Modeling susceptibility to drug-induced long QT with a panel of subject-specific induced pluripotent stem cells

Francesca Stillitano, Jens Hansen, Chi-Wing Kong, Ioannis Karakikes, Christian Funck-Brentano, Lin Geng, Stuart Scott, Stephan Reynier, Ma Wu, Yannick Valogne, Carole Desseaux, Joe-Elie Salem, Dorota Jeziorowska, Noël Zahr, Ronald Li, Ravi Iyengar, Roger J Hajjar, Jean-Sébastien Hulot, Francesca Stillitano, Jens Hansen, Chi-Wing Kong, Ioannis Karakikes, Christian Funck-Brentano, Lin Geng, Stuart Scott, Stephan Reynier, Ma Wu, Yannick Valogne, Carole Desseaux, Joe-Elie Salem, Dorota Jeziorowska, Noël Zahr, Ronald Li, Ravi Iyengar, Roger J Hajjar, Jean-Sébastien Hulot

Abstract

A large number of drugs can induce prolongation of cardiac repolarization and life-threatening cardiac arrhythmias. The prediction of this side effect is however challenging as it usually develops in some genetically predisposed individuals with normal cardiac repolarization at baseline. Here, we describe a platform based on a genetically diverse panel of induced pluripotent stem cells (iPSCs) that reproduces susceptibility to develop a cardiotoxic drug response. We generated iPSC-derived cardiomyocytes from patients presenting in vivo with extremely low or high changes in cardiac repolarization in response to a pharmacological challenge with sotalol. In vitro, the responses to sotalol were highly variable but strongly correlated to the inter-individual differences observed in vivo. Transcriptomic profiling identified dysregulation of genes (DLG2, KCNE4, PTRF, HTR2C, CAMKV) involved in downstream regulation of cardiac repolarization machinery as underlying high sensitivity to sotalol. Our findings offer novel insights for the development of iPSC-based screening assays for testing individual drug reactions.

Trial registration: ClinicalTrials.gov NCT01338441.

Keywords: arrhythmia; cardiotoxicity; human; human biology; induced pluripotent stem cells; medicine.

Conflict of interest statement

SR: Current or former employee of the Cellectis Company. MW: Current or former employee of the Cellectis Company. YV: Current or former employee of the Cellectis Company. CD: Current or former employee of the Cellectis Company. The other authors declare that no competing interests exist.

