22
Compound and Digenic Heterozygosity Contributes to Arrhythmogenic Right Ventricular Cardiomyopathy Tianhong Xu, PhD *,1 , Zhao Yang, MD, PhD *,2,3 , Matteo Vatta, PhD 3 , Alessandra Rampazzo, MD, PhD 4 , Giorgia Beffagna, PhD 3,4 , Kalliopi Pillichou, PhD 4 , Steven E. Scherer, PhD 1 , Jeffrey Saffitz, MD, PhD 5 , Joshua Kravitz 3 , Wojciech Zareba, MD 6 , Gian Antonio Danieli, PhD 4 , Alessandra Lorenzon, PhD 4 , Andrea Nava, MD 8 , Barbara Bauce, MD, PhD 8 , Gaetano Thiene, MD 9 , Cristina Basso, MD, PhD 9 , Hugh Calkins, MD 7 , Kathy Gear, RN 10 , Frank Marcus, MD 10 , and Jeffrey A. Towbin, MD 11 for the Multidisciplinary Study of Right Ventricular Dysplasia Investigators 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 2 Department of Medicine (Cardiovascular Sciences), Baylor College of Medicine, Houston, TX 3 Department of Pediatrics (Section of Cardiology), Baylor College of Medicine, Houston, TX 4 Department of Biology, University of Padua Medical School, Padua, Italy 5 Department of Pathology, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA 6 Department of Medicine, University of Rochester Medical Center, Rochester, NY 7 Department of Cardiology, Johns Hopkins School of Medicine and ARVD Program, Baltimore, MD 8 Department of Cardiothoracic-Vascular Sciences, University of Padua Medical School, Padua, Italy 9 Department of Medico-Diagnostic Sciences, University of Padua Medical School, Padua, Italy 10 Department of Medicine, University of Arizona, Tucson, AZ 11 The Heart Institute and Department of Pediatrics (Pediatric Cardiology), Cincinnati Children's Hospital Medical Center, Cincinnati, OH. Abstract Objective: To define the genetic basis of arrhythmogenic right ventricular cardiomyopathy. Background: Arrhythmogenic right ventricular cardiomyopathy (ARVC), characterized by right ventricular fibrofatty replacement and arrhythmias, causes sudden death. Autosomal dominant Inheritance, reduced penetrance, and 7 desmosome-encoding causative genes are known. The basis of low penetrance is poorly understood. © 2009 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved. Address for Reprints: Jeffrey A. Towbin, M.D. Executive Co-Director, The Heart Institute Professor and Chief, Pediatric Cardiology Cincinnati Children's Hospital Medical Center 333 Burnet Avenue Cincinnati, Ohio 45229 Phone: (513) 636-3049 Fax: (513) 636-4181 [email protected]. * These authors contributed equally Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. No conflicts. NIH Public Access Author Manuscript J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9. Published in final edited form as: J Am Coll Cardiol. 2010 February 9; 55(6): 587–597. doi:10.1016/j.jacc.2009.11.020. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Compound and Digenic Heterozygosity Contributes to Arrhythmogenic Right Ventricular Cardiomyopathy

Embed Size (px)

Citation preview

Compound and Digenic Heterozygosity Contributes toArrhythmogenic Right Ventricular Cardiomyopathy

Tianhong Xu, PhD*,1, Zhao Yang, MD, PhD*,2,3, Matteo Vatta, PhD3, Alessandra Rampazzo,MD, PhD4, Giorgia Beffagna, PhD3,4, Kalliopi Pillichou, PhD4, Steven E. Scherer, PhD1,Jeffrey Saffitz, MD, PhD5, Joshua Kravitz3, Wojciech Zareba, MD6, Gian Antonio Danieli,PhD4, Alessandra Lorenzon, PhD4, Andrea Nava, MD8, Barbara Bauce, MD, PhD8, GaetanoThiene, MD9, Cristina Basso, MD, PhD9, Hugh Calkins, MD7, Kathy Gear, RN10, FrankMarcus, MD10, and Jeffrey A. Towbin, MD11 for the Multidisciplinary Study of RightVentricular Dysplasia Investigators1Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX2Department of Medicine (Cardiovascular Sciences), Baylor College of Medicine, Houston, TX3Department of Pediatrics (Section of Cardiology), Baylor College of Medicine, Houston, TX4Department of Biology, University of Padua Medical School, Padua, Italy5Department of Pathology, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA6Department of Medicine, University of Rochester Medical Center, Rochester, NY7Department of Cardiology, Johns Hopkins School of Medicine and ARVD Program, Baltimore, MD8Department of Cardiothoracic-Vascular Sciences, University of Padua Medical School, Padua, Italy9Department of Medico-Diagnostic Sciences, University of Padua Medical School, Padua, Italy10Department of Medicine, University of Arizona, Tucson, AZ11The Heart Institute and Department of Pediatrics (Pediatric Cardiology), Cincinnati Children'sHospital Medical Center, Cincinnati, OH.

AbstractObjective: To define the genetic basis of arrhythmogenic right ventricular cardiomyopathy.

Background: Arrhythmogenic right ventricular cardiomyopathy (ARVC), characterized by rightventricular fibrofatty replacement and arrhythmias, causes sudden death. Autosomal dominantInheritance, reduced penetrance, and 7 desmosome-encoding causative genes are known. The basisof low penetrance is poorly understood.

© 2009 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.Address for Reprints: Jeffrey A. Towbin, M.D. Executive Co-Director, The Heart Institute Professor and Chief, Pediatric CardiologyCincinnati Children's Hospital Medical Center 333 Burnet Avenue Cincinnati, Ohio 45229 Phone: (513) 636-3049 Fax: (513) [email protected].*These authors contributed equallyPublisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customerswe are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resultingproof before it is published in its final citable form. Please note that during the production process errors may be discovered which couldaffect the content, and all legal disclaimers that apply to the journal pertain.No conflicts.

NIH Public AccessAuthor ManuscriptJ Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

Published in final edited form as:J Am Coll Cardiol. 2010 February 9; 55(6): 587–597. doi:10.1016/j.jacc.2009.11.020.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Methods: ARVC probands and family members were enrolled, blood obtained, lymphoblastoidcell lines immortalized, DNA extracted, PCR amplification of desmosome-encoding genesperformed, PCR products sequenced and diseased tissue samples studied for intercellular junctionprotein distribution using confocal immunofluorescence microscopy and antibodies against keyproteins.

Results: We identified 21 variants in plakophilin-2 (PKP2) in 38 of 198 probands (19%), includingmissense, nonsense, splice site, and deletion/insertion mutations. Pedigrees showed wide intra-familial variability (severe early-onset disease to asymptomatic individuals). In 9/38 probands,PKP2 variants were identified that were encoded in trans (compound heterozygosity). The 38probands hosting PKP2 variants were screened for other desmosomal genes mutations; secondvariants (digenic heterozygosity) were identified in 16/38 subjects with PKP2 variants (42%)including desmoplakin (DSP, n=6), desmoglein-2 (DSG2, n=5), plakophilin-4 (PKP4, n=1), anddesmocollin-2 (DSC2, n=1). Heterozygous mutations in non-PKP 2desmosomal genes occurred in14/198 subjects (7%), including DSP (n=4), DSG2 (n=5), DSC2 (n=3), and junctional plakoglobin(JUP, n=2). All variants occurred in conserved regions; none were identified in 700 ethnic-matchedcontrols.

Immunohistochemical analysis demonstrated abnormalities of protein architecture.

Conclusions: These data suggest that the genetic basis of ARVC includes reduced penetrance withcompound and digenic heterozygosity. Disturbed junctional cytoarchitecture in subjects withdesmosomal mutations confirms that ARVC is a disease of the desmosome and cell junction.

