Ankyrin-B dysfunction predisposes to arrhythmogenic cardiomyopathy and is amenable to therapy

Jason D Roberts, Nathaniel P Murphy, Robert M Hamilton, Ellen R Lubbers, Cynthia A James, Crystal F Kline, Michael H Gollob, Andrew D Krahn, Amy C Sturm, Hassan Musa, Mona El-Refaey, Sara Koenig, Meriam Åström Aneq, Edgar T Hoorntje, Sharon L Graw, Robert W Davies, Muhammad Arshad Rafiq, Tamara T Koopmann, Shabana Aafaqi, Meena Fatah, David A Chiasson, Matthew Rg Taylor, Samantha L Simmons, Mei Han, Chantal Jm van Opbergen, Loren E Wold, Gianfranco Sinagra, Kirti Mittal, Crystal Tichnell, Brittney Murray, Alberto Codima, Babak Nazer, Duy T Nguyen, Frank I Marcus, Nara Sobriera, Elisabeth M Lodder, Maarten P van den Berg, Danna A Spears, John F Robinson, Philip C Ursell, Anna K Green, Allan C Skanes, Anthony S Tang, Martin J Gardner, Robert A Hegele, Toon Ab van Veen, Arthur Am Wilde, Jeff S Healey, Paul Ml Janssen, Luisa Mestroni, J Peter van Tintelen, Hugh Calkins, Daniel P Judge, Thomas J Hund, Melvin M Scheinman, Peter J Mohler, Jason D Roberts, Nathaniel P Murphy, Robert M Hamilton, Ellen R Lubbers, Cynthia A James, Crystal F Kline, Michael H Gollob, Andrew D Krahn, Amy C Sturm, Hassan Musa, Mona El-Refaey, Sara Koenig, Meriam Åström Aneq, Edgar T Hoorntje, Sharon L Graw, Robert W Davies, Muhammad Arshad Rafiq, Tamara T Koopmann, Shabana Aafaqi, Meena Fatah, David A Chiasson, Matthew Rg Taylor, Samantha L Simmons, Mei Han, Chantal Jm van Opbergen, Loren E Wold, Gianfranco Sinagra, Kirti Mittal, Crystal Tichnell, Brittney Murray, Alberto Codima, Babak Nazer, Duy T Nguyen, Frank I Marcus, Nara Sobriera, Elisabeth M Lodder, Maarten P van den Berg, Danna A Spears, John F Robinson, Philip C Ursell, Anna K Green, Allan C Skanes, Anthony S Tang, Martin J Gardner, Robert A Hegele, Toon Ab van Veen, Arthur Am Wilde, Jeff S Healey, Paul Ml Janssen, Luisa Mestroni, J Peter van Tintelen, Hugh Calkins, Daniel P Judge, Thomas J Hund, Melvin M Scheinman, Peter J Mohler

Abstract

Arrhythmogenic cardiomyopathy (ACM) is an inherited arrhythmia syndrome characterized by severe structural and electrical cardiac phenotypes, including myocardial fibrofatty replacement and sudden cardiac death. Clinical management of ACM is largely palliative, owing to an absence of therapies that target its underlying pathophysiology, which stems partially from our limited insight into the condition. Following identification of deceased ACM probands possessing ANK2 rare variants and evidence of ankyrin-B loss of function on cardiac tissue analysis, an ANK2 mouse model was found to develop dramatic structural abnormalities reflective of human ACM, including biventricular dilation, reduced ejection fraction, cardiac fibrosis, and premature death. Desmosomal structure and function appeared preserved in diseased human and murine specimens in the presence of markedly abnormal β-catenin expression and patterning, leading to identification of a previously unknown interaction between ankyrin-B and β-catenin. A pharmacological activator of the WNT/β-catenin pathway, SB-216763, successfully prevented and partially reversed the murine ACM phenotypes. Our findings introduce what we believe to be a new pathway for ACM, a role of ankyrin-B in cardiac structure and signaling, a molecular link between ankyrin-B and β-catenin, and evidence for targeted activation of the WNT/β-catenin pathway as a potential treatment for this disease.

Keywords: Arrhythmias; Cardiology; Cardiovascular disease; Cell Biology; Genetic diseases.