Figures

Figure 1.. QTcf changes following Sotalol administration…
Figure 1.. QTcf changes following Sotalol administration in healthy volunteers.
(A) Flow chart of the clinical study. (B) Distribution of QTcf duration before (blue) and 3 hr after sotalol intake (red). (C) Distribution of delta change in QTcf showing the wide inter-individual variability in response to the same pharmacological stimulation. Subjects with the most extreme responses were selected as low sensitive (low-S) or high- sensitive (high-S) as indicated in green and red respectively. (D) Average delta change in QTcf in the two groups of selected subjects. ***p<0.001. (E) Typical ECG recordings before and after sotalol intake in a high-S subject (upper panels) and in a low-S subject (lower panels). There is one figure supplement. DOI:http://dx.doi.org/10.7554/eLife.19406.003
Figure 1—figure supplement 1.. Overview of the…
Figure 1—figure supplement 1.. Overview of the clinical and experimental study and role of each partner.
This translational study started with a prospective clinical study performed in a clinical investigation center near Paris, France. The objective was to identify healthy subjects with extreme responses to Sotalol 80 mg. Skin punch biopsies were performed in a total of twenty subjects, 10 with low-sensitivity and 10 with high-sensitivity to Sotalol. The fresh skin biopsies were then transferred at the Ectycell company (Romainville, France) in close vicinity of the clinical investigation center. The objective was to culture and bank human dermal fibroblasts and to derive and characterize hiPSC. iPSC clones were then anonymized before any further cardiac experiments were performed. This procedure was agreed by all partners to avoid potential biases in interpreting the recording at the cellular level. iPSCs clones were successfully generated for 17 subjects using retroviral infection and were investigated in this study. For the remaining three subjects (Patients 4, 14 and 20), iPSCs clones were secondarily generated using episomal plasmids and were thus not investigated. The 17 iPSCs clones were then transferred to the Cardiovascular Research Center at Icahn School of Medicine, New York, USA. The objective was to differentiate iPSCs into cardiac myocytes and to perform MEA recordings and drug testing. All required authorizations were obtained. DOI:http://dx.doi.org/10.7554/eLife.19406.004
Figure 2.. Expression of sarcomeric proteins and…
Figure 2.. Expression of sarcomeric proteins and ion channels in human iPSC-CMs.
(A) Confocal microscopy imaging of Troponin T (top), alpha-actinin and connexin 43 (bottom) in single generated iPSC-CMs (from line P11015). Nuclei are stained with DAPI (Blue). (B) Gene expression of cardiac ion channel KCNH2 (encoding hERG) by quantitative PCR; Adult LV tissue is used as a positive control and the level of expression in human ESC-derived cardiomyocytes as a comparator. (C) Example of monolayer of iPSC-CMs seeded and attached on a 6-well MEA chip, each well containing nine microelectrodes (black). See also Video 1. (D) Representative field potential duration (FPD) recorded before and after application of the hERG blocker E4031 (from line P11007). There are five figure supplements. DOI:http://dx.doi.org/10.7554/eLife.19406.007
Figure 2—figure supplement 1.. Expression of exogenous…
Figure 2—figure supplement 1.. Expression of exogenous and endogenous pluripotency genes.
Representative gel showing silencing of the four exogenous reprogramming transgenes (OCT3/4; KLF4; SOX2 ; c-MYC) and quantitative PCR results for endogenous pluripotent stem cell genes (OCT3/4; NANOG; SOX2) in the generated hiPSCs. DOI:http://dx.doi.org/10.7554/eLife.19406.010
Figure 2—figure supplement 2.. MEA arrays.
Figure 2—figure supplement 2.. MEA arrays.
(A) Picture of a 6-well MEA array as used in the study. Each well contains nine micro-electrodes and a limited volume capacity allowing easier contact between cells and micro-electrodes after re-seeding and rapid response to drugs after application in a limited volume of medium. (B) Typical micro-electrodes map of the six well array. (C) Dose-curve responses to the hERG blocker E4031. Increasing concentrations of E4031 were applied as described in Materials and methods section. Individual data were plotted and a hill equation was fitted allowing estimates of EC50 for each cell lines. Results were obtained in 15 cell lines as two cell lines display no changes in FPD in response to E4031, suggesting expression of a non-functional hERG in these iPSC-CMs. For graphical purposes, cell lines from the low-S group are shown in green and from the high-S group in red. DOI:http://dx.doi.org/10.7554/eLife.19406.011