KeywordsArrhythmias; Cardiomyopathies; Desmosomes; Intercalated Disks; Genetic Mutations

INTRODUCTIONArrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC) has been defined as aprimary right ventricular cardiomyopathy initially described by Marcus, Fontaine andcolleagues in the early 1980's.1 It is characterized by fibrofatty infiltration of the rightventricular (RV) myocardium and clinically presents with ventricular arrhythmias, heartfailure, syncope, and sudden death.1,2 The left ventricle (LV) may also be affected.3 In familialcases of ARVC, autosomal dominant inheritance with reduced penetrance has been reportedand is believed to account for approximately 30% of cases.4 In the remaining sporadic cases,the etiology may be an acquired cause such as myocarditis5,6 or an unidentified inheriteddisorder. To date, multiple genetic loci and 7 genes including desmoplakin (DSP),7plakophilin-2 (PKP2),8 desmoglein-2 (DSG2),9 desmocollin-2 (DSC2), 10 transforminggrowth factor β3 (TGF β3),11 ryanodine receptor 2 (RYR2)12, and TransmenbraneProtein-43 (TMEM43) 13 have been identified in ARVC patients. In addition, two complexcardiocutaneous disorders with autosomal recessive inheritance in which cardiomyopathy isassociated with woolly hair and palmoplantar keratoderma have been reported. These includeNaxos disease with ARVC14 and Carvajal syndrome associated with an LV cardiomyopathy.15 The genes identified for these disorders include homozygous mutations in junctionalplakoglobin (JUP) in Naxos disease,16 as well as homozygous mutations in DSP in Carvajalsyndrome.17 The most common gene variants identified in ARVC is PKP2, initially reportedby Gerull and colleagues to be mutated in ~25% of patients with autosomal dominant inheriteddisease.8 In the analysis of PKP2, an essential armadillo-repeat protein of the cardiacdesmosome, they identified heterozygous mutations in 32 of 120 unrelated individuals withARVC. 8 Other investigations have confirmed these findings.18-21 The majority of genesidentified to date causing ARVC encode desmosomal proteins. Within cardiomyocytes, twotypes of cell adhesion junctions are responsible for intercellular adhesion: the desmosomes and

Xu et al. Page 2

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

fascia adherens junctions.22 These cell adhesion junctions are both located at thecardiomyocyte intercalated disk and both contain intracellular proteins that link thecytoplasmic domains of cadherins to components of the cytoskeleton. In cardiomyocytes, avariety of proteins interact to form functional cell-cell junctions. DSGs and DSCs are connectedto the desmin cytoskeleton by DSP, JUP (γ-catenin), and PKP2. These latter proteins, JUP andPKP2, are members of the armadillo family of nuclear and junctional proteins.23, 24 PKP-2interacts with DSP, DSG and intermediate filament proteins at sites within its N-terminus. DSPand PKP2 are located only in desmosomes, whereas JUP participates as a linker in bothdesmosomes and adherens junctions. The adherens junctions are located at the ends ofsarcomeres and are linked to sarcomeric actin through intracellular linker proteins, mostnotably members of the catenin family including JUP, β-catenin, αcatenin and p120 catenin.

In this study, we analyzed probands and family members for ARVC using a standardizedclinical protocol either developed as part of the North American ARVD Registry25 or usingthe standard Task Force criteria (Table 1).26 All individuals were screened for mutations in allof genes encoding proteins involved in desmosomal function, even if a variant were alreadyidentified in any of these desmosome-encoding genes. We report identification of multiplemutations in these genes, including autosomal dominant heterozygous mutations in 26% ofsubjects (52/198), including 38 in PKP2 and 14 in other desmosome-encoding genes.

Additionally, compound heterozygous mutations and digenic mutations were identified in 42%of the subjects (16/38) in whom PKP2 mutations were identified. In addition, we demonstratethat many PKP2 mutations have low penetrance and in many cases may not be the primarycause of the disease, contradicting the previously reported contention that PKP2 is the majorARVC causing gene, accounting for the cause of disease in 25% of ARVC patients.

METHODSPatient Evaluation

After informed consent, probands were evaluated by noninvasive and invasive studiesincluding physical examination and history/family history, chest radiography, 12-leadelectrocardiogram (ECG), echocardiography and cardiac magnetic resonance imaging (cMRI).

In most cases, the clinical evaluation followed the protocol of the NIH-funded North AmericanARVD Registry,25 which included invasive studies including cardiac catheterization,ventricular angiography, and endomyocardial biopsy. In these subjects, all studies (noninvasiveand invasive testing) were analyzed by Core Laboratories. Family members were evaluatedusing the noninvasive studies only (ECG, cMRI, echocardiogram, chest X-ray, and physicalexamination with history/family history). In the subjects in whom genetic studies wereperformed but who declined enrollment in the Registry, or international subjects not eligibleto enroll in the Registry, the Task Force diagnostic criteria (Table 1) were used. 26 Diagnosticcriteria previously described by McKenna et al. were used to determine affectation status.26

After informed consent, blood for DNA extraction and lymphoblastoid cell lineimmortalization was obtained, as approved by the Baylor College of Medicine InstitutionalReview Board (IRB).

DNA Sequencing AnalysisGenomic DNA samples of the 143 U.S. and 55 Italian ARVC index cases (n=198) wereobtained from blood samples and immortalized lymphoblastoid cell lines as previouslydescribed27 and amplified by PCR using primers designed to amplify the coding exons ofdesmosome-encoding genes PKP2, DSP, JUP, DSC2, DSG2, plakophilin-4 (PKP4), and theintermediate filament-encoding gene desmin (DES). Other non-desmosomal genes were

Xu et al. Page 3

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

excluded from this analysis. PCR products from the U.S. cohort were sequenced using BigDye Terminator version 3.1 chemistry (Applied Biosystems; ABI) and analyzed using an ABI3730 DNA sequencer (Applied Biosystems; ABI). In addition, the 55 Italian ARVC indexcases were screened for mutations by denaturing high-performance liquid chromatography(DHPLC) and direct sequencing. DHPLC analysis was performed with the use of WAVENucleic Acid Fragment Analysis System 3500HT with DNASep HT cartridge (TransgenomicLtd NE). Temperatures for sample analysis were selected with the use of WAVEMAKERsoftware. In all 198 subjects, therefore, we analyzed the entire coding sequence and thesurrounding intronic sequences of DES and of the desmosomal genes including PKP2, DSP,DSC2, DSG2, JUP, DSC2, DSG2 and PKP4.

Cloning and Sequencing of PKP2 cDNATotal RNA was isolated from lymphoblastoid cell lines using an RNeasy Mini Kit (Qiagen,Stanford, CA) and subjected to random hexamer-primed cDNA synthesis using Superscript II(Invitrogen). PKP2 cDNA was amplified by PCR with oligonucleotides specific for the cDNAsequence of PKP2 (5′-CCAGCTGAGTACGGCTACATC-3′; 5′-TCAGTCTTTAAGGGAGTGGT-3′), cloned into TA-vectors (Topo TA Cloning Kit;Invitrogen, Carlsbad, CA) and then introduced into TOP10 cells using a One Shot ChemicalTransformation kit (Invitrogen). For each patient sample, plasmid DNA was isolated and theinsert was sequenced using the same oligonucleotides.

