Cargando…

Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells

Bone marrow mesenchymal stromal cells (BMSCs) are multipotent cells able to self-renew and differentiate, depending on the microenvironment, into adipocytes and osteoblasts. These cells have a limited number of replications and enter replicative senescence during in vitro expansion. The role of DNA...

Descripción completa

Detalles Bibliográficos
Autores principales: Giuliani, Angelica, Bacalini, Maria Giulia, Ramini, Deborah, Mensà, Emanuela, Giordani, Chiara, Xumerle, Luciano, Garagnani, Paolo, Olivieri, Fabiola, Procopio, Antonio Domenico, Rippo, Maria Rita, Sabbatinelli, Jacopo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9857206/
https://www.ncbi.nlm.nih.gov/pubmed/36672222
http://dx.doi.org/10.3390/cells12020285
_version_ 1784873814360850432
author Giuliani, Angelica
Bacalini, Maria Giulia
Ramini, Deborah
Mensà, Emanuela
Giordani, Chiara
Xumerle, Luciano
Garagnani, Paolo
Olivieri, Fabiola
Procopio, Antonio Domenico
Rippo, Maria Rita
Sabbatinelli, Jacopo
author_facet Giuliani, Angelica
Bacalini, Maria Giulia
Ramini, Deborah
Mensà, Emanuela
Giordani, Chiara
Xumerle, Luciano
Garagnani, Paolo
Olivieri, Fabiola
Procopio, Antonio Domenico
Rippo, Maria Rita
Sabbatinelli, Jacopo
author_sort Giuliani, Angelica
collection PubMed
description Bone marrow mesenchymal stromal cells (BMSCs) are multipotent cells able to self-renew and differentiate, depending on the microenvironment, into adipocytes and osteoblasts. These cells have a limited number of replications and enter replicative senescence during in vitro expansion. The role of DNA methylation (DNAm) assumes importance in cell function and commitment; however, its exact contribution to BMSC differentiation and replicative senescence is still unclear. We performed a genome-wide DNAm analysis on BMSCs cultured in vitro at early passages and induced to differentiate into adipocytes and osteoblasts, and on replicative senescent BMSCs and HUVECs, to identify DNAm patterns of senescence and differentiation. We also compared BMSCs and HUVECs in replicative senescence and found that, in both cellular systems, genome-wide hypomethylation was accompanied by a higher-than-expected overlap of differentially methylated positions (DMPs) and concordance in terms of direction of the change. A Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis on lineage-independent senescence-associated DMPs revealed 16 common pathways, including Insulin resistance, Molecule adhesion, and Wnt/β-catenin signaling. In both adipogenesis and osteogenesis, we observed a general demethylation of CpG sites compared with undifferentiated BMSCs with a higher number of DMPs in osteogenesis. KEGG analysis resulted in 30 pathways enriched in osteoblasts and only 2 in adipocytes when compared to undifferentiated cells. When comparing differentiated BMSCs with senescent ones, osteogenesis exhibited a greater overlap with senescence in terms of number of DMPs and direction of methylation change compared to adipogenesis. In conclusion, this study may be useful for future research on general mechanisms that occur in replicative senescence and furthermore to identify trajectories of BMSC differentiation and common aspects of differentiated and senescent cells.
format Online
Article
Text
id pubmed-9857206
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-98572062023-01-21 Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells Giuliani, Angelica Bacalini, Maria Giulia Ramini, Deborah Mensà, Emanuela Giordani, Chiara Xumerle, Luciano Garagnani, Paolo Olivieri, Fabiola Procopio, Antonio Domenico Rippo, Maria Rita Sabbatinelli, Jacopo Cells Article Bone marrow mesenchymal stromal cells (BMSCs) are multipotent cells able to self-renew and differentiate, depending on the microenvironment, into adipocytes and osteoblasts. These cells have a limited number of replications and enter replicative senescence during in vitro expansion. The role of DNA methylation (DNAm) assumes importance in cell function and commitment; however, its exact contribution to BMSC differentiation and replicative senescence is still unclear. We performed a genome-wide DNAm analysis on BMSCs cultured in vitro at early passages and induced to differentiate into adipocytes and osteoblasts, and on replicative senescent BMSCs and HUVECs, to identify DNAm patterns of senescence and differentiation. We also compared BMSCs and HUVECs in replicative senescence and found that, in both cellular systems, genome-wide hypomethylation was accompanied by a higher-than-expected overlap of differentially methylated positions (DMPs) and concordance in terms of direction of the change. A Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis on lineage-independent senescence-associated DMPs revealed 16 common pathways, including Insulin resistance, Molecule adhesion, and Wnt/β-catenin signaling. In both adipogenesis and osteogenesis, we observed a general demethylation of CpG sites compared with undifferentiated BMSCs with a higher number of DMPs in osteogenesis. KEGG analysis resulted in 30 pathways enriched in osteoblasts and only 2 in adipocytes when compared to undifferentiated cells. When comparing differentiated BMSCs with senescent ones, osteogenesis exhibited a greater overlap with senescence in terms of number of DMPs and direction of methylation change compared to adipogenesis. In conclusion, this study may be useful for future research on general mechanisms that occur in replicative senescence and furthermore to identify trajectories of BMSC differentiation and common aspects of differentiated and senescent cells. MDPI 2023-01-11 /pmc/articles/PMC9857206/ /pubmed/36672222 http://dx.doi.org/10.3390/cells12020285 Text en © 2023 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Giuliani, Angelica
Bacalini, Maria Giulia
Ramini, Deborah
Mensà, Emanuela
Giordani, Chiara
Xumerle, Luciano
Garagnani, Paolo
Olivieri, Fabiola
Procopio, Antonio Domenico
Rippo, Maria Rita
Sabbatinelli, Jacopo
Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells
title Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells
title_full Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells
title_fullStr Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells
title_full_unstemmed Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells
title_short Genome-Wide Methylation Changes Associated with Replicative Senescence and Differentiation in Endothelial and Bone Marrow Mesenchymal Stromal Cells
title_sort genome-wide methylation changes associated with replicative senescence and differentiation in endothelial and bone marrow mesenchymal stromal cells
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9857206/
https://www.ncbi.nlm.nih.gov/pubmed/36672222
http://dx.doi.org/10.3390/cells12020285
work_keys_str_mv AT giulianiangelica genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT bacalinimariagiulia genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT raminideborah genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT mensaemanuela genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT giordanichiara genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT xumerleluciano genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT garagnanipaolo genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT olivierifabiola genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT procopioantoniodomenico genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT rippomariarita genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells
AT sabbatinellijacopo genomewidemethylationchangesassociatedwithreplicativesenescenceanddifferentiationinendothelialandbonemarrowmesenchymalstromalcells