Figure 2—figure supplement 3.. Quality of FPD…
Figure 2—figure supplement 3.. Quality of FPD adjustment.
(A) Plot of adjusted FPD using the Bazett’s formula against the individual inter-beat interval. This plot shows the miss-correction achieved using the Bazett’s formula with a significant dependence of FPD to beating rate even after adjustment. (B) Plot of adjusted FPD against the individual inter-beat interval after re-estimation of the correction factor to the data set. The plot shows perfect correction with linear regression indicating the lack of influence of the beating rate on the adjusted FPD. DOI:http://dx.doi.org/10.7554/eLife.19406.012
Figure 3.. Differences in iPSC-CMs responses to…
Figure 3.. Differences in iPSC-CMs responses to Sotalol stimulation according to clinical sensitivity to Sotalol.
(A) Adjusted FPD (aFPD) measured in iPSC-CMs derived from subjects with low-sensitivity (green) vs. high-sensitivity (red) in response to increasing concentrations of sotalol. aFPD are normalized to baseline values to account for inter-lines variability in aFPD values. Two-way analysis of variance demonstrates a significant influence of sotalol concentrations (p<0.0005) and of the sensitivity group (p<0.02). *p<0.05 for post-hoc comparison between groups; high-S vs. low-S. N = 2–5 recordings per cell lines per concentrations. (B) Maximal change in aFPD observed during sotalol stimulation. (C) Proportion of observed arrhythmias after sotalol application. (D) Data plot graph showing the correlation between aFPD observed in iPSC-CMs and the DeltaQTcf observed in donors. The aFPD data are reported for sotalol 30 μM concentration. Data points are clustered in two distinct groups. Except for two lines (one line in each group), a threshold a 25% in aFPD change (dashed line) correctly discriminates cells from both groups. DOI:http://dx.doi.org/10.7554/eLife.19406.015
Figure 4.. Patch-clamp analysis of action potential…
Figure 4.. Patch-clamp analysis of action potential (AP) in representative iPSC-CMs.
(A) Representative AP tracings of the iPSC-CMs generated from the low-S (P11009) and high-S (P11029) lines in control and sotalol-treated conditions. (B) bar chart summarizing the APD90 in control and sotalol-treated conditions for the hiPSC-CMs generated from both low-S (P11009) and high-S (P11029) hiPSC cell lines (n = 5 for each condition). *p<0.05; **p<0.01, ANOVA, followed by Tukey's, sotalol-treated versus respective control without sotalol application. DOI:http://dx.doi.org/10.7554/eLife.19406.016
Figure 5.. Identification of dysregulated genes as…
Figure 5.. Identification of dysregulated genes as direct neighbors of QT-associated network in high-S iPSC-CMs.
(A) Known LQTS genes were used as seed nodes (green squares) in the human interactome and five differentially expressed direct neighbors were identified (circles) (path length 1). Up-regulated genes are colored orange, down-regulated genes are in blue. (B) Relative expression of identified genes in each group. Males are represented in blue and females in red. Individual samples are represented by the same symbols in all diagrams. (C) Comparison of the log2-fold changes between the high-S and the low-S groups with the normalized counts of how often a gene (blue or orange dots) was found to be up- or down-regulated between the male and female groups. As there are more females in the high-S group and more males in the low-S groups, report of genes in the lower left or upper right quadrants indicate a gender-specific effect while the lower right and upper left quadrants argue for the lack of gender-specific effect. Except for HTR2C, dysregulation of all other candidate genes was suspected to occur independently of gender. There is one figure supplement. DOI:http://dx.doi.org/10.7554/eLife.19406.018
Figure 5—figure supplement 1.. Prediction of sex…
Figure 5—figure supplement 1.. Prediction of sex hormones-related transcription factors.
Predicted regulatory transcription factors of the identified DEGs (DLG2, KCNE4, CAMKV, PTRF, HTR2C) based on two transcription factor target databases (Chea background and Transfac) and ranked by significance. The sex-hormone related transcription factors (in orange) were ranked 16 (AR), 20 (ESR1) and 24 (ESR2) or 27 (PGR) and 180 (ESR1) while the top 10 candidates were independent of sex hormones influence (in blue). DOI:http://dx.doi.org/10.7554/eLife.19406.019
Author response image 1.
Author response image 1.
DOI:http://dx.doi.org/10.7554/eLife.19406.021

