30
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Maximum Diastolic Potential of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Depends Critically on I Kr

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) hold promise for therapeutic applications. To serve these functions, the hiPSC-CM must recapitulate the electrophysiologic properties of native adult cardiomyocytes. This study examines the electrophysiologic characteristics of hiPSC-CM between 11 and 121 days of maturity. Embryoid bodies (EBs) were generated from hiPS cell line reprogrammed with Oct4, Nanog, Lin28 and Sox2. Sharp microelectrodes were used to record action potentials (AP) from spontaneously beating clusters (BC) micro-dissected from the EBs (n = 103; 37°C) and to examine the response to 5 µM E-4031 (n = 21) or BaCl 2 (n = 22). Patch-clamp techniques were used to record I Kr and I K1 from cells enzymatically dissociated from BC (n = 49; 36°C). Spontaneous cycle length (CL) and AP characteristics varied widely among the 103 preparations. E-4031 (5 µM; n = 21) increased Bazett-corrected AP duration from 291.8±81.2 to 426.4±120.2 msec (p<0.001) and generated early afterdepolarizations in 8/21 preparations. In 13/21 BC, E-4031 rapidly depolarized the clusters leading to inexcitability. BaCl 2, at concentrations that selectively block I K1 (50–100 µM), failed to depolarize the majority of clusters (13/22). Patch-clamp experiments revealed very low or negligible I K1 in 53% (20/38) of the cells studied, but presence of I Kr in all (11/11). Consistent with the electrophysiological data, RT-PCR and immunohistochemistry studies showed relatively poor mRNA and protein expression of I K1 in the majority of cells, but robust expression of I Kr. In contrast to recently reported studies, our data point to major deficiencies of hiPSC-CM, with remarkable diversity of electrophysiologic phenotypes as well as pharmacologic responsiveness among beating clusters and cells up to 121 days post-differentiation (dpd). The vast majority have a maximum diastolic potential that depends critically on I Kr due to the absence of I K1. Thus, efforts should be directed at producing more specialized and mature hiPSC-CM for future therapeutic applications.

          Related collections

          Most cited references15

          • Record: found
          • Abstract: found
          • Article: not found

          Mutations in Kir2.1 cause the developmental and episodic electrical phenotypes of Andersen's syndrome.

          Andersen's syndrome is characterized by periodic paralysis, cardiac arrhythmias, and dysmorphic features. We have mapped an Andersen's locus to chromosome 17q23 near the inward rectifying potassium channel gene KCNJ2. A missense mutation in KCNJ2 (encoding D71V) was identified in the linked family. Eight additional mutations were identified in unrelated patients. Expression of two of these mutations in Xenopus oocytes revealed loss of function and a dominant negative effect in Kir2.1 current as assayed by voltage-clamp. We conclude that mutations in Kir2.1 cause Andersen's syndrome. These findings suggest that Kir2.1 plays an important role in developmental signaling in addition to its previously recognized function in controlling cell excitability in skeletal muscle and heart.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Multipotent flk-1+ cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages.

            Cell-tracing studies in the mouse indicate that the cardiac lineage arises from a population that expresses the vascular endothelial growth factor receptor 2 (VEGFR2, Flk-1), suggesting that it may develop from a progenitor with vascular potential. Using the embryonic stem (ES) cell differentiation model, we have identified a cardiovascular progenitor based on the temporal expression of the primitive streak (PS) marker brachyury and Flk-1. Comparable progenitors could also be isolated from head-fold stage embryos. When cultured with cytokines known to function during cardiogenesis, individual cardiovascular progenitors generated colonies that displayed cardiomyocyte, endothelial, and vascular smooth muscle (VSM) potential. Isolation and characterization of this previously unidentified population suggests that the mammalian cardiovascular system develops from multipotential progenitors.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Cardiomyocyte differentiation of human induced pluripotent stem cells.

