You are on page 1of 18

Cardiomyopathy Compendium

Circulation Research Compendium on Cardiomyopathies


Cardiomyopathies: An Overview
Classification, Epidemiology, and Global Burden of Cardiomyopathies
Dilated Cardiomyopathy: Genetic Determinants and Mechanisms
Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy
Hypertrophic Obstructive Cardiomyopathy: Surgical Myectomy and Septal Ablation
Arrhythmogenic Cardiomyopathy
Inflammatory Cardiomyopathic Syndromes
Restrictive Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy
Cardiomyopathies Due to Left Ventricular Noncompaction, Mitochondrial and Storage Diseases, and Inborn Errors of
Metabolism
Pediatric Cardiomyopathies
Modern Imaging Techniques in Cardiomyopathies
Editors: Eugene Braunwald and Ali J. Marian

Dilated Cardiomyopathy
Genetic Determinants and Mechanisms
Elizabeth M. McNally, Luisa Mestroni

Abstract: Nonischemic dilated cardiomyopathy (DCM) often has a genetic pathogenesis. Because of the large
Downloaded from http://ahajournals.org by on September 29, 2018

number of genes and alleles attributed to DCM, comprehensive genetic testing encompasses ever-increasing
gene panels. Genetic diagnosis can help predict prognosis, especially with regard to arrhythmia risk for certain
subtypes. Moreover, cascade genetic testing in family members can identify those who are at risk or with early
stage disease, offering the opportunity for early intervention. This review will address diagnosis and management
of DCM, including the role of genetic evaluation. We will also overview distinct genetic pathways linked to DCM
and their pathogenetic mechanisms. Historically, cardiac morphology has been used to classify cardiomyopathy
subtypes. Determining genetic variants is emerging as an additional adjunct to help further refine subtypes of
DCM, especially where arrhythmia risk is increased, and ultimately contribute to clinical management.   (Circ Res.
2017;121:731-748. DOI: 10.1161/CIRCRESAHA.116.309396.)
Key Words: cardiomyopathy, dilated ■ genetic testing ■ heart failure ■ mutation ■ sarcomeres ■ therapeutics

Prevalence and Pathogenesis by an enlarged and poorly contractile LV. DCM can be attrib-
of Dilated Cardiomyopathy uted to genetic and nongenetic causes, including hypertension,
Cardiomyopathies are defined as myocardial disorders in valve disease, inflammatory/infectious causes, and toxins.3
which the heart is structurally and functionally abnormal. Even these nongenetic forms of cardiomyopathy can be in-
Morphologically defined subtypes include hypertrophic car- fluenced by an individual’s genetic profile and, furthermore,
diomyopathy (HCM), dilated cardiomyopathy (DCM), ar- mixed pathogeneses may be present. In DCM, the degree of
rhythmogenic cardiomyopathy, and left ventricular (LV) LV systolic dysfunction is variable, and LV systolic dysfunc-
noncompaction cardiomyopathy,1,2 and each of these subtypes tion is often progressive. DCM is a major risk factor for de-
can be genetically mediated (Figure 1). DCM is characterized veloping heart failure (HF) as the presence of reduced systolic

From the Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL (E.M.M.); and Cardiovascular Institute,
University of Colorado Anschutz Medical Campus, Aurora (L.M.).
Correspondence to Elizabeth M. McNally, MD, PhD, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL.
E-mail elizabeth.mcnally@northwestern.edu; or Luisa Mestroni, MD, Cardiovascular Institute, University of Colorado Anschutz Medical Campus, 12700
E, 19th Ave # F442, Aurora, CO 80045-2507. E-mail Luisa.Mestroni@ucdenver.edu
© 2017 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.116.309396

731
732  Circulation Research  September 15, 2017

In this study, individuals with nonischemic DCM were more


Nonstandard Abbreviations and Acronyms
likely to be women, nonwhite, and younger that those with
AHA American Heart Association ischemic cardiomyopathy.
ARVC arrhythmogenic right ventricular cardiomyopathy The true prevalence of DCM, and of genetically mediated
CMR cardiac magnetic resonance DCM, is not fully known. An early estimate of DCM preva-
DCM dilated cardiomyopathy lence derived from a study performed from 1975 to 1984 in
EMB endomyocardial biopsy Olmstead County, MN.5 This epidemiological study relied on
ESC European Society of Cardiology echocardiography, angiography, or autopsy evaluation of DCM
HCM hypertrophic cardiomyopathy cases and resulted in a prevalence of 36.5/100 000 individuals
HF heart failure or 1 in 2700 with a men to women ratio of 3:4 in a European-
ICD implantable cardioverter defibrillator
American population.5 The prevalence of DCM varies likely
iPSC induced pluripotent stem cell
reflecting geographic and ethnic differences, as well as the
methodologies used.6–10 Studies from England (8.3/100 000),11
LV left ventricle
Italy (7.0/100 000),12 and Japan (14/100 000)9 report similar
LVEF left ventricular ejection fraction
DCM prevalence. However, the prevalence of DCM is likely
SCD sudden cardiac death
underestimated. The prior studies relied on older less sensitive
VT ventricular tachycardia
imaging modalities.13 More recently, Hershberger et al14 used a
different approach to estimate DCM prevalence, based on the
function does not imply symptoms. Notably, DCM is often as- known ratio of idiopathic DCM to HCM of ≈2:1, prevalence
sociated with an increased risk of severe arrhythmia, indicating estimates of HF, and prevalence estimates of LV dysfunction as
the pathological involvement of the cardiac conduction system. a surrogate for DCM. With this approach, a much higher preva-
Randomized clinical HF trials typically report 30% to 40% lence of DCM is estimated, in the range of 1:250. Similarly,
of subjects with a nonischemic DCM compared with ischemic estimates of familial DCM prevalence varies: a meta-analysis
DCM.3 Clinical trials are evaluating interventions to reduce of 23 studies found a prevalence estimate of 23% with a range
congestive heart failure symptoms, and these studies may fo- of 2% to 65%, indicating a significant heterogeneity in diag-
cus on the later stages of disease. Similarly, a recent survey of nostic criteria, and a frequency progressively increasing over
hospitalized patients in the United States in which the mean time because of more systematic clinical screening.15 In the
age was 75 years (n=156 013) found that ischemic cardiomy- clinical practice and in the current guidelines, the prevalence
opathy was more common than nonischemic (59% compared of familial DCM is assumed to be ≈30% to 50%.1,3,13,16–18 In
Downloaded from http://ahajournals.org by on September 29, 2018

with 41%).4 Of nonischemic DCM, hypertension accounted patients with familial DCM, ≈40% has an identifiable genetic
for 48%, and idiopathic was the next most common at 31%. cause.17 Also in sporadic DCM, pathogenic genetic variants

Figure 1. Echocardiography demonstrates


forms of cardiomyopathy. Left ventricular
noncompaction cardiomyopathy (LVNC) is
shown in the upper right (arrows indicate deep
trabeculations in the left ventricle [LV]). Dilated
cardiomyopathy (DCM) is defined by enlarged
LV diameters (dashed double sided arrow).
Hypertrophic cardiomyopathy (HCM) is defined
with a thickened LV, including the septum
(marked with double sided arrow).
McNally and Mestroni   Dilated Cardiomyopathy Genetics and Mechanisms   733

can be identified although the frequency of genetic causes in of these studies examine gene positive individuals at one point
this population is not well defined.17 in time, a full view of DCM progression has not been delin-
eated. Definitive studies on DCM progression in genetically
Clinical Diagnosis of DCM at-risk individuals must span many years. Imaging studies
DCM has been defined by the presence of (1) fractional short- have identified chamber size dimensions, strain abnormalities,
ening <25% (>2 SD) or ejection fraction <45% (>2 SD), and and contrast enhancement each as features of early DCM.25
(2) LV end-diastolic diameter >117% (>2 SD of the predicted
value of 112% corrected for age and body surface area), ex- Imaging: Echocardiography
cluding any known cause of myocardial disease.19 In the con- To diagnose DCM, LV measurements can be determined us-
text of a familial DCM, these criteria are used to diagnose the ing multiple imaging modalities. M-mode and 2-dimensional
proband in a family,19 and the strategy for evaluation is shown echocardiography are frequently used to determine LV inter-
in Figure 2. Familial DCM is defined by the presence of (1) ≥2 nal dimensions in systole and diastole (Figure 1). It was origi-
affected relatives with DCM meeting the above criteria, or (2) nally thought that LV dilation occurs in response to reduced
a relative of a DCM patient with unexplained sudden death be- function. However, in genetic DCM, where genetic markers
fore the age of 35 years.19 Family members may be classified make it feasible to monitor LV dimensions for many years, in-
as affected, unaffected, or unknown when subtle cardiac ab- creased LV dimensions typically precede detectable reduction
normalities are present but not sufficient for a definitive diag-
in function.27–29 This state of LV enlargement is recognized as
nosis. Less common forms of primary cardiomyopathies are
a prodome to DCM. Enlarged LV dimensions contrast with
peripartum, tachycardia-induced, stress-provoked Takotsubo
what is seen in HCM, where the earliest findings in genetical-
cardiomyopathy and myocarditis, according to the 2006
ly mediated, sarcomere-positive HCM are reduced LV dimen-
American Heart Association (AHA) definition and classifi-
sions.30 Strain and strain rate differences can be detected by
cation.2 Interestingly, myocarditis and peripartum cardiomy-
echocardiography in first-degree relatives of those with newly
opathy can occur in a familial setting and are believed to have
a genetic component.22–24 Secondary forms of cardiomyopa- diagnosed nonischemic DCM, indicating that LV dimensions
thies, in which cardiomyopathy arises from systemic disor- are not the earliest detectable differences in familial DCM.31,32
ders, such as amyloidosis, hemochromatosis, and because of
toxicity from agents like doxorubicin, are also under genetic Imaging: Cardiac Magnetic Resonance
influence or may arise due to primary genetic mutations.25,26 LV chamber dimensions and function, including strain mea-
Neuromuscular disease is also commonly associated with surements, are also accurately determined by cardiac mag-
Downloaded from http://ahajournals.org by on September 29, 2018

cardiomyopathy, and cardiomyopathy typically arises from netic resonance (CMR) imaging. Contrast agents, mainly
the primary responsible genetic mutation exerting pathologi- gadolinium, are used to evaluate fibrosis and therefore pro-
cal effects directly in the myocardium (see below). With the vide additional information on myocardial tissue quality. In
increase in using genetic testing, especially testing in family DCM, the degree of fibrosis, marked by delayed gadolinium
members, it is now possible to use cardiac imaging modalities enhancement, is a predictor of all-cause mortality and need for
to ascertain early features of disease in gene mutation-positive future hospitalization.33 Specifically, delayed enhancement is
individual who do not fully manifest disease. Because many associated with increased risk for ventricular arrhythmias.34–36

Proband with DCM

Family history
≥ 3 generations

Figure 2. Algorithm for the management


Familial CM Sporadic DCM of a patient with nonischemic dilated
≥ 2 affected family members Non-familial cardiomyopathy (DCM). Patients with DCM
should undergo an accurate family history
Physical exam
examination. A comprehensive examination
Electrocardiogram
should include serum creatine kinase (CK)
Echocardiogram
to evaluate skeletal muscle involvement.
Holter
First-degree relatives of patients with DCM
CMR should be examined (physical examination,
Serum CK ECG, imaging). Young DCM patients or those
with family history should be offered genetic
Cascade genetic screening counseling and should consider genetic testing.
• Genetic testing and counseling A positive genetic testing in the proband offers
the possibility of a confirmatory genetic testing
• Screening of 1st degree relatives in relatives, which may guide follow-up and
need of further testing. CMR indicates cardiac
DCM/gene + or - Unaffected/gene + Unaffected/gene - magnetic resonance. See references 14,19–21.

Clinical follow up
• Every 1-2 y in children/adolescents
• Every 3-5 years adults or when onset of symptoms
734  Circulation Research  September 15, 2017

Delayed enhancement may also reflect features beyond fibro- transmitted data, as well as triggered monitoring. External loop
sis, including edema and inflammatory infiltrate.37 recorders and now implantable loop recorders offer long-term
DCM can also be associated with LV noncompaction seen information. Those with familial DCM are likely to have more
using either echocardiography or CMR. A ratio of >2.3:1 for findings of ventricular ectopy and ventricular tachycardia (VT)
the noncompacted to compacted layer of LV myocardium is on monitoring.47 For primary prevention of SCD in DCM, risk
considered abnormal but notably can be detected in normal stratification often relies on evaluating the specific genetic con-
hearts.38 Recent studies have suggested that hypertrabecula- tribution (see below), family history, delayed enhancement, and
tion is seen in a high fraction (36%) of adult DCM although presence of VT on monitoring.
this was not associated with adverse outcomes compared with
DCM without these noncompaction features.39 Clinical Manifestations Including
Neuromuscular Findings
Endomyocardial Biopsy The range of clinical manifestations in DCM ranges from none
Endomyocardial biopsy (EMB) has been used to confirm di- to overt HF. With the increase in familial and genetic screen-
agnosis in some forms of DCM although with improved car- ing, it is now more common to identify the minimally to mildly
diac imaging, EMB is less frequently used. In some settings, affected stage of DCM in younger individuals. Using genetic
for example, iron overload, amyloid, and other infiltrative pro- markers, strain defects can be detected by echo or CMR. LGE
cesses, myocardial biopsy may still be highly useful.40 The (late gadolinium enhancement) may be present even when the
complication rates with EMB range from 1% to 3%, and seri- heart appears still normal, suggesting that disease is ongoing.
ous complications including perforation and tamponade occur There is an emerging view that this represents an early stage of
at 0.5%.40 EMB has been used to evaluate myocarditis and in disease and one in which early institution of treatment should
the setting of unexplained HF.41,42 The nonuniform nature of benefit. Although it is generally thought that arrhythmia risk
infiltrative disease limits the sensitivity of myocardial biopsy scales with degree of LV dysfunction, for several subtypes of
because the right ventricular septum is targeted for sampling.43 genetic cardiomyopathy, arrhythmias may be the earliest mani-
For the majority of genetic cardiomyopathies, genetic testing festation.48–51 Specifically, in LMNA- and SCN5A-mediated
is favored over EMB. Of the genetic cardiomyopathies, ar- cardiomyopathies, arrhythmias including atrial fibrillation with
rhythmogenic right ventricular cardiomyopathy (ARVC) may slow ventricular response or ventricular arrhythmias may be the
be evaluated by EMB although more recent work suggests that presenting finding. There is little in the clinical evaluation that
alternative and more easily accessible cell types can be used to makes it possible to distinguish one genetic subtype of DCM
diagnose ARVC and avoid EMB.44,45 from another. This phenocopying is what has driven gene panel
Downloaded from http://ahajournals.org by on September 29, 2018

testing because with this approach, multiple genes are screened


Noninvasive Arrhythmia Monitoring at the same time. A DCM gene panel is shown in Figure 3, and
DCM is associated with an increased risk for cardiac arrhyth- a comprehensive list of DCM genes is provided in the Table.
mias and sudden cardiac death (SCD),46 and specific genetic Neuromuscular disease may accompany cardiomyopathy,
DCM subtypes are especially prone to arrhythmias. CMR, es- and in some forms of neuromuscular disease, the present-
pecially the identification of delayed enhancement, can help risk ing feature may be irregular heart rhythms. LMNA mutations
stratify for sudden death.36 Because of increased risk for SCD, can present with or without muscle disease, and the muscle
there is need for arrhythmia surveillance to more appropriately disease ranges from limb-girdle muscular dystrophy to
deploy device management, including pacemakers and implant- Emery–Dreifuss muscular dystrophy, which is typically asso-
able cardioverter defibrillators (ICDs). Symptomatic and even ciated with contractures of the elbows and Achilles tendons.54
life-threatening bradycardia and tachycardia may occur in ge- LMNA mutations are inherited in an autosomal manner, seen
netic DCM. Personal history of syncope or near syncope should as multiple affected family members in each generation. Both
be ascertained, and patient education to increase awareness of X-linked and autosomal neuromuscular diseases can also as-
symptoms is needed. Holter monitoring, for its ease, remains a sociate with cardiomyopathy, and this includes Duchenne
mainstay using 24- to 48-hour sampling. Other external event muscular dystrophy, as well as the autosomal recessive forms
recorders are similarly transcutaneous and provide real-time of sarcoglycanopathies.55 In these disorders, skeletal muscle