ImmunohistochemistryWhen available, formalin-fixed or snap-frozen cardiac tissues from affected patients werestudied for the distribution of intercellular junction proteins using confocalimmunofluorescence microscopy, as previously described28,29 Antigens were exposed inparaffin-embedded sections using microwave antigen-recovery techniques28,29 and thenstained with commercial rabbit polyclonal antibodies against connexin 43 (Cx43 Invitrogen(Zymed), the C-terminal domain of DSP (Serotec, Raleigh, NC), a conserved sequence in theN-cadherins (Sigma-Aldrich, St. Louis MO), and DES (ScyTek Laboratories, Logan, UT), aswell as mouse monoclonal antibodies against JUP (Sigma-Aldrich), DSC 2/3 (Invitrogen(Zymed)), PKP-2 (Biodesign International, Saco, ME), α-catenin (Invitrogen (Zymed)), β-catenin (Invitrogen (Zymed)), and the C-terminus of JUP (Research Diagnostics Inc., Concord,MA). The sections were then stained with appropriate secondary antibodies conjugated withCY3 and visualized by confocal microscopy as described previously.28,29 Patient samples werestained simultaneously with myocardial sections prepared from three age-matched controlswith no clinical or autopsy evidence of heart disease. The amount of immunoreactive signalfor each protein at the intercalated disks was evaluated in a blinded fashion and scored as beingstrongly present, weakly present or absent.

RESULTSGENETIC ANALYSIS: PLAKOPHILIN-2 MUTATIONS AND COMPOUND HETEROZYOSITY

After informed consent, 198 probands (143 North American, 55 Italian) meeting clinicalcriteria for the diagnosis of ARVC using the Task Force criteria (Table 1) and/or ARVDRegistry criteria were enrolled in the genetic analysis study. All subjects were screened formutations in all desmosome-encoding genes, with complete sequencing of the entire geneperformed in all cases. 21 variants in PKP2 were identified in 38 of the 198 probands (19%),a frequency similar to previous reports.8, 18-21 The variants identified included 8 missense, 4nonsense, 2 splice site mutations, and 8 deletion/insertion mutations; the deletion/insertionvariants each predict a protein frame shift (Table 2). Several variants were identified in multipleprobands, including nonsense and frame-shift variants (Table 2). All of the missensesubstitutions resulted in changes at residues that are conserved between species (Supplemental

Xu et al. Page 4

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 1). Among the variants identified, the splice site substitution detected in intron 10, andthe 2509delA in exon 13 have been previously reported.8 The remaining variants are novel.Despite the fact that several of these variants were identified in multiple individuals, analysisof 700 ethnically matched control individuals (1,400 chromosomes) identified only one of thesevariants (A372P) in the control population (1 in 700).

Analysis of the family pedigrees shows that there is significantly reduced penetrance of thesePKP2 variants, similar to that reported by Gerull et al. (Figure 1A and B).8 However, in 9 ofthe probands with PKP2 variants, we identified 2 distinct PKP2 variants (Table 3, patients 5,7, 8, 11, 12, 14, 15, 16 and 17), consistent with compound heterozygosity. One of theseprobands (patient 5, Table 3; Figure 1C) was a member of a family with a history of ARVC inhis generation (Figure 1C). DNA was available from the living affected subject (patient 6,Table 3; Figure 1C) and unaffected siblings of the proband, as well as the phenotypically normalparents, paternal uncle and paternal grandparents. All family members were clinicallyevaluated using the Task Force criteria (Table 1). Only the clinically affected brother carriedboth variants (Figure 1C). The parents were carriers of individual PKP2 variants and theclinically unaffected (not meeting Task Force criteria) sibling hosted only the varianttransmitted by his father and paternal grandfather (Figure 1C). RT-PCR of PKP2 mRNA wasperformed using samples obtained from five other affected probands and sequencing revealedthat in each case the variants were encoded in trans. Thus, these variants were either inheritedindependently from the phenotypically normal patients or were de novo. As noted in Table 3,disparities between the clinical phenotypes and genetic findings appear somewhat common.Notably, all subjects had RV involvement, several had biventricular disease, and all hadassociated arrhythmias. Importantly, not all subjects met full Task Force Criteria, therebypointing to imperfections in these criteria and necessitating modifications of these criteria(Marcus et al, Circulation, in press) 30.

These findings led us to consider the possibility that ARVC may be due to another form of“compound heterozygosity” with mutations/genetic variants in two desmosomal proteinencoding genes required for clinical disease, called “digenic heterozygosity”.

GENETIC ANALYSIS: DIGENIC HETEROZYOSITYIn all 198 probands, the desmosome-encoding genes DSP, DSC2, DSG2, JUP, DSC2,DSG2,PKP4, and DES were sequenced in addition to PKP2 sequencing. This sequencing identified13 variants in second desmosomal genes in 13 subjects with PKP2 variants, including DSP in6 cases, DSG2 in 5 cases, PKP4 in 1 subject, and DSC2 in 1 subject (Table 3). None of thesevariants were identified in at least 700 ethnic-matched controls (>1,400 chromosomes). Thus,of the 38 probands with PKP2 variants, compound or digenic heterozygosity was identified in16 (42%), including 6 probands with 3 variants: in two of these cases, one of the PKP2 variantswas the A372P polymorphism identified in the control population. Five probands hadadditional family members available for clinical and genetic evaluation including the followingprobands: patient 1 with the Q90R DSP variant (Table 3), the DSP W207X variant (patient 2,Table 3), the DSG2 V56M variant (patient 14, Table 3), the DSG2 R146H variant (patient 15,Table 3) and the DSP R1255K variant (patient 16, Table 3). The DSP Q90R variant was a denovo substitution and therefore could be pathogenic alone or in combination with the PKP2variant (Figure 1A). In patient 12, a 4609C>T DSP substitution (R1537C) was also identified,a variant previously reported as a single nucleotide polymorphism(http://www.ncbi.nlm.nih.gov/SNP/snp_ref.cgi?type=rs&rs=rs28763967). Whether thisvariant becomes clinically relevant in combination with the PKP2 variants and is needed forthe development of clinical features is speculation until animal models are completed. In thefamily of Patient 2 (Figure 2A), the PKP2 I531S variant was identified in three of the foursiblings available for study. All family members were clinically evaluated using the Task Force

Xu et al. Page 5

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

criteria (Table 1) and one of the three siblings was clinically unaffected (and also has twincarrier daughters) while the other two siblings were clinically affected. Only the proband hostedthe second variant, DSP W207X. In this case, we propose three potential interpretations ofthese data. Since the proband had the most severe phenotype (sudden death at age 25 years andsevere ARVC on autopsy), it is possible that the DSP W207X mutation was required to manifestsevere clinical disease. Secondly, this mutation may have no involvement in the developmentof disease and another, as yet identified mutation, is carried by the affected siblings but not bythe unaffected sibling. Thirdly, the I531S mutation may be disease-causing alone withextremely reduced penetrance, as has been described in other forms of cardiomyopathy, andrequires other factors, such as acquired agents, to manifest and contribute to clinical expression.In this regard, it should be noted that the proband developed mononucleosis two years prior todeath.

As previously noted, Patient 14 hosted a DSG2 V56M variant; in addition, however, this subjectalso had two additional PKP2 variants (Figure 2B). Based upon evaluation of the pedigree, itis difficult to be certain as to the role of each of these variants; however, it appears that thecombination of PKP2 S140F and DSG2 V56M, which was only detected in the proband, ledto a more severe clinical phenotype (Figure 2B).

Patient 15, a 51 year old male, was diagnosed with a severe form of ARVC at the age of 39years after an episode of ventricular fibrillation. He was found to host three different variants(PKP2 S209R, PKP2 T816fsX825 and DSG2 R146H) (Figure 3A). The PKP2 T816fsX825was inherited from his mother (II,2). Since his father's DNA was not available, it was notpossible to establish if the two missense variations (PKP2 S209R and DSG2 R146H) wereinherited from the father or are de novo mutations. His mother and maternal aunt, who bothcarried the PKP2 frameshift variant, were clinically unaffected by Task Force criteria andasymptomatic.

