Your browser doesn't support javascript.
loading
Transfer of cardiomyocyte-derived extracellular vesicles to neighboring cardiac cells requires tunneling nanotubes during heart development.
Chen, Ting; Ellman, Ditte Gry; Fang, Shu; Bak, Sara Thornby; Nørgård, Mikkel Ørnfeldt; Svenningsen, Per; Andersen, Ditte Caroline.
Affiliation
  • Chen T; Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark.
  • Ellman DG; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.
  • Fang S; Department of Urology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
  • Bak ST; Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark.
  • Nørgård MØ; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.
  • Svenningsen P; Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark.
  • Andersen DC; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.
Theranostics ; 14(10): 3843-3858, 2024.
Article in En | MEDLINE | ID: mdl-38994028
ABSTRACT
Rationale Extracellular vesicles (EVs) are thought to mediate intercellular communication during development and disease. Yet, biological insight to intercellular EV transfer remains elusive, also in the heart, and is technically challenging to demonstrate. Here, we aimed to investigate biological transfer of cardiomyocyte-derived EVs in the neonatal heart.

Methods:

We exploited CD9 as a marker of EVs, and generated two lines of cardiomyocyte specific EV reporter mice Tnnt2-Cre; double-floxed inverted CD9/EGFP and αMHC-MerCreMer; double-floxed inverted CD9/EGFP. The two mouse lines were utilized to determine whether developing cardiomyocytes transfer EVs to other cardiac cells (non-myocytes and cardiomyocytes) in vitro and in vivo and investigate the intercellular transport pathway of cardiomyocyte-derived EVs.

Results:

Genetic tagging of cardiomyocytes was confirmed in both reporter mouse lines and proof of concept in the postnatal heart showed that, a fraction of EGFP+/MYH1- non-myocytes exist firmly demonstrating in vivo cardiomyocyte-derived EV transfer. However, two sets of direct and indirect EGFP +/- cardiac cell co-cultures showed that cardiomyocyte-derived EGFP+ EV transfer requires cell-cell contact and that uptake of EGFP+ EVs from the medium is limited. The same was observed when co-cultiring with mouse macrophages. Further mechanistic insight showed that cardiomyocyte EV transfer occurs through type I tunneling nanotubes.

Conclusion:

While the current notion assumes that EVs are transferred through secretion to the surroundings, our data show that cardiomyocyte-derived EV transfer in the developing heart occurs through nanotubes between neighboring cells. Whether these data are fundamental and relate to adult hearts and other organs remains to be determined, but they imply that the normal developmental process of EV transfer goes through cell-cell contact rather than through the extracellular compartment.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cell Communication / Coculture Techniques / Myocytes, Cardiac / Extracellular Vesicles Limits: Animals Language: En Journal: Theranostics Year: 2024 Document type: Article Affiliation country: Denmark

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cell Communication / Coculture Techniques / Myocytes, Cardiac / Extracellular Vesicles Limits: Animals Language: En Journal: Theranostics Year: 2024 Document type: Article Affiliation country: Denmark