ABCC9 ACTC1 ACTN2 AGL1 ALMS1 ALPK3 ANKRD1 BAG3 BRAF


CACNA1C CALR3 CASQ2 CAV3 CHRM2 CRYAB CSRP3 CTF1 CTNNA3
DES DMD DOLK DSC2 DSCG2 DSP DTNA EMD EYA4 Figure 3. Dilated cardiomyopathy (DCM)
FHL1 FHL2 FKRP FKTN FLNC FXN GATAD1 GATA4 GATA6 gene panels are used for genetic testing.
GAA GLA HCN4 HRAS ILK JPH2 JUP KRAS LAMA4 Shown is a list of 111 genes offered from
LAMP2 LDB3 LMNA LRRC10 MAP2K1 MAP2K2 MIB1 MTND1 MTND5 multiple commercial testing laboratories for
MTND6 MTTD MTTG MTTH MTTI MTTK MTTL1 MTTL2 MTTM the evaluation of DCM. Those shown in black
MTTQ MTTS1 MTTS2 MURC MYBPC3 MYH6 MYH7 MYL2 MYL3 are commonly found on DCM panels from
MYLK2 MYOM1 MYOZ2 MYPN NEBL NEXN NKX2-5 NPPA NRAS multiple sources, whereas those shown in gray
PDLIM3 PKP2 PLKHM2 PLN PRDM16 PRKAG2 PTPN11 RAF1 RBM2 are found on only some panels reflecting their
RIT1 RYR2 SCN5A SGCD SLC22A5 SOS1 TAZ TBX20 TCAP role in syndromic cardiomyopathy, such as
TGFB3 TMEM43 TMPO TNNC1 TNNI3 TNNT2 TPM1 TRDN TTN Noonan syndrome, neuromuscular disease, and
TTR TXNRD2 VCL mitochondrial myopathies.

Found only on one panel and reflect genes implicated in Noonan, mitochondrial, neuromuscular
McNally and Mestroni   Dilated Cardiomyopathy Genetics and Mechanisms   735

Table.  Frequency and Phenotype Correlates of Definitive and Table. Continued


Putative DCM Genes
Frequency and Overlapping
Frequency and Overlapping Gene Protein Phenotypes
Gene Protein Phenotypes
Dystrophin Sarcolemma, structural
Sarcomere Force generation/ complex integrity
transmission
 DMD† Dystrophin Duchenne/Becker
 MYH6 α-Myosin heavy chain HCM, CHD, Sick sinus muscular dystrophy
syndrome
 DTNA α-Dystrobrevin LVNC
 MYH7*,† β-Myosin heavy chain 3%–4% of DCM; HCM,
 SGCA† α-Sarcoglycan LGMD2D
LVNC
 SGCB† β-Sarcoglycan LGMD2E
 TPM1† α-Tropomyosin 1%–2% of DCM; HCM,
LVNC  SGCD† δ-Sarcoglycan LGMD2F
 ACTC1† α-Cardiac actin HCM, LVNC  SGCG γ-Sarcoglycan LGMD2C
 TNNT2*,† Cardiac troponin T 3% of DCM; HCM, LVNC  CAV3 Caveolin HCM, LGMD1C, distal
myopathy
 TNNC1† Cardiac troponin C HCM, LVNC
 ILK Integrin-linked kinase
 TNNI3† Cardiac troponin I HCM
 FKTN Fukutin Dystroglycanopathy,
 MYBPC3 Myosin-binding protein C HCM, LVNC
congenital muscular
 TTN*,† Titin 12%–25% of DCM; HCM, dystrophy
tibial muscle dystrophy
 FKRP Fukutin-related protein Dystroglycanopathy, LGMD
 TNNI3K† Troponin I–interacting Conduction defect, atrial
Cytoskeleton Mechanotransduction/mechanosignaling/structural
kinase fibrillation
integrity
 MYL2 Myosin light chain 2, HCM
 DES† Desmin <1% of DCM;
regulatory
desminopathies,
 MYL3 Myosin light chain 3 HCM myofibrillar myopathy
 MYLK2 Myosin light chain kinase HCM; in panels, not  VCL† Metavinculin 1% of DCM
2 reported as DCM gene
Downloaded from http://ahajournals.org by on September 29, 2018

 FLNC† Filamin C 1% of DCM, AR-DCM;


 MYOM1 Myomesin 1 Myofibrillar myopathy; in myofibrillar myopathy,
panels, not reported as HCM, RCM
DCM gene
 SYNM Desmulin
 MYOZ2 Myozenin 2 HCM
 PDLIM3 PDZ LIM domain protein 3
Z disk Mechanosensing/
mechanosignaling  PLEC1 Plectin-1 LGMD2Q, epidermolysis
bullosa
 ACTN2† α-Actinin 2 LVNC
Desmosomes Cell–cell adhesion/mechanotransmission/
 BAG3† BCL2-associated Myofibrillar myopathy mechanosignaling
athanogene 3
 DSC2 Desmocollin 2 ARVC, mild palmoplantar
 CRYAB† α-B-crystallin Protein aggregation keratoderma
myopathy
 DSG2 Desmoglein 2 ARVC
 TCAP† Titin-cap/telethonin LGMD2G
 DSP*,† Desmoplakin 2% of DCM; ARVC
 MYPN Myopalladin HCM, RCM
 PKP2 Plakophilin 2 ARVC
 CSRP3† Muscle LIM protein HCM
 CTNNA3 Catenin α 3 ARVC; in panels, not
 NEXN Nexilin HCM reported as DCM gene
 FHL1 Four-and-a-half LIM EDMD, HCM Sarcoplasmic Ca homeostasis,
protein1 reticulum and contractility modulation,
 FHL2 Four-and-a-half LIM HCM cytoplasm signaling
protein 2  PLN† Phospholamban ARVC, HCM
 ANKRD1† Cardiac ankyrin repeat Congenital heart disease  RYR2 Ryanodine receptor 2, Ca ARVC, CPVT
protein channel
 MURC Muscle-related coiled coil  CALR3 Calreticulin-3 ARVC, HCM; in panels,
protein not reported as
 LDB3† Cypher/ZASP LVNC DCM gene

 NEBL† Nebulette LVNC, HCM  JPH2 Junctophilin 2 HCM

(Continued ) (Continued )
736  Circulation Research  September 15, 2017

Table. Continued Table. Continued
Frequency and Overlapping Frequency and Overlapping
Gene Protein Phenotypes Gene Protein Phenotypes
 DOLK Dolichol kinase Congenital disorder of  GATA5 GATA-binding protein 6 Atrial, ventricular septal
glycosylation defects, Fallot
 MAP2K1 Mitogen-activated protein Noonan syndrome; in  GATAD1 GATA zinc finger domain
kinase kinase 1 panels, not reported as protein 1
DCM gene
 NKX2-5 Cardiac-specific Atrial, ventricular septal
 MAP2K2 Mitogen-activated protein HCM, Noonan syndrome; homeobox 1 defects, Fallot, hypoplastic
kinase kinase 2 in panels, not reported as left heart
DCM gene
 ALSM1 Centrosome and basal Alstrom syndrome
 NRAS Neuroblastoma RAS viral HCM, Noonan syndrome body–associated protein (phenocopy)
oncogene homolog
 ALPK3 α-Kinase 3 Pediatric cardiomyopathy
 PRKAG2 Protein kinase AMP- HCM, WPW
 LRRC10 Leucine-rich repeat
activated noncatalytic
containing 10
subunit γ 2
 NPPA Natriuretic paptide A Atrial fibrillation
 PTPN11 Protein tyrosine HCM, Noonan and Leopard
phosphatase, nonreceptor syndromes  PLEKHM2 Pleckstrin homology LVNC
type 11 domain
 RAF1 Proto-oncogene HCM, Noonan and Leopard  TGFB3 Transforming growth ARVC; in panels, not
syndromes factor β 3 reported as DCM gene
 RIT1 Ras-like protein Noonan syndrome; in  TMEM43 Transmembrane protein ARVC, EDMD; in panels,
panels, not reported as 43 not reported as DCM gene
DCM gene
Ion channels
 SOS1 SOS Ras/Rac guanine HCM, Noonan syndrome;
 SCN5A*,† Type V voltage-gated 2%–3% of DCM; LQT,
nucleotide exchange in panels, not reported as
cardiac Na channel Brugada, atrial fibrillation,
factor 1 DCM gene
conduction defects
 TRDN Triadin CVPT; in panels, not
Downloaded from http://ahajournals.org by on September 29, 2018

 ABCC9† Sulfonylurea receptor Atrial fibrillation,


reported as DCM gene
2A, component of ATP- osteochondrodysplasia
Nuclear Nuclear structural integrity, sensitive potassium
envelope mechanotransduction, channel
mechanosignaling
 KCNQ1† Potassium channel Atrial fibrillation,
 LMNA*,† Lamin A/C 4%–8% of DCM; multiple LQT1, short QT1, Jervell
phenotypes, LGMD1B, and Lange-Nielsen
EDMD, progeria syndrome
 EMD† Emerin EDMD  CACNA1C Calcium voltage-gated Brugada syndrome,
channel subunit α1 C Timothy syndrome
 LAP2/TMPO Lamin-associated
polypeptide 2  HCN4 Hyperpolarization- Brugada, Sick sinus
activated cyclic syndrome
 SYNE1/2 Nesprin 1/2 EDMD, ataxia
nucleotide-gated
Nucleus Transcription cofactors, potassium channel 4
gene expression
Mitochondria Supply and regulation of
 EYA4 Eyes absent 4 Deafness energy metabolism
 FOXD4 Forkhead box D4  CPT2 Carnitine Carnitine deficiency,
palmitoyltransferase 2 myopathy, lethal neonatal
 HOPX Homeobox only protein
 FRDA/FXN Frataxin Fredreich ataxia
 NFKB1 NF-κ B1
 DNAJC19† HSP40 homolog, C19 3-methylglutaconic
 PRDM16 PR domain containing 16 LVNC
aciduria, type V
 TBX20 T-box 20 Atrial septal defect
 SDHA Succinate dehydrogenase Leigh syndrome
 ZBTB17 Zinc finger and BTB domain containing protein 17
 SOD2 Superoxide dismutase
 RBM20*,† RNA-binding protein 20 2% of DCM; RNA-binding
 TAZ/G4.5† Tafazzin LVNC, Barth syndrome,
protein of spliceosome of
endocardial
TTN and other proteins
fibroelastosis 2
 GATA4 GATA-binding protein 4 Atrial, ventricular septal
 CTF1 Cardiotrophin 1 cytokine
defects, Fallot

(Continued ) (Continued )
McNally and Mestroni   Dilated Cardiomyopathy Genetics and Mechanisms   737

Table. Continued disease usually appears in childhood with a typical DCM aris-


ing in the teenage years or early twenties. DCM in neuromus-
Frequency and Overlapping
Gene Protein Phenotypes cular disease is highly amenable to treatment and responds
well to guideline-directed medical therapy. Both forms of
 mtDNA Mitochondrial-encoded Typically syndromic,
myotonic muscular dystrophy, type 1 and type 2, can also be
TRNA genes mitochondial
myopathy; in panels, not associated with DCM.56–59 Atrial and ventricular arrhythmias
always reported as DCM are common in these tri- and tetra-nucleotide repeat expan-
genes sion disorders and should be aggressively managed. Myotonic
 TXNRD2 Thioredoxin reductase 2
dystrophy type 2 usually present in older individuals, and in
this case, genetic testing panels usually do not include these
Extracellular Cell adhesion and genes and thus the diagnosis can be easily missed especially if
matrix mechanosignaling
neuromuscular symptoms are not so pronounced.
 LAMA2 Laminin 2, merosin Congenital muscular
dystrophy DCM Genetics
 LAMA4 Laminin 4 The majority of genetic DCM is inherited in an autosomal
Lysosome
dominant pattern with variable expressivity and penetrance
(Figure 4) although specific forms of autosomal recessive,
 LAMP2 Lysosome-associated Danon disease
X-linked recessive, and mitochondrial inheritance each oc-
membrane protein 2
cur.14,19,60 De novo mutations also contribute to genetic car-
 AGL Amylo-α-1, 6-glucosidase, Glycogen storage disease diomyopathy and are defined when neither biological parent
4-α-glucanotransferase carries the offspring’s mutation. De novo mutations have been
 BRAF B-Raf proto-oncogene, Cardiofaciocutaneous, described in many different genes, and the presence of a de
serine/threonine kinase Leopard, Noonan novo variant can be used to define the pathogenic status of
syndromes genetic variants because the frequency of de novo variation in
 GAA Glucosidase α, acid Glycogen storage disease each genome is exceedingly rare. Thus, a novel mutation in-
 GLA α-Galactosidase Fabry disease troducing a protein-altering change in a cardiomyopathy gene
is typically considered pathogenic.
Other
Interpreting whether genetic variants are pathogenic is in-
 PSEN1 Presenillin 1, γ secretase Alzheimer disease creasing complex because of the vast amount of rare variation in
Downloaded from http://ahajournals.org by on September 29, 2018

