Mouse models of arrhythmogenic cardiovascular disease: challenges and opportunities.

Arrhythmogenic cardiovascular disease is associated with significant morbidity and mortality and, in spite of therapeutic advances, remains an enormous public health burden. The scope of this problem motivates efforts to delineate the molecular, cellular and systemic mechanisms underlying increased arrhythmia risk in inherited and acquired cardiac and systemic disease. The mouse is used increasingly in these efforts owing to the ease with which genetic strategies can be exploited and mechanisms can be probed. The question then arises whether the mouse has proven to be a useful model system to delineate arrhythmogenic cardiovascular disease mechanisms. Rather than trying to provide a definite answer, the goal here is to consider the issues that arise when using mouse models and to highlight the opportunities.

[1]  Ronald W. Alfa,et al.  Mouse model of Timothy syndrome recapitulates triad of autistic traits , 2011, Proceedings of the National Academy of Sciences.

[2]  A. Wilde,et al.  UvA-DARE ( Digital Academic Repository ) Variable Na ( v ) 1 . 5 Protein Expression from the Wild-Type Allele Correlates with the Penetrance of Cardiac Conduction Disease in the Scn 5 a ( + /-) Mouse Model , 2010 .

[3]  S. Chugh Early identification of risk factors for sudden cardiac death , 2010, Nature Reviews Cardiology.

[4]  J. Nerbonne,et al.  Stabilization of Kv4 protein by the accessory K+ channel interacting protein 2 (KChIP2) subunit is required for the generation of native myocardial fast transient outward K+ currents , 2013, The Journal of physiology.

[5]  E. Schulze-Bahr,et al.  Arrhythmia-associated cardiac Ca2+ cycling proteins and gene mutations , 2012, Wiener Medizinische Wochenschrift.

[6]  M. Papadakis,et al.  Sudden cardiac death in young athletes: practical challenges and diagnostic dilemmas. , 2013, Journal of the American College of Cardiology.

[7]  W. Shen,et al.  Atrial fibrillation , 2013, Journal of biomedical research.

[8]  J. Bigger,et al.  Sudden cardiac death in patients with diabetes , 2004, Current diabetes reports.

[9]  Mark E. Anderson,et al.  Sudden Cardiac Death Prediction and Prevention: Report From a National Heart, Lung, and Blood Institute and Heart Rhythm Society Workshop , 2010, Circulation.

[10]  E. Zaklyazminskaya,et al.  Cardiac channelopathies: genetic and molecular mechanisms. , 2013, Gene.

[11]  M. Lei,et al.  Pathophysiological Mechanisms of Sino-Atrial Dysfunction and Ventricular Conduction Disease Associated with SCN5A Deficiency: Insights from Mouse Models , 2012, Front. Physio..

[12]  Mehmet K. Aktas,et al.  Congenital Long and Short QT Syndromes , 2012, Cardiology.

[13]  J. Nerbonne,et al.  Distinct Cellular and Molecular Mechanisms Underlie Functional Remodeling of Repolarizing K+ Currents With Left Ventricular Hypertrophy , 2008, Circulation research.

[14]  Can Yuan,et al.  Cellular mechanisms of ventricular arrhythmias in a mouse model of Timothy syndrome (long QT syndrome 8). , 2014, Journal of molecular and cellular cardiology.

[15]  Elisabetta Cerbai,et al.  Role of potassium currents in cardiac arrhythmias. , 2008, Europace : European pacing, arrhythmias, and cardiac electrophysiology : journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology.

[16]  Sara Negri,et al.  Arrhythmogenesis in catecholaminergic polymorphic ventricular tachycardia: insights from a RyR2 R4496C knock-in mouse model. , 2006, Circulation research.

[17]  X. Wehrens,et al.  Animal models of arrhythmogenic cardiomyopathy , 2009, Disease Models & Mechanisms.

[18]  D. Christini,et al.  Targeted deletion of kcne2 impairs ventricular repolarization via disruption of IK,slow1 and Ito,f , 2008, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[19]  Michael D. Schneider,et al.  Conduction Slowing and Sudden Arrhythmic Death in Mice With Cardiac-Restricted Inactivation of Connexin43 , 2001, Circulation research.

[20]  S. Lehnart Understanding the physiology of heart failure through cellular and in vivo models‐towards targeting of complex mechanisms , 2013, Experimental physiology.

[21]  S. Priori,et al.  Inherited calcium channelopathies in the pathophysiology of arrhythmias , 2012, Nature Reviews Cardiology.

