Your browser doesn't support javascript.
loading
Stem cell-derived cardiomyocytes expressing a dominant negative pacemaker HCN4 channel do not reduce the risk of graft-related arrhythmias.
Wulkan, Fanny; Romagnuolo, Rocco; Qiang, Beiping; Valdman Sadikov, Tamilla; Kim, Kyung-Phil; Quesnel, Elya; Jiang, Wenlei; Andharia, Naaz; Weyers, Jill J; Ghugre, Nilesh R; Ozcan, Bilgehan; Alibhai, Faisal J; Laflamme, Michael A.
Affiliation
  • Wulkan F; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
  • Romagnuolo R; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
  • Qiang B; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
  • Valdman Sadikov T; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
  • Kim KP; BlueRock Therapeutics, Toronto, ON, Canada.
  • Quesnel E; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
  • Jiang W; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
  • Andharia N; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
  • Weyers JJ; Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.
  • Ghugre NR; Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.
  • Ozcan B; Schulich Heart Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
  • Alibhai FJ; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
  • Laflamme MA; McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
Front Cardiovasc Med ; 11: 1374881, 2024.
Article in En | MEDLINE | ID: mdl-39045008
ABSTRACT

Background:

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) show tremendous promise for cardiac regeneration following myocardial infarction (MI), but their transplantation gives rise to transient ventricular tachycardia (VT) in large-animal MI models, representing a major hurdle to translation. Our group previously reported that these arrhythmias arise from a focal mechanism whereby graft tissue functions as an ectopic pacemaker; therefore, we hypothesized that hPSC-CMs engineered with a dominant negative form of the pacemaker ion channel HCN4 (dnHCN4) would exhibit reduced automaticity and arrhythmogenic risk following transplantation.

Methods:

We used CRISPR/Cas9-mediated gene-editing to create transgenic dnHCN4 hPSC-CMs, and their electrophysiological behavior was evaluated in vitro by patch-clamp recordings and optical mapping. Next, we transplanted WT and homozygous dnHCN4 hPSC-CMs in a pig MI model and compared post-transplantation outcomes including the incidence of spontaneous arrhythmias and graft structure by immunohistochemistry.

Results:

In vitro dnHCN4 hPSC-CMs exhibited significantly reduced automaticity and pacemaker funny current (I f ) density relative to wildtype (WT) cardiomyocytes. Following transplantation with either dnHCN4 or WT hPSC-CMs, all recipient hearts showed transmural infarct scar that was partially remuscularized by scattered islands of human myocardium. However, in contrast to our hypothesis, both dnHCN4 and WT hPSC-CM recipients exhibited frequent episodes of ventricular tachycardia (VT).

Conclusions:

While genetic silencing of the pacemaker ion channel HCN4 suppresses the automaticity of hPSC-CMs in vitro, this intervention is insufficient to reduce VT risk post-transplantation in the pig MI model, implying more complex mechanism(s) are operational in vivo.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Front Cardiovasc Med Year: 2024 Document type: Article Affiliation country: Canada Country of publication: Switzerland

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Front Cardiovasc Med Year: 2024 Document type: Article Affiliation country: Canada Country of publication: Switzerland