intramembrane protease each human genome. The emerging consensus around interpre-
complex
tation of genetic variation and its effect on phenotype rely on a
 PSEN2 Presenillin 2, γ secretase Alzheimer disease classification system ranging from pathogenic, likely pathogen-
intramembrane protease ic, variant of uncertain significance, likely benign, and benign.61
complex The availability of large control cohorts provides invaluable in-
 CHRM2 Cholinergic receptor formation of the frequency of variants, and the largest available
muscarinic 2 data set is currently Exome Aggregation Consortium (http://
 HFE Hemochromatosis Phenocopy exac.broadinstitute.org), which collected exome sequencing
data of >60 000 individuals from a series of studies including
 HRAS HRas proto-oncogene, HCM, Costello syndrome;
GTPase in panels, not reported as the 1000 Genomes Project and the Exome Sequencing Project
DCM gene (http://evs.gs.washington.edu/EVS). The current trend is to con-
sider putative pathogenic variants as those that are either unique
 KRAS KRAS proto-oncogene, HCM, Costello syndrome;
GTPase in panels, not reported as to the DCM patient or family,52 or extremely rare (minor allele
DCM gene frequency <1×10–4).62 The recently adopted more stringent cri-
teria for genetic testing have prompted the reclassification of
 MIB1 Mitogen-activated protein LVNC; in panels, not
kinase kinase 2 reported as DCM gene
variants and indicate the needs of a continuous reanalysis of
data.52 The use of whole-exome/-genome sequencing in clinical
 SLC22A5 Cation/carnitine Skeletal myopathy laboratories warrants strong criteria to discriminate common
transporter
variants. At present, genetic testing typically relies on self-re-
 TTR Transthyretin Amyloidosis (phenocopy) ported ethnicity testing, and it is important to match ethnicity
AR-DCM indicates arrhythmogenic dilated cardiomyopathy; ARVC, between the proband and testing databases. However, at this
arrhythmogenic right ventricular cardiomyopathy; CHD, coronary heart point, this integration of common and rare variation is not rou-
disease; CPVT, cathecolaminergic polymorphic ventricular tachycardia; DCM, tinely being used in cardiomyopathy genetic testing, potentially
dilated cardiomyopathy; EDMD, Emery–Dreifuss muscular dystrophy; HCM,
contributing to false-positive interpretation.63
hypertrophic cardiomyopathy; LGMD, limb-girdle muscular dystrophy; LVNC,
left ventricular noncompaction; NF, nuclear factor; and RCM, restrictive DCM is genetically heterogeneous, and DCM genes en-
cardiomyopathy. code proteins of broad cellular functions. Mutations in genes
Frequent (*), definitive (†), and putative DCM genes,14,21,25,52,53 OMIM (www. encoding cytoskeletal, sarcomeric, mitochondrial, desmo-
omim.org, accessed January 19, 2017); GeneCards (www.genecards.org, somal, nuclear membrane, and RNA-binding proteins have all
accessed January 19, 2017). been linked to DCM. Thus, the pathological mechanisms that
lead to DCM are diverse. The genes below are listed in order
738  Circulation Research  September 15, 2017

Figure 4. A typical dilated cardiomyopathy


(DCM) pedigree is shown highlighting
variable expressivity. Most DCM is inherited
in an autosomal dominant pattern. Affected
individuals with DCM are shown in black. A
gene panel revealed the previously reported
pathogenic TPM1 E54K variant. The proband
TPM1 E54K+ TPM1 E54K+ (arrow) presented in early life requiring heart
EF45% EF45%
transplant during early childhood. Other members
of the family are in their third to sixth decade
with left ventricular ejection fraction (EF) 45%,
demonstrating variable expressivity of the primary
TPM1 E54K+ TPM1 E54K+ mutation. Environmental and additional genetic
CHF at 6 wks EF45%
Transplant age 3
factors may contribute to variable expressivity.
CHF indicates congestive heart failure.

of frequency for their contribution to genetic DCM with focus study showed that truncating variants in the general popula-
on the most commonly implicated genes and their mechanism tion are linked to eccentric cardiac remodeling, suggesting
of action if known. that TTN truncations may be at-risk alleles.74
TTN Peripartum cardiomyopathy is a heterogeneous syn-
The discovery of the role of TTN-truncating variants in DCM drome of mixed pathogenesis. Yet a subset of peripartum
has been major advance.64 The TTN gene encodes the giant cardiomyopathy is attributable to TTN-truncating variants.75,76
protein titin, which is the largest known protein expressed Peripartum cardiomyopathy can be associated with recovered
in the heart. Titin functions as a spring, providing passive LV function after pregnancy. Moreover, the observation that
force and regulating sarcomere contraction and signaling.65 TTN-truncating variants can be associated with recovery of
Titin is a large ≈35 000 amino acid protein that spans half function in DCM after LV assist device placement also sug-
the length of the sarcomere from Z disc to M band and is re- gests a dynamic state of TTN-truncating variants.77 Additional
ferred to as a third filament with the thin and thick filaments genes with mutations beyond TTN have also been described
that comprise the sarcomere. Proposed as a molecular rule in peripartum cardiomyopathy.23 Overall, the presence of
for the sarcomere, titin has domains that can accommodate TTN-truncating variants in the general population argues for
passive stiffness.66 Titin’s I band region includes the proline–
caution in interpreting these variants and again underscores
Downloaded from http://ahajournals.org by on September 29, 2018

glutamate–valine–lysine repetitive region, which is thought


the importance of familial segregation analysis. At this point,
to directly regulate passive tension. The I band region of the
TTN gene is encoded by 220 of TTN’s 360 exons. The large until more is known, the presence of a TTN truncation variant
size, repetitive nature, and extensive alternative splicing of
TTN make it challenging for genetic analysis. The proline–
glutamate–valine–lysine region is just carboxyl to the N2A
and N2B regions that interact with the four and half LIM
protein, identified as a modifier for HCM.67 Notably, TTN
is differentially spliced throughout heart development and
adaptively to distinct physiological states, including HF.68
The larger N2A form is associated with a more compliant
ventricle (Figure 5). In contrast, the smaller N2B form lacks
more of the repetitive units and is associated with stiffer
heart. Deep RNA sequencing of TTN from failed hearts sug-
gests highly variable exon usage in this region consistent
with even subtler defects in cardiac elasticity that may be
variable across regions of the LV.69
Using a TTN-specific array designed to capture all TTN
exons, it was shown that truncating variants of TTN contribute
to 20% to 25% of nonischemic DCM.64 Before this, only a few
missense TTN variants had been described linked to DCM.70
Induced pluripotent stem cells (iPSC) differentiated into car-
diomyocytes in culture demonstrate a paucity of sarcomeres,
suggesting that force may be impaired directly through sar-
Figure 5. Shown in the top is a schematic of the sarcomere
comere loss in TTN truncations.71 In these studies and oth- with the position of the thick myosin-containing filaments
ers, it has been shown that TTN truncations are observed at and the thin actin-containing filaments. Titin is considered a
a low frequency in the general population, ranging from 1% third filament of the sarcomere because its spans from Z disk
to 3%.64,69,72 There is a tendency for TTN truncations in DCM to M band. The lower schematics show the major splice forms
of titin (N2BA, N2B, N2A). The green box represents a unique
to distribute to the A band, rather than the I band,69 and TTN sequence domain. The PEVK region is named for the repetitive
truncations can also be associated with mild DCM.73 A recent amino acid sequences (proline, glutamine, valine, lysine).
McNally and Mestroni   Dilated Cardiomyopathy Genetics and Mechanisms   739

should trigger at least intermittent cardiac imaging and man- LMNA mutations associate frequently with a signature
agement aimed at reducing other stressors to the heart. of dysrhythmias that includes sinus node dysfunction, atrial
TTN has a high prevalence of missense variants, both rare fibrillation, atrioventricular node dysfunction, VT, ventricu-
and common.78 TTN missense variants have been reported in lar fibrillation, and SCD.48,49 Notably, cardiac conduction
ARVC and other forms of cardiomyopathy.70,78–81 In addition, system disease may precede the development of LV dilation
TTN missense variants have also been reported in skeletal my- and dysfunction, and the presence of early conduction system
opathy, including the common tibial myopathy.82,83 The enor- disease may suggest LMNA mutation. Aspects of the arrhyth-
mous number of TTN missense variants makes these variants mia and LV dysfunction phenotypes are not fully replicated in
exceedingly complex to interpret in the context of broad ge- the mouse models; namely, atrial fibrillation and ventricular
netic testing on individuals with DCM. arrhythmias are not frequently seen, and a homozygous mu-
Zebrafish have been used successfully to model myopa- tation is often needed to generate DCM. Heterozygous trun-
thies because of TTN mutations, demonstrating both cardiac cating LMNA mutations have a higher arrhythmia risk than
and skeletal muscle defects.84 A mouse model with an in-frame missense variants,49 and a prolonged PR interval indicates car-
deletion in the proline–glutamate–valine–lysine region of diac conduction system disease in laminopathy.100 The suscep-
TTN develops diastolic dysfunction, consistent with the com- tibility of the cardiac conduction system to LMNA mutations
plex role of titin for both systolic and diastolic dysfunctions.85 is not well understood.
In both rats and mice with heterozygous TTN truncation muta-
PLN
tions, additional stressors, such as transaortic constriction, are
The PLN gene encodes phospholamban, a 52 amino acid
used to promote the development of DCM.74,86
residue transmembrane protein that, when unphosphory-
LMNA lated, inhibits sarcoplasmic reticulum Ca2+-ATPase. Several
LMNA missense and truncating mutations account for 5% to dominant mutations in PLN have been associated with DCM,
8% of genetic DCM.87,88 Like TTN, LMNA mutations are in- including the R14del mutation that is a founder mutation in
herited in an autosomal dominant manner. The single LMNA the Netherlands and Germany. Thus, in some populations,
gene encodes lamins A and C, and differential splicing at the the percentage of DCM due to PLN mutations is high. The
3′ end results in 2 proteins that are identical across their first phenotype with PLN mutations is variable. Early onset DCM
566 amino acids; mutations in LMNA lead to a constellation with lethal ventricular arrhythmias was described.101,102
of diseases from premature aging to myopathies and DCM.89 Similarly, individuals from the Netherlands with the R14del
Mutations that alter processing of lamin A lead to accumu- founder mutation have a severe phenotype.103 However,
other reports suggest a milder phenotype.104,105 Identifying
Downloaded from http://ahajournals.org by on September 29, 2018

lation of prelamin A (sometimes called progerin), and these


have been associated with the premature aging syndrome the same primary mutation(s) with a range of phenotype de-
Hutchinson–Gilford progeria.90 LMNA mutations linked to pendent on genetic background supports that other factors,
autosomal dominant DCM are both missense and frameshift- including genetic factors, may modify the outcome of PLN-
ing in nature, and these mutations may occur anywhere along mediated DCM.
the length of the coding region. DCM-associated mutations iPSCs with the R14del PLN mutation were generated and
are not specifically associated with prelamin A accumulation; found to display features of cellular cardiomyopathy, includ-
thus, the basic mechanisms underlying the premature aging ing aberrant Ca2+ handling after caffeine and a higher per-
syndrome versus DCM seem to be distinct. The mechanisms centage of irregular Ca2+ transients, and these features were
responsible for autosomal dominant DCM LMNA mutations reversed after gene editing to correct the primary mutation.106
may be a mix of multiple defects, including dominant-nega- These same cells were used to engineer 3-dimensional human
heart tissues, and in this setting, more clear cut cardiomyop-
tive function and haploinsufficiency.91,92 Lamins A and C are
athic features were seen including reduced developed force
implicated in many different cellular processes from regulat-
that was improved after genetic correction.107 In iPSC-derived
ing gene expression, mechanosensing, DNA replication, and
cardiomyocytes and in hearts from PLN mutation carriers,
nuclear to cytoplasmic transport.
aggregates of phospholamban were seen in a perinuclear and
Loss of LMNA leads to a defect in mechanosignaling.93,94
cytoplasmic pattern, suggesting that aggregated phospholam-
Mechanosignaling defects were observed in cell with a homo-
ban contributes to the pathology possibly through aberrant
zygous deletion in the mouse Lmna gene. Male mice hetero-
autophagy.108
zygously deleted for Lmna exhibit cardiomyopathy features
in later life, suggesting that mice can be used to model lami- RBM20
nopathy.95 In LMNA-associated adult-onset DCM, the mTOR RNA-binding motif 20 is an RNA-binding protein expressed
pathway can be activated, and in animal models, inhibition of highly in both atria and ventricle. Dominant mutations in the
mTOR by temsirolimus or rapamycin was able to rescue the RBM20 gene were first described in DCM, where they con-
DCM phenotype.96,97 Mitogen-activated protein kinase signal- tribute to 1% to 5% of DCM.109,110 RBM20 is 1227 amino acid
ing is increased in these models, leading to clinical trials test- in length and contains a ribonucleic acid recognition motif
ing compounds aimed at reducing this signaling.98 Recently, domain between residues 525 and 600 amino acid. A second
early phase, encouraging results were reported from a phase conserved domain is found between 650 and 725 amino acid,
2 registration trial on A797 (Array Biopharma), an oral, se- and the mutations originally described in nonischemic DCM
lective p38 mitogen-activated protein kinase inhibitor in 12 fall within or near these domains. More recently, mutations in
patients with LMNA-associated DCM.99 a third conserved region were identified in a glutamate-rich
740  Circulation Research  September 15, 2017

region.111 As an RNA-binding protein, RBM20 is implicated F1759A in mice leads to atrial fibrillation and persistent sodi-
in tissue-specific splicing relevant to development and adap- um currents in atria and ventricles.121 Along with atrial fibrilla-
tation to disease states. In the heart, RBM20 regulates car- tion, these mice have progressive reduced LV ejection fraction
diac splicing, including the splicing of TTN.112–114 Thus, the (LVEF) consistent with a model for DCM. Thus, the distinct
downstream molecular consequences of RBM20 mutations roles of SCN5A in the myocardium and the conduction system
may share similarities to those occurring from TTN-truncating lead to a combination of arrhythmia and myocyte dysfunction.
variants.
iPSC-derived cardiomyocytes with the RBM20 R636S Cytoskeletal Genes
mutation develop a gene expression and splicing profile con- Genes encoding cardiac cytoskeletal proteins have been impli-
sistent with cardiomyopathy affecting not only TTN but also cated in DCM (Figure 6). For example, mutations in dystro-
the CAMK2D and CACNA1C genes.115 Sarcomeres within phin link to X-linked DCM and cardiomyopathy in Duchenne
these RBM20 mutant lines were thinner, similar to what was muscular dystrophy.122–124 Along with dystrophin mutations,
described for TTN mutant iPSC-cardiomyocytes.71 Recently, mutations in the sarcoglycan genes produce cardiomyopathy,
RBM20 was implicated in the production of circular RNAs usually associated with muscular dystrophy.55,125 In these dis-
from the TTN locus, and mice deleted for RBM20 failed to orders, the gene products play a normal and essential role in
produce these Ttn-derived circular RNAs.116 Although the managing sarcolemmal stability. Thus, in the absence of these
function of circular RNAs is not known, the authors described genes, the sarcolemma becomes unstable, leading to cardio-
that a subset of Ttn-derived circular RNAs were misregulated myocytes loss and heart dysfunction. Several emerging thera-
in DCM. pies for restoring dystrophin expression are being tested or
have been approved recently.126,127 Antisense oligonucleotides
SCN5A are being delivered to produce internally truncated dystrophin
SCN5A encodes the major sodium channel expressed in the proteins, and stop codon suppression compounds promote
heart, and heterozygous dominant mutations in SCN5A are read through of premature stop codons. The degree to which
also found in primary arrhythmia syndromes, including the these drugs access the human heart is not well-known, and
long QT and Brugada syndromes. Missense mutations in ongoing studies will be in a position to assess this in humans.
SCN5A have also been described in familial DCM, and these More recently, FLNC mutations, in the gene-encoding fila-
mutations carry a higher risk for arrhythmias.50,51 There is min C, have been described in DCM.128–130 Filamin C interacts
considerable genetic heterogeneity in the SCN5A gene in the with the dystrophin complex, and deletion of Flnc from the
general population, making it challenging to interpret rare mouse leads to skeletal myopathy.131,132 In humans, truncat-
variation in the SCN5A gene.62 Genotype–phenotype asso-
Downloaded from http://ahajournals.org by on September 29, 2018

ing mutations lead to cardiomyopathy that is associated with a


ciation studies may guide genotype-based therapies. For ex- high rate of ventricular arrhythmias and SCD, suggesting that
ample, the SCN5A R222Q falls within the S4 voltage sensor filamin C has a role in the cardiac conduction system in addi-
and is thought to enhance excitability. Treatment with lido- tion to the cardiomyocyte.
caine was observed to suppress the bigeminy associated with
cardiomyopathy.117 Mitochondrial Mutations
iPSCs have been used to model SCN5A mutations as- Both nuclear-encoded and mitochondrial-encoded mitochon-
sociated with primary human arrhythmia syndrome.118–120 drial genes lead to cardiomyopathy.133–136 Mutations in the mi-
Transgenic-directed inducible expression of the Scn5A tochondrial genomes may be difficult to identify and interpret

