• Title/Summary/Keyword: cell senescence

Search Result 191, Processing Time 0.03 seconds

Replicative Senescence in Cellular Aging and Oxidative Stress (세포 노화에 있어서 복제 세네센스 현상과 산화적 스트레스의 영향)

  • 박영철
    • Toxicological Research
    • /
    • v.19 no.3
    • /
    • pp.161-172
    • /
    • 2003
  • Explanted mammalian cells perform a limited number of cell division in vitro and than are arrested in a state known as replicative senescence. Such cells are irreversibly blocked, mostly in the G1 phase of cell cycle, and are no longer sensitive to growth factor stimulation. Thus replicative senescence is defined as a permanent and irreversible loss of replicative potential of cells. For this characteristic, replicative senescence seems to evolve to protect mammalian organism from cancer. However, senescence also contributes to aging. It seems to decrease with age of the cell donor and, as a form of cell senescence, is thought to underlie the aging process. Extensive evidence supports the idea that progressive telomere loss contributes to the phenomenon of cell senescence. Telomeres are repetitive structures of the sequence (TTAGGG)n at the ends of linear chromosomes. It has been shown that the average length of telomere repeats in human somatic cells decreases by 30∼200 bp with each cell division. It is generally believed that when telomeres reach a critical length, a signal is activated to initiate the senescent program. This has given rise to the hypothesis that telomeres act as mitotic clocks to regulate lifespan. One proposes that cumulative oxidative stress, mainly reactive oxygen species generated from mitochondria, may mainly cause telomere shortening, accelerating aging. Here, the biological importance and mechanism of replicative senescence were briefly reviewed. Also it was summarized that how oxidative stress affects replicative senescence and telomere shortening.

Exploiting tumor cell senescence in anticancer therapy

  • Lee, Minyoung;Lee, Jae-Seon
    • BMB Reports
    • /
    • v.47 no.2
    • /
    • pp.51-59
    • /
    • 2014
  • Cellular senescence is a physiological process of irreversible cell-cycle arrest that contributes to various physiological and pathological processes of aging. Whereas replicative senescence is associated with telomere attrition after repeated cell division, stress-induced premature senescence occurs in response to aberrant oncogenic signaling, oxidative stress, and DNA damage which is independent of telomere dysfunction. Recent evidence indicates that cellular senescence provides a barrier to tumorigenesis and is a determinant of the outcome of cancer treatment. However, the senescence-associated secretory phenotype, which contributes to multiple facets of senescent cancer cells, may influence both cancer-inhibitory and cancer-promoting mechanisms of neighboring cells. Conventional treatments, such as chemo- and radiotherapies, preferentially induce premature senescence instead of apoptosis in the appropriate cellular context. In addition, treatment-induced premature senescence could compensate for resistance to apoptosis via alternative signaling pathways. Therefore, we believe that an intensive effort to understand cancer cell senescence could facilitate the development of novel therapeutic strategies for improving the efficacy of anticancer therapies. This review summarizes the current understanding of molecular mechanisms, functions, and clinical applications of cellular senescence for anticancer therapy.

Effects of 5-Aza-2'-Deoxycytidine, Bromodeoxyuridine, Interferons and Hydrogen Peroxide on Cellular Senescence in Cholangiocarcinoma Cells

  • Moolmuang, Benchamart;Singhirunnusorn, Pattama;Ruchirawat, Mathuros
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.17 no.3
    • /
    • pp.957-963
    • /
    • 2016
  • Cellular senescence, a barrier to tumorigenesis, controls aberrant proliferation of cells. We here aimed to investigate cellular senescence in immortalized cholangiocyte and cholangiocarcinoma cell lines using five different inducing agents: 5-aza-2'deoxycytidine, bromodeoxyuridine, interferons ($IFN{\beta}$ and $IFN{\gamma}$), and hydrogen peroxide. We analyzed senescence characteristics, colony formation ability, expression of genes involved in cell cycling and interferon signaling pathways, and protein levels. Treatment with all five agents decreased cell proliferation and induced cellular senescence in immortalized cholangiocyte and cholangiocarcinoma cell lines with different degrees of growth-inhibitory effects depending on cell type and origin. Bromodeoxyuridine gave the strongest stimulus to inhibit growth and induce senescence in most cell lines tested. Expression of p21 and interferon related genes was upregulated in most conditions. The fact that bromodeoxyuridine had the strongest effects on growth inhibition and senescence induction implies that senescence in cholangiocarcinoma cells is likely controlled by DNA damage response pathways relating to the p53/p21 signaling. In addition, interferon signaling pathways may partly regulate this mechanism in cholangiocarcinoma cells.