[22]  Burcu Duygu,et al.  Genetics and epigenetics of arrhythmia and heart failure , 2013, Front. Genet..

[23]  F. Charpentier,et al.  Mouse models of SCN5A-related cardiac arrhythmias. , 2008, Progress in biophysics and molecular biology.

[24]  Carlo Napolitano,et al.  Abnormal Propagation of Calcium Waves and Ultrastructural Remodeling in Recessive Catecholaminergic Polymorphic Ventricular Tachycardia , 2013, Circulation research.

[25]  J. Kalman,et al.  Update on the management of atrial fibrillation , 2013, The Medical journal of Australia.

[26]  J. Baizer,et al.  A Mouse Model of Timothy Syndrome: a Complex Autistic Disorder Resulting from a Point Mutation in Cav1.2. , 2012, North American journal of medicine & science.

[27]  G. Breithardt,et al.  Effect of pacing and mexiletine on dispersion of repolarisation and arrhythmias in DeltaKPQ SCN5A (long QT3) mice. , 2003, Cardiovascular research.

[28]  Linuo Zhou,et al.  Prevalence, incidence and risk factors of chronic heart failure in the type 2 diabetic population: systematic review. , 2009, Current diabetes reviews.

[29]  D. McEwen,et al.  Sodium channel Scn1b null mice exhibit prolonged QT and RR intervals. , 2007, Journal of molecular and cellular cardiology.

[30]  J. Nerbonne,et al.  Enhanced cardiac PI3Kα signalling mitigates arrhythmogenic electrical remodelling in pathological hypertrophy and heart failure. , 2012, Cardiovascular research.

[31]  K. Willecke,et al.  Deletion of the last five C-terminal amino acid residues of connexin43 leads to lethal ventricular arrhythmias in mice without affecting coupling via gap junction channels , 2013, Basic Research in Cardiology.

[32]  J. Epstein,et al.  Notch signaling regulates murine atrioventricular conduction and the formation of accessory pathways. , 2011, The Journal of clinical investigation.

[33]  李永军,et al.  Atrial Fibrillation , 1999 .

[34]  D. Escande,et al.  Cardiac channelopathies: from men to mice , 2004, Annals of medicine.

[35]  Jie Zhang,et al.  Contactin-2 Expression in the Cardiac Purkinje Fiber Network , 2010, Circulation. Arrhythmia and electrophysiology.

[36]  B. London,et al.  Cardiac Arrhythmias: From (Transgenic) Mice to Men , 2001, Journal of cardiovascular electrophysiology.

[37]  B. Gersh,et al.  Sudden cardiac death: epidemiology and risk factors , 2010, Nature Reviews Cardiology.

[38]  W. Catterall,et al.  Sudden unexpected death in a mouse model of Dravet syndrome. , 2013, The Journal of clinical investigation.

[39]  J. Nerbonne Studying cardiac arrhythmias in the mouse--a reasonable model for probing mechanisms? , 2004, Trends in cardiovascular medicine.

[40]  M. Faggioni,et al.  Calsequestrin 2 and arrhythmias. , 2012, American journal of physiology. Heart and circulatory physiology.

[41]  A. Buxton,et al.  Left Ventricular Ejection Fraction for Sudden Death Risk Stratification and Guiding Implantable Cardioverter-defibrillators Implantation , 2010, Journal of cardiovascular pharmacology.

[42]  J. Ross,et al.  A Defect in the Kv Channel-Interacting Protein 2 (KChIP2) Gene Leads to a Complete Loss of I to and Confers Susceptibility to Ventricular Tachycardia , 2001, Cell.

[43]  J. Epstein,et al.  Myocardial Notch Signaling Reprograms Cardiomyoctes to a Conduction‐Like Phenotype , 2012, Circulation.

[44]  Willem Flameng,et al.  Abrupt rate accelerations or premature beats cause life-threatening arrhythmias in mice with long-QT3 syndrome , 2001, Nature Medicine.

[45]  J. Nerbonne,et al.  Molecular physiology of cardiac repolarization. , 2005, Physiological reviews.

[46]  S. Demolombe,et al.  Early ion-channel remodeling and arrhythmias precede hypertrophy in a mouse model of complete atrioventricular block. , 2011, Journal of molecular and cellular cardiology.

[47]  D. Terentyev,et al.  A mutation in calsequestrin, CASQ2D307H, impairs Sarcoplasmic Reticulum Ca2+ handling and causes complex ventricular arrhythmias in mice. , 2007, Cardiovascular research.