Figure 6. Shown are major components


within the cardiomyocyte with emphasis on
compartments that contribute to genetically
mediated dilated cardiomyopathy (DCM).
The extracellular matrix is shown in gray.
The dystrophin complex that includes the
sarcoglycans (green) is mutated in forms
of DCM with neuromuscular disease. The
sarcomeres (pink) include components
that are mutated in both hypertrophic
cardiomyopathy and DCM. Z band (dark
red) is a mechanosensing hub that serves to
transmit force from the sarcomeres. Mutations
in both mitochondrial-encoded (purple) and
nuclear-encoded mitochondria proteins
lead to cardiomyopathy. The nuclear lamina
include lamins A and C, and the gene LMNA is
commonly mutated in DCM (Illustration Credit:
Ben Smith).
McNally and Mestroni   Dilated Cardiomyopathy Genetics and Mechanisms   741

given the role of heteroplasmy and the fact that most genetic Clinical Cascade Screening
testing relies on peripheral blood DNA, which may or may Clinical cascade screening of relatives is recommended per
not match what occurs in the heart. Nuclear-encoded mito- AHA and European Society of Cardiology (ESC) guide-
chondrial genes follow either autosomal dominant or reces- lines.3,143 First-degree relatives of affected family members
sive inheritance, whereas mitochondrial-encoded genes show should be clinically evaluated (Figure 2). First line of screen-
maternal inheritance. ing usually relies on ECG and echocardiography to evaluate
ventricular size and function. Clinical history should evalu-
Additional Genetic Mutations in DCM ate signs and symptoms for arrhythmias and any history of
The proteins encoding the sarcomere, the unit of contraction, neuromuscular disease. Other cost-effective tools to consider
are also implicated in DCM. Recent data from clinical genetic in family member screening include ascertaining arrhythmia
testing indicate that MYH7, TNNT2, and TPM1 are the most history and Holter monitoring.47 A diagnosis of familial DCM
frequent mutated sarcomere genes in DCM, ranging from ≈2% is made where ≥2 family members are affected by DCM.19
to 4%, while MYBPC3 mutations are rare.52 Recently, trunca-
tions of the gene-encoding obscurin have been found in both Clinical Genetic Testing
LV noncompaction and DCM phenotypes.137 DCM may be According to the 2016 AHA Scientific Statement on DCM,3
best considered as a cardiomyopathy of mixed origin, familial genetic testing is recommended (with moderate level of con-
in ≈30% to 50% of patients, consistent with its being a ge- sensus) in patients with familial (Level of Evidence A) and
netic disease. Nonfamilial or idiopathic DCM may still have nonfamilial idiopathic cardiomyopathy in conjunction with
a genetic origin albeit a complex one. Because all DCM mu- genetic counseling (Level of Evidence B) although there is
tations have variable expressivity, this may support a model strong level of consensus in recommending mutation-specific
of oligogenic contribution along with environmental or other genetic testing for family members after the identification of
pathogenic stimuli. As it currently stands, not all DCM genes a DCM-causative mutation in the proband (Level of Evidence
have been discovered. Thus, the pathogenesis in nonfamilial B). Reflecting lack of full consensus in the field, the 2016 ESC
cases could be as yet undiscovered genes, low penetrance, de Position Statement143 is slightly different and recommends ge-
novo mutations, missing heritability because of multiple genes netic testing in all familial DCM or nonfamilial with clinical
with weaker effect, copy number variations, enhancer region clues (such as atrioventricular block or creatine kinase eleva-
mutations, and intronic variants or may be the result of the in- tion). Interestingly, the ESC Position Statement recommends
teraction between modifier genes and the environment.13,138 that genetic testing should be oriented by clinical diagnostic
clues and restricted to genes known to cause DCM although
Downloaded from http://ahajournals.org by on September 29, 2018

Digenic/Oligogenic Pathogenesis considering large panels of genes only when the family struc-
Occasionally, digenic variants have been reported; while it ture is large enough to permit segregation analysis.
is possible that some of these variants may in fact be variant Multiple commercial and academic vendors provide ge-
of uncertain significance or benign, in other cases the digenic netic testing gene panels under certification of the Clinical
condition have been found to segregate in informative kindred Laboratory Improvement Amendment. A typical DCM gene
and associate with a more severe phenotype. This is the case panel includes ≈40 to 50 genes (Figure 3).52,144 The probabil-
of a large family cosegregating a LMNA mutation, where rela- ity of positive genetic testing in familial DCM is overall in
tives with an additional TTN truncation showed worse outcome the range of 40% with the current next-generation sequenc-
and distinct pathological changes.139 With the growth of gene ing panels and seems not different compared with sporadic
panels for cardiomyopathy genetic testing, it is not uncommon cases.13,52 A pancardiomyopathy panel, as opposed to a DCM
to identify >1 potential pathogenic variant. Segregation anal- panel, is chosen when the phenotype is unclear, and a more
ysis may be helpful to clarify primary versus other variants. comprehensive screening is preferred. The level of evidence to
Compound mutations MYBPC3 lead to early onset, sometime support testing for some genes has been questioned, and this
neonatal cardiomyopathy.140–142 Thus, in families with multiple is largely based on a high level of genetic variation in those
affected members with DCM, a broad gene panel on a younger, genes in the population at large.145 Larger panels may yield
affected proband may provide a more comprehensive view of greater difficulty in interpreting the results because of variant
potential variation. Segregation testing for variants of interest of unknown significance. In a survey of 766 patients screened
can then help clarify those variants with greatest effect. in a clinical laboratory, with the introduction of next-genera-
tion sequencing technology and larger panels, while the rate
Genetic Testing of positive testing increased from 10% to 40%, the number of
variant of uncertain significance increased 10-fold.13,52,146
Family Screening
Genetic evaluation of DCM should begin with extensive and Genetic Counseling
accurate evaluations of the patient’s family history, involving Genetic counselors, especially those with experience in car-
at least 3 generations and including history of cardiomyopathy diovascular testing, provide both pre- and post-test counseling.
and history of sudden unexpected death at young age (<35 With the increasing complexity of cardiomyopathy genetic
years).3,19,20 This information will guide genetic testing, pro- testing, referral to specialized cardiovascular genetic clinics
vide good care to family members, and aid in the interpre- should be considered.20 Pre-test genetic counseling should in-
tation of the results and help identifying relatives at risk of volve discussion of potential genetic results (pathogenic mu-
disease. tations, variants of uncertain significance, and benign genetic
742  Circulation Research  September 15, 2017

variants). This counseling should also discuss the impact on to reduce morbidity in patients with sinus rhythm and a heart
insurability, reproduction, and lifestyle. Post-test counseling rate >70 bmp.3,150 The treatment of patients with an LVEF be-
focuses on variant interpretation/possible reinterpretation, re- tween 40% and 50%, defined in the ESC guidelines HF with
productive risks to offspring, and family testing. midrange ejection fraction and in the American College of
If a genetic mutation is identified, genetic cascade screen- Cardiology/AHA/Heart Failure Society of America guidelines
ing can be conducted with family members. Cascade genetic HF with improved ejection fraction, remains less clear.153
testing evaluates the specific family mutation rather than a
gene panel. Site-specific testing is of low cost and rapid turn- Arrhythmia Management
around so that this can be a cost-effective strategy to elimi- Arrhythmia management in genetic DCM patients follows
nate the need to serially follow gene mutation-negative family the general recommendations for prevention of SCD and ICD
members. For gene mutation-positive family members, cur- implantation based on the reduced LVEF (<35%). However,
rent guidelines suggest an annual clinical follow-up with ECG there are notable exceptions. First, a subset of patients with
and echocardiography.3,20,143 DCM present early in the disease course with life-threatening
In the absence of an informative genetic testing, asymp- ventricular arrhythmias (2%)156 or with frequent ventricular
tomatic first-degree relatives should be examined every 3 to 5 arrhythmias (30%), which are unrelated to the severity of
years.20 This strategy may allow prompt therapeutic measures LV dysfunction.47 These patients mirror the arrhythmogenic
in carriers showing sign of myocardial dysfunction. Even in presentation of ARVC and are described as arrhythmogenic
the absence of a positive genetic testing, longitudinal studies DCM.47 Arrhythmogenic DCM patients who present with
have shown the benefit of family screening and monitoring. syncope, nonsustained ventricular tachycardia, and frequent
In ≈10% of cases, mild myocardial dysfunction may progress premature ventricular contractions show a higher incidence of
into overt DCM within 5 years.29,147 Furthermore, clinical fam- life-threatening arrhythmic events (SCD, sustained VT, and
ily screening in DCM helps to identify affected relatives at cardiac arrest) compared with the other patients with DCM
earlier stages of disease, and this associates with improved while they show no difference in outcome of HF. The coex-
survival as compared with sporadic DCM.148 istence of a family history of SCD and the arrhythmogenic
DCM phenotype predicts a high risk of SCD events (Figure 7).
Management of DCM These recent data suggest that ventricular arrhythmias should
be systematically and carefully evaluated with monitoring and
Established Medical Therapies
that family history of ventricular arrhythmias predicts a poor
Management of DCM is focused on (1) LV dimension and
prognosis and increased risk of SCD.
function, (2) arrhythmia surveillance and treatment, and
Downloaded from http://ahajournals.org by on September 29, 2018

Arrhythmogenic DCM can be genetically determined,


(3) reducing congestive symptoms if present. Symptomatic
and a clear association between risk of SCD and gene
DCM and HF with reduced LVEF is managed following
current AHA/American College of Cardiology and ESC has been established with the LMNA gene. The 2015 ESC
guidelines. Guideline-directed therapy includes angiotensin- guidelines for the management of patients with ventricular
converting enzyme inhibitors or angiotensin receptor block- arrhythmias and the prevention of SCD157 recommend an
ers, in association with β-blockers, aldosterone antagonists, ICD in patients with DCM and a confirmed disease-causing
and in selected cases, vasodilators.149–151 Medications should LMNA mutation and clinical risk factors (nonsustained ven-
be titrated to the dose used in clinical trials unless limited tricular tachycardia during ambulatory ECG monitoring,
by side effects.152 Patients with DCM on optimal therapy LVEF 45%, male sex, and truncating mutations [Class IIa;
with complete left bundle branch block may benefit from Level of Evidence B]). Likewise, the Heart Rhythm Society/
cardiac resynchronization.149,150,153,154 The improvement of American College of Cardiology/AHA Expert Consensus
survival with ICDs in patients with LVEF <35% is also well Statement on the use of ICD therapy inpatients highlighted
established.149,150 Patients with refractory HF may require the increased risk for SCD in LMNA carriers.158 Risk fac-
advanced therapies, including LV assist devices or cardiac tors for SCD identified in 2 large LMNA carrier cohorts in
transplant.149,150 Europe49 and in the United States159 include nonsustained
Two newer medications for those not responding to opti- ventricular tachycardia during ambulatory ECG monitor-
mal medical therapy include the angiotensin receptor-nepryli- ing,49 LVEF <45%49 to 50%,159 male sex, and truncating
sin inhibitor (valsartan/sacubitril) and the sinoatrial modulator, mutations.49,159 Kumar et al159 reported life-threatening ven-
ivabradine.3,150,153 Updated guidelines recommend switching tricular arrhythmia rates of 3% to 7% per year, which is
from angiotensin-converting enzyme inhibitor/angiotensin re- higher or comparable to other known groups of high-risk
ceptor blockers to angiotensin receptor-neprylisin inhibitor in patients including those with LVEF<25%, ARVC, HCM,
class II to III patients who are not responding to optimal medi- and high-risk ischemic cardiomyopathy.
cal therapy (ESC guidelines150) or even in those responding to DCM also carries an increased stroke risk although less
optimal therapy, considering the evidence of superior benefit is known about the specific risk in genetic cardiomyopathy.
of angiotensin receptor-neprylisin inhibitor over angiotensin- As such, anticoagulation should be considered to reduce the
converting enzyme inhibitor/angiotensin receptor blockers in risk of stroke in DCM with atrial fibrillation in particular in
terms of mortality and morbidity (AHA/American College of the presence of additional risk factor for cardioembolic events,
Cardiology/Heart Failure Society of America guidelines155). such as history of hypertension, diabetes mellitus, previous
Ivabradine instead can be added to optimal medical therapy stroke or transient ischemic attack, or ≥75 years of age.149
McNally and Mestroni   Dilated Cardiomyopathy Genetics and Mechanisms   743

Figure 7. Sudden cardiac death


(SCD) and life-threatening ventricular
arrhythmias in dilated cardiomyopathy
(DCM). Cox-estimated event-free
survival stratified by 2 risk factors, family
history of SCD or ventricular arrhythmias
(sustained ventricular tachycardia
[SVT] or ventricular fibrillation [VF])
and arrhythmogenic DCM (AR-DCM)
diagnosis, in a cohort of 285 patients with
DCM. The AR-DCM phenotype (P=0.02)
and family history of SCD or ventricular
arrhythmias (SCD/SVT/VF; P=0.038)
showed an additive prognostic effect
on mortality for arrhythmic events. See
reference Spezzacatene et al.47

