• 제목/요약/키워드: Cellular senescence

검색결과 166건 처리시간 0.024초

Vorinostat Induces Cellular Senescence in Fibroblasts Derived from Young and Aged Dogs

  • Kim, Min-Jung;Oh, Hyun-Ju;Setyawan, Erif Maha Nugraha;Choi, Yoo-Bin;Lee, Seok-Hee;Lee, Byeong-Chun
    • 한국임상수의학회지
    • /
    • 제34권1호
    • /
    • pp.27-33
    • /
    • 2017
  • Although HDACIs affect ubiquitously expressed histone deacetylase and increase cellular senescence, there has been little study on the effect of age on treatment with HDACIs. Accordingly, the purpose of this study was to compare cellular senescence status and vorinostat-induced senescence in fibroblasts derived from aged dogs compared to young dogs. Skin tissues were taken from young (1-year-old) and aged (7-year-old) male dogs, and fibroblasts were cultured without (control) or with 10 uM of vorinostat for 24 hr. Beta-galactosidase activity was assessed, and real-time polymerase chain reaction and western blotting were performed to analyze the expression levels of transcripts and proteins related to cellular senescence. Beta-galactosidase activity was higher in aged dogs compared to young dogs in the control group, and was increased by vorinostat treatment. Expression of p21, p53 and p16 transcripts was higher in the aged than in the young group, and all transcripts were affected by vorinostat in both young and aged groups. Western blot results showed lower H3K9 acetylation in the aged dogs compared to the young dogs, and the acetylation was increased by vorinostat treatment in both groups. However, there was no significant difference between the transcript or protein alterations induced by vorinostat.

The p53-p21Cip1/WAF1 Pathway Is Necessary for Cellular Senescence Induced by the Inhibition of Protein Kinase CKII in Human Colon Cancer Cells

  • Kang, Ji-Young;Kim, Jin Joo;Jang, Seok Young;Bae, Young-Seuk
    • Molecules and Cells
    • /
    • 제28권5호
    • /
    • pp.489-494
    • /
    • 2009
  • We have previously shown that the down-regulation of protein kinase CKII activity is tightly associated with cellular senescence of human fibroblast IMR-90 cells. Here, we examined the roles of p53 and $p21^{Cip1/WAF1}$ in senescence development induced by CKII inhibition using wild-type, isogenic p53-/- and isogenic p21-/- HCT116 human colon cancer cell lines. A senescent marker appeared after staining for senescence-associated ${\beta}$-galactosidase activity in wild-type HCT116 cells treated with CKII inhibitor or $CKII{\alpha}$ siRNA, but this response was almost abolished in p53- or $p21^{Cip1/WAF1}$-null cells. Increased cellular levels of p53 and $p21^{Cip1/WAF1}$ protein occurred with the inhibition of CKII. CKII inhibition upregulated p53 and $p21^{Cip1/WAF1}$ expression at post-transcriptional level and transcription level, respectively. RB phosphorylation significantly decreased in cells treated with CKII inhibitor. Taken together, this study shows that the activation of the $p53-p21^{Cip1/WAF1}$ pathway acts as a major mediator of cellular senescence induced by CKII inhibition.

NF-κB in Cellular Senescence and Cancer Treatment

  • Jing, Hua;Lee, Soyoung
    • Molecules and Cells
    • /
    • 제37권3호
    • /
    • pp.189-195
    • /
    • 2014
  • The NF-${\kappa}B$ pathway transcriptionally controls a large set of target genes that play important roles in cell survival, inflammation, and immune responses. While many studies showed anti-tumorigenic and pro-survival role of NF-${\kappa}B$ in cancer cells, recent findings postulate that NF-${\kappa}B$ participates in a senescence-associated cytokine response, thereby suggesting a tumor restraining role of NF-${\kappa}B$. In this review, we discuss implications of the NF-${\kappa}B$ signaling pathway in cancer. Particularly, we emphasize the connection of NF-${\kappa}B$ with cellular senescence as a response to chemotherapy, and furthermore, present examples how distinct oncogenic network contexts surrounding NF-${\kappa}B$ produce fundamentally different treatment outcomes in aggressive B-cell lymphomas as an example.