References

    1. Behr ER, Roden D. Drug-induced arrhythmia: pharmacogenomic prescribing? European Heart Journal. 2013;34:89–95. doi: 10.1093/eurheartj/ehs351.
    1. Berger SI, Ma'ayan A, Iyengar R. Systems pharmacology of arrhythmias. Science Signaling. 2010;3:ra30. doi: 10.1126/scisignal.2000723.
    1. Budnitz DS, Pollock DA, Weidenbach KN, Mendelsohn AB, Schroeder TJ, Annest JL. National surveillance of emergency department visits for outpatient adverse drug events. JAMA. 2006;296:1858–1866. doi: 10.1001/jama.296.15.1858.
    1. Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD, Lan F, Diecke S, Huber B, Mordwinkin NM, Plews JR, Abilez OJ, Cui B, Gold JD, Wu JC. Chemically defined generation of human cardiomyocytes. Nature Methods. 2014;11:855–860. doi: 10.1038/nmeth.2999.
    1. Cerrone M, Priori SG. Genetics of sudden death: focus on inherited channelopathies. European Heart Journal. 2011;32:2109–2118. doi: 10.1093/eurheartj/ehr082.
    1. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, Clark NR, Ma'ayan A. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013;14:128. doi: 10.1186/1471-2105-14-128.
    1. Chen EY, Xu H, Gordonov S, Lim MP, Perkins MH, Ma'ayan A. Expression2Kinases: mRNA profiling linked to multiple upstream regulatory layers. Bioinformatics. 2012;28:105–111. doi: 10.1093/bioinformatics/btr625.
    1. Cirulli ET, Goldstein DB. Uncovering the roles of rare variants in common disease through whole-genome sequencing. Nature Reviews Genetics. 2010;11:415–425. doi: 10.1038/nrg2779.
    1. Crotti L, Johnson CN, Graf E, De Ferrari GM, Cuneo BF, Ovadia M, Papagiannis J, Feldkamp MD, Rathi SG, Kunic JD, Pedrazzini M, Wieland T, Lichtner P, Beckmann BM, Clark T, Shaffer C, Benson DW, Kääb S, Meitinger T, Strom TM, Chazin WJ, Schwartz PJ, George AL. Calmodulin mutations associated with recurrent cardiac arrest in infants. Circulation. 2013;127:1009–1017. doi: 10.1161/CIRCULATIONAHA.112.001216.
    1. Darpo B, Karnad DR, Badilini F, Florian J, Garnett CE, Kothari S, Panicker GK, Sarapa N. Are women more susceptible than men to drug-induced QT prolongation? Concentration-QTc modelling in a phase 1 study with oral rac-sotalol. British Journal of Clinical Pharmacology. 2014;77:522–531. doi: 10.1111/bcp.12201.
    1. Fermini B, Fossa AA. The impact of drug-induced QT interval prolongation on drug discovery and development. Nature Reviews Drug Discovery. 2003;2:439–447. doi: 10.1038/nrd1108.
    1. Funck-Brentano C, Jaillon P. Rate-corrected QT interval: techniques and limitations. The American Journal of Cardiology. 1993;72:17B–22. doi: 10.1016/0002-9149(93)90035-B.
    1. Gintant G. An evaluation of hERG current assay performance: Translating preclinical safety studies to clinical QT prolongation. Pharmacology & Therapeutics. 2011;129:109–119. doi: 10.1016/j.pharmthera.2010.08.008.
    1. Giorgi MA, Bolaños R, Gonzalez CD, Di Girolamo G. QT interval prolongation: preclinical and clinical testing arrhythmogenesis in drugs and regulatory implications. Current Drug Safety. 2010;5:54–57. doi: 10.2174/157488610789869148.
    1. Hulot JS, Démolis JL, Rivière R, Strabach S, Christin-Maitre S, Funck-Brentano C. Influence of endogenous oestrogens on QT interval duration. European Heart Journal. 2003;24:1663–1667. doi: 10.1016/S0195-668X(03)00436-6.
    1. Inoue H, Yamanaka S. The use of induced pluripotent stem cells in drug development. Clinical Pharmacology & Therapeutics. 2011;89:655–661. doi: 10.1038/clpt.2011.38.
