Your browser doesn't support javascript.
loading
Mitochondria-Rich Extracellular Vesicles From Autologous Stem Cell-Derived Cardiomyocytes Restore Energetics of Ischemic Myocardium.
Ikeda, Gentaro; Santoso, Michelle R; Tada, Yuko; Li, Albert M; Vaskova, Evgeniya; Jung, Ji-Hye; O'Brien, Connor; Egan, Elizabeth; Ye, Jiangbin; Yang, Phillip C.
Afiliação
  • Ikeda G; Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA. Electronic address: https://twitter.com/gentaro_ikeda.
  • Santoso MR; Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA. Electronic address: https://twitter.com/meeshsantoso.
  • Tada Y; Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.
  • Li AM; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA.
  • Vaskova E; Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.
  • Jung JH; Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.
  • O'Brien C; Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA.
  • Egan E; Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA.
  • Ye J; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA.
  • Yang PC; Stanford Cardiovascular Institute and Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA. Electronic address: phillip@stanford.edu.
J Am Coll Cardiol ; 77(8): 1073-1088, 2021 03 02.
Article em En | MEDLINE | ID: mdl-33632482
ABSTRACT

BACKGROUND:

Mitochondrial dysfunction results in an imbalance between energy supply and demand in a failing heart. An innovative therapy that targets the intracellular bioenergetics directly through mitochondria transfer may be necessary.

OBJECTIVES:

The purpose of this study was to establish a preclinical proof-of-concept that extracellular vesicle (EV)-mediated transfer of autologous mitochondria and their related energy source enhance cardiac function through restoration of myocardial bioenergetics.

METHODS:

Human-induced pluripotent stem cell-derived cardiomyocytes (iCMs) were employed. iCM-conditioned medium was ultracentrifuged to collect mitochondria-rich EVs (M-EVs). Therapeutic effects of M-EVs were investigated using in vivo murine myocardial infarction (MI) model.

RESULTS:

Electron microscopy revealed healthy-shaped mitochondria inside M-EVs. Confocal microscopy showed that M-EV-derived mitochondria were transferred into the recipient iCMs and fused with their endogenous mitochondrial networks. Treatment with 1.0 × 108/ml M-EVs significantly restored the intracellular adenosine triphosphate production and improved contractile profiles of hypoxia-injured iCMs as early as 3 h after treatment. In contrast, isolated mitochondria that contained 300× more mitochondrial proteins than 1.0 × 108/ml M-EVs showed no effect after 24 h. M-EVs contained mitochondrial biogenesis-related messenger ribonucleic acids, including proliferator-activated receptor γ coactivator-1α, which on transfer activated mitochondrial biogenesis in the recipient iCMs at 24 h after treatment. Finally, intramyocardial injection of 1.0 × 108 M-EVs demonstrated significantly improved post-MI cardiac function through restoration of bioenergetics and mitochondrial biogenesis.

CONCLUSIONS:

M-EVs facilitated immediate transfer of their mitochondrial and nonmitochondrial cargos, contributing to improved intracellular energetics in vitro. Intramyocardial injection of M-EVs enhanced post-MI cardiac function in vivo. This therapy can be developed as a novel, precision therapeutic for mitochondria-related diseases including heart failure.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Traumatismo por Reperfusão Miocárdica / Miócitos Cardíacos / Células-Tronco Pluripotentes Induzidas / Vesículas Extracelulares / Mitocôndrias Limite: Animals / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Traumatismo por Reperfusão Miocárdica / Miócitos Cardíacos / Células-Tronco Pluripotentes Induzidas / Vesículas Extracelulares / Mitocôndrias Limite: Animals / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article