Your browser doesn't support javascript.
loading
Hypoxic extracellular vesicles from hiPSCs protect cardiomyocytes from oxidative damage by transferring antioxidant proteins and enhancing Akt/Erk/NRF2 signaling.
Bobis-Wozowicz, Sylwia; Paw, Milena; Sarna, Michal; Kedracka-Krok, Sylwia; Nit, Kinga; Blazowska, Natalia; Dobosz, Anna; Hammad, Ruba; Cathomen, Toni; Zuba-Surma, Ewa; Tyszka-Czochara, Malgorzata; Madeja, Zbigniew.
Affiliation
  • Bobis-Wozowicz S; Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland. sylwia.bobis@uj.edu.pl.
  • Paw M; Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland.
  • Sarna M; Faculty of Biochemistry, Biophysics and Biotechnology, Department of Biophysics, Jagiellonian University, Krakow, Poland.
  • Kedracka-Krok S; Faculty of Biochemistry, Biophysics and Biotechnology, Department of Physical Biochemistry, Jagiellonian University, Krakow, Poland.
  • Nit K; Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland.
  • Blazowska N; Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland.
  • Dobosz A; Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland.
  • Hammad R; Freiburg iPS Core Facility, Institute for Transfusion Medicine and Gene Therapy, Medical Center- University of Freiburg, Freiburg, Germany.
  • Cathomen T; Center for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany.
  • Zuba-Surma E; Freiburg iPS Core Facility, Institute for Transfusion Medicine and Gene Therapy, Medical Center- University of Freiburg, Freiburg, Germany.
  • Tyszka-Czochara M; Center for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany.
  • Madeja Z; Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biology, Jagiellonian University, Kraków, Poland.
Cell Commun Signal ; 22(1): 356, 2024 Jul 09.
Article in En | MEDLINE | ID: mdl-38982464
ABSTRACT

BACKGROUND:

Stem cell-derived extracellular vesicles (EVs) are an emerging class of therapeutics with excellent biocompatibility, bioactivity and pro-regenerative capacity. One of the potential targets for EV-based medicines are cardiovascular diseases (CVD). In this work we used EVs derived from human induced pluripotent stem cells (hiPSCs; hiPS-EVs) cultured under different oxygen concentrations (21, 5 and 3% O2) to dissect the molecular mechanisms responsible for cardioprotection.

METHODS:

EVs were isolated by ultrafiltration combined with size exclusion chromatography (UF + SEC), followed by characterization by nanoparticle tracking analysis, atomic force microscopy (AFM) and Western blot methods. Liquid chromatography and tandem mass spectrometry coupled with bioinformatic analyses were used to identify differentially enriched proteins in various oxygen conditions. We directly compared the cardioprotective effects of these EVs in an oxygen-glucose deprivation/reoxygenation (OGD/R) model of cardiomyocyte (CM) injury. Using advanced molecular biology, fluorescence microscopy, atomic force spectroscopy and bioinformatics techniques, we investigated intracellular signaling pathways involved in the regulation of cell survival, apoptosis and antioxidant response. The direct effect of EVs on NRF2-regulated signaling was evaluated in CMs following NRF2 inhibition with ML385.

RESULTS:

We demonstrate that hiPS-EVs derived from physiological hypoxia at 5% O2 (EV-H5) exert enhanced cytoprotective function towards damaged CMs compared to EVs derived from other tested oxygen conditions (normoxia; EV-N and hypoxia 3% O2; EV-H3). This resulted from higher phosphorylation rates of Akt kinase in the recipient cells after transfer, modulation of AMPK activity and reduced apoptosis. Furthermore, we provide direct evidence for improved calcium signaling and sustained contractility in CMs treated with EV-H5 using AFM measurements. Mechanistically, our mass spectrometry and bioinformatics analyses revealed differentially enriched proteins in EV-H5 associated with the antioxidant pathway regulated by NRF2. In this regard, EV-H5 increased the nuclear translocation of NRF2 protein and enhanced its transcription in CMs upon OGD/R. In contrast, inhibition of NRF2 with ML385 abolished the protective effect of EVs on CMs.

CONCLUSIONS:

In this work, we demonstrate a superior cardioprotective function of EV-H5 compared to EV-N and EV-H3. Such EVs were most effective in restoring redox balance in stressed CMs, preserving their contractile function and preventing cell death. Our data support the potential use of hiPS-EVs derived from physiological hypoxia, as cell-free therapeutics with regenerative properties for the treatment of cardiac diseases.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Signal Transduction / Myocytes, Cardiac / Proto-Oncogene Proteins c-akt / NF-E2-Related Factor 2 / Induced Pluripotent Stem Cells / Extracellular Vesicles / Antioxidants Limits: Animals / Humans Language: En Journal: Cell Commun Signal Year: 2024 Document type: Article Affiliation country: Poland

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Signal Transduction / Myocytes, Cardiac / Proto-Oncogene Proteins c-akt / NF-E2-Related Factor 2 / Induced Pluripotent Stem Cells / Extracellular Vesicles / Antioxidants Limits: Animals / Humans Language: En Journal: Cell Commun Signal Year: 2024 Document type: Article Affiliation country: Poland