Stem Cell Research & Therapy (Jan 2024)

Critical contribution of mitochondria in the development of cardiomyopathy linked to desmin mutation

  • Yeranuhi Hovhannisyan,
  • Zhenlin Li,
  • Domitille Callon,
  • Rodolphe Suspène,
  • Vivien Batoumeni,
  • Alexis Canette,
  • Jocelyne Blanc,
  • Hakim Hocini,
  • Cécile Lefebvre,
  • Nora El-Jahrani,
  • Maria Kitsara,
  • Aurore L’honoré,
  • Ekaterini Kordeli,
  • Paul Fornes,
  • Jean-Paul Concordet,
  • Gérard Tachdjian,
  • Anne-Marie Rodriguez,
  • Jean-Pierre Vartanian,
  • Anthony Béhin,
  • Karim Wahbi,
  • Pierre Joanne,
  • Onnik Agbulut

DOI
https://doi.org/10.1186/s13287-023-03619-7
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 23

Abstract

Read online

Abstract Background Beyond the observed alterations in cellular structure and mitochondria, the mechanisms linking rare genetic mutations to the development of heart failure in patients affected by desmin mutations remain unclear due in part, to the lack of relevant human cardiomyocyte models. Methods To shed light on the role of mitochondria in these mechanisms, we investigated cardiomyocytes derived from human induced pluripotent stem cells carrying the heterozygous DES E439K mutation that were either isolated from a patient or generated by gene editing. To increase physiological relevance, cardiomyocytes were either cultured on an anisotropic micropatterned surface to obtain elongated and aligned cardiomyocytes, or as a cardiac spheroid to create a micro-tissue. Moreover, when applicable, results from cardiomyocytes were confirmed with heart biopsies of suddenly died patient of the same family harboring DES E439K mutation, and post-mortem heart samples from five control healthy donors. Results The heterozygous DES E439K mutation leads to dramatic changes in the overall cytoarchitecture of cardiomyocytes, including cell size and morphology. Most importantly, mutant cardiomyocytes display altered mitochondrial architecture, mitochondrial respiratory capacity and metabolic activity reminiscent of defects observed in patient’s heart tissue. Finally, to challenge the pathological mechanism, we transferred normal mitochondria inside the mutant cardiomyocytes and demonstrated that this treatment was able to restore mitochondrial and contractile functions of cardiomyocytes. Conclusions This work highlights the deleterious effects of DES E439K mutation, demonstrates the crucial role of mitochondrial abnormalities in the pathophysiology of desmin-related cardiomyopathy, and opens up new potential therapeutic perspectives for this disease.

Keywords