              The ability to derive human induced pluripotent stem (hiPS) cell lines by reprogramming of adult fibroblasts with a set of transcription factors offers unique opportunities for basic and translational cardiovascular research. In the present study, we aimed to characterize the cardiomyocyte differentiation potential of hiPS cells and to study the molecular, structural, and functional properties of the generated hiPS-derived cardiomyocytes. Cardiomyocyte differentiation of the hiPS cells was induced with the embryoid body differentiation system. Gene expression studies demonstrated that the cardiomyocyte differentiation process of the hiPS cells was characterized by an initial increase in mesoderm and cardiomesoderm markers, followed by expression of cardiac-specific transcription factors and finally by cardiac-specific structural genes. Cells in the contracting embryoid bodies were stained positively for cardiac troponin-I, sarcomeric alpha-actinin, and connexin-43. Reverse-transcription polymerase chain reaction studies demonstrated the expression of cardiac-specific sarcomeric proteins and ion channels. Multielectrode array recordings established the development of a functional syncytium with stable pacemaker activity and action potential propagation. Positive and negative chronotropic responses were induced by application of isoproterenol and carbamylcholine, respectively. Administration of quinidine, E4031 (I(Kr) blocker), and chromanol 293B (I(Ks) blocker) significantly affected repolarization, as manifested by prolongation of the local field potential duration. hiPS cells can differentiate into myocytes with cardiac-specific molecular, structural, and functional properties. These results, coupled with the potential of this technology to generate patient-specific hiPS lines, hold great promise for the development of in vitro models of cardiac genetic disorders, for drug discovery and testing, and for the emerging field of cardiovascular regenerative medicine.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS One
                PLoS ONE
                plos
                plosone
                PLoS ONE
                Public Library of Science (San Francisco, USA )
                1932-6203
                2012
                5 July 2012
                13 July 2012
                : 7
                : 7
                : e40288
                Affiliations
                [1 ]Stem Cell Research and Genomics, Masonic Medical Research Laboratory, Utica, New York, United States of America
                [2 ]Experimental Cardiology, Masonic Medical Research Laboratory, Utica, New York, United States of America
                [3 ]Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
                University of Milan, Italy
                Author notes

                Conceived and designed the experiments: MXD JMDD AS CA. Performed the experiments: MXD JMDD RG YW JMC VVN HBM DH JU MD JT. Analyzed the data: MXD JMDD JMC VVN HBM. Wrote the paper: MXD JMDD JMC VVN HBM AS CA. Conceived, designed strategies to improve the yield and quality of hiPS-CMs, performed molecular biology assays, supervised the project, and analyzed the immunohistochemical studies: MXD. Conceived, designed and performed the EP experiments on beating clusters and supervised the project: JMDD. Performed the experiments on beating clusters: RJG. Did RNA isolations and preparation of reagents: YW. Made the mathematical model: VVN. Performed the EP experiments on single cells: JMC HBM DH JU. Performed the immunohistochemical studies: MD. Prepared the embryoid bodies and beating clusters: JAT. Designed strategies to improve the yield and quality of hiPS-CMs: AS.

                Article
                PONE-D-12-03570
                10.1371/journal.pone.0040288
                3396384
                22815737
                96e0abfb-3777-43aa-95b4-d9302a26ce32
                Doss et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
                History
                : 3 February 2012
                : 4 June 2012
                Page count
                Pages: 17
                Categories
                Research Article
                Biology
                Biotechnology
                Developmental Biology
                Stem Cells
                Induced Pluripotent Stem Cells
                Immunology
                Immunologic Techniques
                Immunohistochemical Analysis
                Molecular Cell Biology
                Cellular Types
                Myocytes
                Medicine
                Anatomy and Physiology
                Cell Physiology
                Electrophysiology
                Integrative Physiology
                Cardiovascular
                Arrhythmias
                Electrophysiology

                Uncategorized
                Uncategorized

                Comments

                Comment on this article