Management of Genotype-Positive/Phenotype- cardiomyopathy. iPSCs can be more readily generated from


Negative Patients patients with DCM, and these cells can be differentiated into
Genetic testing identifies genotype-positive/phenotype- cardiomyocytes for study. A major limitation of iPSC-derived
negative family members, and this information is useful for cardiomyocytes is their relative immaturity and variabil-
prevention strategies, lifestyle recommendations, including ity from culture to culture, but nonetheless, these cells can
participation in competitive sports, and possible arrhythmia be used to study cellular properties reflective of DCM fea-
Downloaded from http://ahajournals.org by on September 29, 2018

management. The current guidelines recommend observation tures. As yet, these stem cells are not yet sufficiently mature
in asymptomatic at-risk relatives with yearly clinical assess- for treating cardiomyopathy161 although they are yielding an
ment.20 There is less consensus on the medical management, important platform for understanding mechanisms and testing
timing, and the type of intervention in these patients. Current therapies.165
guidelines recommend control of risk factors in this stage,
such as hypertension.149 ESC and AHA suggest restriction Conclusions and Future Directions
from competitive sports in DCM genotype-positive/pheno- Better understanding of the DCM phenome and recent im-
type-negative although evidence is lacking to support these provements in sequencing technology to define DCM genome
recommendations.160 When initial signs of ventricular dys- will eventually improve the diagnosis, prevention, and ther-
function present during follow-up, earlier institution of medi- apy of this disease. Next-generation sequencing technology
cal therapy can begin although the exact timing of this is not provides a cost effective and accurate diagnostic method to
known.20 yield biomarkers that indicate disease risk, especially within
families. With this progress, criteria for pathogenic muta-
Cardiac Regeneration for DCM? tions are evolving and becoming more and more stringent
DCM is usually associated with cardiomyocytes loss, and the and may require re-evaluation of the molecular diagnosis over
human heart has limited regenerative capacity. Strategies for time. Several questions still remain in DCM management that
regeneration and repair include the application of a cell suspen- prompt future investigations, such as the interpretation of ge-
sion, growth factors, miRNAs, and the implantation of an engi- netic testing, the correct treatment of pre-clinical asymptom-
neered tissue.161–163 Various studies and clinical trials have tested atic DCM gene carriers, and the development of gene- and
cardiac progenitor cells, bone marrow–derived stem cells, and mechanism-specific therapies.
pluripotent stem cells.164 Frustratingly, these clinical interven-
tions demonstrated safety but often failed to prove functional Sources of Funding
improvement. The mechanism underlying the potential effects This work was supported by National Institutes of Health (NIH)
HL128075 to E.M. McNally and NIH UL1 TR001082, R01 HL69071,
of bone marrow–derived stem cells is unclear; the injected cells R01 116906, and in part by a Trans-Atlantic Network of Excellence
do not appear to remain in the cardiac tissue but may release grant from the Leducq Foundation (14-CVD 03) to L. Mestroni.
paracrine factors and recruit cardiac progenitor cells.161
Stem cells are now being used to provide cellular mod- Disclosures
els of DCM. The absence of human cardiomyocyte cell E.M. McNally is a consultant to Invitae and L. Mestroni is a consul-
lines has been a problem for advancing research in human tant to Array Biopharma.
744  Circulation Research  September 15, 2017

References 18. Mestroni L, Rocco C, Gregori D, Sinagra G, Di Lenarda A, Miocic S,


Vatta M, Pinamonti B, Muntoni F, Caforio AL, McKenna WJ, Falaschi
1. Elliott P, Andersson B, Arbustini E, et al. Classification of the cardiomy-
A, Giacca M, Camerini. Familial dilated cardiomyopathy: evidence for
opathies: a position statement from the European Society of Cardiology
genetic and phenotypic heterogeneity. Heart Muscle Disease Study Group.
Working Group on Myocardial and Pericardial Diseases. Eur Heart J.
J Am Coll Cardiol. 1999;34:181–190.
2008;29:270–276. doi: 10.1093/eurheartj/ehm342.
19. Mestroni L, Maisch B, McKenna WJ, Schwartz K, Charron P, Rocco
2. Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D,
C, Tesson F, Richter A, Wilke A, Komajda M. Guidelines for the study
Moss AJ, Seidman CE, Young JB; American Heart Association; Council
of familial dilated cardiomyopathies. Collaborative Research Group of
on Clinical Cardiology, Heart Failure and Transplantation Committee;
the European Human and Capital Mobility Project on Familial Dilated
Quality of Care and Outcomes Research and Functional Genomics and
Cardiomyopathy. Eur Heart J. 1999;20:93–102.
Translational Biology Interdisciplinary Working Groups; Council on
20. Hershberger RE, Lindenfeld J, Mestroni L, Seidman CE, Taylor MR,
Epidemiology and Prevention. Contemporary definitions and classifica-
Towbin JA; Heart Failure Society of America. Genetic evaluation of
tion of the cardiomyopathies: an American Heart Association Scientific
cardiomyopathy–a Heart Failure Society of America practice guideline. J
Statement from the Council on Clinical Cardiology, Heart Failure and
Card Fail. 2009;15:83–97. doi: 10.1016/j.cardfail.2009.01.006.
Transplantation Committee; Quality of Care and Outcomes Research and
Functional Genomics and Translational Biology Interdisciplinary Working 21. Burke MA, Cook SA, Seidman JG, Seidman CE. Clinical and mecha-
Groups; and Council on Epidemiology and Prevention. Circulation. nistic insights into the genetics of cardiomyopathy. J Am Coll Cardiol.
2006;113:1807–1816. doi: 10.1161/CIRCULATIONAHA.106.174287. 2016;68:2871–2886. doi: 10.1016/j.jacc.2016.08.079.
3. Bozkurt B, Colvin M, Cook J, et al; American Heart Association 22. Lopez-Ayala JM, Pastor-Quirante F, Gonzalez-Carrillo J, Lopez-Cuenca
Committee on Heart Failure and Transplantation of the Council on D, Sanchez-Munoz JJ, Oliva-Sandoval MJ, Gimeno JR. Genetics of myo-
Clinical Cardiology; Council on Cardiovascular Disease in the Young; carditis in arrhythmogenic right ventricular dysplasia. Heart Rhythm.
Council on Cardiovascular and Stroke Nursing; Council on Epidemiology 2015;12:766–773. doi: 10.1016/j.hrthm.2015.01.001.
and Prevention; and Council on Quality of Care and Outcomes Research. 23. Morales A, Painter T, Li R, Siegfried JD, Li D, Norton N, Hershberger
Current diagnostic and treatment strategies for specific dilated cardio- RE. Rare variant mutations in pregnancy-associated or peripartum
myopathies: a scientific statement from the American Heart Association. cardiomyopathy. Circulation. 2010;121:2176–2182. doi: 10.1161/
Circulation. 2016;134:e579–e646. doi: 10.1161/CIR.0000000000000455. CIRCULATIONAHA.109.931220.
4. Shore S, Grau-Sepulveda MV, Bhatt DL, Heidenreich PA, Eapen ZJ, 24. van Spaendonck-Zwarts KY, van Tintelen JP, van Veldhuisen DJ, van
Hernandez AF, Yancy CW, Fonarow GC. Characteristics, treatments, and der Werf R, Jongbloed JD, Paulus WJ, Dooijes D, van den Berg MP.
outcomes of hospitalized heart failure patients stratified by etiologies Peripartum cardiomyopathy as a part of familial dilated cardiomyopathy.
of cardiomyopathy. JACC Heart Fail. 2015;3:906–916. doi: 10.1016/j. Circulation. 2010;121:2169–2175. doi: 10.1161/CIRCULATIONAHA.
jchf.2015.06.012. 109.929646.
5. Codd MB, Sugrue DD, Gersh BJ, Melton LJ 3rd. Epidemiology of idio- 25. Japp AG, Gulati A, Cook SA, Cowie MR, Prasad SK. The diagnosis and
pathic dilated and hypertrophic cardiomyopathy. A population-based study evaluation of dilated cardiomyopathy. J Am Coll Cardiol. 2016;67:2996–
in Olmsted County, Minnesota, 1975-1984. Circulation. 1989;80:564–572. 3010. doi: 10.1016/j.jacc.2016.03.590.
6. Torp A. Incidence of congestive cardiomyopathy. Postgrad Med J. 26. Burridge PW, Li YF, Matsa E, et al. Human induced pluripotent stem cell-
1978;54:435–439. derived cardiomyocytes recapitulate the predilection of breast cancer pa-
7. Gillum RF. Idiopathic cardiomyopathy in the United States, 1970-1982. tients to doxorubicin-induced cardiotoxicity. Nat Med. 2016;22:547–556.
Am Heart J. 1986;111:752–755. doi: 10.1038/nm.4087.
Downloaded from http://ahajournals.org by on September 29, 2018

8. Manolio TA, Baughman KL, Rodeheffer R, Pearson TA, Bristow JD, 27. Fatkin D; Members of the CSANZ Cardiac Genetic Diseases Council
Michels VV, Abelmann WH, Harlan WR. Prevalence and etiology of idio- Writing Group. Guidelines for the diagnosis and management of famil-
pathic dilated cardiomyopathy (summary of a National Heart, Lung, and ial dilated cardiomyopathy. Heart Lung Circ. 2011;20:691–693. doi:
Blood Institute workshop). Am J Cardiol. 1992;69:1458–1466. 10.1016/j.hlc.2011.07.008.
9. Miura K, Nakagawa H, Morikawa Y, Sasayama S, Matsumori A, Hasegawa 28. Crispell KA, Hanson EL, Coates K, Toy W, Hershberger RE. Periodic re-
K, Ohno Y, Tamakoshi A, Kawamura T, Inaba Y. Epidemiology of idio- screening is indicated for family members at risk of developing familial
pathic cardiomyopathy in Japan: results from a nationwide survey. Heart. dilated cardiomyopathy. J Am Coll Cardiol. 2002;39:1503–1507.
2002;87:126–130. 29. Mahon NG, Murphy RT, MacRae CA, Caforio AL, Elliott PM, McKenna
10. Amoah AG, Kallen C. Aetiology of heart failure as seen from a National WJ. Echocardiographic evaluation in asymptomatic relatives of patients
Cardiac Referral Centre in Africa. Cardiology. 2000;93:11–18. doi: 6996. with dilated cardiomyopathy reveals preclinical disease. Ann Intern Med.
11. Williams DG, Olsen EG. Prevalence of overt dilated cardiomyopathy in 2005;143:108–115.
two regions of England. Br Heart J. 1985;54:153–155. 30. Captur G, Lopes LR, Mohun TJ, et al. Prediction of sarcomere mutations
12. Rakar S, Sinagra G, Di Lenarda A, Poletti A, Bussani R, Silvestri F, in subclinical hypertrophic cardiomyopathy. Circ Cardiovasc Imaging.
Camerini F. Epidemiology of dilated cardiomyopathy. A prospective 2014;7:863–871. doi: 10.1161/CIRCIMAGING.114.002411.
post-mortem study of 5252 necropsies. The Heart Muscle Disease Study 31. Sefa Okten M, Tuluce K, Yakar Tuluce S, Kilic S, Soner Kemal H, Sayin
Group. Eur Heart J. 1997;18:117–123. A, Vuran O, Yagmur B, Mutlu I, Simsek E, Soydas Cinar C, Gurgun C.
13. Sweet M, Taylor MR, Mestroni L. Diagnosis, prevalence, and screen- Screening first-degree relatives of patients with idiopathic dilated cardio-
ing of familial dilated cardiomyopathy. Expert Opin Orphan Drugs. myopathy [published online ahead of print November 10, 2016]. Herz.
2015;3:869–876. doi: 10.1517/21678707.2015.1057498. doi: 10.1007/s00059-016-4498-1. https://link.springer.com/article/10.
14. Hershberger RE, Hedges DJ, Morales A. Dilated cardiomyopathy:
1007%2Fs00059-016-4498-1.
the complexity of a diverse genetic architecture. Nat Rev Cardiol. 32. Cordero-Reyes AM, Youker K, Estep JD, Torre-Amione G, Nagueh SF.
2013;10:531–547. doi: 10.1038/nrcardio.2013.105. Molecular and cellular correlates of cardiac function in end-stage DCM:
15. Petretta M, Pirozzi F, Sasso L, Paglia A, Bonaduce D. Review and
a study using speckle tracking echocardiography. JACC Cardiovasc
metaanalysis of the frequency of familial dilated cardiomyopathy. Am J Imaging. 2014;7:441–452. doi: 10.1016/j.jcmg.2013.11.010.
Cardiol. 2011;108:1171–1176. doi: 10.1016/j.amjcard.2011.06.022. 33. Assomull RG, Prasad SK, Lyne J, Smith G, Burman ED, Khan M,

