Genetics and heart failure: a concise guide for the clinician

Cecile Skrzynia, Jonathan S Berg, Monte S Willis, Brian C Jensen, Cecile Skrzynia, Jonathan S Berg, Monte S Willis, Brian C Jensen

Abstract

The pathogenesis of heart failure involves a complex interaction between genetic and environmental factors. Genetic factors may influence the susceptibility to the underlying etiology of heart failure, the rapidity of disease progression, or the response to pharmacologic therapy. The genetic contribution to heart failure is relatively minor in most multifactorial cases, but more direct and profound in the case of familial dilated cardiomyopathy. Early studies of genetic risk for heart failure focused on polymorphisms in genes integral to the adrenergic and renin-angiotensin-aldosterone system. Some of these variants were found to increase the risk of developing heart failure, and others appeared to affect the therapeutic response to neurohormonal antagonists. Regardless, each variant individually confers a relatively modest increase in risk and likely requires complex interaction with other variants and the environment for heart failure to develop. Dilated cardiomyopathy frequently leads to heart failure, and a genetic etiology increasingly has been recognized in cases previously considered to be "idiopathic". Up to 50% of dilated cardiomyopathy cases without other cause likely are due to a heritable genetic mutation. Such mutations typically are found in genes encoding sarcomeric proteins and are inherited in an autosomal dominant fashion. In recent years, rapid advances in sequencing technology have improved our ability to diagnose familial dilated cardiomyopathy and those diagnostic tests are available widely. Optimal care for the expanding population of patients with heritable heart failure involves counselors and physicians with specialized training in genetics, but numerous online genetics resources are available to practicing clinicians.

Figures

Fig. (1)
Fig. (1)
Heart failure arises from a complex interaction between genetic and environmental factors. Genetic factors may influence the susceptibility to the underlying etiology of heart failure, the progression of the disease, or the response to pharmacologic therapy. Dilated cardiomyopathy (DCM) can result directly from a mutation in a single gene, but the genetic component of heart failure typically consists of interacting variants in many genes with minor individual effects.
Fig. (2)
Fig. (2)
Selected sarcomeric gene mutations in familial dilated cardiomyopathy. Mutations in over 50 genes can cause familial dilated cardiomyopathy. Most of those mutations occur in genes encoding sarcomeric proteins, and a selected number of the most commonly affected genes are listed.