Patient 16, a 40 year old male, was diagnosed with a severe form of ARVC after an episodeof ventricular fibrillation that occurred during a sports activity. After resuscitation, he receivedan implantable cardioverter defibrillator (ICD). His family history included a male cousin whodied suddenly at 30 years of age while hiking. No autopsy was performed. The proband (II,1)inherited the DSP missense mutation and PKP2 stop mutation from his mother (I,2), and themissense PKP2 mutation from the father (I,1) (Figure 3B). At least one mutation was alsodetected in the proband's sons. In this family, all mutation carriers, with the exception of theproband, were completely asymptomatic and clinically unaffected by Task Force criteria.

Overall, there was more VF and exercise-induced VT in those subjects having compound ordigenic heterozygosity compared to those subjects with single heterozygous PKP2 mutations.In addition, the age of onset of symptomatic ARVC, most commonly ventricular arrhythmiasor syncope, was earlier in those subjects with multiple genetic variants in PKP2 or in PKP2plus other desmosomal genes (mean age 31.5 years) compared to those subjects with singleheterozygous gene mutations (mean age 39.5 years). Therefore, it appears that compound anddigenic heterozygosity leads to more clinically apparent and severe ARVC with an earlier ageof onset than those hosting heterozygous mutations in these genes.

GENETIC ANALYSIS: SINGLE GENE MUTATIONSIn 14 of the 198 subjects analyzed (7%), single heterozygous mutations were identified indesmosome-encoding genes other than PKP2. The heterozygous mutations identified included4 variants in DSP, 5 variants in DSG2, 3 variants in DSC2, and 2 variants in JUP (Table 4).All variants were identified in conserved regions and none of these variants were identified in700 ethnic-matched controls (1,400 chromosomes).

Xu et al. Page 6

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

FUNCTIONAL ANALYSISMyocardial autopsy samples were available from two individuals with PKP2 variants,including one patient (PG) with a single heterozygous PKP2 W528X nonsense mutation (nosecond variant has been identified to date) and a second patient (M686) with digenicheterozygosity including the PKP2 I531S variant as well as a DSP W207X mutation. Thesemyocardial samples were sectioned and stained for a variety of junctional proteins, includingPKP2, DSP, JUP, N-cadherin, and DSC2/3, as well as for Cx43 and DES (Figure 4). Stainingof samples from patient PG was very weak or absent for each of these proteins except for DSP,which was normal. In contrast, staining of samples from patient M686 was essentially normalfor each antibody, except for Cx43, which was absent (Figure 4), and DSP, which appeared tostain weakly. These data suggest that the DSP nonsense mutation in patient M686 does noteffect DSP localization and that these variants do not grossly affect the desmosomal junctions.In contrast, both the desmosomal and cadherin junctions appear to be affected in patient PG,with loss of staining for N-cadherin and JUP, in addition to PKP2. Signal intensities werecomparable in samples obtained from both left ventricle and right ventricle and in areasrelatively unaffected by fibrofatty replacement.

DISCUSSIONArrhythmogenic right ventricular cardiomyopathy (ARVC) has emerged as a significant causeof sudden death, heart failure and the need for heart transplantation over the past decade.2,31The incidence was initially thought to be particularly high in the Veneto region of Italy andin other parts of Europe, but low elsewhere.2 More recently, the disease has been increasinglyrecognized throughout the world.3,32,33 It is believed to be inherited in a moderate percentageof cases, with autosomal dominant inheritance predominating.4 Multiple genes, mostly thoseencoding desmosomal proteins, have been identified as causative in ARVC, with PKP2reported to be responsible for approximately 25% of all cases.8, 18-21 In the work presentedherein, however, interpretation of these data are questioned and the concepts of low penetrance,as well as compound and digenic heterozygosity are proposed as potential determinants of theclinical presentation in subjects carrying mutations and in their family members. In addition,identification of novel heterozygous mutations in other desmosome-encoding genes, includingnovel mutations in JUP, further supports the notion that the “final common pathway” forARVC is the cell-cell junctions. 34

In this report, we demonstrate that, while variants in PKP2 are relatively common (identifiedin 38/198 or 19% of probands), harboring one PKP2 variant may not by itself be sufficient todetermine overt clinical disease. In at least 16/38 (42%) cases, concomitant causes such aseither a “second hit” in the same gene (compound heterozygosity) or in a second desmosome-encoding gene (digenic heterozygosity) or an acquired disruption of these proteins orenvironmental factors, has been shown to be required for the overt clinical phenotype todevelop or for modification of disease severity. Hence, while PKP2 “mutations” are relativelycommon, a second variant in PKP2 or in DSP, DSC2, DSG2, PKP4, JUP or other interactingjunction protein-encoding gene appears to be important for the disease and its clinicalconsequences to be manifest. Other “second hit” genes or interactors are likely to be discoveredin the future. In addition, a variety of heterozygous mutations in all of the known desmosome-encoding genes were identified to cause ARVC and its associated clinical signs and symptoms.

Interestingly, the subjects harboring more than one variant in PKP2 and/or PKP2 plus otherdesmosome-encoding genes appear to have earlier onset of disease and more clinical severitythan those individuals harboring heterozygous mutations alone. Interpretation of these data,however, is confounded by the small number of large pedigrees available for analysis. In orderto track segregation of the genotype with disease, large families with clinical ARVC arerequired but, in our study, small families and sporadic cases were predominantly seen.

Xu et al. Page 7

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

However, in the families in which multiple individuals were enrolled and genetically screened,a single genetic variant commonly did not lead to overt clinical disease using the Task Forcecriteria (Table 1) or the stringent diagnostic evaluation used by the ARVD Registry. It will beinteresting to determine if the carriers previously phenotyped as unaffected will be found tohave any signs of early ARVC upon detailed examination (MRI, echocardiogram, etc) orwhether later development of clinical signs due to an otherwise concealed pathological processwill occur over time. These genetic findings should encourage physicians caring for patientswith ARVC to screen as many families as possible, irrespective of clinical presentation.Clearly, longitudinal follow-up of patients and carrier family members will be critical indetermining the influence of these gene variants and, more importantly, will be critical inproviding excellent preventive care. As noted, we have only identified second variants in about40% of the probands in whom PKP2 variants were identified. What about the remainder?Firstly, many genes that encode proteins that either directly or indirectly contributes to thefunction and integrity of cell adhesion junctions remain to be screened, such as the genesencoding α- or β-catenin. Mutations in proteins of the myocyte cytoarchitecture which arelinked to these proteins, such as actin, α-actinin-2, metavinculin, or Z-disk proteins could beimportant and will also be evaluated. Secondly, some mutations could result in dominantnegative proteins giving rise to autosomal dominant inheritance, the mode of inheritancewidely held as the most common in patients suffering from ARVC. In addition to the compoundheterozygous and digenic mutations noted, we also identified apparent isolated heterozygousmutations in desmosome-encoding genes in another 7% of subjects. We would speculate thatother relevant genes have yet to be discovered. An interesting observation from these studiesis that nonsense or frame-shift mutations alone in PKP2 often do not result in clinical diseasein these families. These data would suggest that haploinsufficiency for PKP2 is not criticallyimportant or even sufficient or that compensatory mechanisms occur. Only when the remainingcopy of PKP2 is mutated or there is a mutation in another gene, does overt clinical diseasedevelop. It also suggests that the description of these genetic variants as “disease-causingmutations” may be inaccurate and that defining affectation status by genetic analysis alonemay not be entirely appropriate. For instance, on face value, the I531S variant would appearto be a polymorphism. However, the detection of this variant in 5 of the 143 probands (fromdifferent regions of the U.S.), but in none of the 700 controls, would suggest that this variantis linked to disease. On the other hand, nonsense mutations in PKP2 would be expected toresult in overt clinical disease but, in many cases, does not. For this reason, we would cautionpotential “fee-for-service” laboratories offering “clinical testing” for ARVC from definitivestatements regarding cause-and effect relationships, particularly in subjects in whom PKP2variants are identified, especially if the remaining genes are not screened. In this circumstance,not only is the affected subject at risk to have a mistaken causative gene assigned, but “at-risk”family members may be either mistakenly diagnosed with a causative mutation or, moreconcerning, be given a negative result. In the latter case, this could lead to discharge fromfollow-up despite actually carrying a disease causing mutation in another gene not analyzed.Thia could lead to tragic outcomes. Grossman et al.35 ablated the PKP2 gene in mice, causinglethal alteration in heart morphogenesis at mid-gestation characterization by reducedtrabeculations, disarrayed cytoskeleton, rupture of the cardiac wall, and hemopericardium. Inthe absence of PKP2, the cytoskeletal linker protein desmoplakin dissociates from thejunctional plaques that connect cardiomyocytes, resulting in reduced architectural stability ofintercalated disks. Constant mechanical stress, as seen in the contracting heart, is likely to bedeleterious to the weakened intercellular junctions, leading to disruption, dilation anddysfunction. However, heterozygous mice were healthy and fertile. This is consistent with ournotion that haploinsufficiency for PKP2 is not sufficient to cause disease. The right ventricle,based on its geometry, architecture, and role in cardiac function, dilates to a variety of volumeand pressure abnormalities and would likely develop structural and functional disease earlierthan the left ventricle. This was clearly shown in the DSP transgenic mouse model of Yang etal, who demonstrated early right ventricular dilation with later left ventricular dilation, as well