Links between accelerated replicative cellular senescence and down-regulation of SPHK1 transcription

  • Kim, Min Kyung;Lee, Wooseong;Yoon, Gang-Ho;Chang, Eun-Ju;Choi, Sun-Cheol;Kim, Seong Who
    • BMB Reports
    • /
    • 제52권3호
    • /
    • pp.220-225
    • /
    • 2019
  • We have identified a mechanism to diminish the proliferative capacity of cells during cell expansion using human adipose-derived stromal cells (hAD-SCs) as a model of replicative senescence. hAD-SCs of high-passage numbers exhibited a reduced proliferative capacity with accelerated cellular senescence. Levels of key bioactive sphingolipids were significantly increased in these senescent hAD-SCs. Notably, the transcription of sphingosine kinase 1 (SPHK1) was down-regulated in hAD-SCs at high-passage numbers. SPHK1 knockdown as well as inhibition of its enzymatic activity impeded the proliferation of hAD-SCs, with concomitant induction of cellular senescence and accumulation of sphingolipids, as seen in high-passage cells. SPHK1 knockdown-accelerated cellular senescence was attenuated by co-treatment with sphingosine-1-phosphate and an inhibitor of ceramide synthesis, fumonisin $B_1$, but not by treatment with either one alone. Together, these results suggest that transcriptional down-regulation of SPHK1 is a critical inducer of altered sphingolipid profiles and enhances replicative senescence during multiple rounds of cell division.

Duck Oil-loaded Nanoemulsion Inhibits Senescence of Angiotensin II-treated Vascular Smooth Muscle Cells by Upregulating SIRT1

  • Kang, Eun Sil;Kim, Hyo Juong;Han, Sung Gu;Seo, Han Geuk
    • 한국축산식품학회지
    • /
    • 제40권1호
    • /
    • pp.106-117
    • /
    • 2020
  • Cellular senescence is associated with age-related vascular disorders and has been implicated in vascular dysfunctions. Here, we show that duck oil-loaded nanoemulsion (DO-NE) attenuates premature senescence of vascular smooth muscle cells (VSMCs) triggered by angiotensin II (Ang II). Compared with control nanoemulsion (NE), DO-NE significantly inhibited the activity of senescence-associated β-galactosidase, which is a biomarker of cellular senescence, in Ang II-treated VSMCs. SIRT1 protein expression was dose- and time-dependently induced in VSMCs exposed to DO-NE, but not in those exposed to NE, and SIRT1 promoter activity was also elevated. Consistently, DO-NE also dose-dependently rescued Ang II-induced repression of SIRT1 expression, indicating that SIRT1 is linked to the anti-senescence action of DO-NE in VSMCs treated with Ang II. Furthermore, the SIRT1 agonist resveratrol potentiated the effects of DO-NE on VSMCs exposed to Ang II, whereas the SIRT1 inhibitor sirtinol elicited the opposite effect. These findings indicate that DO-NE inhibits senescence by upregulating SIRT1 and thereby impedes vascular aging triggered by Ang II.

Erratum to: From cell senescence to age-related diseases: differential mechanisms of action of senescence-associated secretory phenotypes

  • Byun, Hae-Ok;Lee, Young-Kyoung;Kim, Jeong-Min;Yoon, Gyesoon
    • BMB Reports
    • /
    • 제49권11호
    • /
    • pp.641-650
    • /
    • 2016
  • Cellular senescence is a process by which cells enter a state of permanent cell cycle arrest. It is commonly believed to underlie organismal aging and age-associated diseases. However, the mechanism by which cellular senescence contributes to aging and age-associated pathologies remains unclear. Recent studies showed that senescent cells exert detrimental effects on the tissue microenvironment, generating pathological facilitators or aggravators. The most significant environmental effector resulting from senescent cells is the senescence-associated secretory phenotype (SASP), which is constituted by a strikingly increased expression and secretion of diverse pro-inflammatory cytokines. Careful investigation into the components of SASPs and their mechanism of action, may improve our understanding of the pathological backgrounds of age-associated diseases. In this review, we focus on the differential expression of SASP-related genes, in addition to SASP components, during the progress of senescence. We also provide a perspective on the possible action mechanisms of SASP components, and potential contributions of SASP-expressing senescent cells, to age-associated pathologies.