References

    1. Lee DS, Pencina MJ, Benjamin EJ , et al. Association of parental heart failure with risk of heart failure in offspring. N Engl J Med. 2006;355:138–47.
    1. Gonzaga-Jauregui C, Lupski Jr, Gibbs RA. Human genome sequencing in health and disease. Annu Rev Med. 2012;63:35–61.
    1. Conrad DF, Jakobsson M, Coop G , et al. A worldwide survey of haplotype variation and linkage disequilibrium in the human genome. Nat Genet. 2006;38:1251–60.
    1. Golbus Jr, Puckelwartz MJ, Fahrenbach JP , et al. Population-based variation in cardiomyopathy genes. Circ Cardiovasc Genet. 2012;5:391–9.
    1. Norton N, Robertson PD, Rieder MJ , et al. Evaluating pathogenicity of rare variants from dilated cardiomyopathy in the exome era. Circ Cardiovasc Genet. 2012;5:167–74.
    1. Anderson S, Bankier AT, Barrell BG , et al. Sequence and organization of the human mitochondrial genome. Nature. 1981;290(5806):457–65.
    1. Musunuru K, Kathiresan S. HapMap and mapping genes for cardiovascular disease. Circ Cardiovasc Genet. 2008;1:66–71.
    1. Manolio TA, Collins FS. The HapMap and genome-wide association studies in diagnosis and therapy. Annu Rev Med. 2009;60:443–56.
    1. Hirschhorn JN, Gajdos ZK. Genome-wide association studies results from the first few years and potential implications for clinical medi-cine. Annu Rev Med. 2011;62:11–24.
    1. Roden DM, Johnson JA, Kimmel SE , et al. Cardiovascular pharmacogenomics. Circ Res. 2011;109:807–20.
    1. Talameh JA, Lanfear DE. Pharmacogenetics in chronic heart failure new developments and current challenges. Curr Heart Fail Rep. 2012;9:23–32.
    1. Cappola TP, Dorn GW. 2nd Clinical considerations of heritable factors in common heart failure. Circ Cardiovasc Genet. 2011:4701–9.
    1. Dorn GW. 2nd. The genomic architecture of sporadic heart failure. Circ Res. 2011;108:1270–83.
    1. Mialet Perez J, Rathz DA, Petrashevskaya NN , et al. Beta 1-adrenergic receptor polymorphisms confer differential function and pre-disposition to heart failure. Nat Med. 2003;9:1300–5.
    1. Liggett SB, Mialet-Perez J, Thaneemit-Chen S , et al. A polymorphism within a conserved beta(1)-adrenergic receptor motif alters cardiac function and beta-blocker response in human heart failure. Proc Natl Acad Sci USA. 2006;103:11288–93.
    1. Cresci S, Kelly RJ, Cappola TP , et al. Clinical and genetic modifiers of long-term survival in heart failure. J Am Coll Cardiol. 2009;54:432–44.
    1. White HL, de Boer RA, Maqbool A , et al. An evaluation of the beta-1 adrenergic receptor Arg389Gly polymorphism in individuals with heart failure a MERIT-HF sub-study. Eur J Heart Fail. 2003;5:463–8.
    1. Lobmeyer MT, Gong Y, Terra SG , et al. Synergistic polymorphisms of beta1 and alpha2C-adrenergic receptors and the influence on left ventricular ejection fraction response to beta-blocker therapy in heart failure. Pharmacogenet Genomics. 2007;17:277–82.
    1. Bristow MR, Murphy GA, Krause-Steinrauf H , et al. An alpha2C-adrenergic receptor polymorphism alters the norepinephrine-lowering effects and therapeutic response of the beta-blocker bucindolol in chronic heart failure. Circ Heart Fail. 2010;3:21–8.
    1. Liggett SB, Wagoner LE, Craft LL , et al. The Ile164 beta2-adrenergic receptor polymorphism adversely affects the outcome of con-gestive heart failure. J Clin Invest. 1998;102:1534–9.
    1. Turki J, Lorenz JN, Green SA , et al. Myocardial signaling defects and impaired cardiac function of a human beta 2-adrenergic recep-tor polymorphism expressed in transgenic mice. Proc Natl Acad Sci USA. 1996;93:10483–8.
    1. Brodde OE, Buscher R, Tellkamp R , et al. Blunted cardiac responses to receptor activation in subjects with Thr164Ile beta(2)-adrenoceptors. Circulation. 2001;103:1048–50.
    1. Wang WC, Mihlbachler KA, Bleecker ER , et al. A polymorphism of G-protein coupled receptor kinase5 alters agonist-promoted desensitization of beta2-adrenergic receptors. Pharmacogenet Genomics. 2008;18:729–32.
    1. Liggett SB, Cresci S, Kelly RJ , et al. A GRK5 polymorphism that inhibits beta-adrenergic receptor signaling is protective in heart failure. Nat Med. 2008;14:510–7.
    1. Lobmeyer MT, Wang L, Zineh I , et al. Polymorphisms in genes coding for GRK2 and GRK5 and response differences in antihyper-tensive-treated patients. Pharmacogenet Genomics. 2001;21:42–9.