Xu et al. Page 8

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

as intercalated disk disruption and loss of desmosomes.29 In addition, further support isprovided by the fact that the left ventricle develops late-onset disease in some patients withARVC.3, 36 In addition to the genetic mutations, mechanical stress and stretch forces on thedisturbed desmosome and intercalated disk, likely plays a role in disease development andseverity.34,37

Together, the genetic and functional data provided here should help to define the nature of thegenetic and clinical basis of ARVC. This work could significantly impact on clinical geneticscreening, as simple single gene analysis (particularly for PKP2) would be inappropriate andthe potential for inaccurate interpretation based on single (or even multiple) gene analysisappears to be high. More detailed information regarding the true clinical relevance of PKP2mutations is needed. Further, the moderate number of mutations in the other desmosome-encoding genes strongly suggests that all genes in this pathway should be screened in allsubjects. Identification of heterozygous mutations in JUP, as well as the potential mutationwithin the PKP4 gene adds to the spectrum of affected genes involving the desmosome. Furtherstudies of PKP4 in subjects with ARVC, as well as functional analysis of models with mutantPKP4 will be needed to clearly state that this is a potential disease-causing gene in ARVC.

The lack of functional studies of the variants described is a limitation of the study. Cellularand animal models incorporating these mutations individually and together could help toresolve these issues to some extent but these approaches may not necessarily recapitulate thehuman condition. In attempt to better understand the mechanisms involved in the development(or lack of development) of the clinical phenotype associated with these variants, we are in theprocess of developing these models for study. In addition to the models, the addition ofmechanical stretch and stress on the cells and animals could facilitate the development ofphenotypic differences from non-stressed models and provide greater insight.

Supplementary MaterialRefer to Web version on PubMed Central for supplementary material.

REFERENCES1. Marcus FI, Fontaine GH, Guiraudon G, Frank R, Laurenceau JL, Malergue C, Grosgogeat Y. Right

ventricular dysplasia: a report of 24 adult cases. Circulation 1982;65:384–398. [PubMed: 7053899]2. Corrado D, Basso C, Thiene G, McKenna WJ, Davies MJ, Fontaliran F, Nava A, Silvestri F,

Blomstrom-Lundqvist C, Wlodarska EK, Fontaine G, Camerini F. Spectrum of clinicopathologicmanifestations of arrhythmogenic right ventricular cardiomyopathy/dysplasia: a multicenter study. J.Am. Coll. Cardiol 1997;30:1512–1520. [PubMed: 9362410]

3. Horimoto M, Funayama N, Satoh M, Igarashi T, Sekiguchi M. Histologic evidence of left ventricularinvolvement in arrhythmogenic right ventricular dysplasia. Jpn Circ J 1989;53:1530–1534. [PubMed:2632822]

4. Towbin JA. Molecular genetics of sudden cardiac death. Cardiovasc Pathol 2001;10:283–295.[PubMed: 11755374]

5. Bowles NE, Ni J, Marcus F, Towbin JA. The detection of cardiotropic viruses in the myocardium ofpatients with arrhythmogenic right ventricular dysplasia/cardiomyopathy. J.Am. Coll. Cardiol2002;39:892–895. [PubMed: 11869858]

6. Basso C, Thiene G, Corrado D, Angelini A, Nava A, Valente M. Arrhythmogenic right ventricularcardiomyopathy. Dysplasia, dystrophy, or myocarditis? Circulation 1996;94:983–991. [PubMed:8790036]

7. Rampazzo A, Nava A, Malacrida S, Beffagna G, Bauce B, Rossi V, Zimbello R, Simionati B, BassoC, Thiene G, Towbin JA, Danieli GA. Mutation in human desmoplakin domain binding to plakoglobincauses a dominant form of arrhythmogenic right ventricular cardiomyopathy. Am J Hum Genet2002;71:1200–1206. [PubMed: 12373648]

Xu et al. Page 9

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

8. Gerull B, Heuser A, Wichter T, Paul M, Basson CT, McDermott DA, Lerman BB, Markowitz SM,Ellinor PT, MacRae CA, Peters S, Grossman KS, Drenckhahn J, Michely B, Sasse-Klaassen S,Birchmeier W, Dietz R, Breithardt G, Schulze-Bahr E, Thierfelder L. Mutations in the desmosomalprotein plakophilin-2 are common in arrhythmogenic right ventricular cardiomyopathy. Nat Genet2004;36:1162–1164. [PubMed: 15489853]

9. Pilichou K, Nava A, Basso C, Beffagna G, Bauce B, Lorenzon A, Frigo G, Vettori A, Valente M,Towbin JA, Thiene G, Danieli GA, Rampazzo A. Mutations in desmoglein-2 gene are associated witharrhythmogenic right ventricular cardiomyopathy. Circulation 2006;113:1171–1179. [PubMed:16505173]

10. Syrris P, Ward D, Evans A, Asimaki A, Gandjbakhch E, Sen-Chowdhry S, McKenna WJ.Arrhythmogenic right ventricular dysplasia/cardiomyopathy associated with mutations in thedesmosomal gene desmocollin-2. Am J. Hum Genet 2006;79:978–984. [PubMed: 17033975]

11. Beffagna G, Occhi G, Nava A, Vitiello A, Ditadi A, Basso C, Bauce B, Carraro G, Thiene G, TowbinJA, Danieli GA, Rampazzo A. Regulatory mutations in transforming growth factor-beta3 gene causearrhythmogenic right ventricular cardiomyopathy type 1. Cardiovasc Res 2005;65:366–73.[PubMed: 15639475]

12. Tiso N, Stephan DA, Nava A, Bagattin A, Devaney JM, Stanchi F, Larderet G, Brahmbhatt B, BrownK, Bauce B, Muriago M, Basso C, Thiene G, Danieli GA, Rampazzo A. Identification of mutationsin the cardiac ryanodine receptor gene in families affected with arrhythmogenic right ventricularcardiomyopathy type 2 (ARVD2). Hum Mol Genet 2001;10:189–194. [PubMed: 11159936]

13. Merner ND, Hodgkinson KA, Haywood AF, Connors S, French VM, Drenckhahn JD, Kupprion C,Ramadanova K, Thierfelder L, McKenna W, Gallagher B, Morris-Larkin L, Bassett AS, Parfrey PS,Young TL. Arrhythmogenic right ventricular cardiomyopathy type 5 is a fully penetrant, lethalarrhythmic disorder caused by a missense mutation in the TMEM43 gene. Am J Hum Genet2008;82:809–821. [PubMed: 18313022]

14. Coonar AS, Protonotarios N, Tsatsopoulou A, Needham EW, Houlston RS, Cliff S, Otter MI, MurdayVA, Mattu RK, McKenna WJ. Gene for arrhythmogenic right ventricular cardiomyopathy withdiffuse nonepidermolytic palmoplantar keratoderma and woolly hair (Naxos disease) maps to 17q21.Circulation 1998;97:2049–58. 1. [PubMed: 9610536]

15. Carvajal-Huerta L. Epidermolytic palmoplantar keratoderma with woolly hair and dilatedcardiomyopathy. Am. Acad. Dermatol 1998;39:418–421.