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1. Deceased proband harboring AnkB loss-of-function…
Figure 1. Deceased proband harboring AnkB loss-of-function p.Glu1458Gly variant exhibits arrhythmogenic cardiomyopathy.
(A) Surface 12-lead ECG. (B) Loop recording demonstrating nonsustained polymorphic VT. (C) Cardiac magnetic resonance short-axis image revealing moderate biventricular dilation; the yellow ring highlights focal “crinkling” of the subtricuspid region of the right ventricular free wall consistent with the “accordion sign.” (D) Delayed enhancement imaging revealed scarring in the right ventricular (RV) free wall in a short-axis view (arrows) and (E) the left ventricular (LV) lateral wall (arrow) in a long-axis view. (F) Mid-transverse section of the autopsied heart revealing severe right ventricular dilation and wall thinning as well as concentric hypertrophy of the left ventricle. Histology of the (G) right ventricle showing severe fibrofatty infiltration of the free wall in association with extensive ventricular interstitial fibrosis and (H) left ventricle showing moderate hypertrophy with focal areas of fibrofatty muscular infiltration and widespread interstitial fibrosis. Scale bars: 160 μm (G) and 60 μm (H).
Figure 2. Deceased proband possessing AnkB loss-of-function…
Figure 2. Deceased proband possessing AnkB loss-of-function p.Met1988Thr variant exhibits arrhythmogenic cardiomyopathy.
(A) Apical transverse section of the explanted heart of the deceased AnkB-p.Met1988Thr proband revealing fatty infiltration and localized thinning of the anterior and lateral aspects of the right ventricular free wall. Scale bar: 1 cm. (B) Open view of the right ventricle revealing moderate dilation. Scale bar: 1 cm. Histology of the anteroapical right ventricular free wall at (C) ×1 and (D) ×40 original magnification, revealing marked fatty infiltration of the myocardium with patchy interstitial fibrosis and myocardial disorganization. Scale bars: 5 mm (C) and 300 μm (D). (E) Four-lead surface ECG of paternal cousin following a treadmill test, revealing VT.
Figure 3. Cardiomyocyte-specific deletion of AnkB results…
Figure 3. Cardiomyocyte-specific deletion of AnkB results in ventricular remodeling, cardiac arrhythmias, and reduced survival.
Immunoblotting for AnkB (A) in heart tissue from Ank2-cKO (n = 4) and Ank2fl/fl (n = 5) mice (complete immunoblots are shown in Supplemental Figure 3). (B and C) Quantification of (B) left ventricular ejection fraction of Ank2fl/fl mice (n = 4) and Ank2-cKO mice (n = 9) and (C) right ventricular fractional shortening in Ank2fl/fl (n = 6) and Ank2-cKO (n = 7) mice. (D) Kaplan-Meier survival curves for Ank2fl/fl (n = 36) and Ank2-cKO (n = 69) mice. (E and F) Representative ECGs for Ank2-cKO mice following injection of 2.0 mg/kg epinephrine. Arrows denote (E) VT and (F) trigeminy. (G) Heart weight/tibia length (HW/TL) ratios of Ank2fl/fl (n = 8) and Ank2-cKO (n = 9) mice. (H and I) Representative Masson’s trichrome–stained whole slide views of (H) Ank2fl/fl and (I) Ank2-cKO and (J) magnified Ank2fl/fl and (K) Ank2-cKO hearts. Images are representative of 4 hearts from mice of each genotype. Scale bars: 2.5 mm (H and I) and 25 μm (J and K). Data represent the mean ± SEM. Statistical significance for AC and G was determined using a 2-tailed parametric t test at 95% CI and a log-rank test for D.
Figure 4. Ank2 -cKO murine hearts show…
Figure 4. Ank2-cKO murine hearts show misclocalization of β-catenin.