    1. Rigat B, Hubert C, Alhenc-Gelas F , et al. An insertion/deletion polymorphism in the angiotensin I-converting enzyme gene account-ing for half the variance of serum enzyme levels. J Clin Invest. 1990;86:1343–6.
    1. Huang W, Xie C, Zhou H , et al. Association of the angiotensin-converting enzyme gene polymorphism with chronic heart failure in Chinese Han patients. Eur J Heart Fail. 2004;6:23–7.
    1. Palmer BR, Pilbrow AP, Yandle TG , et al. Angiotensin-converting enzyme gene polymorphism interacts with left ventricular ejection fraction and brain natriuretic peptide levels to predict mortality after myocardial infarction. J Am Coll Cardiol. 2003;41:729–36.
    1. Bai Y, Wang L, Hu S, Wei Y. Association of angiotensin-converting enzyme I/D polymorphism with heart failure a meta-analysis. Mol Cell Biochem. 2012;361:297–304.
    1. Iwai N, Ohmichi N, Nakamura Y, Kinoshita M. DD genotype of the angiotensin-converting enzyme gene is a risk factor for left ventricular hypertrophy. Circulation. 1994;90:2622–8.
    1. Schunkert H, Hense HW, Holmer SR , et al. Association between a deletion polymorphism of the angiotensin-converting-enzyme gene and left ventricular hypertrophy. N Engl J Med. 1994;330:1634–8.
    1. Andersson B, Sylven C. The DD genotype of the angiotensin-converting enzyme gene is associated with increased mortality in idio-pathic heart failure. J Am Coll Cardiol. 1996;28:162–7.
    1. McNamara DM, Holubkov R, Postava L , et al. Pharmacogenetic interactions between angiotensin-converting enzyme inhibitor thera-py and the angiotensin-converting enzyme deletion polymorphism in patients with congestive heart failure. J Am Coll Cardiol. 2004;44:2019–26.
    1. Wu CK, Luo JL, Tsai CT , et al. Demonstrating the pharmacogenetic effects of angiotensin-converting enzyme inhibitors on long-term prognosis of diastolic heart failure. Pharmacogenomics J. 2010;10:46–53.
    1. Cappola TP, Li M, He J , et al. Common variants in HSPB7 and FRMD4B associated with advanced heart failure. Circ Cardiovasc Genet. 2010;3:147–54.
    1. Smith NL, Felix JF, Morrison AC , et al. Association of genome-wide variation with the risk of incident heart failure in adults of European and African ancestry a prospective meta-analysis from the cohorts for heart and aging research in genomic epidemiology (CHARGE) consortium. Circ Cardiovasc Genet. 2010;3:256–66.
    1. Callis TE, Jensen BC, Weck KE, Willis MS. Evolving molecular diagnostics for familial cardiomyopathies at the heart of it all. Expert Rev Mol Diagn. 2010;10:329–51.
    1. McNally EM, Golbus Jr, Puckelwartz MJ. Genetic mutations and mechanisms in dilated cardiomyopathy. J Clin Invest. 2013;123:19–26.
    1. Herman DS, Lam L, Taylor MR , et al. Truncations of titin causing dilated cardiomyopathy. N Engl J Med. 2012;366:619–28.
    1. Hershberger RE, Siegfried JD. Update 2011: clinical and genetic issues in familial dilated cardiomyopathy. J Am Coll Cardiol. 2011;57:1641–9.
    1. Van Rijsingen IA, Arbustini E, Elliott PM , et al. Risk factors for malignant ventricular arrhythmias in lamin a/c mutation carriers a European cohort study. J Am Coll Cardiol. 2012;59:493–500.
    1. Hershberger RE, Lindenfeld J, Mestroni L , et al. Genetic evaluation of cardiomyopathy--a Heart Failure Society of America practice guideline. J Card Fail. 2009;15:83–97.
    1. Lakdawala NK, Funke BH, Baxter S , et al. Genetic testing for dilated cardiomyopathy in clinical practice. J Card Fail. 2012;18:296–303.
    1. Hershberger RE, Cowan J, Morales A, Siegfried JD. Progress with genetic cardiomyopathies screening, counseling, and testing in dilated, hypertrophic, and arrhythmogenic right ventricular dysplasia/cardiomyopathy. Circ Heart Fail. 2009;2:253–61.
    1. Judge DP. Use of genetics in the clinical evaluation of cardiomyopathy. JAMA. 2009;302:2471–6.
    1. Task F, Resta R, Biesecker BB , et al. National Society of Genetic Counselors' Definition A new definition of Genetic Counseling National Society of Genetic Counselors' Task Force report. J Genet Couns. 2006;15:77–83.
    1. Teekakirikul P, Kelly MA, Rehm HL , et al. Inherited cardiomyopathies molecular genetics and clinical genetic testing in the post-genomic era. J Mol Diagn. 2013;15:158–70.
    1. Patterson C. A good idea a physician's perspective on genetic counseling for hypertrophic cardiomyopathy. J Cardiovasc Transl Res. 2009;2:508–9.
    1. Velagaleti RS, O'Donnell CJ. Genomics of heart failure. Heart Fail Clin. 2010;6:115–24.

Source: PubMed

3
Abonner