16. McKoy G, Protonotarios N, Crosby A, Tsatsopoulou A, Anastasakis A, Coonar A, Norman M,Baboonian C, Jeffery S, McKenna WJ. Identification of a deletion in plakoglobin in arrhythmogenicright ventricular cardiomyopathy with palmoplantar keratoderma and woolly hair (Naxos disease).Lancet 2000;355:2119–24. [PubMed: 10902626]

17. Norgett EE, Hatsell SJ, Carvajal-Huerta L, Cabezas JC, Common J, Purkis PE, Whittock N, LeighIM, Stevens HP, Kelsell DP. Recessive mutation in desmoplakin disrupts desmoplakin-intermediatefilament interactions and causes dilated cardiomyopathy, woolly hair and keratoderma. Hum MolGenet 2000;9:2761–6. [PubMed: 11063735]

18. Syrris P, Ward D, Asimaki A, Sen-Chowdhry S, Ebrahim HY, Evans A, Hitomi N, Norman M,Pantazis A, Shaw AL, Elliott PM, McKenna WJ. Clinical expression of plakophilin-2 mutations infamilial arrhythmogenic right ventricular cardiomyopathy. Circulation 2006;113:356–364.[PubMed: 16415378]

19. Van Tintelen JP, Entius MM, Bhuiyan ZA, Jongbloed R, Wiesfeld AC, Wilde AA, van der Smagt J,Boven LG, Mannens MM, van Langen IM, van Hofstra RM, Otterspoor RC, Doevendens PA,Rodriguez LM, van Gelder IC, Hauer RN. Plakophilin-2 mutations are the major determinant ofarrhythmogenic right ventricular dysplasia/cardiomyopathy. Circulation 2006;113:1650–1658.[PubMed: 16567567]

20. Dalal D, Molin LH, Piccini J, Tichnell C, James C, Bomma C, Prakasa K, Towbin JA, Marcus FI,Spevak PJ, Bluemke DA, Abraham T, Russell SD, Calkins H, Judge DP. Clinical features ofarrhythmogenic right ventricular dysplasia/cardiomyopathy associated with mutations inplakophilin-2. Circulation 2006;113:1641–1649. [PubMed: 16549640]

21. Dalal D, James C, Devanagondi R, Tichnell C, Tucker A, Prakasa K, Spevak PJ, Bluemke DA,Abraham T, Russell SD, Calkins H, Judge DP. Penetrance of mutations in plakophilin-2 among

Xu et al. Page 10

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

families with arrhythmogenic right ventricular dysplasia/cardiomyopathy. J. Am. Coll. Cardiol2006;48:1416–1424. [PubMed: 17010805]

22. Garrod DR, Merritt AJ, Nie Z. Desmosomal adhesion: structural basis, molecular mechanism andregulation. Mol Membr Biol 2002;19:81–94. [PubMed: 12126234]

23. Peifer M, Berg S, Reynolds AB. A repeating amino acid motif shared by proteins with diverse cellularroles. Cell 1994;76:789–91. [PubMed: 7907279]

24. Yin T, Green KJ. Regulation of desmosome assembly and adhesion. Semin Cell Dev Biol2004;15:665–77. [PubMed: 15561586]

25. Marcus F, Towbin JA, Zareba W, Moss A, Calkins H, Brown M, Gear K, ARVD/C Investigators.Arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C): a multidisciplinary study:design and protocol. Circulation 2003;107:2975–2978. [PubMed: 12814984]

26. McKenna WJ, Thiene G, Nava A, Fontaliran F, Blomstrom-Lundqvist C, Fontaine G, Camerini F.Diagnosis of arrhythmogenic right ventricular dysplasia/cardiomyopathy: Task Force of the WorkingGroup Myocardial and Pericardial Disease of the European Society of Cardiology and of theScientific Council on Cardiomyopathies of the International Society and Federation of Cardiology.Br Heart J 1994;71:215–218. [PubMed: 8142187]

27. Vatta M, Mohapatra B, Jimenez S, Sanchez X, Faulkner G, Perles Z, Sinagra G, Lin JH, Vu TM,Zhou Q, Bowles KR, Di Lenarda A, Schimenti L, Fox M, Chrisco MAA, Murphy RT, McKenna W,Elliott P, Bowles NE, Chen J, Valle G, Towbin JA. Mutations in Cypher/ZASP in patients with dilatedcardiomyopathy and left ventricular non-compaction. J. Am. Coll. Cardiol 2003;42:2014–2027.[PubMed: 14662268]

28. Kaplan SR, Gard JJ, Carvajal-Huerta L, Ruiz-Cabezas JC, Thiene G, Saffitz JE. Structural andmolecular pathology of the heart in Carvajal syndrome. Cardiovasc Pathol 2004;13:26–32. [PubMed:14761782]

29. Yang Z, Bowles NE, Scherer SE, Taylor MD, Kearney DL, Ge S, Nadvoretskiy VV, Defreitas G,Carabello B, Brandon LI, Godsel LM, Green KJ, Saffitz JE, Li H, Danieli GA, Calkins H, MarcusF, Towbin JA. Desmosomal dysfunction due to mutations in desmoplakin causes arrhythmogenicright ventricular dysplasia/cardiomyopathy. Circ Res 2006;99:646–655. [PubMed: 16917092]

30. Marcus FI, McKenna WJ, Sherrill D, Basso C, Bauce B, Bluemke DA, Caulkins H, Corrado DA,Cox MGPJ, Daubert J, Fontaine G, Gear K, Hauer RNW, Nava A, Picard MH, Protonotarios N,Saffitz JE, Yoerger Sanborn DM, Steinberg JS, Tandri HM, Thiene G, Towbin JA, Tsatsopoulou A,Wichter T, Zareba W. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia(ARVC/D): Proposed modification of Task Force Criteria. Circulation. 2009 in press.