Representative IF images of (A) desmin and N-cadherin, (B) plakoglobin and α-actinin, (C) connexin-43 and α-actinin, (D) desmoplakin and α-actinin, and (E) AnkB and β-catenin in Ank2fl/fl and Ank2-cKO cardiac cryoslices. Staining was completed in tissue from 3 hearts for each stain, per genotype, with 3 images taken per heart. Scale bars: 20 μm.
Figure 5. AnkB and β-catenin are molecular…
Figure 5. AnkB and β-catenin are molecular partners.
(A and B) Representative images of (A) plakoglobin and β-catenin staining from control and AnkB p.Met1988Thr human right ventricular tissue, and (B) β-catenin staining of control and AnkB p.Glu1458Gly human right ventricular tissue. Scale bars: 20 μm. (C) Co-IP assay of AnkB IgG and β-catenin in human ventricular lysate. (D) GST-pulldown assay of in vitro–translated AnkB MBD and β-catenin in mouse cardiac lysate (complete images are shown in Supplemental Figure 8). The binding experiments were replicated 3 times. (E) Representative immunoblot of β-catenin and (F) quantification of full-length β-catenin in Ank2fl/fl (n = 4) and Ank2-cKO (n = 5) mouse cardiac lysate (complete images are shown in Supplemental Figure 8). Data represent the mean ± SEM. Statistical analysis for F was performed using a 2-tailed parametric t test at 95% CI. Binding assays were completed in triplicate.
Figure 6. GSK3-β inhibition is sufficient to…
Figure 6. GSK3-β inhibition is sufficient to prevent cardiac remodeling associated with cardiac deletion of AnkB.
(A) Diagrammatic representation of GSK3-βi prevention study in Ank2fl/fl and Ank2-cKO mice. Echocardiograms were performed at the time points indicated by arrows. (B) Heart weight/tibia length ratios. n = 3. (C) Levels of p–β-catenin by IB of Ank2-cKO cardiac tissue lysates. n = 3. (D and E) Ejection fraction and fractional shortening at baseline. Ejection fraction (F) and fractional shortening (G) after 8 weeks of drug therapy in vehicle- and GSK3-βi–treated Ank2fl/fl and Ank2-cKO mice in the prevention study. n = 5 (D and E) and n = 9 (F and G). (H) Representative Masson’s trichrome–stained heart sections from vehicle-treated Ank2fl/fl mice. (I) Ank2fl/fl GSK-βi–treated, (J) Ank2-cKO vehicle-treated, and (K) Ank2-cKO GSK3-βi–treated mice after 8 weeks of drug therapy in the prevention study. Scale bars: 25 μm (IK). Images are representative of 2 hearts from mice of each genotype and treatment condition. Data represent the mean ± SEM. Statistical analysis for C was done with a 2-tailed parametric t test at 95% CI. Statistical analysis for B and DG was performed with a 2-way ANOVA followed by Tukey’s post hoc test.
Figure 7. GSK3-β inhibition is sufficient to…
Figure 7. GSK3-β inhibition is sufficient to reverse cardiac remodeling associated with cardiac deletion of AnkB.
(A) Diagrammatic representation of the GSK3-βi rescue study involving Ank2fl/fl and Ank2-cKO mice. Echocardiograms were performed at the time points indicated by arrows. Ejection fraction (B) and fractional shortening (C) at 12 weeks of age and ejection fraction (D) and fractional shortening (E) after 4 weeks of drug therapy in vehicle- and GSK3-βi–treated Ank2fl/fl and Ank2-cKO mice. n = 3 (BE). (F and G) Myocyte cross-sectional area in Ank2fl/fl (n = 3 for each treatment) and Ank2-cKO (n = 3 for each treatment) mice following treatment with vehicle or GSK3-βi. (HK) Representative cross-sectional images of Ank2fl/fl and Ank2-cKO hearts following vehicle and GSK3-βi therapy. Images are representative of 3 hearts from mice of each genotype and treatment condition. Scale bars: 25 μm. Data represent the mean ± SEM. Statistical analysis for BF was performed with a 2-way ANOVA followed by Tukey’s post hoc test.

