Recessive cardiac phenotypes in induced pluripotent stem cell models of Jervell and Lange-Nielsen syndrome: Disease mechanisms and pharmacological rescue

Significance There are few laboratory models that recapitulate human cardiac disease. Here, we created human cell models for Jervell and Lange-Nielsen syndrome (JLNS) in vitro, based on human induced pluripotent stem cells (hiPSCs). JLNS is one of the most severe disorders of heart rhythm and can cause sudden death in young patients. JLNS is inherited recessively and is caused by homozygous mutations in the slow component of the delayed rectifier potassium current, IKs. Cardiomyocytes (CMs) from two independent sets of patient-derived and engineered hiPSCs showed electrophysiological defects that reflect the severity of the condition in patients. Our work allowed better understanding of the mechanisms of recessive inheritance. Furthermore, JLNS-CMs showed increased sensitivity to proarrhythmic drugs, which could be rescued pharmacologically, demonstrating the potential of hiPSC-CMs in drug testing. Jervell and Lange-Nielsen syndrome (JLNS) is one of the most severe life-threatening cardiac arrhythmias. Patients display delayed cardiac repolarization, associated high risk of sudden death due to ventricular tachycardia, and congenital bilateral deafness. In contrast to the autosomal dominant forms of long QT syndrome, JLNS is a recessive trait, resulting from homozygous (or compound heterozygous) mutations in KCNQ1 or KCNE1. These genes encode the α and β subunits, respectively, of the ion channel conducting the slow component of the delayed rectifier K+ current, IKs. We used complementary approaches, reprogramming patient cells and genetic engineering, to generate human induced pluripotent stem cell (hiPSC) models of JLNS, covering splice site (c.478-2A>T) and missense (c.1781G>A) mutations, the two major classes of JLNS-causing defects in KCNQ1. Electrophysiological comparison of hiPSC-derived cardiomyocytes (CMs) from homozygous JLNS, heterozygous, and wild-type lines recapitulated the typical and severe features of JLNS, including pronounced action and field potential prolongation and severe reduction or absence of IKs. We show that this phenotype had distinct underlying molecular mechanisms in the two sets of cell lines: the previously unidentified c.478-2A>T mutation was amorphic and gave rise to a strictly recessive phenotype in JLNS-CMs, whereas the missense c.1781G>A lesion caused a gene dosage-dependent channel reduction at the cell membrane. Moreover, adrenergic stimulation caused action potential prolongation specifically in JLNS-CMs. Furthermore, sensitivity to proarrhythmic drugs was strongly enhanced in JLNS-CMs but could be pharmacologically corrected. Our data provide mechanistic insight into distinct classes of JLNS-causing mutations and demonstrate the potential of hiPSC-CMs in drug evaluation.

[1]  M. Keating,et al.  Molecular basis of the long-QT syndrome associated with deafness. , 1997, The New England journal of medicine.

[2]  P. Coumel,et al.  A novel mutation in the potassium channel gene KVLQT1 causes the Jervell and Lange-Nielsen cardioauditory syndrome , 1997, Nature Genetics.

[3]  H. R. Lu,et al.  Repolarization reserve determines drug responses in human pluripotent stem cell derived cardiomyocytes. , 2013, Stem Cell Research.

[4]  G. Gemme,et al.  [RARE CARDIAC ARRYTHMIAS OF THE PEDIATRIC AGE. II. SYNCOPAL ATTACKS DUE TO PAROXYSMAL VENTRICULAR FIBRILLATION. (PRESENTATION OF 1ST CASE IN ITALIAN PEDIATRIC LITERATURE)]. , 1963, La Clinica pediatrica.

[5]  U. Ravens,et al.  Activation of Human ether-a-go-go-Related Gene Potassium Channels by the Diphenylurea 1,3-Bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea (NS1643) , 2006, Molecular Pharmacology.

[6]  M. Carter,et al.  A Regulatory Mechanism That Detects Premature Nonsense Codons in T-cell Receptor Transcripts in Vivo Is Reversed by Protein Synthesis Inhibitors in Vitro* , 1995, The Journal of Biological Chemistry.

[7]  M Bitner-Glindzicz,et al.  Jervell and Lange-Nielsen syndrome: a Norwegian perspective. , 1999, American journal of medical genetics.

[8]  M. Pembrey,et al.  Mutational spectrum in the cardioauditory syndrome of Jervell and Lange-Nielsen , 2000, Human Genetics.