16. Grünig E, Tasman JA, Kücherer H, Franz W, Kübler W, Katus HA.
Sheppard MN, Poole-Wilson PA, Pennell DJ. Cardiovascular magnetic
Frequency and phenotypes of familial dilated cardiomyopathy. J Am Coll resonance, fibrosis, and prognosis in dilated cardiomyopathy. J Am Coll
Cardiol. 1998;31:186–194. Cardiol. 2006;48:1977–1985.
17. Ganesh SK, Arnett DK, Assimes TL, et al; American Heart Association 34. Gao P, Yee R, Gula L, Krahn AD, Skanes A, Leong-Sit P, Klein GJ, Stirrat
Council on Functional Genomics and Translational Biology; American J, Fine N, Pallaveshi L, Wisenberg G, Thompson TR, Prato F, Drangova
Heart Association Council on Epidemiology and Prevention; American M, White JA. Prediction of arrhythmic events in ischemic and dilated
Heart Association Council on Basic Cardiovascular Sciences; American cardiomyopathy patients referred for implantable cardiac defibrillator:
Heart Association Council on Cardiovascular Disease in the Young; evaluation of multiple scar quantification measures for late gadolinium
American Heart Association Council on Cardiovascular and Stroke enhancement magnetic resonance imaging. Circ Cardiovasc Imaging.
Nursing; American Heart Association Stroke Council. Genetics and ge- 2012;5:448–456. doi: 10.1161/CIRCIMAGING.111.971549.
nomics for the prevention and treatment of cardiovascular disease: update: 35. Perazzolo Marra M, De Lazzari M, Zorzi A, et al. Impact of the pres-
a scientific statement from the American Heart Association. Circulation. ence and amount of myocardial fibrosis by cardiac magnetic reso-
2013;128:2813–2851. doi: 10.1161/01.cir.0000437913.98912.1d. nance on arrhythmic outcome and sudden cardiac death in nonischemic
McNally and Mestroni   Dilated Cardiomyopathy Genetics and Mechanisms   745

dilated cardiomyopathy. Heart Rhythm. 2014;11:856–863. doi: 10.1016/j. dilated cardiomyopathy and commonly localize to the voltage-sensing
hrthm.2014.01.014. mechanism. J Am Coll Cardiol. 2011;57:2160–2168. doi: 10.1016/j.
36. Gulati A, Jabbour A, Ismail TF, et al. Association of fibrosis with mortality jacc.2010.09.084.
and sudden cardiac death in patients with nonischemic dilated cardiomy- 51. Mann SA, Castro ML, Ohanian M, Guo G, Zodgekar P, Sheu A,

opathy. JAMA. 2013;309:896–908. doi: 10.1001/jama.2013.1363. Stockhammer K, Thompson T, Playford D, Subbiah R, Kuchar D,
37. Ordovas KG, Higgins CB. Delayed contrast enhancement on MR images Aggarwal A, Vandenberg JI, Fatkin D. R222Q SCN5A mutation is associ-
of myocardium: past, present, future. Radiology. 2011;261:358–374. doi: ated with reversible ventricular ectopy and dilated cardiomyopathy. J Am
10.1148/radiol.11091882. Coll Cardiol. 2012;60:1566–1573. doi: 10.1016/j.jacc.2012.05.050.
38. Dawson DK, McLernon DJ, Raj VJ, Maceira AM, Prasad S, Frenneaux 52. Pugh TJ, Kelly MA, Gowrisankar S, et al. The landscape of genetic varia-
MP, Pennell DJ, Kilner PJ. Cardiovascular magnetic resonance determi- tion in dilated cardiomyopathy as surveyed by clinical DNA sequencing.
nants of left ventricular noncompaction. Am J Cardiol. 2014;114:456– Genet Med. 2014;16:601–608. doi: 10.1038/gim.2013.204.
462. doi: 10.1016/j.amjcard.2014.05.017. 53. Marian AJ, van Rooij E, Roberts R. Genetics and genomics of single-gene
39. Amzulescu MS, Rousseau MF, Ahn SA, Boileau L, de Meester de
cardiovascular diseases: common hereditary cardiomyopathies as proto-
Ravenstein C, Vancraeynest D, Pasquet A, Vanoverschelde JL, Pouleur AC, types of single-gene disorders. J Am Coll Cardiol. 2016;68:2831–2849.
Gerber BL. Prognostic impact of hypertrabeculation and noncompaction doi: 10.1016/j.jacc.2016.09.968.
phenotype in dilated cardiomyopathy: a CMR study. JACC Cardiovasc 54. Brown CA, Lanning RW, McKinney KQ, et al. Novel and recurrent muta-
Imaging. 2015;8:934–946. doi: 10.1016/j.jcmg.2015.04.015. tions in lamin A/C in patients with Emery-Dreifuss muscular dystrophy.
40. Gil KE, Pawlak A, Gil RJ, Frontczak-Baniewicz M, Bil J. The role of in- Am J Med Genet. 2001;102:359–367.
vasive diagnostics and its impact on the treatment of dilated cardiomyopa- 55. McNally EM. New approaches in the therapy of cardiomyopathy in mus-
thy: a systematic review. Adv Med Sci. 2016;61:331–343. doi: 10.1016/j. cular dystrophy. Annu Rev Med. 2007;58:75–88. doi: 10.1146/annurev.
advms.2016.07.001. med.58.011706.144703.
41. Caforio AL, Pankuweit S, Arbustini E, et al; European Society of
56. Tanawuttiwat T, Wagner KR, Tomaselli G, Nazarian S. Left ventricu-
Cardiology Working Group on Myocardial and Pericardial Diseases. lar dysfunction and conduction disturbances in patients with myotonic
Current state of knowledge on aetiology, diagnosis, management, and muscular dystrophy type I and II. JAMA Cardiol. 2017;2:225–228. doi:
therapy of myocarditis: a position statement of the European Society of 10.1001/jamacardio.2016.4145.
Cardiology Working Group on Myocardial and Pericardial Diseases. Eur 57. Sommerville RB, Vincenti MG, Winborn K, Casey A, Stitziel NO,

Heart J. 2013;34:2636–2648, 2648a. doi: 10.1093/eurheartj/eht210. Connolly AM, Mann DL. Diagnosis and management of adult hereditary
42. Cooper LT, Baughman KL, Feldman AM, Frustaci A, Jessup M, Kuhl cardio-neuromuscular disorders: a model for the multidisciplinary care of
U, Levine GN, Narula J, Starling RC, Towbin J, Virmani R; American complex genetic disorders. Trends Cardiovasc Med. 2017;27:51–58. doi:
Heart Association; American College of Cardiology; European Society 10.1016/j.tcm.2016.06.005.
of Cardiology. The role of endomyocardial biopsy in the management of 58. McNally EM, Sparano D. Mechanisms and management of the heart
cardiovascular disease: a scientific statement from the American Heart in myotonic dystrophy. Heart. 2011;97:1094–1100. doi: 10.1136/
Association, the American College of Cardiology, and the European hrt.2010.214197.
Society of Cardiology. Circulation. 2007;116:2216–2233. doi: 10.1161/ 59. Wahbi K, Babuty D, Probst V, et al. Incidence and predictors of sudden death,
CIRCULATIONAHA.107.186093. major conduction defects and sustained ventricular tachyarrhythmias in 1388
43. Cooper LT, Baughman KL, Feldman AM, Frustaci A, Jessup M, Kuhl patients with myotonic dystrophy type 1. Eur Heart J. 2017;38:751–758.
U, Levine GN, Narula J, Starling RC, Towbin J, Virmani R. The role of 60. McNally EM, Golbus JR, Puckelwartz MJ. Genetic mutations and mecha-
Downloaded from http://ahajournals.org by on September 29, 2018

endomyocardial biopsy in the management of cardiovascular disease: a nisms in dilated cardiomyopathy. J Clin Invest. 2013;123:19–26. doi:
scientific statement from the American Heart Association, the American 10.1172/JCI62862.
College of Cardiology, and the European Society of Cardiology Endorsed 61. McNally EM, Barefield DY, Puckelwartz MJ. The genetic landscape of
by the Heart Failure Society of America and the Heart Failure Association cardiomyopathy and its role in heart failure. Cell Metab. 2015;21:174–
of the European Society of Cardiology. Eur Heart J. 2007;28:3076–3093. 182. doi: 10.1016/j.cmet.2015.01.013.
doi: 10.1093/eurheartj/ehm456. 62. Walsh R, Thomson KL, Ware JS, et al. Reassessment of Mendelian gene
44. Asimaki A, Tandri H, Huang H, Halushka MK, Gautam S, Basso C, pathogenicity using 7,855 cardiomyopathy cases and 60,706 reference
Thiene G, Tsatsopoulou A, Protonotarios N, McKenna WJ, Calkins H, samples. Genet Med. 2017;19:192–203. doi: 10.1038/gim.2016.90.
Saffitz JE. A new diagnostic test for arrhythmogenic right ventricular 63. Li Q, Gruner C, Chan RH, Care M, Siminovitch K, Williams L, Woo
cardiomyopathy. N Engl J Med. 2009;360:1075–1084. doi: 10.1056/ A, Rakowski H. Genotype-positive status in patients with hypertro-
NEJMoa0808138. phic cardiomyopathy is associated with higher rates of heart fail-
45. Asimaki A, Protonotarios A, James CA, Chelko SP, Tichnell C, Murray ure events. Circ Cardiovasc Genet. 2014;7:416–422. doi: 10.1161/
B, Tsatsopoulou A, Anastasakis A, te Riele A, Kléber AG, Judge DP, CIRCGENETICS.113.000331.
Calkins H, Saffitz JE. Characterizing the molecular pathology of arrhyth- 64. Herman DS, Lam L, Taylor MR, et al. Truncations of titin causing di-
mogenic cardiomyopathy in patient buccal mucosa cells. Circ Arrhythm lated cardiomyopathy. N Engl J Med. 2012;366:619–628. doi: 10.1056/
Electrophysiol. 2016;9:e003688. doi: 10.1161/CIRCEP.115.003688. NEJMoa1110186.
46. Felker GM, Thompson RE, Hare JM, Hruban RH, Clemetson DE, Howard 65. LeWinter MM, Granzier HL. Cardiac titin and heart disease. J Cardiovasc
DL, Baughman KL, Kasper EK. Underlying causes and long-term sur- Pharmacol. 2014;63:207–212. doi: 10.1097/FJC.0000000000000007.
vival in patients with initially unexplained cardiomyopathy. N Engl J Med. 66. Anderson BR, Granzier HL. Titin-based tension in the cardiac sarcomere:
2000;342:1077–1084. doi: 10.1056/NEJM200004133421502. molecular origin and physiological adaptations. Prog Biophys Mol Biol.
47. Spezzacatene A, Sinagra G, Merlo M, Barbati G, Graw SL, Brun F, 2012;110:204–217. doi: 10.1016/j.pbiomolbio.2012.08.003.
Slavov D, Di Lenarda A, Salcedo EE, Towbin JA, Saffitz JE, Marcus FI, 67. Christodoulou DC, Wakimoto H, Onoue K, et al. 5’RNA-Seq iden-

Zareba W, Taylor MR, Mestroni L; Familial Cardiomyopathy Registry. tifies Fhl1 as a genetic modifier in cardiomyopathy. J Clin Invest.
Arrhythmogenic phenotype in dilated cardiomyopathy: natural his- 2014;124:1364–1370. doi: 10.1172/JCI70108.
tory and predictors of life-threatening arrhythmias. J Am Heart Assoc. 68. Nagueh SF, Shah G, Wu Y, Torre-Amione G, King NM, Lahmers S,
2015;4:e002149. doi: 10.1161/JAHA.115.002149. Witt CC, Becker K, Labeit S, Granzier HL. Altered titin expression,
48. van Berlo JH, de Voogt WG, van der Kooi AJ, van Tintelen JP, Bonne myocardial stiffness, and left ventricular function in patients with di-
G, Yaou RB, Duboc D, Rossenbacker T, Heidbüchel H, de Visser M, lated cardiomyopathy. Circulation. 2004;110:155–162. doi: 10.1161/01.
Crijns HJ, Pinto YM. Meta-analysis of clinical characteristics of 299 car- CIR.0000135591.37759.AF.
riers of LMNA gene mutations: do lamin A/C mutations portend a high 69. Roberts AM, Ware JS, Herman DS, et al. Integrated allelic, transcrip-
risk of sudden death? J Mol Med (Berl). 2005;83:79–83. doi: 10.1007/ tional, and phenomic dissection of the cardiac effects of titin trunca-
s00109-004-0589-1. tions in health and disease. Sci Transl Med. 2015;7:270ra6. doi: 10.1126/
49. van Rijsingen IA, Arbustini E, Elliott PM, et al. Risk factors for malignant scitranslmed.3010134.
ventricular arrhythmias in lamin a/c mutation carriers a European cohort 70. Gerull B, Gramlich M, Atherton J, McNabb M, Trombitás K, Sasse-
study. J Am Coll Cardiol. 2012;59:493–500. doi: 10.1016/j.jacc.2011.08.078. Klaassen S, Seidman JG, Seidman C, Granzier H, Labeit S, Frenneaux M,
50. McNair WP, Sinagra G, Taylor MR, Di Lenarda A, Ferguson DA, Salcedo Thierfelder L. Mutations of TTN, encoding the giant muscle filament titin,
EE, Slavov D, Zhu X, Caldwell JH, Mestroni L; Familial Cardiomyopathy cause familial dilated cardiomyopathy. Nat Genet. 2002;30:201–204. doi:
Registry Research Group. SCN5A mutations associate with arrhythmic 10.1038/ng815.
746  Circulation Research  September 15, 2017

71. Hinson JT, Chopra A, Nafissi N, et al. HEART DISEASE. Titin mutations 88. Parks SB, Kushner JD, Nauman D, Burgess D, Ludwigsen S, Peterson
in iPS cells define sarcomere insufficiency as a cause of dilated cardiomy- A, Li D, Jakobs P, Litt M, Porter CB, Rahko PS, Hershberger RE. Lamin
opathy. Science. 2015;349:982–986. doi: 10.1126/science.aaa5458. A/C mutation analysis in a cohort of 324 unrelated patients with idio-
72. Golbus JR, Puckelwartz MJ, Fahrenbach JP, Dellefave-Castillo LM,
pathic or familial dilated cardiomyopathy. Am Heart J. 2008;156:161–
Wolfgeher D, McNally EM. Population-based variation in cardiomy- 169. doi: 10.1016/j.ahj.2008.01.026.
opathy genes. Circ Cardiovasc Genet. 2012;5:391–399. doi: 10.1161/ 89. Lu JT, Muchir A, Nagy PL, Worman HJ. LMNA cardiomyopathy:
CIRCGENETICS.112.962928. cell biology and genetics meet clinical medicine. Dis Model Mech.
73. Jansweijer JA, Nieuwhof K, Russo F, et al. Truncating titin mutations are 2011;4:562–568. doi: 10.1242/dmm.006346.
associated with a mild and treatable form of dilated cardiomyopathy. Eur 90. Young SG, Meta M, Yang SH, Fong LG. Prelamin A farnesylation
J Heart Fail. 2017;19:512–521. doi: 10.1002/ejhf.673. and progeroid syndromes. J Biol Chem. 2006;281:39741–39745. doi:
74. Schafer S, de Marvao A, Adami E, et al. Titin-truncating variants affect 10.1074/jbc.R600033200.
heart function in disease cohorts and the general population. Nat Genet. 91. Narula N, Favalli V, Tarantino P, et al. Quantitative expression of the
2017;49:46–53. mutated lamin A/C gene in patients with cardiolaminopathy. J Am Coll
75. van Spaendonck-Zwarts KY, Posafalvi A, van den Berg MP, Hilfiker- Cardiol. 2012;60:1916–1920. doi: 10.1016/j.jacc.2012.05.059.
Kleiner D, Bollen IA, Sliwa K, Alders M, Almomani R, van Langen 92. Dittmer TA, Sahni N, Kubben N, Hill DE, Vidal M, Burgess RC, Roukos
IM, van der Meer P, Sinke RJ, van der Velden J, Van Veldhuisen DJ, van V, Misteli T. Systematic identification of pathological lamin A interac-
Tintelen JP, Jongbloed JD. Titin gene mutations are common in families tors. Mol Biol Cell. 2014;25:1493–1510. doi: 10.1091/mbc.E14-02-0733.
with both peripartum cardiomyopathy and dilated cardiomyopathy. Eur 93. Nikolova V, Leimena C, McMahon AC, Tan JC, Chandar S, Jogia D,
Heart J. 2014;35:2165–2173. doi: 10.1093/eurheartj/ehu050. Kesteven SH, Michalicek J, Otway R, Verheyen F, Rainer S, Stewart CL,
76. Ware JS, Li J, Mazaika E, et al; IMAC-2 and IPAC Investigators. Shared Martin D, Feneley MP, Fatkin D. Defects in nuclear structure and func-
genetic predisposition in peripartum and dilated cardiomyopathies. N Engl tion promote dilated cardiomyopathy in lamin A/C-deficient mice. J Clin
J Med. 2016;374:233–241. doi: 10.1056/NEJMoa1505517. Invest. 2004;113:357–369. doi: 10.1172/JCI19448.
77. Felkin LE, Walsh R, Ware JS, Yacoub MH, Birks EJ, Barton PJ, Cook SA. 94. Lammerding J, Schulze PC, Takahashi T, Kozlov S, Sullivan T, Kamm
Recovery of cardiac function in cardiomyopathy caused by titin truncation. RD, Stewart CL, Lee RT. Lamin A/C deficiency causes defective nuclear
JAMA Cardiol. 2016;1:234–235. doi: 10.1001/jamacardio.2016.0208. mechanics and mechanotransduction. J Clin Invest. 2004;113:370–378.
78. Norton N, Li D, Rampersaud E, Morales A, Martin ER, Zuchner S, Guo doi: 10.1172/JCI19670.
S, Gonzalez M, Hedges DJ, Robertson PD, Krumm N, Nickerson DA, 95. Chandar S, Yeo LS, Leimena C, et al. Effects of mechanical stress and
Hershberger RE; National Heart, Lung, and Blood Institute GO Exome carvedilol in lamin A/C-deficient dilated cardiomyopathy. Circ Res.
Sequencing Project and the Exome Sequencing Project Family Studies 2010;106:573–582. doi: 10.1161/CIRCRESAHA.109.204388.
Project Team. Exome sequencing and genome-wide linkage analysis in 96. Ramos FJ, Chen SC, Garelick MG, Dai DF, Liao CY, Schreiber KH,
17 families illustrate the complex contribution of TTN truncating variants MacKay VL, An EH, Strong R, Ladiges WC, Rabinovitch PS, Kaeberlein
to dilated cardiomyopathy. Circ Cardiovasc Genet. 2013;6:144–153. doi: M, Kennedy BK. Rapamycin reverses elevated mTORC1 signaling in
10.1161/CIRCGENETICS.111.000062. lamin A/C-deficient mice, rescues cardiac and skeletal muscle function,
79. Gigli M, Begay RL, Morea G, Graw SL, Sinagra G, Taylor MR, Granzier and extends survival. Sci Transl Med. 2012;4:144ra103. doi: 10.1126/
H, Mestroni L. A review of the giant protein titin in clinical molecular scitranslmed.3003802.
diagnostics of cardiomyopathies. Front Cardiovasc Med. 2016;3:21. doi: 97. Choi JC, Muchir A, Wu W, Iwata S, Homma S, Morrow JP, Worman
10.3389/fcvm.2016.00021. HJ. Temsirolimus activates autophagy and ameliorates cardiomyopathy
Downloaded from http://ahajournals.org by on September 29, 2018