References

    1. Corrado D, Link MS, Calkins H. Arrhythmogenic right ventricular cardiomyopathy. N Engl J Med. 2017;376(15):1489–1490. doi: 10.1056/NEJMc1701400.
    1. Sen-Chowdhry S, et al. Left-dominant arrhythmogenic cardiomyopathy: an under-recognized clinical entity. J Am Coll Cardiol. 2008;52(25):2175–2187. doi: 10.1016/j.jacc.2008.09.019.
    1. Corrado D, et al. Treatment of arrhythmogenic right ventricular cardiomyopathy/dysplasia: an International Task Force consensus statement. Circulation. 2015;132(5):441–453. doi: 10.1161/CIRCULATIONAHA.115.017944.
    1. McKoy G, et al. Identification of a deletion in plakoglobin in arrhythmogenic right ventricular cardiomyopathy with palmoplantar keratoderma and woolly hair (Naxos disease) Lancet. 2000;355(9221):2119–2124. doi: 10.1016/S0140-6736(00)02379-5.
    1. Gerull B, et al. Mutations in the desmosomal protein plakophilin-2 are common in arrhythmogenic right ventricular cardiomyopathy. Nat Genet. 2004;36(11):1162–1164. doi: 10.1038/ng1461.
    1. Awad MM, et al. DSG2 mutations contribute to arrhythmogenic right ventricular dysplasia/cardiomyopathy. Am J Hum Genet. 2006;79(1):136–142. doi: 10.1086/504393.
    1. Garcia-Gras E, et al. Suppression of canonical Wnt/beta-catenin signaling by nuclear plakoglobin recapitulates phenotype of arrhythmogenic right ventricular cardiomyopathy. J Clin Invest. 2006;116(7):2012–2021. doi: 10.1172/JCI27751.
    1. Chen SN, Gurha P, Lombardi R, Ruggiero A, Willerson JT, Marian AJ. The hippo pathway is activated and is a causal mechanism for adipogenesis in arrhythmogenic cardiomyopathy. Circ Res. 2014;114(3):454–468. doi: 10.1161/CIRCRESAHA.114.302810.
    1. Delmar M, McKenna WJ. The cardiac desmosome and arrhythmogenic cardiomyopathies: from gene to disease. Circ Res. 2010;107(6):700–714. doi: 10.1161/CIRCRESAHA.110.223412.
    1. Marcus FI, et al. Right ventricular dysplasia: a report of 24 adult cases. Circulation. 1982;65(2):384–398. doi: 10.1161/01.CIR.65.2.384.
    1. Mohler PJ, et al. Ankyrin-B mutation causes type 4 long-QT cardiac arrhythmia and sudden cardiac death. Nature. 2003;421(6923):634–639. doi: 10.1038/nature01335.
    1. Mohler PJ, et al. A cardiac arrhythmia syndrome caused by loss of ankyrin-B function. Proc Natl Acad Sci U S A. 2004;101(24):9137–9142. doi: 10.1073/pnas.0402546101.
    1. Cunha SR, et al. Defects in ankyrin-based membrane protein targeting pathways underlie atrial fibrillation. Circulation. 2011;124(11):1212–1222. doi: 10.1161/CIRCULATIONAHA.111.023986.
    1. Le Scouarnec S, et al. Dysfunction in ankyrin-B-dependent ion channel and transporter targeting causes human sinus node disease. Proc Natl Acad Sci U S A. 2008;105(40):15617–15622. doi: 10.1073/pnas.0805500105.
    1. Scotland P, Zhou D, Benveniste H, Bennett V. Nervous system defects of AnkyrinB (-/-) mice suggest functional overlap between the cell adhesion molecule L1 and 440-kD AnkyrinB in premyelinated axons. J Cell Biol. 1998;143(5):1305–1315. doi: 10.1083/jcb.143.5.1305.
    1. Dalal D, et al. Morphologic variants of familial arrhythmogenic right ventricular dysplasia/cardiomyopathy a genetics-magnetic resonance imaging correlation study. J Am Coll Cardiol. 2009;53(15):1289–1299. doi: 10.1016/j.jacc.2008.12.045.
    1. Lek M, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536(7616):285–291. doi: 10.1038/nature19057.
    1. Kirchhof P, et al. Age- and training-dependent development of arrhythmogenic right ventricular cardiomyopathy in heterozygous plakoglobin-deficient mice. Circulation. 2006;114(17):1799–1806. doi: 10.1161/CIRCULATIONAHA.106.624502.
    1. Cerrone M, et al. Plakophilin-2 is required for transcription of genes that control calcium cycling and cardiac rhythm. Nat Commun. 2017;8(1):106. doi: 10.1038/s41467-017-00127-0.
    1. Chelko SP, et al. Central role for GSK3β in the pathogenesis of arrhythmogenic cardiomyopathy. JCI Insight. 2016;1(5):e85923
    1. Moncayo-Arlandi J, et al. Molecular disturbance underlies to arrhythmogenic cardiomyopathy induced by transgene content, age and exercise in a truncated PKP2 mouse model. Hum Mol Genet. 2016;25(17):3676–3688. doi: 10.1093/hmg/ddw213.
    1. Cruz FM, et al. Exercise triggers ARVC phenotype in mice expressing a disease-causing mutated version of human plakophilin-2. J Am Coll Cardiol. 2015;65(14):1438–1450. doi: 10.1016/j.jacc.2015.01.045.