[9]  J. Saenen,et al.  Molecular aspects of the congenital and acquired Long QT Syndrome: clinical implications. , 2008, Journal of molecular and cellular cardiology.

[10]  G. Breithardt,et al.  KCNE1 mutations cause Jervell and Lange-Nielsen syndrome , 1997, Nature Genetics.

[11]  Salih Coşkun,et al.  Romano-Ward sendromunda KCNQ1 geninde bir duplikasyon mutasyonu , 2015 .

[12]  Joseph J. Babcock,et al.  Modulation of hERG potassium channel gating normalizes action potential duration prolonged by dysfunctional KCNQ1 potassium channel , 2012, Proceedings of the National Academy of Sciences.

[13]  B. Attali,et al.  Structural Basis of Slow Activation Gating in the Cardiac IKs Channel Complex , 2011, Cellular Physiology and Biochemistry.

[14]  A. Pfeufer,et al.  Long QT Syndrome–Associated Mutations in KCNQ1 and KCNE1 Subunits Disrupt Normal Endosomal Recycling of IKs Channels , 2008, Circulation research.

[15]  David A. Scott,et al.  Double Nicking by RNA-Guided CRISPR Cas9 for Enhanced Genome Editing Specificity , 2013, Cell.

[16]  M. Bitner-Glindzicz,et al.  A spectrum of functional effects for disease causing mutations in the Jervell and Lange-Nielsen syndrome. , 2001, Cardiovascular research.

[17]  Bernhard M. Schuldt,et al.  A bioinformatic assay for pluripotency in human cells , 2011, Nature Methods.

[18]  A J Moss,et al.  Spectrum of Mutations in Long-QT Syndrome Genes: KVLQT1, HERG, SCN5A, KCNE1, and KCNE2 , 2000, Circulation.

[19]  Karl-Ludwig Laugwitz,et al.  Patient-specific induced pluripotent stem-cell models for long-QT syndrome. , 2010, New England Journal of Medicine.

[20]  P. Lambiase,et al.  Cellular mechanisms underlying the increased disease severity seen for patients with long QT syndrome caused by compound mutations in KCNQ1. , 2014, The Biochemical journal.

[21]  Bba,et al.  Clinical Aspects of Type-1 Long-QT Syndrome by Location, Coding Type, and Biophysical Function of Mutations Involving the KCNQ1 Gene , 2007, Circulation.

[22]  S. Olesen,et al.  The KCNQ1 potassium channel: from gene to physiological function. , 2005, Physiology.

[23]  S. Priori,et al.  CaV1.2 Calcium Channel Dysfunction Causes a Multisystem Disorder Including Arrhythmia and Autism , 2004, Cell.

[24]  Ward Oc A NEW FAMILIAL CARDIAC SYNDROME IN CHILDREN. , 1964 .

[25]  Lauri Toivonen,et al.  The Jervell and Lange-Nielsen Syndrome: Natural History, Molecular Basis, and Clinical Outcome , 2006, Archives des maladies du coeur et des vaisseaux.

[26]  Oscar Casis,et al.  Mechanism of Action of a Novel Human ether-a-go-go-Related Gene Channel Activator , 2006, Molecular Pharmacology.

[27]  B Attali,et al.  A recessive C‐terminal Jervell and Lange‐Nielsen mutation of the KCNQ1 channel impairs subunit assembly , 2000, The EMBO journal.

[28]  H. Nakauchi,et al.  Development of Defective and Persistent Sendai Virus Vector , 2010, The Journal of Biological Chemistry.

[29]  Stanley Nattel,et al.  Innovative approaches to anti-arrhythmic drug therapy , 2006, Nature Reviews Drug Discovery.

[30]  Shinsuke Yuasa,et al.  Disease characterization using LQTS-specific induced pluripotent stem cells. , 2012, Cardiovascular research.

[31]  Simona Casini,et al.  Isogenic human pluripotent stem cell pairs reveal the role of a KCNH2 mutation in long-QT syndrome , 2013, The EMBO journal.

[32]  Andrew J. Wilson,et al.  Abnormal KCNQ1 trafficking influences disease pathogenesis in hereditary long QT syndromes (LQT1). , 2005, Cardiovascular research.

[33]  Teng Hong Tan,et al.  Modeling type 3 long QT syndrome with cardiomyocytes derived from patient-specific induced pluripotent stem cells. , 2013, International journal of cardiology.

