Cargando…

Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis

Adipose-derived mesenchymal stem cells (ASCs) represent a valid therapeutic option for clinical application in several diseases, due to their ability to repair damaged tissues and to mitigate the inflammatory/immune response. A better understanding of the underlying mechanisms regulating ASC biology...

Descripción completa

Detalles Bibliográficos
Autores principales: Ceccarelli, S., Gerini, G., Megiorni, F., Pontecorvi, P., Messina, E., Camero, S., Anastasiadou, E., Romano, E., Onesti, M. G., Napoli, C., Marchese, C.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9478209/
https://www.ncbi.nlm.nih.gov/pubmed/36120582
http://dx.doi.org/10.3389/fcell.2022.926180
_version_ 1784790517884649472
author Ceccarelli, S.
Gerini, G.
Megiorni, F.
Pontecorvi, P.
Messina, E.
Camero, S.
Anastasiadou, E.
Romano, E.
Onesti, M. G.
Napoli, C.
Marchese, C.
author_facet Ceccarelli, S.
Gerini, G.
Megiorni, F.
Pontecorvi, P.
Messina, E.
Camero, S.
Anastasiadou, E.
Romano, E.
Onesti, M. G.
Napoli, C.
Marchese, C.
author_sort Ceccarelli, S.
collection PubMed
description Adipose-derived mesenchymal stem cells (ASCs) represent a valid therapeutic option for clinical application in several diseases, due to their ability to repair damaged tissues and to mitigate the inflammatory/immune response. A better understanding of the underlying mechanisms regulating ASC biology might represent the chance to modulate their in vitro characteristics and differentiation potential for regenerative medicine purposes. Herein, we investigated the effects of the demethylating agent 5-azacytidine (5-aza) on proliferation, clonogenicity, migration, adipogenic differentiation and senescence of ASCs, to identify the molecular pathways involved. Through functional assays, we observed a detrimental effect of 5-aza on ASC self-renewal capacity and migration, accompanied by actin cytoskeleton reorganization, with decreased stress fibers. Conversely, 5-aza treatment enhanced ASC adipogenic differentiation, as assessed by lipid accumulation and expression of lineage-specific markers. We analyzed the involvement of the Akt/mTOR, MAPK and Wnt/β-catenin pathways in these processes. Our results indicated impairment of Akt and ERK phosphorylation, potentially explaining the reduced cell proliferation and migration. We observed a 5-aza-mediated inhibition of the Wnt signaling pathway, this potentially explaining the pro-adipogenic effect of the drug. Finally, 5-aza treatment significantly induced ASC senescence, through upregulation of the p53/p21 axis. Our data may have important translational implications, by helping in clarifying the potential risks and advantages of using epigenetic treatment to improve ASC characteristics for cell-based clinical approaches.
format Online
Article
Text
id pubmed-9478209
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-94782092022-09-17 Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis Ceccarelli, S. Gerini, G. Megiorni, F. Pontecorvi, P. Messina, E. Camero, S. Anastasiadou, E. Romano, E. Onesti, M. G. Napoli, C. Marchese, C. Front Cell Dev Biol Cell and Developmental Biology Adipose-derived mesenchymal stem cells (ASCs) represent a valid therapeutic option for clinical application in several diseases, due to their ability to repair damaged tissues and to mitigate the inflammatory/immune response. A better understanding of the underlying mechanisms regulating ASC biology might represent the chance to modulate their in vitro characteristics and differentiation potential for regenerative medicine purposes. Herein, we investigated the effects of the demethylating agent 5-azacytidine (5-aza) on proliferation, clonogenicity, migration, adipogenic differentiation and senescence of ASCs, to identify the molecular pathways involved. Through functional assays, we observed a detrimental effect of 5-aza on ASC self-renewal capacity and migration, accompanied by actin cytoskeleton reorganization, with decreased stress fibers. Conversely, 5-aza treatment enhanced ASC adipogenic differentiation, as assessed by lipid accumulation and expression of lineage-specific markers. We analyzed the involvement of the Akt/mTOR, MAPK and Wnt/β-catenin pathways in these processes. Our results indicated impairment of Akt and ERK phosphorylation, potentially explaining the reduced cell proliferation and migration. We observed a 5-aza-mediated inhibition of the Wnt signaling pathway, this potentially explaining the pro-adipogenic effect of the drug. Finally, 5-aza treatment significantly induced ASC senescence, through upregulation of the p53/p21 axis. Our data may have important translational implications, by helping in clarifying the potential risks and advantages of using epigenetic treatment to improve ASC characteristics for cell-based clinical approaches. Frontiers Media S.A. 2022-09-02 /pmc/articles/PMC9478209/ /pubmed/36120582 http://dx.doi.org/10.3389/fcell.2022.926180 Text en Copyright © 2022 Ceccarelli, Gerini, Megiorni, Pontecorvi, Messina, Camero, Anastasiadou, Romano, Onesti, Napoli and Marchese. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Cell and Developmental Biology
Ceccarelli, S.
Gerini, G.
Megiorni, F.
Pontecorvi, P.
Messina, E.
Camero, S.
Anastasiadou, E.
Romano, E.
Onesti, M. G.
Napoli, C.
Marchese, C.
Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis
title Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis
title_full Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis
title_fullStr Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis
title_full_unstemmed Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis
title_short Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis
title_sort inhibiting dna methylation as a strategy to enhance adipose-derived stem cells differentiation: focus on the role of akt/mtor and wnt/β-catenin pathways on adipogenesis
topic Cell and Developmental Biology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9478209/
https://www.ncbi.nlm.nih.gov/pubmed/36120582
http://dx.doi.org/10.3389/fcell.2022.926180
work_keys_str_mv AT ceccarellis inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT gerinig inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT megiornif inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT pontecorvip inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT messinae inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT cameros inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT anastasiadoue inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT romanoe inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT onestimg inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT napolic inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis
AT marchesec inhibitingdnamethylationasastrategytoenhanceadiposederivedstemcellsdifferentiationfocusontheroleofaktmtorandwntbcateninpathwaysonadipogenesis