Cargando…

Downregulation of IGFBP5 contributes to replicative senescence via ERK2 activation in mouse embryonic fibroblasts

Insulin-like growth factor (IGF)-binding proteins (IGFBPs) are secretory proteins that regulate IGF signaling. In this study, we investigated the role of IGFBP5 in replicative senescence in embryonic mouse fibroblasts (MEFs). During passages according to the 3T3 method, MEFs underwent senescence aft...

Descripción completa

Detalles Bibliográficos
Autores principales: Nojima, Iyori, Hosoda, Ryusuke, Toda, Yuki, Saito, Yoshiki, Ueda, Naohiro, Horimoto, Kouhei, Iwahara, Naotoshi, Horio, Yoshiyuki, Kuno, Atsushi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9037271/
https://www.ncbi.nlm.nih.gov/pubmed/35378512
http://dx.doi.org/10.18632/aging.203999
_version_ 1784693698638905344
author Nojima, Iyori
Hosoda, Ryusuke
Toda, Yuki
Saito, Yoshiki
Ueda, Naohiro
Horimoto, Kouhei
Iwahara, Naotoshi
Horio, Yoshiyuki
Kuno, Atsushi
author_facet Nojima, Iyori
Hosoda, Ryusuke
Toda, Yuki
Saito, Yoshiki
Ueda, Naohiro
Horimoto, Kouhei
Iwahara, Naotoshi
Horio, Yoshiyuki
Kuno, Atsushi
author_sort Nojima, Iyori
collection PubMed
description Insulin-like growth factor (IGF)-binding proteins (IGFBPs) are secretory proteins that regulate IGF signaling. In this study, we investigated the role of IGFBP5 in replicative senescence in embryonic mouse fibroblasts (MEFs). During passages according to the 3T3 method, MEFs underwent senescence after the 5th passage (P5) based on cell growth arrest, an increase in the number of cells positive for senescence-associated β-galactosidase (SA-β-GAL) staining, and upregulation of p16 and p19. In P8 MEFs, IGFBP5 mRNA level was markedly reduced compared with that in P2 MEFs. Downregulation of IGFBP5 via siRNA in P2 MEFs increased the number of SA-β-GAL-positive cells, upregulated p16 and p19, and inhibited cell growth. Incubation of MEFs with IGFBP5 during serial passage increased the cumulative population doubling and decreased SA-β-GAL positivity compared with those in vehicle-treated cells. IGFBP5 knockdown in P2 MEFs increased phosphorylation levels of ERK1 and ERK2. Silencing of ERK2, but not that of ERK1, blocked the increase in the number of SA-β-GAL-positive cells in IGFBP5-knockdown cells. The reduction in the cell number and upregulation of p16 and p21 in IGFBP5-knockdown cells were attenuated by ERK2 knockdown. Our results suggest that downregulation of IGFBP5 during serial passage contributes to replicative senescence via ERK2 in MEFs.
format Online
Article
Text
id pubmed-9037271
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Impact Journals
record_format MEDLINE/PubMed
spelling pubmed-90372712022-04-26 Downregulation of IGFBP5 contributes to replicative senescence via ERK2 activation in mouse embryonic fibroblasts Nojima, Iyori Hosoda, Ryusuke Toda, Yuki Saito, Yoshiki Ueda, Naohiro Horimoto, Kouhei Iwahara, Naotoshi Horio, Yoshiyuki Kuno, Atsushi Aging (Albany NY) Research Paper Insulin-like growth factor (IGF)-binding proteins (IGFBPs) are secretory proteins that regulate IGF signaling. In this study, we investigated the role of IGFBP5 in replicative senescence in embryonic mouse fibroblasts (MEFs). During passages according to the 3T3 method, MEFs underwent senescence after the 5th passage (P5) based on cell growth arrest, an increase in the number of cells positive for senescence-associated β-galactosidase (SA-β-GAL) staining, and upregulation of p16 and p19. In P8 MEFs, IGFBP5 mRNA level was markedly reduced compared with that in P2 MEFs. Downregulation of IGFBP5 via siRNA in P2 MEFs increased the number of SA-β-GAL-positive cells, upregulated p16 and p19, and inhibited cell growth. Incubation of MEFs with IGFBP5 during serial passage increased the cumulative population doubling and decreased SA-β-GAL positivity compared with those in vehicle-treated cells. IGFBP5 knockdown in P2 MEFs increased phosphorylation levels of ERK1 and ERK2. Silencing of ERK2, but not that of ERK1, blocked the increase in the number of SA-β-GAL-positive cells in IGFBP5-knockdown cells. The reduction in the cell number and upregulation of p16 and p21 in IGFBP5-knockdown cells were attenuated by ERK2 knockdown. Our results suggest that downregulation of IGFBP5 during serial passage contributes to replicative senescence via ERK2 in MEFs. Impact Journals 2022-04-04 /pmc/articles/PMC9037271/ /pubmed/35378512 http://dx.doi.org/10.18632/aging.203999 Text en Copyright: © 2022 Nojima et al. https://creativecommons.org/licenses/by/3.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/3.0/) (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Nojima, Iyori
Hosoda, Ryusuke
Toda, Yuki
Saito, Yoshiki
Ueda, Naohiro
Horimoto, Kouhei
Iwahara, Naotoshi
Horio, Yoshiyuki
Kuno, Atsushi
Downregulation of IGFBP5 contributes to replicative senescence via ERK2 activation in mouse embryonic fibroblasts
title Downregulation of IGFBP5 contributes to replicative senescence via ERK2 activation in mouse embryonic fibroblasts
title_full Downregulation of IGFBP5 contributes to replicative senescence via ERK2 activation in mouse embryonic fibroblasts
title_fullStr Downregulation of IGFBP5 contributes to replicative senescence via ERK2 activation in mouse embryonic fibroblasts
title_full_unstemmed Downregulation of IGFBP5 contributes to replicative senescence via ERK2 activation in mouse embryonic fibroblasts
title_short Downregulation of IGFBP5 contributes to replicative senescence via ERK2 activation in mouse embryonic fibroblasts
title_sort downregulation of igfbp5 contributes to replicative senescence via erk2 activation in mouse embryonic fibroblasts
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9037271/
https://www.ncbi.nlm.nih.gov/pubmed/35378512
http://dx.doi.org/10.18632/aging.203999
work_keys_str_mv AT nojimaiyori downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts
AT hosodaryusuke downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts
AT todayuki downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts
AT saitoyoshiki downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts
AT uedanaohiro downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts
AT horimotokouhei downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts
AT iwaharanaotoshi downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts
AT horioyoshiyuki downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts
AT kunoatsushi downregulationofigfbp5contributestoreplicativesenescenceviaerk2activationinmouseembryonicfibroblasts