80. Akinrinade O, Ollila L, Vattulainen S, Tallila J, Gentile M, Salmenperä caused by lamin A/C gene mutation. Sci Transl Med. 2012;4:144ra102.
P, Koillinen H, Kaartinen M, Nieminen MS, Myllykangas S, Alastalo TP, doi: 10.1126/scitranslmed.3003875.
Koskenvuo JW, Heliö T. Genetics and genotype-phenotype correlations in 98. Wu W, Muchir A, Shan J, Bonne G, Worman HJ. Mitogen-activated
Finnish patients with dilated cardiomyopathy. Eur Heart J. 2015;36:2327– protein kinase inhibitors improve heart function and prevent fibrosis in
2337. doi: 10.1093/eurheartj/ehv253. cardiomyopathy caused by mutation in lamin A/C gene. Circulation.
81. Begay RL, Graw S, Sinagra G, Merlo M, Slavov D, Gowan K, Jones KL, 2011;123:53–61. doi: 10.1161/CIRCULATIONAHA.110.970673.
Barbati G, Spezzacatene A, Brun F, Di Lenarda A, Smith JE, Granzier HL, 99. MacRae C, Taylor MRG, Mestroni L, Moses, JR, Ashley EA, Wheeler
Mestroni L, Taylor M. Role of titin missense variants in dilated cardiomy- MT, Lakdawala NK, Hershberger RE, Ptaszynski M, Sandor V, Saunders
opathy. J Am Heart Assoc. 2015;4. doi: 10.1161/JAHA.115.002645. ME, Oliver C, Lee PA, Judge DP. Phase 2 study of a797, an oral, selec-
82. Ceyhan-Birsoy O, Agrawal PB, Hidalgo C, et al. Recessive truncating titin tive p38 mitogen-activated protein kinase inhibitor, in patients with lamin
gene, TTN, mutations presenting as centronuclear myopathy. Neurology. a/c-related dilated cardiomyopathy. Eur Heart J. 2016;37:1011.
2013;81:1205–1214. doi: 10.1212/WNL.0b013e3182a6ca62. 100. Hasselberg NE, Edvardsen T, Petri H, Berge KE, Leren TP, Bundgaard
83. Hackman P, Vihola A, Haravuori H, Marchand S, Sarparanta J, De Seze J, H, Haugaa KH. Risk prediction of ventricular arrhythmias and myo-
Labeit S, Witt C, Peltonen L, Richard I, Udd B. Tibial muscular dystrophy cardial function in Lamin A/C mutation positive subjects. Europace.
is a titinopathy caused by mutations in TTN, the gene encoding the gi- 2014;16:563–571. doi: 10.1093/europace/eut291.
ant skeletal-muscle protein titin. Am J Hum Genet. 2002;71:492–500. doi: 101. Haghighi K, Kolokathis F, Gramolini AO, Waggoner JR, Pater L, Lynch
10.1086/342380. RA, Fan GC, Tsiapras D, Parekh RR, Dorn GW 2nd, MacLennan DH,
84. Zou J, Tran D, Baalbaki M, et al. An internal promoter underlies the Kremastinos DT, Kranias EG. A mutation in the human phospholamban
difference in disease severity between N- and C-terminal truncation gene, deleting arginine 14, results in lethal, hereditary cardiomyopathy. Proc
mutations of Titin in zebrafish. Elife. 2015;4:e09406. doi: 10.7554/ Natl Acad Sci U S A. 2006;103:1388–1393. doi: 10.1073/pnas.0510519103.
eLife.09406. 102. Liu GS, Morales A, Vafiadaki E, Lam CK, Cai WF, Haghighi K, Adly G,
85. Granzier HL, Radke MH, Peng J, Westermann D, Nelson OL, Rost K, Hershberger RE, Kranias EG. A novel human R25C-phospholamban muta-
King NM, Yu Q, Tschöpe C, McNabb M, Larson DF, Labeit S, Gotthardt tion is associated with super-inhibition of calcium cycling and ventricular
M. Truncation of titin’s elastic PEVK region leads to cardiomyopathy arrhythmia. Cardiovasc Res. 2015;107:164–174. doi: 10.1093/cvr/cvv127.
with diastolic dysfunction. Circ Res. 2009;105:557–564. doi: 10.1161/ 103. van der Zwaag PA, van Rijsingen IA, Asimaki A, et al. Phospholamban
CIRCRESAHA.109.200964. R14del mutation in patients diagnosed with dilated cardiomyopathy or
86. Zhou Q, Kesteven S, Wu J, Aidery P, Gawaz M, Gramlich M, Feneley MP, arrhythmogenic right ventricular cardiomyopathy: evidence support-
Harvey RP. Pressure overload by transverse aortic constriction induces ing the concept of arrhythmogenic cardiomyopathy. Eur J Heart Fail.
maladaptive hypertrophy in a titin-truncated mouse model. Biomed Res 2012;14:1199–1207. doi: 10.1093/eurjhf/hfs119.
Int. 2015;2015:163564. doi: 10.1155/2015/163564. 104. DeWitt MM, MacLeod HM, Soliven B, McNally EM. Phospholamban
87. van Tintelen JP, Hofstra RM, Katerberg H, Rossenbacker T, Wiesfeld AC, R14 deletion results in late-onset, mild, hereditary dilated cardio-
du Marchie Sarvaas GJ, Wilde AA, van Langen IM, Nannenberg EA, van myopathy. J Am Coll Cardiol. 2006;48:1396–1398. doi: 10.1016/j.
der Kooi AJ, Kraak M, van Gelder IC, van Veldhuisen DJ, Vos Y, van den jacc.2006.07.016.
Berg MP; Working Group on Inherited Cardiac Disorders, Line 27/50, 105. Truszkowska GT, Bilińska ZT, Kosińska J, et al. A study in Polish pa-
Interuniversity Cardiology Institute of The Netherlands. High yield of tients with cardiomyopathy emphasizes pathogenicity of phospholamban
LMNA mutations in patients with dilated cardiomyopathy and/or con- (PLN) mutations at amino acid position 9 and low penetrance of hetero-
duction disease referred to cardiogenetics outpatient clinics. Am Heart J. zygous null PLN mutations. BMC Med Genet. 2015;16:21. doi: 10.1186/
2007;154:1130–1139. doi: 10.1016/j.ahj.2007.07.038. s12881-015-0167-0.
McNally and Mestroni   Dilated Cardiomyopathy Genetics and Mechanisms   747

106. Karakikes I, Stillitano F, Nonnenmacher M, et al. Correction of human remodeling of dilated cardiomyopathy in muscular dystrophy. Circulation.
phospholamban R14del mutation associated with cardiomyopathy using 2005;112:2799–2804. doi: 10.1161/CIRCULATIONAHA.104.528281.
targeted nucleases and combination therapy. Nat Commun. 2015;6:6955. 124. Ortiz-Lopez R, Li H, Su J, Goytia V, Towbin JA. Evidence for a dystro-
doi: 10.1038/ncomms7955. phin missense mutation as a cause of X-linked dilated cardiomyopathy.
107. Stillitano F, Turnbull IC, Karakikes I, Nonnenmacher M, Backeris P, Circulation. 1997;95:2434–2440.
Hulot JS, Kranias EG, Hajjar RJ, Costa KD. Genomic correction of fa- 125. Tsubata S, Bowles KR, Vatta M, Zintz C, Titus J, Muhonen L, Bowles
milial cardiomyopathy in human engineered cardiac tissues. Eur Heart NE, Towbin JA. Mutations in the human delta-sarcoglycan gene in famil-
J. 2016;37:3282–3284. doi: 10.1093/eurheartj/ehw307. ial and sporadic dilated cardiomyopathy. J Clin Invest. 2000;106:655–
108. Te Rijdt WP, van Tintelen JP, Vink A, van der Wal AC, de Boer RA, van 662. doi: 10.1172/JCI9224.
den Berg MP, Suurmeijer AJ. Phospholamban p.Arg14del cardiomyopa- 126. Haas M, Vlcek V, Balabanov P, Salmonson T, Bakchine S, Markey G,
thy is characterized by phospholamban aggregates, aggresomes, and au- Weise M, Schlosser-Weber G, Brohmann H, Yerro CP, Mendizabal MR,
tophagic degradation. Histopathology. 2016;69:542–550. doi: 10.1111/ Stoyanova-Beninska V, Hillege HL. European Medicines Agency review
his.12963. of ataluren for the treatment of ambulant patients aged 5 years and older
109. Brauch KM, Karst ML, Herron KJ, de Andrade M, Pellikka PA,
with Duchenne muscular dystrophy resulting from a nonsense muta-
Rodeheffer RJ, Michels VV, Olson TM. Mutations in ribonucleic acid tion in the dystrophin gene. Neuromuscul Disord. 2015;25:5–13. doi:
binding protein gene cause familial dilated cardiomyopathy. J Am Coll 10.1016/j.nmd.2014.11.011.
Cardiol. 2009;54:930–941. doi: 10.1016/j.jacc.2009.05.038. 127. Niks EH, Aartsma-Rus A. Exon skipping: a first in class strategy for
110. Li D, Morales A, Gonzalez-Quintana J, Norton N, Siegfried JD, Hofmeyer duchenne muscular dystrophy. Expert Opin Biol Ther. 2017;17:225–236.
M, Hershberger RE. Identification of novel mutations in RBM20 in pa- 128. Begay RL, Tharp CA, Martin A, et al. FLNC gene splice mutations cause
tients with dilated cardiomyopathy. Clin Transl Sci. 2010;3:90–97. doi: dilated cardiomyopathy. JACC Basic Transl Sci. 2016;1:344–359. doi:
10.1111/j.1752-8062.2010.00198.x. 10.1016/j.jacbts.2016.05.004.
111. Beqqali A, Bollen IA, Rasmussen TB, van den Hoogenhof MM, van 129. Ortiz-Genga MF, Cuenca S, Dal Ferro M, et al. Truncating FLNC muta-
Deutekom HW, Schafer S, Haas J, Meder B, Sørensen KE, van Oort RJ, tions are associated with high-risk dilated and arrhythmogenic cardio-
Mogensen J, Hubner N, Creemers EE, van der Velden J, Pinto YM. A myopathies. J Am Coll Cardiol. 2016;68:2440–2451. doi: 10.1016/j.
mutation in the glutamate-rich region of RNA-binding motif protein 20 jacc.2016.09.927.
causes dilated cardiomyopathy through missplicing of titin and impaired 130. Golbus JR, Puckelwartz MJ, Dellefave-Castillo L, Fahrenbach JP,

Frank-Starling mechanism. Cardiovasc Res. 2016;112:452–463. doi: Nelakuditi V, Pesce LL, Pytel P, McNally EM. Targeted analy-
10.1093/cvr/cvw192. sis of whole genome sequence data to diagnose genetic cardiomy-
112. Guo W, Schafer S, Greaser ML, et al. RBM20, a gene for hereditary car- opathy. Circ Cardiovasc Genet. 2014;7:751–759. doi: 10.1161/
diomyopathy, regulates titin splicing. Nat Med. 2012;18:766–773. doi: CIRCGENETICS.113.000578.
10.1038/nm.2693. 131. Thompson TG, Chan YM, Hack AA, Brosius M, Rajala M, Lidov HG,
113. Li S, Guo W, Dewey CN, Greaser ML. Rbm20 regulates titin alternative McNally EM, Watkins S, Kunkel LM. Filamin 2 (FLN2): a muscle-spe-
splicing as a splicing repressor. Nucleic Acids Res. 2013;41:2659–2672. cific sarcoglycan interacting protein. J Cell Biol. 2000;148:115–126.
doi: 10.1093/nar/gks1362. 132. Dalkilic I, Schienda J, Thompson TG, Kunkel LM. Loss of filamin C
114. Maatz H, Jens M, Liss M, et al. RNA-binding protein RBM20 repress- (FLNc) results in severe defects in myogenesis and myotube structure.
es splicing to orchestrate cardiac pre-mRNA processing. J Clin Invest. Mol Cell Biol. 2006;26:6522–6534. doi: 10.1128/MCB.00243-06.
2014;124:3419–3430. doi: 10.1172/JCI74523. 133. Arbustini E, Diegoli M, Fasani R, Grasso M, Morbini P, Banchieri N,
Downloaded from http://ahajournals.org by on September 29, 2018