    1. Krusche CA, et al. Desmoglein 2 mutant mice develop cardiac fibrosis and dilation. Basic Res Cardiol. 2011;106(4):617–633. doi: 10.1007/s00395-011-0175-y.
    1. Asimaki A, et al. A new diagnostic test for arrhythmogenic right ventricular cardiomyopathy. N Engl J Med. 2009;360(11):1075–1084. doi: 10.1056/NEJMoa0808138.
    1. Noorman M, et al. Remodeling of the cardiac sodium channel, connexin43, and plakoglobin at the intercalated disk in patients with arrhythmogenic cardiomyopathy. Heart Rhythm. 2013;10(3):412–419. doi: 10.1016/j.hrthm.2012.11.018.
    1. Asimaki A, et al. Identification of a new modulator of the intercalated disc in a zebrafish model of arrhythmogenic cardiomyopathy. Sci Transl Med. 2014;6(240):240ra74. doi: 10.1126/scitranslmed.3008008.
    1. Swayne LA, et al. Novel variant in the ANK2 membrane-binding domain is associated with ankyrin-B syndrome and structural heart disease in a First Nations population with a high rate of long QT syndrome. Circ Cardiovasc Genet. 2017;10(1):e001537.
    1. Marcus FI, et al. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the task force criteria. Circulation. 2010;121(13):1533–1541. doi: 10.1161/CIRCULATIONAHA.108.840827.
    1. Wolfe RS. My kind of biology. Annu Rev Microbiol. 1991;45:1–35. doi: 10.1146/annurev.mi.45.100191.000245.
    1. Little SC, et al. Protein phosphatase 2A regulatory subunit B56α limits phosphatase activity in the heart. Sci Signal. 2015;8(386):ra72. doi: 10.1126/scisignal.aaa5876.
    1. Zhou J, et al. Upregulation of gamma-catenin compensates for the loss of beta-catenin in adult cardiomyocytes. Am J Physiol Heart Circ Physiol. 2007;292(1):H270–H276. doi: 10.1152/ajpheart.00576.2006.
    1. James CA, et al. Exercise increases age-related penetrance and arrhythmic risk in arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated desmosomal mutation carriers. J Am Coll Cardiol. 2013;62(14):1290–1297. doi: 10.1016/j.jacc.2013.06.033.
    1. Xu T, et al. Compound and digenic heterozygosity contributes to arrhythmogenic right ventricular cardiomyopathy. J Am Coll Cardiol. 2010;55(6):587–597. doi: 10.1016/j.jacc.2009.11.020.
    1. Sen-Chowdhry S, Syrris P, Pantazis A, Quarta G, McKenna WJ, Chambers JC. Mutational heterogeneity, modifier genes, and environmental influences contribute to phenotypic diversity of arrhythmogenic cardiomyopathy. Circ Cardiovasc Genet. 2010;3(4):323–330. doi: 10.1161/CIRCGENETICS.109.935262.
    1. Haggerty CM, et al. Electronic health record phenotype in subjects with genetic variants associated with arrhythmogenic right ventricular cardiomyopathy: a study of 30,716 subjects with exome sequencing. Genet Med. 2017;19(11):1245–1252. doi: 10.1038/gim.2017.40.
    1. Hall CL, Sutanto H, Dalageorgou C, McKenna WJ, Syrris P, Futema M. Frequency of genetic variants associated with arrhythmogenic right ventricular cardiomyopathy in the genome aggregation database. Eur J Hum Genet. 2018;26(9):1312–1318. doi: 10.1038/s41431-018-0169-4.
    1. Sawant AC, et al. Exercise has a disproportionate role in the pathogenesis of arrhythmogenic right ventricular dysplasia/cardiomyopathy in patients without desmosomal mutations. J Am Heart Assoc. 2014;3(6):e001471.
    1. Musa H, et al. Common human ANK2 variant confers in vivo arrhythmia phenotypes. Heart Rhythm. 2016;13(9):1932–1940. doi: 10.1016/j.hrthm.2016.06.012.
    1. Huq AJ, et al. A novel mechanism for human cardiac ankyrin-B syndrome due to reciprocal chromosomal translocation. Heart Lung Circ. 2017;26(6):612–618. doi: 10.1016/j.hlc.2016.09.013.
    1. Cheng HW, et al. Assessment of right ventricular structure and function in mouse model of pulmonary artery constriction by transthoracic echocardiography. J Vis Exp. 2014;(84):e51041.
    1. Adzhubei IA, et al. A method and server for predicting damaging missense mutations. Nat Methods. 2010;7(4):248–249. doi: 10.1038/nmeth0410-248.
    1. Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4(7):1073–1081. doi: 10.1038/nprot.2009.86.
    1. Schwarz JM, Rödelsperger C, Schuelke M, Seelow D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat Methods. 2010;7(8):575–576. doi: 10.1038/nmeth0810-575.
    1. Kircher M, Witten DM, Jain P, O’Roak BJ, Cooper GM, Shendure J. A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet. 2014;46(3):310–315. doi: 10.1038/ng.2892.

Source: PubMed

3
Sottoscrivi