    1. Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, Feldman O, Gepstein A, Arbel G, Hammerman H, Boulos M, Gepstein L. Modelling the long QT syndrome with induced pluripotent stem cells. Nature. 2011;471:225–229. doi: 10.1038/nature09747.
    1. Kannankeril P, Roden DM, Darbar D. Drug-induced long QT syndrome. Pharmacological Reviews. 2010;62:760–781. doi: 10.1124/pr.110.003723.
    1. Kannankeril PJ, Roden DM, Norris KJ, Whalen SP, George AL, Murray KT. Genetic susceptibility to acquired long QT syndrome: pharmacologic challenge in first-degree relatives. Heart Rhythm. 2005;2:134–140. doi: 10.1016/j.hrthm.2004.10.039.
    1. Karakikes I, Senyei GD, Hansen J, Kong CW, Azeloglu EU, Stillitano F, Lieu DK, Wang J, Ren L, Hulot JS, Iyengar R, Li RA, Hajjar RJ. Small molecule-mediated directed differentiation of human embryonic stem cells toward ventricular cardiomyocytes. Stem Cells Translational Medicine. 2014;3:18–31. doi: 10.5966/sctm.2013-0110.
    1. Keller GA, Di Girolamo G. Prokinetic agents and QT prolongation: a familiar scene with new actors. Current Drug Safety. 2010;5:73–78. doi: 10.2174/157488610789869166.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nature Methods. 2012;9:357–359. doi: 10.1038/nmeth.1923.
    1. Leonoudakis D, Conti LR, Anderson S, Radeke CM, McGuire LM, Adams ME, Froehner SC, Yates JR, Vandenberg CA. Protein trafficking and anchoring complexes revealed by proteomic analysis of inward rectifier potassium channel (Kir2.x)-associated proteins. Journal of Biological Chemistry. 2004;279:22331–22346. doi: 10.1074/jbc.M400285200.
    1. Levy DI, Cepaitis E, Wanderling S, Toth PT, Archer SL, Goldstein SA. The membrane protein MiRP3 regulates Kv4.2 channels in a KChIP-dependent manner. The Journal of Physiology. 2010;588:2657–2668. doi: 10.1113/jphysiol.2010.191395.
    1. Liang P, Lan F, Lee AS, Gong T, Sanchez-Freire V, Wang Y, Diecke S, Sallam K, Knowles JW, Wang PJ, Nguyen PK, Bers DM, Robbins RC, Wu JC. Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation. 2013;127:1677–1691. doi: 10.1161/CIRCULATIONAHA.113.001883.
    1. Liu Z, Liu X, Yu H, Pei J, Zhang Y, Gong J, Pu J. Common Variants in TRDN and CALM1 Are Associated with Risk of Sudden Cardiac Death in Chronic Heart Failure Patients in Chinese Han Population. PLOS One. 2015;10:e0132459. doi: 10.1371/journal.pone.0132459.
    1. Lundy SD, Zhu WZ, Regnier M, Laflamme MA. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells and Development. 2013;22:1991–2002. doi: 10.1089/scd.2012.0490.
    1. Maguy A, Hebert TE, Nattel S. Involvement of lipid rafts and caveolae in cardiac ion channel function. Cardiovascular Research. 2006;69:798–807. doi: 10.1016/j.cardiores.2005.11.013.
    1. Makkar RR. Female Gender as a Risk Factor for Torsades de Pointes Associated With Cardiovascular Drugs. JAMA. 1993;270:2590–2597. doi: 10.1001/jama.1993.03510210076031.
    1. Mann DA. Human induced pluripotent stem cell-derived hepatocytes for toxicology testing. Expert Opinion on Drug Metabolism & Toxicology. 2015;11:1–5. doi: 10.1517/17425255.2015.981523.
    1. McIntyre LM, Lopiano KK, Morse AM, Amin V, Oberg AL, Young LJ, Nuzhdin SV. RNA-seq: technical variability and sampling. BMC Genomics. 2011;12:293. doi: 10.1186/1471-2164-12-293.
    1. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flügel L, Dorn T, Goedel A, Höhnke C, Hofmann F, Seyfarth M, Sinnecker D, Schömig A, Laugwitz KL. Patient-specific induced pluripotent stem-cell models for long-QT syndrome. New England Journal of Medicine. 2010;363:1397–1409. doi: 10.1056/NEJMoa0908679.
