Extending Human Induced Pluripotent Stem Cell Technology to Infectious Diseases: New Model for Viral Myocarditis Daniel Sinnecker, Karl-Ludwig Laugwitz and Alessandra Moretti Circ Res. 2014;115:537-539 doi: 10.1161/CIRCRESAHA.114.304786 Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231 Copyright © 2014 American Heart Association, Inc. All rights reserved. Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the World Wide Web at: http://circres.ahajournals.org/content/115/6/537

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for which permission is being requested is located, click Request Permissions in the middle column of the Web page under Services. Further information about this process is available in the Permissions and Rights Question and Answer document. Reprints: Information about reprints can be found online at: http://www.lww.com/reprints Subscriptions: Information about subscribing to Circulation Research is online at: http://circres.ahajournals.org//subscriptions/

Downloaded from http://circres.ahajournals.org/ at Tulane University on September 1, 2014

Editorial Extending Human Induced Pluripotent Stem Cell Technology to Infectious Diseases New Model for Viral Myocarditis Daniel Sinnecker, Karl-Ludwig Laugwitz, Alessandra Moretti

I

nfections by viruses, such as adenoviruses, enteroviruses, or parvoviruses, can affect the heart, resulting in myocardial inflammation.1 Viral myocarditis encompasses a wide spectrum of clinical presentations, ranging from oligosymptomatic infections to the rapid development of terminal heart failure.

Article, see p 556 Clinical diagnosis of viral myocarditis remains a challenge. Although modern imaging modalities, such as late gadolinium enhancement MRI, can be used to estimate the likelihood of viral heart disease in suspected cases, the definite diagnosis still requires histopathologic examination of endomyocardial biopsies.2 Viral heart disease seems to account for a relevant proportion of otherwise unexplained dilated cardiomyopathies. In large series of patients with this diagnosis who underwent endomyocardial biopsy, myocarditis was found in ≈10% of cases.3,4 Because of the unavailability of specific noninvasive tests, the exact prevalence of viral myocarditis is unknown, and it is likely that many cases remain undiagnosed.5 One of the most prevalent and most intensively studied myocarditis-causing viruses is coxsackievirus B3, a member of the enterovirus genus of unenveloped singlestranded positive-sense RNA viruses. The entry of coxsackieviruses into cardiomyocytes depends on surface expression of the coxsackievirus and adenovirus receptor, a transmembrane protein with 2 extracellular immunoglobulin domains that is highly expressed on the cell membrane of cardiomyocytes.6 Upon replication in the host myocardium, a protein called enteroviral protease 2A is synthesized, which was demonstrated to cleave the sarcolemmal protein dystrophin specifically, disrupting the cytoskeletal integrity of the cardiomyocytes.7 Much of our knowledge of the pathomechanisms contributing to the onset and progression of human viral myocarditis is based on studies of experimental cellular The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association. From the I. Medical Department, Cardiology, Klinikum rechts der Isar—Technische Universität München, Munich, Germany (D.S., K.L.L., A.M.); and DZHK (German Centre for Cardiovascular Research)– Partner Site Munich Heart Alliance, Munich, Germany (K.-L.L., A.M.). Correspondence to Alessandra Moretti, PhD, Klinikum rechts der Isar, I. Medizinische Klinik und Poliklinik, Molecular Cardiology, Ismaninger Str. 22, 81675 München, Germany. E-mail [email protected] (Circ Res. 2014;115:537-539.) © 2014 American Heart Association, Inc. Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.114.304786