From cell senescence to age-related diseases: differential mechanisms of action of senescence-associated secretory phenotypes

  • Byun, Hae-Ok;Lee, Young-Kyoung;Kim, Jeong-Min;Yoon, Gyesoon
    • BMB Reports
    • /
    • 제48권10호
    • /
    • pp.549-558
    • /
    • 2015
  • Cellular senescence is a process by which cells enter a state of permanent cell cycle arrest. It is commonly believed to underlie organismal aging and age-associated diseases. However, the mechanism by which cellular senescence contributes to aging and age-associated pathologies remains unclear. Recent studies showed that senescent cells exert detrimental effects on the tissue microenvironment, generating pathological facilitators or aggravators. The most significant environmental effector resulting from senescent cells is the senescence-associated secretory phenotype (SASP), which is constituted by a strikingly increased expression and secretion of diverse pro-inflammatory cytokines. Careful investigation into the components of SASPs and their mechanism of action, may improve our understanding of the pathological backgrounds of age-associated diseases. In this review, we focus on the differential expression of SASP-related genes, in addition to SASP components, during the progress of senescence. We also provide a perspective on the possible action mechanisms of SASP components, and potential contributions of SASP-expressing senescent cells, to age-associated pathologies.

Analysis of Gene Expression in Human Dermal Fibroblasts Treated with Senescence-Modulating COX Inhibitors

  • Han, Jeong A.;Kim, Jong-Il
    • Genomics & Informatics
    • /
    • 제15권2호
    • /
    • pp.56-64
    • /
    • 2017
  • We have previously reported that NS-398, a cyclooxygenase-2 (COX-2)-selective inhibitor, inhibited replicative cellular senescence in human dermal fibroblasts and skin aging in hairless mice. In contrast, celecoxib, another COX-2-selective inhibitor, and aspirin, a non-selective COX inhibitor, accelerated the senescence and aging. To figure out causal factors for the senescence-modulating effect of the inhibitors, we here performed cDNA microarray experiment and subsequent Gene Set Enrichment Analysis. The data showed that several senescence-related gene sets were regulated by the inhibitor treatment. NS-398 up-regulated gene sets involved in the tumor necrosis factor ${\beta}$ receptor pathway and the fructose and mannose metabolism, whereas it down-regulated a gene set involved in protein secretion. Celecoxib up-regulated gene sets involved in G2M checkpoint and E2F targets. Aspirin up-regulated the gene set involved in protein secretion, and down-regulated gene sets involved in RNA transcription. These results suggest that COX inhibitors modulate cellular senescence by different mechanisms and will provide useful information to understand senescence-modulating mechanisms of COX inhibitors.

Antioxidant effects of selenocysteine on replicative senescence in human adipose-derived mesenchymal stem cells

  • Suh, Nayoung;Lee, Eun-bi
    • BMB Reports
    • /
    • 제50권11호
    • /
    • pp.572-577
    • /
    • 2017
  • In most clinical applications, human mesenchymal stem cells (hMSCs) are expanded in large scale before their administration. Prolonged culture in vitro results in cellular senescence-associated phenotypes, including accumulation of reactive oxygen species (ROS) and decreased cell viabilities. Profiling of stem cell-related genes during in vitro expansion revealed that numerous canonical pathways were significantly changed. To determine the effect of selenocysteine (Sec), a rare amino acid found in several antioxidant enzymes, on the replicative senescence in hMSCs, we treated senescent hMSCs with Sec. Supplementation of Sec in the culture medium in late-passage hMSCs reduced ROS levels and improved the survival of hMSCs. In addition, a subset of key antioxidant genes and Sec-containing selenoproteins showed increased mRNA levels after Sec treatment. Furthermore, ROS metabolism and inflammation pathways were predicted to be downregulated. Taken together, our results suggest that Sec has antioxidant effects on the replicative senescence of hMSCs.