Discovery of genetic variation on
with desmin gene defects. Eur. J. Heart Fail. 8, sequencing: single-​gene, gene panel, or exome/ chromosome 5q22 associated with mortality in heart
477–483 (2006). genome sequencing. Genet. Med. 17, 444–451 failure. PLOS Genet. 12, e1006034 (2016).
74. Begay, R. L. et al. FLNC gene splice mutations cause (2015). 122. Morrison, A. C. et al. Genomic variation associated
dilated cardiomyopathy. JACC Basic Transl. Sci. 1, 99. Yang, Y. et al. Clinical whole-​exome sequencing for the with mortality among adults of European and
344–359 (2016). diagnosis of mendelian disorders. N. Engl. J. Med. African ancestry with heart failure: the Cohorts for
75. Ortiz-​Genga, M. F. et al. Truncating FLNC mutations are 369, 1502–1511 (2013). Heart and Aging Research in Genomic Epidemiology
associated with high-​risk dilated and arrhythmogenic 100. Bagnall, R. D. et al. Whole genome sequencing Consortium. Circ. Cardiovasc. Genet. 3, 248–255
cardiomyopathies. J. Am. Coll. Cardiol. 68, improves outcomes of genetic testing in patients with (2010).
2440–2451 (2016). hypertrophic cardiomyopathy. J. Am. Coll. Cardiol. 72, 123. Myers, V. D. et al. Association of variants in BAG3 with
76. Begay, R. L. et al. Filamin C truncation mutations 419–429 (2018). cardiomyopathy outcomes in African American
are associated with arrhythmogenic dilated 101. Biesecker, L. G. & Green, R. C. Diagnostic clinical individuals. JAMA Cardiol. 3, 929–938 (2018).
cardiomyopathy and changes in the cell–cell adhesion genome and exome sequencing. N. Engl. J. Med. 370, 124. Liggett, S. B. et al. A polymorphism within a conserved
structures. JACC Clin. Electrophysiol. 4, 504–514 2418–2425 (2014). β1-adrenergic receptor motif alters cardiac function
(2018). 102. Mohammed, S. et al. Genetic insurance discrimination and β-​blocker response in human heart failure. Proc.
77. Selcen, D. et al. Mutation in BAG3 causes severe in sudden arrhythmia death syndromes empirical Natl Acad. Sci. USA 103, 11288–11293 (2006).
dominant childhood muscular dystrophy. Ann. Neurol. evidence from a cross-​sectional survey in North 125. Bruck, H. et al. The Arg389Gly β1-adrenoceptor
65, 83–89 (2009). America. Circ. Cardiovasc. Genet. 10, e001442 polymorphism and catecholamine effects on plasma-​
78. Elliott, P. et al. Prevalence of desmosomal protein gene (2017). renin activity. J. Am. Coll. Cardiol. 46, 2111–2115
mutations in patients with dilated cardiomyopathy. 103. Joly, Y., Braker, M. & Le Huynh, M. Genetic (2005).
Circ. Cardiovasc. Genet. 3, 314–322 (2010). discrimination in private insurance: global 126. Terra, S. G. et al. β1-Adrenergic receptor
79. Garcia-​Pavia, P. et al. Desmosomal protein gene perspectives. New Genet. Soc. 29, 351–368 (2010). polymorphisms and left ventricular remodeling
mutations in patients with idiopathic dilated 104. Bland, A. et al. Clinically impactful differences in changes in response to β-​blocker therapy.
cardiomyopathy undergoing cardiac transplantation: variant interpretation between clinicians and testing Pharmacogenet. Genomics 15, 227–234 (2005).

Nature Reviews | Cardiology


Reviews
127. White, H. et al. An evaluation of the β1 adrenergic a retrospective cohort study. PLOS ONE 10, 1–16 human ischemic heart failure. Lab. Invest. 99,
receptor Arg389Gly polymorphism in individuals (2015). 371–386 (2018).
at risk of coronary events: a WOSCOPS substudy. 133. Pare, G. et al. Genetic variants associated with 141. Zierhut, H. A., MacFarlane, I. M., Ahmed, Z. & Davies, J.
Eur. Heart J. 23, 1087–1092 (2002). angiotensin-​converting enzyme inhibitor-​associated Genetic counselors’ experiences and interest in
128. White, H. L. et al. An evaluation of the β1 adrenergic angioedema. Pharmacogenet. Genomics 23, telegenetics and remote counseling. J. Genet. Couns.
receptor Arg389Gly polymorphism in individuals with 470–478 (2013). 27, 329–338 (2018).
heart failure: a MERIT-​HF sub-​study. Eur. J. Heart Fail. 134. Pereira, N. L. & Weinshilboum, R. M. Cardiovascular 142. Du, L. & Becher, S. I. Genetic and genomic
5, 463–468 (2003). pharmacogenomics and individualized drug therapy. consultation: are we ready for direct-​to-consumer
129. Andersson, B., Blange, I., Sylven, C. & Sylvén, C. Nat. Rev. Cardiol. 6, 632–638 (2009). telegenetics? Front. Genet. 9, 550 (2018).
Angiotensin-​II type 1 receptor gene polymorphism 135. Merlo, M. et al. Evolving concepts in dilated cardio­
and long-​term survival in patients with idiopathic myopathy. Eur. J. Heart Fail. 20, 228–239 (2017). Author contributions
congestive heart failure. Eur. J. Heart Fail. 1, 136. Van Spaendonck-​Zwarts, K. Y. et al. Peripartum All the authors researched data for the article, wrote the man-
363–369 (1999). cardiomyopathy as a part of familial dilated uscript and reviewed and edited it before submission. A.N.R.
130. Huang, W., Xie, C., Zhou, H., Yang, T. & Sun, M. cardiomyopathy. Circulation 121, 2169–2175 (2010). and N.L.P. contributed to discussions about the article
Association of the angiotensin-​converting enzyme 137. Blauwet, L. A. & Cooper, L. T. Diagnosis and content.
gene polymorphism with chronic heart failure in management of peripartum cardiomyopathy. Heart
Chinese Han patients. Eur. J. Heart Fail. 6, 23–27 97, 1970–1981 (2011). Competing interests
(2004). 138. Scott, E., Hasbullah, J. S., Ross, C. J. & Carleton, B. C. The authors declare no competing interests.
131. McNamara, D. M. et al. Pharmacogenetic interactions Reducing anthracycline-​induced cardiotoxicity through
between angiotensin-​converting enzyme inhibitor pharmacogenetics. Pharmacogenomics 19, 1147–1150 Peer review information
therapy and the angiotensin-​converting enzyme (2018). Nature Reviews Cardiology thanks L. Mestroni, A. Morales,
deletion polymorphism in patients with congestive 139. Hazebroek, M. R. et al. Prognostic relevance of gene-​ G. Sinagra and M. van den Berg for their contribution to the
heart failure. J. Am. Coll. Cardiol. 44, 1644–1649 environment interactions in patients with dilated peer review of this work.
(2004). cardiomyopathy applying the MOGE(S) classification.
132. Nelveg-​Kristensen, K. E. et al. Pharmacogenetic risk J. Am. Coll. Cardiol. 66, 1313–1323 (2015). Publisher’s note
stratification in angiotensin-​converting enzyme 140. Pepin, M. E. et al. Genome-​wide DNA methylation Springer Nature remains neutral with regard to jurisdictional
inhibitor-​treated patients with congestive heart failure: encodes cardiac transcriptional reprogramming in claims in published maps and institutional affiliations.

www.nature.com/nrcardio

You might also like