and murine models. Such models comprise immortalized, proliferative cell lines, such as HEK293T and HeLa cells, or genetically susceptible mice that are infected with coxsackievirus B3.8–10 In this issue of Circulation Research, Sharma et al11 report on a different approach at modeling viral myocarditis. They obtained either skin fibroblasts or mononuclear blood cells from healthy individuals and, by viral transduction with a specific cocktail of reprogramming factors, reprogrammed them to induced pluripotent stem cells (iPSCs). These iPSCs, which can be propagated in culture indefinitely, were subjected to cardiac differentiation to obtain human spontaneously beating cardiomyocytes (Figure [A]). The coxsackievirus and adenovirus receptor was present on the plasma membrane of iPSC-derived cardiomyocytes, albeit mRNA expression level was ≈30-fold lower than in primary human myocardial tissue. The iPSC-derived cardiomyocytes were then infected with a coxsackievirus B3 strain that was modified to induce expression of luciferase to monitor viral gene expression by luciferase luminescence indirectly. Within 6 hours of infection, the cardiomyocytes started to beat irregularly and displayed disrupted intracellular calcium transients. Moreover, they ceased to beat after ≈12 hours and detached from the culture dishes after 24 hours. When the number of virus particles applied to the cells was reduced, these cytopathic effects developed during a longer time frame. However, even low virus numbers eventually resulted in the death of all iPSC-derived cardiomyocytes. In addition, the authors used this system to investigate the effect of several drugs on virus replication as measured by luciferase luminescence. Interferon beta 1 (IFNβ1), ribavirin, fluoxetine, and pyrrolidine dithiocarbamate all reduced virus replication in iPSC-derived cardiomyocytes. The small-molecule compounds ribavirin and pyrrolidine dithiocarbamate, in contrast to IFNβ1, were also effective if administered concurrently with or even after infection with the virus. IFNβ1 was more effective if given 12 hours in advance, presumably because its mechanism of action involves the activation of transcription of downstream target genes. To elucidate the role of IFNβ1-mediated transcriptional regulation in its antiviral activity further, gene expression analysis was performed on coxsackievirus B3–infected iPSC-derived cardiomyocytes either pretreated with IFNβ1 or untreated. Among the 139 genes found to be differentially expressed, 22 genes were previously implicated in viral elimination pathways, suggesting that this approach might be suitable to identify additional genes with a role in the cellular defense against cardiac coxsackievirus infection.

Downloaded from http://circres.ahajournals.org/ 537 at Tulane University on September 1, 2014

538  Circulation Research  August 29, 2014

Figure.   A, Human skin fibroblasts and peripheral blood mononuclear cells (PBMCs) were converted into human induced pluripotent stem cells (hiPSCs) by viral-meditated reprogramming of the OKSM factors (OCT4, KLF1, SOX2, and cMYC). Differentiation of hiPSCs into the cardiovascular lineage allowed obtaining unlimited number of highly pure human cardiomyocytes. B, Infection of hiPSCderived cardiomyocytes with a luciferaseexpressing coxsackievirus B3 strain (CVB3-Luc) as a novel human system to model the pathogenic processes of coxsackievirus-induced myocarditis, screen antiviral therapeutics, and dissect drug mechanisms.

The study by Sharma et al11 extends the field of iPSC-based cardiovascular disease models to infectious diseases. To date, genetically caused channelopathies and cardiomyopathies have been the focus of this emerging technology.12 Although animal models are an important fundament of cardiovascular research, it is often desirable to investigate disease mechanisms or drug effects in human cells to exclude that species differences result in wrong conclusions. Primary human myocardial tissue can be obtained only by invasive procedures and cannot be propagated or kept in culture for a long time period. Cardiomyocytes derived from iPSC are one possible solution to these obstacles because they represent a theoretically unlimited source of human cardiomyocytes. In their study, Sharma et al11 have shown that, in the case of coxsackievirus B3–mediated myocarditis, this system can be used not only to model the disease in vitro but also as a platform to evaluate drugs that interfere with the disease, and—by gene expression analysis—to investigate the mechanism of drug action further (Figure [B]). In principle, iPSC technology would offer the possibility to investigate other important aspects of viral myocarditis. It is well known that the susceptibility to viral myocarditis is genetically determined.13 The progression of viral myocarditis to dilated cardiomyopathy may be also influenced by genetic susceptibility factors.14 By deriving iPSCs not only from healthy individuals but also from patients affected by viral myocarditis (assuming their susceptibility to the disease) and from patients who have developed dilated cardiomyopathy after viral myocarditis, it might be possible to elucidate the molecular mechanisms of this susceptibility further. Knowledge of the signal transduction pathways that confer resistance to virus-mediated myocardial damage might inform the development of specific drugs to prevent the potentially devastating consequences of this disease.

Nevertheless, it should be noted that the study by Sharma et al11 models only 1 aspect of viral myocarditis—the cellautonomous interaction between the cardiomyocytes and the virus particles. Earlier studies in experimental models have suggested 3 still controversial mechanisms by which viral myocarditis causes damage to the heart: (1) direct virus-induced cardiomyocyte injury; (2) destruction of the myocardium by infiltrating immune cells targeting virus-infected cardiomyocytes; (3) autoimmune-mediated destruction of cardiac cells by circulating autoantibodies and autoreactive immune cells.15 In its present state, the iPSC-derived experimental myocarditis model only allows investigation of the first aspect. It may be an advantage to be able to study the virus–cardiomyocyte interaction in an isolated fashion. However, to gain a complete picture of the pathogenesis of viral myocarditis, human disease models that also capture the other above-mentioned aspects of the disease will be necessary in the future.