31. Thiene G, Nava A, Corrado D, Rossi L, Pennelli N. Right ventricular cardiomyopathy and suddendeath in young people. N Engl J Med 1988;318:129–33. [PubMed: 3336399]

32. Dalal D, Nasir K, Bomma C, Prakasa K Tandri H, Piccini J, Roquin A, Tichnell C, James C, RussellSD, Judge DP, Abraham T, Spevak PJ, Bluemke DA, Calkins H. Arrhythmogenic right ventriculardysplasia: a United States experience. Circulation 2005;112:3823–3832. [PubMed: 16344387]

33. Cho Y, Park T, Shin D, Lee JH, Ryu HM, Jang GL, Lee DY, Park Y, Lee H, Kim H, Shin SC, HeoJH, Kang H, Lee BR, Nah DY, Yang DH, Park HS, Chae SC, Jun JE, Park WH. Clinicalmanifestations of arrhythmogenic right ventricular cardiomyopathy in Korean patients. Int J Cardiol2007;122:137–142. [PubMed: 17254652]

34. Vatta M, Marcus FI, Towbin JA. Arrhythmogenic right ventricular cardiomyopathy: a “final commonpathway that defines clinical phenotype”. Eur Heart J 2007;28:529–530. [PubMed: 17303588]

35. Grossmann KS, Grund C, Huesken J, Behrend M, Erdmann B, Franke WW, Birchmeier W.Requirement of plakophilin 2 for heart morphogenesis and cardiac junction formation. J Cell Biol2004;167:149–160. [PubMed: 15479741]

36. Takahashi N, Ishida Y, Maeno M, Hirose Y, Kawano S, Fukuoka S, Hayashida K, Kuribayashi S,Hamada S, Yamada N, Takamiya M, Shimomura K, Ohne T. Noninvasive identification of leftventricular involvements in arrhythmogenic right ventricular dysplasia: comparison of 123I-MIB G,201 TICI, magnetic resonance imaging and ultrafast computed tomography. Ann Nucl Med1997;11:233–241. [PubMed: 9310173]

37. Towbin JA, Bowles NE. Dilated cardiomyopathy: a tale of cytoskeletal proteins and beyond. JCardiovasc Electrophysiol 2006;17:919–926. [PubMed: 16764708]

Xu et al. Page 11

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 1. Pedigrees of three families with PKP2 variants and incomplete penetrance

Xu et al. Page 12

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Panel A: L404fsX409 PKP2 was identified in the proband and unaffected sister and wasinherited from the unaffected mother (+; in blue). In addition, a de novo variant, DSP Q90R,was identified in the proband (◆). The arrow indicates the proband. Panel B: K456NfsX458was identified in the proband and inherited from the unaffected mother (+). The arrow indicatesthe proband. NA: Not available for genetic analysis. Panel C: Pedigree of a family withcompound heterozygous PKP2 variants. V837fsX930 (+) was identified in the proband andboth of his siblings and was inherited from the unaffected father, who in turn inherited it fromhis unaffected father. R388W (◆) was identified in the proband and his affected brother andwas inherited from his phenotypically normal mother. The arrow indicates the proband. NA:Not available for genetic analysis.

Xu et al. Page 13

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 2. Pedigree of two U.S. families with compound desmosomal mutations (digenicheterozygosity)Panel A: PKP2 I531S (+) was identified in the proband and two of her siblings. DSP W207X(◆) was identified in the proband who experienced sudden death at 25 years of age. Heraffected living brother was diagnosed by cardiac MRI and is still alive at the age of 42 years.Her unaffected sister carrying the I531S variant is now 51years of age and has twin unaffecteddaughters, aged 19 years, both of whom carry the variant. Individuals without a definitivediagnosis of ARVC, but in whom signs or symptoms compatible with this diagnosis werereported, are identified in blue. The arrow indicates the proband. Panel B: PKP2 S140F (+),PKP2 IVS10-1G>C (◆) and DSG2 V56M (♣) were detected in the proband. The oldest child

Xu et al. Page 14

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

of the proband, who is 19 years old, does not meet full Task Force criteria of ARVC but hasarrhythmias and symptoms. The arrow indicates the proband. SD: Sudden death; ABN Echo:Abnormal echocardiogram; CHF: Congestive Heart Failure; PM: Pacemaker

Xu et al. Page 15

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 3. Pedigree of two Italian families with compound desmosomal mutationsPanel A: PKP2 T816fsX825 (◆) was identified in the proband and two family members.PKP2 S209R (+) and DSG2 R146H (♣) were also identified in the proband. Panel B: PKP2Q211X (+), PKP2 I778T (♣) and DSP R1255K (◆) were detected in the proband. All familymembers carrying different mutations were completely asymptomatic. The probands areindicated by the arrows. SD: Sudden death; aSD: aborted sudden death.

Xu et al. Page 16

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Figure 4. Immunostaining of myocardial sections from a control heart and from the hearts of ARVCpatients PG (PKP2 W538X) and M686 (PKP2 I531S and DSP W207X)The panels show staining for PKP2 (top), DSP (middle), and CX43 (bottom). Bars = 20μm

Xu et al. Page 17

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Xu et al. Page 18

Table 1Task Force Diagnostic Criteria for ARVC.26

The diagnosis of ARVC is fulfilled by the presence of 2 major, 1 major plus 2 minor, or 4 minor criteria from I-VI.25, 26

I. Global and/or regional dysfunction and structural alterations

Major

Severe dilation and reduction of right ventricular ejection fraction with no (or only mild) LVimpairment

Localized right ventricular aneurysms (akinetic or dyskinetic areas with diastolic bulging)

Severe segmental dilation of the RV

Minor

Mild global right ventricular dilation and/or ejection fraction reduction with normal LV

Mild segmental dilation of the RV

Regional right ventricular hypokinesia

II. Tissue characterization of wall

Major

Fibrofatty replacement of myocardium on endomyocardial biopsy

III. Repolarization abnormalities

Minor

Inverted T waves in right precordial leads (V2and V3) in people aged >12 years, in absence ofright bundle-branch block

IV. Depolarization/conduction abnormalities

Major

Epsilon waves or localized prolongation (>110 ms) of the QRS complex in right precordial leads(V1–V3).

Minor

Late potentials (signal-averaged ECG)

V. Arrhythmias

Minor

Left bundle-branch block type ventricular tachycardia (sustained and nonsustained) by ECG,Holter, or exercise testing

Frequent ventricular extrasystoles (>1000/24 hours) (Holter)

VI. Family history

Major

Familial disease confirmed at necropsy or surgery.

Family history of premature sudden death (<35 years) due to suspected right ventriculardysplasia

Minor

Familial history (clinical diagnosis based on present criteria).

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Xu et al. Page 19

Table 2

PKP2 gene variants identified in patients with ARVC

NT change * Exon AA Change** US Probands Italian Probands

145_148delCAGA 1 S50fsX110 4 3

419C>T 3 S140F 1 0

627C>G 3 S209R 0 1

630C>T 3 Q211X 1 1

1114G>C 4 A372P 3 0

1162C>T 4 R388W 1 0

1170-2A>G Intron 4 1 0

1212insT 5 L404fsX409 1 0

1368delA 5 K456fsX458 1 0

1592T>G 7 I531S 3 0

1613G>A 7 W538X 2 0

1643delG 7 G548fsX562 0 1

1760G>A 8 V587I 0 1

1978C>T 10 Q660X 1 0

2009delC 10 N670fsX683 0 1

2119C>T 10 Q707X 0 1

2146-1G>C Intron 10 3 0

2197_2202insGdelCACACC 11 H733fsX740 1 0

2359C>T 12 L787F 1 0

2447_2448delCC 12 T816fsX825 0 2

2509delA 13 V837fsX930 3 0

*NT change: Nucleotide change;

**AA change: Amino acid change.

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Xu et al. Page 20

Tabl

e 3

Clin

ical

Dem

ogra

phic

s of A

RV

C S

ubje

cts w

ith D

esm

osom

al G

ene

Mut

atio

ns. N

ote

that

mos

t, bu

t not

all

subj

ects

with

mut

atio

ns m

eet f

ull T

ask

Forc

e D

iagn

ostic

Crit

eria

. All

subj

ects

hav

e rig

ht v

entri

cula

rin

volv

emen

t and

arr

hyth

mia

s and

seve

ral s

ubje

cts a

lso

have

left

vent

ricul

ar in

volv

emen

t.