    1. Moss AJ, Robinson J. Clinical features of the idiopathic long QT syndrome. Circulation. 1992;85:I140–144.
    1. Nabi IR. Cavin fever: regulating caveolae. Nature Cell Biology. 2009;11:789–791. doi: 10.1038/ncb0709-789.
    1. Navarrete EG, Liang P, Lan F, Sanchez-Freire V, Simmons C, Gong T, Sharma A, Burridge PW, Patlolla B, Lee AS, Wu H, Beygui RE, Wu SM, Robbins RC, Bers DM, Wu JC. Screening drug-induced arrhythmia [corrected] using human induced pluripotent stem cell-derived cardiomyocytes and low-impedance microelectrode arrays. Circulation. 2013;128:S3–S13. doi: 10.1161/CIRCULATIONAHA.112.000570.
    1. Nozaki Y, Honda Y, Tsujimoto S, Watanabe H, Kunimatsu T, Funabashi H. Availability of human induced pluripotent stem cell-derived cardiomyocytes in assessment of drug potential for QT prolongation. Toxicology and Applied Pharmacology. 2014;278:72–77. doi: 10.1016/j.taap.2014.04.007.
    1. Odening KE, Koren G. How do sex hormones modify arrhythmogenesis in long QT syndrome? Sex hormone effects on arrhythmogenic substrate and triggered activity. Heart Rhythm. 2014;11:2107–2115. doi: 10.1016/j.hrthm.2014.06.023.
    1. Paulussen AD, Gilissen RA, Armstrong M, Doevendans PA, Verhasselt P, Smeets HJ, Schulze-Bahr E, Haverkamp W, Breithardt G, Cohen N, Aerssens J. Genetic variations of KCNQ1, KCNH2, SCN5A, KCNE1, and KCNE2 in drug-induced long QT syndrome patients. Journal of Molecular Medicine. 2004;82:182–188. doi: 10.1007/s00109-003-0522-z.
    1. Petropoulou E, Jamshidi Y, Behr ER. The genetics of pro-arrhythmic adverse drug reactions. British Journal of Clinical Pharmacology. 2014;77:618–625. doi: 10.1111/bcp.12208.
    1. Pfeufer A, Sanna S, Arking DE, Müller M, Gateva V, Fuchsberger C, Ehret GB, Orrú M, Pattaro C, Köttgen A, Perz S, Usala G, Barbalic M, Li M, Pütz B, Scuteri A, Prineas RJ, Sinner MF, Gieger C, Najjar SS, Kao WH, Mühleisen TW, Dei M, Happle C, Möhlenkamp S, Crisponi L, Erbel R, Jöckel KH, Naitza S, Steinbeck G, Marroni F, Hicks AA, Lakatta E, Müller-Myhsok B, Pramstaller PP, Wichmann HE, Schlessinger D, Boerwinkle E, Meitinger T, Uda M, Coresh J, Kääb S, Abecasis GR, Chakravarti A. Common variants at ten loci modulate the QT interval duration in the QTSCD Study. Nature Genetics. 2009;41:407–414. doi: 10.1038/ng.362.
    1. Rajab A, Straub V, McCann LJ, Seelow D, Varon R, Barresi R, Schulze A, Lucke B, Lützkendorf S, Karbasiyan M, Bachmann S, Spuler S, Schuelke M. Fatal cardiac arrhythmia and long-QT syndrome in a new form of congenital generalized lipodystrophy with muscle rippling (CGL4) due to PTRF-CAVIN mutations. PLoS Genetics. 2010;6:e1000874. doi: 10.1371/journal.pgen.1000874.
    1. Roden DM. Long QT syndrome: reduced repolarization reserve and the genetic link. Journal of Internal Medicine. 2006;259:59–69. doi: 10.1111/j.1365-2796.2005.01589.x.
    1. Roden DM. Cellular basis of drug-induced torsades de pointes. British Journal of Pharmacology. 2008a;154:1502–1507. doi: 10.1038/bjp.2008.238.
    1. Roden DM. Repolarization reserve: a moving target. Circulation. 2008b;118:981–982. doi: 10.1161/CIRCULATIONAHA.108.798918.