[34]  L. Arbour,et al.  LQTS in Northern BC: homozygosity for KCNQ1 V205M presents with a more severe cardiac phenotype but with minimal impact on auditory function , 2014, Clinical genetics.

[35]  Fred H. Gage,et al.  Induced pluripotent stem cells: the new patient? , 2012, Nature Reviews Molecular Cell Biology.

[36]  W. A. Murray,et al.  Long QT syndrome during high-dose cisapride. , 1995, Archives of internal medicine.

[37]  M. Janse,et al.  Electrophysiological changes in heart failure and their relationship to arrhythmogenesis. , 2004, Cardiovascular research.

[38]  C. Mummery,et al.  Pluripotent stem cell models of human heart disease. , 2013, Cold Spring Harbor perspectives in medicine.

[39]  C. Mummery,et al.  Cardiomyocytes Derived From Pluripotent Stem Cells Recapitulate Electrophysiological Characteristics of an Overlap Syndrome of Cardiac Sodium Channel Disease , 2012, Circulation.

[40]  M. Franz,et al.  Targeted disruption of the Kcnq1 gene produces a mouse model of Jervell and Lange– Nielsen Syndrome , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[41]  Thomas Meitinger,et al.  Calmodulin Mutations Associated With Recurrent Cardiac Arrest in Infants , 2013, Circulation.

[42]  L. Maquat Nonsense-mediated mRNA decay: splicing, translation and mRNP dynamics , 2004, Nature Reviews Molecular Cell Biology.

[43]  A. Feinberg,et al.  Human KVLQT1 gene shows tissue-specific imprinting and encompasses Beckwith-Wiedemann syndrome chromosomal rearrangements , 1997, Nature Genetics.

[44]  P. Kowey,et al.  Increasing I(Ks) corrects abnormal repolarization in rabbit models of acquired LQT2 and ventricular hypertrophy. , 2002, American journal of physiology. Heart and circulatory physiology.

[45]  O. C. Ward A NEW FAMILIAL CARDIAC SYNDROME IN CHILDREN. , 1964, Journal of the Irish Medical Association.

[46]  Shinya Yamanaka,et al.  iPS cells: a game changer for future medicine , 2014, The EMBO journal.

[47]  A. Brown,et al.  A mechanism for the proarrhythmic effects of cisapride (Propulsid): high affinity blockade of the human cardiac potassium channel HERG , 1997, FEBS letters.

[48]  G. Breithardt,et al.  Life-threatening Arrhythmias Genotype-phenotype Correlation in the Long-qt Syndrome : Gene-specific Triggers for Genotype-phenotype Correlation in the Long-qt Syndrome Gene-specific Triggers for Life-threatening Arrhythmias , 2022 .

[49]  T. Seufferlein,et al.  Modelling Human Channelopathies Using Induced Pluripotent Stem Cells: A Comprehensive Review , 2013, Stem cells international.

[50]  A. Jervell,et al.  CONGENITAL DEAF‐MUTISM, FUNCTIONAL HEART DISEASE WITH PROLONGATION OF THE Q‐T INTERVAL, AND SUDDEN DEATH , 1999, American heart journal.

[51]  Donald M Bers,et al.  Screening Drug-Induced Arrhythmia Using Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes and Low-Impedance Microelectrode Arrays , 2013, Circulation.

[52]  A. Rivas,et al.  Inner Ear Abnormalities in a Kcnq1 (Kvlqt1) Knockout Mouse: A Model of Jervell and Lange-Nielsen Syndrome , 2005, Otology & neurotology : official publication of the American Otological Society, American Neurotology Society [and] European Academy of Otology and Neurotology.

[53]  Michael J Ackerman,et al.  Impact of genetics on the clinical management of channelopathies. , 2013, Journal of the American College of Cardiology.

[54]  Robert Passier,et al.  Prediction of drug-induced cardiotoxicity using human embryonic stem cell-derived cardiomyocytes. , 2010, Stem cell research.

[55]  A. Wilde,et al.  IKs in heart and hearing, the ear can do with less than the heart. , 2013, Circulation. Cardiovascular genetics.

[56]  Divya Rajamohan,et al.  Current status of drug screening and disease modelling in human pluripotent stem cells , 2012, BioEssays : news and reviews in molecular, cellular and developmental biology.