Your browser doesn't support javascript.
loading
Subtype-specific differentiation of cardiac pacemaker cell clusters from human induced pluripotent stem cells.
Schweizer, Patrick A; Darche, Fabrice F; Ullrich, Nina D; Geschwill, Pascal; Greber, Boris; Rivinius, Rasmus; Seyler, Claudia; Müller-Decker, Karin; Draguhn, Andreas; Utikal, Jochen; Koenen, Michael; Katus, Hugo A; Thomas, Dierk.
Afiliação
  • Schweizer PA; Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120, Heidelberg, Germany. patrick.schweizer@med.uni-heidelberg.de.
  • Darche FF; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, INF 410, D-69120, Heidelberg, Germany. patrick.schweizer@med.uni-heidelberg.de.
  • Ullrich ND; Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120, Heidelberg, Germany.
  • Geschwill P; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, INF 410, D-69120, Heidelberg, Germany.
  • Greber B; Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, Heidelberg University, INF 326, D-69120, Heidelberg, Germany.
  • Rivinius R; Institute of Physiology and Pathophysiology, Division of Neuro- and Sensory Physiology, Heidelberg University, INF 326, D-69120, Heidelberg, Germany.
  • Seyler C; Department of Cell and Developmental Biology, Max-Planck-Institute for Molecular Biomedicine, Röntgenstrasse, 20, D-48149, Münster, Germany.
  • Müller-Decker K; Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120, Heidelberg, Germany.
  • Draguhn A; Department of Cardiology, Medical University Hospital Heidelberg, INF 410, D-69120, Heidelberg, Germany.
  • Utikal J; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, INF 410, D-69120, Heidelberg, Germany.
  • Koenen M; Unit Tumor Models, German Cancer Research Center (DKFZ), Heidelberg, INF 280, D-69120, Heidelberg, Germany.
  • Katus HA; Institute of Physiology and Pathophysiology, Division of Neuro- and Sensory Physiology, Heidelberg University, INF 326, D-69120, Heidelberg, Germany.
  • Thomas D; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, INF 410, D-69120, Heidelberg, Germany.
Stem Cell Res Ther ; 8(1): 229, 2017 10 16.
Article em En | MEDLINE | ID: mdl-29037217
ABSTRACT

BACKGROUND:

Human induced pluripotent stem cells (hiPSC) harbor the potential to differentiate into diverse cardiac cell types. Previous experimental efforts were primarily directed at the generation of hiPSC-derived cells with ventricular cardiomyocyte characteristics. Aiming at a straightforward approach for pacemaker cell modeling and replacement, we sought to selectively differentiate cells with nodal-type properties.

METHODS:

hiPSC were differentiated into spontaneously beating clusters by co-culturing with visceral endoderm-like cells in a serum-free medium. Subsequent culturing in a specified fetal bovine serum (FBS)-enriched cell medium produced a pacemaker-type phenotype that was studied in detail using quantitative real-time polymerase chain reaction (qRT-PCR), immunocytochemistry, and patch-clamp electrophysiology. Further investigations comprised pharmacological stimulations and co-culturing with neonatal cardiomyocytes.

RESULTS:

hiPSC co-cultured in a serum-free medium with the visceral endoderm-like cell line END-2 produced spontaneously beating clusters after 10-12 days of culture. The pacemaker-specific genes HCN4, TBX3, and TBX18 were abundantly expressed at this early developmental stage, while levels of sarcomeric gene products remained low. We observed that working-type cardiomyogenic differentiation can be suppressed by transfer of early clusters into a FBS-enriched cell medium immediately after beating onset. After 6 weeks under these conditions, sinoatrial node (SAN) hallmark genes remained at high levels, while working-type myocardial transcripts (NKX2.5, TBX5) were low. Clusters were characterized by regular activity and robust beating rates (70-90 beats/min) and were triggered by spontaneous Ca2+ transients recapitulating calcium clock properties of genuine pacemaker cells. They were responsive to adrenergic/cholinergic stimulation and able to pace neonatal rat ventricular myocytes in co-culture experiments. Action potential (AP) measurements of cells individualized from clusters exhibited nodal-type (63.4%) and atrial-type (36.6%) AP morphologies, while ventricular AP configurations were not observed.

CONCLUSION:

We provide a novel culture media-based, transgene-free approach for targeted generation of hiPSC-derived pacemaker-type cells that grow in clusters and offer the potential for disease modeling, drug testing, and individualized cell-based replacement therapy of the SAN.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Relógios Biológicos / Diferenciação Celular / Miócitos Cardíacos / Células-Tronco Pluripotentes Induzidas Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Relógios Biológicos / Diferenciação Celular / Miócitos Cardíacos / Células-Tronco Pluripotentes Induzidas Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article