    1. Rolland T, Taşan M, Charloteaux B, Pevzner SJ, Zhong Q, Sahni N, Yi S, Lemmens I, Fontanillo C, Mosca R, Kamburov A, Ghiassian SD, Yang X, Ghamsari L, Balcha D, Begg BE, Braun P, Brehme M, Broly MP, Carvunis AR, Convery-Zupan D, Corominas R, Coulombe-Huntington J, Dann E, Dreze M, Dricot A, Fan C, Franzosa E, Gebreab F, Gutierrez BJ, Hardy MF, Jin M, Kang S, Kiros R, Lin GN, Luck K, MacWilliams A, Menche J, Murray RR, Palagi A, Poulin MM, Rambout X, Rasla J, Reichert P, Romero V, Ruyssinck E, Sahalie JM, Scholz A, Shah AA, Sharma A, Shen Y, Spirohn K, Tam S, Tejeda AO, Trigg SA, Twizere JC, Vega K, Walsh J, Cusick ME, Xia Y, Barabási AL, Iakoucheva LM, Aloy P, De Las Rivas J, Tavernier J, Calderwood MA, Hill DE, Hao T, Roth FP, Vidal M. A proteome-scale map of the human interactome network. Cell. 2014;159:1212–1226. doi: 10.1016/j.cell.2014.10.050.
    1. Sager PT, Gintant G, Turner JR, Pettit S, Stockbridge N. Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium. American Heart Journal. 2014;167:292–300. doi: 10.1016/j.ahj.2013.11.004.
    1. Sarganas G, Garbe E, Klimpel A, Hering RC, Bronder E, Haverkamp W. Epidemiology of symptomatic drug-induced long QT syndrome and Torsade de Pointes in Germany. Europace. 2014;16:101–108. doi: 10.1093/europace/eut214.
    1. Schaaf S, Shibamiya A, Mewe M, Eder A, Stöhr A, Hirt MN, Rau T, Zimmermann WH, Conradi L, Eschenhagen T, Hansen A. Human engineered heart tissue as a versatile tool in basic research and preclinical toxicology. PLoS One. 2011;6:e26397. doi: 10.1371/journal.pone.0026397.
    1. Sinnecker D, Goedel A, Dorn T, Dirschinger RJ, Moretti A, Laugwitz KL. Modeling long-QT syndromes with iPS cells. Journal of Cardiovascular Translational Research. 2013;6:31–36. doi: 10.1007/s12265-012-9416-1.
    1. Sinnecker D, Laugwitz KL, Moretti A. Induced pluripotent stem cell-derived cardiomyocytes for drug development and toxicity testing. Pharmacology & Therapeutics. 2014;143:246–252. doi: 10.1016/j.pharmthera.2014.03.004.
    1. Soyka LF, Wirtz C, Spangenberg RB. Clinical safety profile of sotalol in patients with arrhythmias. The American Journal of Cardiology. 1990;65:74A–81. doi: 10.1016/0002-9149(90)90207-H.
    1. Terrenoire C, Wang K, Tung KW, Chung WK, Pass RH, Lu JT, Jean JC, Omari A, Sampson KJ, Kotton DN, Keller G, Kass RS. Induced pluripotent stem cells used to reveal drug actions in a long QT syndrome family with complex genetics. The Journal of General Physiology. 2013;141:61–72. doi: 10.1085/jgp.201210899.
    1. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25:1105–1111. doi: 10.1093/bioinformatics/btp120.
    1. Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, Salzberg SL, Wold BJ, Pachter L. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nature Biotechnology. 2010;28:511–515. doi: 10.1038/nbt.1621.
    1. Turnbull IC, Karakikes I, Serrao GW, Backeris P, Lee JJ, Xie C, Senyei G, Gordon RE, Li RA, Akar FG, Hajjar RJ, Hulot JS, Costa KD. Advancing functional engineered cardiac tissues toward a preclinical model of human myocardium. The FASEB Journal. 2014;28:644–654. doi: 10.1096/fj.13-228007.
    1. Wang Y, Liang P, Lan F, Wu H, Lisowski L, Gu M, Hu S, Kay MA, Urnov FD, Shinnawi R, Gold JD, Gepstein L, Wu JC. Genome editing of isogenic human induced pluripotent stem cells recapitulates long QT phenotype for drug testing. Journal of the American College of Cardiology. 2014;64:451–459. doi: 10.1016/j.jacc.2014.04.057.
    1. Yang P, Kanki H, Drolet B, Yang T, Wei J, Viswanathan PC, Hohnloser SH, Shimizu W, Schwartz PJ, Stanton M, Murray KT, Norris K, George AL, Roden DM. Allelic variants in long-QT disease genes in patients with drug-associated torsades de pointes. Circulation. 2002;105:1943–1948. doi: 10.1161/01.CIR.0000014448.19052.4C.
    1. Zanetti LA. Sotalol: a new class III antiarrhythmic agent. Clinical Pharmacy. 1993;12:883–891.

Source: PubMed

3
구독하다