PtG

ende

rA

geon

set

FHX

Affe

cted

pare

nts

Nat

iona

lity

Palp

itatio

nsSy

ncop

e/ SC

D

CH

F/N

YH

Acl

ass

Arr

hyth

mia

EC

Gab

nlSA

EC

Gab

nlIC

DT

rans

plan

tE

xer

VT

RV

inyv

olL

Vin

vol

MR

Iab

nD

XC

rite

ria

M/m

His

tolo

gyge

neN

t Cha

nge

aa Cha

nge

1F

25N

No

US

Yes

No/

No

No/

IV

Tape

xR

VO

TY

esY

esN

oN

oN

oY

esN

oY

esY

esY

esY

esPK

P2D

SP12

12in

sT26

9A>G

L404

fsX

409

Q90

R

2F

22Y

No

US

Yes

Yes

/No

No/

IV

Tape

xR

VO

TY

esY

esY

esN

oN

oY

esN

oY

esY

esY

esY

esPK

P2D

SP15

92T>

G62

0G>A

I531

SW

207X

3F

23Y

No

US

Yes

Yes

/No

No/

IV

T R

VO

TY

esY

esY

esN

oY

esY

esY

esY

esY

esY

esY

esPK

P2D

SG2

1592

T>G

146G

>AI5

31S

R49

H

4M

29N

No

US

Yes

No/

No

No/

IV

T ap

exY

esY

esN

oN

oY

esY

esN

oY

esY

esY

esN

oPK

P2D

SG2

145_

148d

elC

AG

A10

03A

>GS5

0fsX

110

T335

A

5* 6M

16Y

No

US

Yes

Yes

/No

No/

IV

T R

VO

TY

esY

esY

esN

oN

oY

esN

oY

esY

esY

esY

esPK

P2PK

P211

62C

>T25

09de

lAR

388W

V83

7fsX

930

M12

YU

SY

esY

es/N

oN

o/I

VT

RV

OT

Yes

Yes

Yes

No

No

Yes

No

Yes

Yes

Yes

Yes

7M

32Y

No

US

Yes

Yes

/No

No/

IV

T R

VO

Tap

exY

esY

esY

esY

esY

esY

esN

oY

esY

esY

esY

esPK

P2PK

P214

5_14

8del

CA

GA

1592

T>G

S50f

sX11

0I5

31S

8M

52N

No

US

Yes

No/

No

No/

IV

T R

VO

TY

esY

esY

esN

oN

oY

esY

esY

esY

esY

esN

oPK

P2PK

P215

92T>

G23

59C

>TI5

31S

L787

F

9**

10M

26Y

No

US

Yes

No/

No

No/

IV

T R

VO

TY

esY

esY

esN

oY

esY

esN

oY

esY

esY

esY

esPK

P2D

SC2

1613

G>A

1914

G>C

W53

8XQ

63SH

F25

YU

SY

esN

o/N

oN

o/I

VT

RV

OT

Yes

No

Yes

No

No

Yes

No

Yes

No

Yes

Yes

11F

30N

No

US

Yes

No/

No

No/

IN

SVT

Yes

No

Yes

No

Yes

Yes

No

Yes

No

Yes

No

PKP2

PKP2

DSP

1613

G>A

1914

G>C

88G

>A

W53

8XQ

638H

V30

M

12M

41N

No

US

Yes

Yes

/No

No/

IV

TY

esY

esY

esN

oY

esY

esN

oY

esY

esN

oN

oPK

P2PK

P2D

SP

1114

G>C

2145

-1G

>C46

09C

>T

A37

2PR

1537

C

13M

30Y

No

US

Yes

Yes

/No

No/

IV

TN

oY

esY

esY

esY

esY

esN

oY

esY

esN

oY

esPK

P2PK

P421

46-1

G>C

2786

A>G

D92

9G

14M

50Y

No

US

Yes

Yes

/Yes

No/

IV

T-R

Vor

igin

Yes

Yes

Yes

No

Yes

Yes

No

Yes

Yes

Yes

Yes

PKP2

PKP2

DSG

2

419C

>T21

46-1

G>C

166

G>A

S140

FV

56M

15M

44Y

No

Italia

nY

esY

es/Y

esN

o/I

VF

Yes

Yes

Yes

No

No

Yes

No

Not

perf

1/3

No

PKP2

PKP2

DSG

2

627C

>G24

47_2

448d

elC

C43

7G>A

S209

RT8

16fs

X82

5R

146H

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Xu et al. Page 21

PtG

ende

rA

geon

set

FHX

Affe

cted

pare

nts

Nat

iona

lity

Palp

itatio

nsSy

ncop

e/ SC

D

CH

F/N

YH

Acl

ass

Arr

hyth

mia

EC

Gab

nlSA

EC

Gab

nlIC

DT

rans

plan

tE

xer

VT

RV

inyv

olL

Vin

vol

MR

Iab

nD

XC

rite

ria

M/m

His

tolo

gyge

neN

t Cha

nge

aa Cha

nge

16M

40Y

No

Italia

nY

esY

es/Y

esN

o/I

VF

Yes

No

Yes

No

Yes

Yes

No

Yes

1/3

No

PKP2

PKP2

DSP

630C

>T23

33T>

C37

64G

>A

Q21

1XI7

78T

R12

55K

17F

31Y

No

Italia

nY

esN

o/N

oN

o/I

NSV

TY

esY

esN

oN

oN

oY

esY

esN

otpe

rf1/

4Y

esPK

P2PK

P2D

SP

2119

C>T

184C

>A49

61T>

C

Q70

7XQ

62K

L165

4P

18M

34Y

One

Italia

nY

esN

o/N

oN

o/I

NSV

TY

esY

esN

oN

oN

oY

esN

oN

otpe

rf1/

4N

oPK

P2D

SG2

145_

148d

elC

AG

A11

15G

>AS5

0fsX

110

V39

1I

Patie

nt 5

(pro

band

) and

6 a

re fr

om sa

me

fam

ily;

Pt=

Patie

nt n

umbe

r; FH

x= F

amily

His

tory

of A

RV

C; S

CD

= Su

dden

Car

diac

Dea

th; C

HF=

Con

gest

ive

Hea

rt Fa

ilure

; NY

HA

= N

ew Y

ork

Hea

rt A

ssoc

iatio

n; E

CG

abn

l= E

lect

roca

rdio

grap

hic

abno

rmal

ities

; SA

ECG

= Si

gnal

Ave

rage

d EC

G; I

CD

= Im

plan

tabl

e C

ardi

over

ter D

efib

rilla

tor;

Dx

Crit

eria

= D

iagn

ostic

Crit

eria

; M/m

= M

ajor

/min

or c

riter

ia; N

t= N

ucle

otid

e; a

a= A

min

o A

cid

**Pa

tient

9 (p

roba

nd) a

nd 1

0 ar

e fr

om sa

me

fam

ily.

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Xu et al. Page 22

Table 4

Heterozygous mutations identified in Desmosomal genes other than PKP2

Patient Gender Gene NT Change* AA Change**

1 M DSC2 874 C>T P292S

2 M DSC2 1026 A>G I342V

3 F DSC2 1660 C>T Q554X

4 M DSG2 146 G>A R49H

5 M DSG2 560 A>G D187G

6 M DSG2 1520 G>A C507Y

7 M DSG2 1003 A>G T335A

8 F DSG2 961 T>A F321I

9 M DSP 8501 G>A R2834H

10 F DSP 1598 T>C I533T

11 M DSP 688 G>A D230N

12 F DSP 1482 A>T Y494F

13 M JUP 392_394delTCA I31delI

14 M JUP 475 G>T V159L

*NT change: Nucleotide change;

**AA change: Amino acid change.

J Am Coll Cardiol. Author manuscript; available in PMC 2011 February 9.