An exosomal-carried short periostin isoform induces cardiomyocyte proliferation.

Details

Ressource 1Request a copy Under indefinite embargo.
UNIL restricted access
State: Public
Version: author
License: CC BY 4.0
Serval ID
serval:BIB_F3AC0F228F68
Type
Article: article from journal or magazin.
Collection
Publications
Institution
Title
An exosomal-carried short periostin isoform induces cardiomyocyte proliferation.
Journal
Theranostics
Author(s)
Balbi C., Milano G., Fertig T.E., Lazzarini E., Bolis S., Taniyama Y., Sanada F., Di Silvestre D., Mauri P., Gherghiceanu M., Lüscher T.F., Barile L., Vassalli G.
ISSN
1838-7640 (Electronic)
ISSN-L
1838-7640
Publication state
Published
Issued date
2021
Peer-reviewed
Oui
Volume
11
Number
12
Pages
5634-5649
Language
english
Notes
Publication types: Journal Article ; Research Support, Non-U.S. Gov't
Publication Status: epublish
Abstract
Although a small number of cardiomyocytes may reenter the cell cycle after injury, the adult mammalian heart is incapable of a robust cardiomyocyte proliferation. Periostin, a secreted extracellular matrix protein, has been implicated as a regulator of cardiomyocyte proliferation; however, this role remains controversial. Alternative splicing of the human periostin gene results in 6 isoforms lacking sequences between exons 17 and 21, in addition to full-length periostin. We previously showed that exosomes (Exo) secreted by human cardiac explant-derived progenitor cells (CPC) carried periostin. Here, we aimed to investigate their cell cycle activity. Methods: CPC were derived as the cellular outgrowth of ex vivo cultured cardiac atrial explants. Exo were purified from CPC conditioned medium using size exclusion chromatography. Exosomal periostin was analyzed by Western blotting using a pair of antibodies (one raised against aa 537-836, and one raised against amino acids mapping at exon 17 of human periostin), by ELISA, and by cryo-EM with immune-gold labeling. Cell cycle activity was assessed in neonatal rat cardiomyocytes, in human induced pluripotent stem cell (iPS)-derived cardiomyocytes, and in adult rat cardiomyocytes after myocardial infarction. The role of periostin in cell cycle activity was investigated by transfecting donor CPC with a siRNA against this protein. Results: Periostin expression in CPC-secreted exosomes was detected using the antibody raised against aa 537-836 of the human protein, but not with the exon 17-specific antibody, consistent with an isoform lacking exon 17. Periostin was visualized on vesicle surfaces by cryo-EM and immune-gold labeling. CPC-derived exosomes induced cell proliferation in neonatal rat cardiomyocytes both in vitro and in vivo, in human iPS-derived cardiomyocytes, and in adult rat cardiomyocytes after myocardial infarction. Exo promoted phosphorylation of focal adhesion kinase (FAK), actin polymerization, and nuclear translocation of Yes-associated protein (YAP) in cardiomyocytes. Knocking down of periostin or YAP, or blocking FAK phosphorylation with PF-573228 nullified Exo-induced proliferation. A truncated human periostin peptide (aa 22-669), but not recombinant human full-length periostin, mimicked the pro-proliferative activity of exosomes. Conclusions: Our results show, for the first time, that CPC-secreted exosomes promote cardiomyocyte cell cycle-reentry via a short periostin isoform expressed on their surfaces, whereas recombinant full-length periostin does not. These findings highlight isoform-specific roles of periostin in cardiomyocyte proliferation.
Keywords
Hippo pathway, cardiomyocyte, exosomes, extracellular vesicles, isoforms, periostin, proliferation
Pubmed
Web of science
Open Access
Yes
Create date
11/05/2021 9:44
Last modification date
12/01/2022 7:35
Usage data