2009
DOI: 10.4161/cc.8.24.10215
|View full text |Cite
|
Sign up to set email alerts
|

Pseudo-DNA damage response in senescent cells

Abstract: Cellular senescence is currently viewed as a response to DNA damage. In this report, we showed that non-damaging agents such as sodium butyrate-induced p21 and ectopic expression of either p21 or p16 cause cellular senescence without detectable DNA breaks. Nevertheless, senescent cells displayed components of DNA damage response (DDR) such as γH2AX foci and uniform nuclear staining for p-ATM. Importantly, there was no accumulation of 53BP1 in γH2AX foci of senescent cells. Consistently, comet assay failed to d… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

10
166
0

Year Published

2010
2010
2021
2021

Publication Types

Select...
8
2

Relationship

1
9

Authors

Journals

citations
Cited by 186 publications
(176 citation statements)
references
References 44 publications
(64 reference statements)
10
166
0
Order By: Relevance
“…45 Rapamycin may also prevent the formation of ATM-phosphorylated H2AX (γ-H2AX), thereby suppressing mTOR-dependent pseudo-DNA damage response in senescent cells. 46 These findings support the notion that mTOR signaling is involved in cell stresses including ribosomal stress. 12 The mTOR kinase therefore may cooperate with E2F1 under ribosomal stress.…”
Section: Discussionsupporting
confidence: 80%
“…45 Rapamycin may also prevent the formation of ATM-phosphorylated H2AX (γ-H2AX), thereby suppressing mTOR-dependent pseudo-DNA damage response in senescent cells. 46 These findings support the notion that mTOR signaling is involved in cell stresses including ribosomal stress. 12 The mTOR kinase therefore may cooperate with E2F1 under ribosomal stress.…”
Section: Discussionsupporting
confidence: 80%
“…Thus, overexpression of the five miRNAs favored DNA damage, and not solely DDR (gH2AX), which can also occur in the absence of DNA damage (pseudo-DDR). 24 Finally, we showed that the mTOR pathway was an essential driver of senescence induced by senescence-associated miRNAs as SA-b-gal staining was significantly reduced by treatment with rapamycin. [20][21][22] Of note, our preliminary observations, by studying the expression levels of the senescence-inducing miRNAs in human skin broblasts from young (age [17][18][19][20][21][22][23][24][25] or old (age 89-94) donors, show that the highest expression levels of miR-376a* and miR-494 were found in one old donor cell line, and expression of miR-486-5p was on average two-fold higher in old with respect to young cells.…”
Section: Discussionmentioning
confidence: 97%
“…[37][38][39][40][41] Therefore, in addition to the critical role of the STAT3-IGFBP5 axis in the IL-6-induced premature senescence, mTOR activity caused by serum growth factors and added IL-6 together with secreted factors during the course is likely to contribute to the premature senescence. However, our preliminary experiments showed that TIG3 cells with a chimeric receptor with truncated gp130 containing only the YRHQ motif, which activates only the STAT3 pathway without activating the ERK1/2 or PI3K/AKT/mTOR pathways, induced premature senescence in TIG3 cells at the extent comparable with that shown in IL-6/sIL-6R stimulation (unpublished data, Kojima H and Nakajima K).…”
Section: Methodsmentioning
confidence: 99%