Acknowledgments This work was supported by grants from the German Research Foundation (Research Unit 923, Mo 2217/1-1 to A. Moretti and La 1238 3-1/4-1 to K.-L. Laugwitz; Si 1747/1-1 to D. Sinnecker), the European Research Council (ERC 261053 to K.-L. Laugwitz), the DZHK (German Centre for Cardiovascular Research—partner site Munich Heart Alliance) and the Else Kröner-Fresenius-Stiftung.

Disclosures None.

References 1. Yajima T, Knowlton KU. Viral myocarditis: from the perspective of the virus. Circulation. 2009;119:2615–2624. 2. Caforio ALP, Pankuweit S, Arbustini E, et al. Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European Society of Cardiology working group on myocardial and pericardial diseases. Eur Heart J. 2013;34:2636–2648.

Downloaded from http://circres.ahajournals.org/ at Tulane University on September 1, 2014

Sinnecker et al   Modeling Viral Myocarditis With iPSCs   539 3. Mason JW, O’Connell JB, Herskowitz A, Rose NR, McManus BM, Billingham ME, Moon TE. A clinical trial of immunosuppressive therapy for myocarditis. The Myocarditis Treatment Trial Investigators. N Engl J Med. 1995;333:269–275. 4. Felker GM, Hu W, Hare JM, Hruban RH, Baughman KL, Kasper E. The spectrum of dilated cardiomyopathy. The Johns Hopkins experience with 1,278 patients. Medicine. 1999;78:270–283. 5. Schultheiss HP, Kühl U, Cooper LT. The management of myocarditis. Eur Heart J. 2011;32:2616–2625. 6. Shi Y, Chen C, Lisewski U, Wrackmeyer U, Radke M, Westermann D, Sauter M, Tschöpe C, Poller W, Klingel K, Gotthardt M. Cardiac deletion of the Coxsackievirus-adenovirus receptor abolishes Coxsackievirus B3 infection and prevents myocarditis in vivo. J Am Coll Cardiol. 2009;53:1219–1226. 7. Badorff C, Lee GH, Lamphear BJ, Martone ME, Campbell KP, Rhoads RE, Knowlton KU. Enteroviral protease 2A cleaves dystrophin: evidence of cytoskeletal disruption in an acquired cardiomyopathy. Nat Med. 1999;5:320–326. 8. Liu W, Dienz O, Roberts B, Moussawi M, Rincon M, Huber SA. IL-21R expression on CD8+ T cells promotes CD8+ T cell activation in coxsackievirus B3 induced myocarditis. Exp Mol Pathol. 2012;92:327–333. 9. Liu W, Moussawi M, Roberts B, Boyson JE, Huber SA. Cross-regulation of T regulatory-cell response after coxsackievirus B3 infection by NKT and gammadelta T cells in the mouse. Am J Pathol. 2013;183:441–449.

10. Mukherjee A, Morosky SA, Delorme-Axford E, Dybdahl-Sissoko N, Oberste MS, Wang T, Coyne CB. The coxsackievirus B 3C protease cleaves MAVS and TRIF to attenuate host type I interferon and apoptotic signaling. PLoS Pathog. 2011;7:e1001311. 11. Sharma A, Marceau C, Hamaguchi R, et al. Human induced pluripotent stem cell-derived cardiomyocytes as an in vitro model for coxsackievirus B3-induced myocarditis and antiviral drug screening platform. Circ Res. 2014;115:556–566. 12. Moretti A, Laugwitz KL, Dorn T, Sinnecker D, Mummery C. Pluripotent stem cell models of human heart disease. Cold Spring Harbor Perspect Med. 2013;3:a014027. 13. Aly M, Wiltshire S, Chahrour G, Osti JC, Vidal SM. Complex genetic control of host susceptibility to coxsackievirus B3-induced myocarditis. Genes Immun. 2007;8:193–204. 14. Gorbea C, Makar KA, Pauschinger M, Pratt G, Bersola JL, Varela J, David RM, Banks L, Huang CH, Li H, Schultheiss HP, Towbin JA, Vallejo JG, Bowles NE. A role for Toll-like receptor 3 variants in host susceptibility to enteroviral myocarditis and dilated cardiomyopathy. J Biol Chem. 2010;285:23208–23223. 15. Esfandiarei M, McManus BM. Molecular biology and pathogenesis of viral myocarditis. Annu Rev Pathol Mech Dis. 2008;3:127–155. Key Words: induced pluripotent stem cells



receptor, Coxsackievirus B3

Downloaded from http://circres.ahajournals.org/ at Tulane University on September 1, 2014

Extending human induced pluripotent stem cell technology to infectious diseases: new model for viral myocarditis.

Extending human induced pluripotent stem cell technology to infectious diseases: new model for viral myocarditis. - PDF Download Free
695KB Sizes 2 Downloads 8 Views