115. Wyles SP, Li X, Hrstka SC, Reyes S, Oommen S, Beraldi R, Edwards Bellini O, Dal Bello B, Pilotto A, Magrini G, Campana C, Fortina P,
J, Terzic A, Olson TM, Nelson TJ. Modeling structural and functional Gavazzi A, Narula J, Viganò M. Mitochondrial DNA mutations and
deficiencies of RBM20 familial dilated cardiomyopathy using human mitochondrial abnormalities in dilated cardiomyopathy. Am J Pathol.
induced pluripotent stem cells. Hum Mol Genet. 2016;25:254–265. doi: 1998;153:1501–1510. doi: 10.1016/S0002-9440(10)65738-0.
10.1093/hmg/ddv468. 134. Ashrafian H, Docherty L, Leo V, et al. A mutation in the mitochon-
116. Khan MA, Reckman YJ, Aufiero S, van den Hoogenhof MM, van der drial fission gene Dnm1l leads to cardiomyopathy. PLoS Genet.
Made I, Beqqali A, Koolbergen DR, Rasmussen TB, van der Velden 2010;6:e1001000. doi: 10.1371/journal.pgen.1001000.
J, Creemers EE, Pinto YM. RBM20 regulates circular RNA produc- 135. Grasso M, Diegoli M, Brega A, Campana C, Tavazzi L, Arbustini E.
tion from the titin gene. Circ Res. 2016;119:996–1003. doi: 10.1161/ The mitochondrial DNA mutation T12297C affects a highly con-
CIRCRESAHA.116.309568. served nucleotide of tRNA(Leu(CUN)) and is associated with dilated
117. Nair K, Pekhletski R, Harris L, Care M, Morel C, Farid T, Backx PH, cardiomyopathy. Eur J Hum Genet. 2001;9:311–315. doi: 10.1038/
Szabo E, Nanthakumar K. Escape capture bigeminy: phenotypic mark- sj.ejhg.5200622.
er of cardiac sodium channel voltage sensor mutation R222Q. Heart 136. Sibbing D, Pfeufer A, Perisic T, et al. Mutations in the mitochondrial
Rhythm. 2012;9:1681–1688.e1. doi: 10.1016/j.hrthm.2012.06.029. thioredoxin reductase gene TXNRD2 cause dilated cardiomyopathy. Eur
118. Davis RP, Casini S, van den Berg CW, Hoekstra M, Remme CA,
Heart J. 2011;32:1121–1133. doi: 10.1093/eurheartj/ehq507.
Dambrot C, Salvatori D, Oostwaard DW, Wilde AA, Bezzina CR, 137. Rowland TJ, Graw SL, Sweet ME, Gigli M, Taylor MR, Mestroni L.
Verkerk AO, Freund C, Mummery CL. Cardiomyocytes derived Obscurin variants in patients with left ventricular noncompaction. J Am
from pluripotent stem cells recapitulate electrophysiological charac- Coll Cardiol. 2016;68:2237–2238.
teristics of an overlap syndrome of cardiac sodium channel disease. 138. Marian AJ. The case of “Missing Causal Genes” and the practice of
Circulation. 2012;125:3079–3091. doi: 10.1161/CIRCULATIONAHA. medicine: a Sherlock Holmes approach of deductive reasoning. Circ Res.
111.066092. 2016;119:21–24. doi: 10.1161/CIRCRESAHA.116.308830.
119. Liang P, Sallam K, Wu H, et al. Patient-specific and genome-edited in- 139. Roncarati R, Viviani Anselmi C, Krawitz P, Lattanzi G, von Kodolitsch
duced pluripotent stem cell-derived cardiomyocytes elucidate single-cell Y, Perrot A, di Pasquale E, Papa L, Portararo P, Columbaro M, Forni A,
phenotype of brugada syndrome. J Am Coll Cardiol. 2016;68:2086– Faggian G, Condorelli G, Robinson PN. Doubly heterozygous LMNA
2096. doi: 10.1016/j.jacc.2016.07.779. and TTN mutations revealed by exome sequencing in a severe form of
120. Malan D, Friedrichs S, Fleischmann BK, Sasse P. Cardiomyocytes
dilated cardiomyopathy. Eur J Hum Genet. 2013;21:1105–1111. doi:
obtained from induced pluripotent stem cells with long-QT syndrome 10.1038/ejhg.2013.16.
3 recapitulate typical disease-specific features in vitro. Circ Res. 140. Wessels MW, Herkert JC, Frohn-Mulder IM, Dalinghaus M, van den
2011;109:841–847. doi: 10.1161/CIRCRESAHA.111.243139. Wijngaard A, de Krijger RR, Michels M, de Coo IF, Hoedemaekers
121. Wan E, Abrams J, Weinberg RL, Katchman AN, Bayne J, Zakharov YM, Dooijes D. Compound heterozygous or homozygous truncating
SI, Yang L, Morrow JP, Garan H, Marx SO. Aberrant sodium influx MYBPC3 mutations cause lethal cardiomyopathy with features of non-
causes cardiomyopathy and atrial fibrillation in mice. J Clin Invest. compaction and septal defects. Eur J Hum Genet. 2015;23:922–928. doi:
2016;126:112–122. doi: 10.1172/JCI84669. 10.1038/ejhg.2014.211.
122. Feng J, Yan J, Buzin CH, Towbin JA, Sommer SS. Mutations in the dys- 141. Zahka K, Kalidas K, Simpson MA, Cross H, Keller BB, Galambos C,
trophin gene are associated with sporadic dilated cardiomyopathy. Mol Gurtz K, Patton MA, Crosby AH. Homozygous mutation of MYBPC3
Genet Metab. 2002;77:119–126. associated with severe infantile hypertrophic cardiomyopathy at high
123. Jefferies JL, Eidem BW, Belmont JW, Craigen WJ, Ware SM,
frequency among the Amish. Heart. 2008;94:1326–1330. doi: 10.1136/
Fernbach SD, Neish SR, Smith EO, Towbin JA. Genetic predictors and hrt.2007.127241.
748  Circulation Research  September 15, 2017

142. Dellefave LM, Pytel P, Mewborn S, Mora B, Guris DL, Fedson S, why is it important? Nat Rev Cardiol. 2016;13:623–628. doi: 10.1038/
Waggoner D, Moskowitz I, McNally EM. Sarcomere mutations in car- nrcardio.2016.134.
diomyopathy with left ventricular hypertrabeculation. Circ Cardiovasc 154. Ruschitzka F, Abraham WT, Singh JP, Bax JJ, Borer JS, Brugada

Genet. 2009;2:442–449. doi: 10.1161/CIRCGENETICS.109.861955. J, Dickstein K, Ford I, Gorcsan J 3rd, Gras D, Krum H, Sogaard P,
143. Pinto YM, Elliott PM, Arbustini E, et al. Proposal for a revised defini- Holzmeister J; EchoCRT Study Group. Cardiac-resynchronization
tion of dilated cardiomyopathy, hypokinetic non-dilated cardiomyopa- therapy in heart failure with a narrow QRS complex. N Engl J Med.
thy, and its implications for clinical practice: a position statement of the 2013;369:1395–1405. doi: 10.1056/NEJMoa1306687.
ESC working group on myocardial and pericardial diseases. Eur Heart J. 155. Yancy CW, Jessup M, Bozkurt B, et al. 2016 ACC/AHA/HFSA focused
2016;37:1850–1858. doi: 10.1093/eurheartj/ehv727. update on new pharmacological therapy for heart failure: an update of
144. van Spaendonck-Zwarts KY, van Rijsingen IA, van den Berg MP,
the 2013 ACCF/AHA guideline for the management of heart failure: a re-
Lekanne Deprez RH, Post JG, van Mil AM, Asselbergs FW, Christiaans port of the American College of Cardiology/American Heart Association
I, van Langen IM, Wilde AA, de Boer RA, Jongbloed JD, Pinto YM, Task Force on Clinical Practice Guidelines and the Heart Failure Society
van Tintelen JP. Genetic analysis in 418 index patients with idiopathic of America. J Am Coll Cardiol. 2016;68:1476–1488. doi: 10.1016/j.
dilated cardiomyopathy: overview of 10 years’ experience. Eur J Heart jacc.2016.05.011.
Fail. 2013;15:628–636. doi: 10.1093/eurjhf/hft013. 156. Losurdo P, Stolfo D, Merlo M, Barbati G, Gobbo M, Gigli M, Ramani R,
145. Garcia J, Tahiliani J, Johnson NM, Aguilar S, Beltran D, Daly A, Decker Pinamonti B, Zecchin M, Finocchiaro G, Mestroni L, Sinagra G. Early
E, Haverfield E, Herrera B, Murillo L, Nykamp K, Topper S. Clinical arrhythmic events in idiopathic dilated cardiomyopathy. JACC Clin EP.
genetic testing for the cardiomyopathies and arrhythmias: a systematic 2016;2:535–543.
framework for establishing clinical validity and addressing genotypic 157. Priori SG, Blomström-Lundqvist C, Mazzanti A, et al. 2015 ESC

and phenotypic heterogeneity. Front Cardiovasc Med. 2016;3:20. doi: Guidelines for the management of patients with ventricular arrhyth-
10.3389/fcvm.2016.00020. mias and the prevention of sudden cardiac death: the task force for the
146. Harakalova M, Kummeling G, Sammani A, Linschoten M, Baas AF, management of patients with ventricular arrhythmias and the prevention
van der Smagt J, Doevendans PA, van Tintelen JP, Dooijes D, Mokry M, of sudden cardiac death of the European Society of Cardiology (ESC).
Asselbergs FW. A systematic analysis of genetic dilated cardiomyopathy Endorsed by: Association for European Paediatric and Congenital
reveals numerous ubiquitously expressed and muscle-specific genes. Eur Cardiology (AEPC). Eur Heart J. 2015;36:2793–2867. doi: 10.1093/
J Heart Fail. 2015;17:484–493. doi: 10.1002/ejhf.255. eurheartj/ehv316.
147. Fatkin D, Yeoh T, Hayward CS, Benson V, Sheu A, Richmond Z,
158. Kusumoto FM, Calkins H, Boehmer J, et al. HRS/ACC/AHA ex-

Feneley MP, Keogh AM, Macdonald PS. Evaluation of left ventricu- pert consensus statement on the use of implantable cardioverter-
lar enlargement as a marker of early disease in familial dilated car- defibrillator therapy in patients who are not included or not well
diomyopathy. Circ Cardiovasc Genet. 2011;4:342–348. doi: 10.1161/ represented in clinical trials. Circulation. 2014;130:94–125. doi:
CIRCGENETICS.110.958918. 10.1161/CIR.0000000000000056.
148. Moretti M, Merlo M, Barbati G, Di Lenarda A, Brun F, Pinamonti B, 159. Kumar S, Baldinger SH, Gandjbakhch E, et al. Long-term arrhythmic
Gregori D, Mestroni L, Sinagra G. Prognostic impact of familial screen- and nonarrhythmic outcomes of lamin A/C mutation carriers. J Am Coll
ing in dilated cardiomyopathy. Eur J Heart Fail. 2010;12:922–927. doi: Cardiol. 2016;68:2299–2307. doi: 10.1016/j.jacc.2016.08.058.
10.1093/eurjhf/hfq093. 160. Saberi S, Day SM. Exercise prescription for the athlete with cardiomy-
149. Yancy CW, Jessup M, Bozkurt B, et al; American College of Cardiology opathy. Cardiol Clin. 2016;34:591–601. doi: 10.1016/j.ccl.2016.06.008.
Foundation; American Heart Association Task Force on Practice 161. Rosen MR, Myerburg RJ, Francis DP, Cole GD, Marbán E. Translating
Downloaded from http://ahajournals.org by on September 29, 2018

Guidelines. 2013 ACCF/AHA guideline for the management of heart stem cell research to cardiac disease therapies: pitfalls and prospects
failure: a report of the American College of Cardiology Foundation/ for improvement. J Am Coll Cardiol. 2014;64:922–937. doi: 10.1016/j.
American Heart Association Task Force on Practice Guidelines. J Am jacc.2014.06.1175.
Coll Cardiol. 2013;62:e147–e239. doi: 10.1016/j.jacc.2013.05.019. 162. Eulalio A, Mano M, Dal Ferro M, Zentilin L, Sinagra G, Zacchigna S,
150. Ponikowski P, Voors AA, Anker SD, et al; Authors/Task Force Members. Giacca M. Functional screening identifies miRNAs inducing cardiac re-
2016 ESC Guidelines for the diagnosis and treatment of acute and chronic generation. Nature. 2012;492:376–381. doi: 10.1038/nature11739.
heart failure: the task force for the diagnosis and treatment of acute and 163. Mann DL, Lee RJ, Coats AJ, Neagoe G, Dragomir D, Pusineri E,
chronic heart failure of the European Society of Cardiology (ESC) devel- Piredda M, Bettari L, Kirwan BA, Dowling R, Volterrani M, Solomon
oped with the special contribution of the Heart Failure Association (HFA) of SD, Sabbah HN, Hinson A, Anker SD. One-year follow-up results from
the ESC. Eur Heart J. 2016;37:2129–2200. doi: 10.1093/eurheartj/ehw128. AUGMENT-HF: a multicentre randomized controlled clinical trial of
151. Zannad F, Gattis Stough W, Rossignol P, et al. Mineralocorticoid receptor the efficacy of left ventricular augmentation with Algisyl in the treat-
antagonists for heart failure with reduced ejection fraction: integrating ment of heart failure. Eur J Heart Fail. 2016;18:314–325. doi: 10.1002/
evidence into clinical practice. Eur Heart J. 2012;33:2782–2795. doi: ejhf.449.
10.1093/eurheartj/ehs257. 164. Feric NT, Radisic M. Strategies and challenges to myocardial replace-
152. Mestroni L, Brun F, Spezzacatene A, Sinagra G, Taylor MR. Genetic ment therapy. Stem Cells Transl Med. 2016;5:410–416. doi: 10.5966/
causes of dilated cardiomyopathy. Prog Pediatr Cardiol. 2014;37:13–18. sctm.2015-0288.
doi: 10.1016/j.ppedcard.2014.10.003. 165. Sun N, Yazawa M, Liu J, et al. Patient-specific induced pluripotent stem
153. Jessup M, Marwick TH, Ponikowski P, Voors AA, Yancy CW. 2016 ESC cells as a model for familial dilated cardiomyopathy. Sci Transl Med.
and ACC/AHA/HFSA heart failure guideline update - what is new and 2012;4:130ra47. doi: 10.1126/scitranslmed.3003552.

You might also like