Cellular senescence: a promising strategy for cancer therapy

  • Lee, Seongju;Lee, Jae-Seon
    • BMB Reports
    • /
    • v.52 no.1
    • /
    • pp.35-41
    • /
    • 2019
  • Cellular senescence, a permanent state of cell cycle arrest, is believed to have originally evolved to limit the proliferation of old or damaged cells. However, it has been recently shown that cellular senescence is a physiological and pathological program contributing to embryogenesis, immune response, and wound repair, as well as aging and age-related diseases. Unlike replicative senescence associated with telomere attrition, premature senescence rapidly occurs in response to various intrinsic and extrinsic insults. Thus, cellular senescence has also been considered suppressive mechanism of tumorigenesis. Current studies have revealed that therapy-induced senescence (TIS), a type of senescence caused by traditional cancer therapy, could play a critical role in cancer treatment. In this review, we outline the key features and the molecular pathways of cellular senescence. Better understanding of cellular senescence will provide insights into the development of powerful strategies to control cellular senescence for therapeutic benefit. Lastly, we discuss existing strategies for the induction of cancer cell senescence to improve efficacy of anticancer therapy.

Screening Methods for Anti-senescence Activity in Dermal Fibroblasts under Pyruvate-deprivation Conditions

  • Kil, In Sup;Shim, Jinsup;Cho, Gayoung;Choi, Sowoong;Son, Eui Dong;Kim, Hyoung-June
    • Korea Journal of Cosmetic Science
    • /
    • v.1 no.1
    • /
    • pp.1-9
    • /
    • 2019
  • The identification of compounds with anti-senescence activity in cell culture system is a first step in aging research. Given that pyruvate can be used energy source by conversion to acetyl-CoA in mitochondria, and protects cultured cell from various stress-induced cell damage and cell death, synthetic media (e.g., DMEM) often includes 1 mM pyruvate, which is very higher than the pyruvate concentration in human blood (approximately 30 ��M). However, the use of medium containing high concentration of pyruvate is not suitable for screening anti-senescence compounds, because pyruvate also protects against the cellular senescence of primary human dermal fibroblasts (NHDFs) through NAD+ generated during conversion to lactate. In this study, four extracts, i.e., Sprouted seed and fruit complex, Poncirus trifoliata fruit extract, Jaum balancing complex, and Prunus mume extract were used for evaluation of different anti-senescence effect in the absence or presence of 0.1 mM pyruvate, similar to the physiological pyruvate concentration. The senescence in NHDFs cultured with DMEM in the presence of 0.1 mM pyruvate (approximately the physiological concentration in human blood) is accelerated, as observed in pyruvate deprivation conditions. The cytotoxicity of the Poncirus trifoliata fruit extract was protected by pyruvate, and Jaum balancing complex and Prunus mume extract had anti-senescence activity in the presence of 0.1 mM pyruvate, but not in the absence of pyruvate. Given that pyruvate is a powerful protector against both cytotoxicity and cellular senescence, the screening of candidate agents for anti-senescence in high pyruvate conditions using an in vitro cell culture system is not valid. Therefore, we recommend the use of a low concentration of pyruvate to evaluate the anti-senescence effects of candidates, which is more similar to in vivo aging conditions than excessive stress-induced senescence models, to exclude the effect of excessive pyruvate in vitro.

Recent Advances in Cellular Senescence, Cancer and Aging

  • Lim, Chang-Su;Judith Campisi
    • Biotechnology and Bioprocess Engineering:BBE
    • /
    • v.6 no.4
    • /
    • pp.231-236
    • /
    • 2001
  • How much do we know about the biology of aging from cell culture studies Most normal somatic cells have a finite potential to divide due to a process termed cellular or replicative senescence. A growing body evidence suggests that senescence evolved to protect higher eu-karyotes, particularly mammals, from developing cancer, We now know that telomere shortening due to the biochemistry of DNA replication, induces replicative senescence in human cells. How-ever in rodent cells, replicative senescence occurs despite very long telomeres. Recent findings suggest that replicative senescence is just the tip of the iceberg of a more general process termed cellular senescence. It appears that cellular senescence is a response to potentially oncogenic in-sults, including oxidative damage. In young orgainsms, growth arrest by cell senescence sup-presses tumor development, but later in life, due to the accumulation of senescent cells which se-cret factors that can disrupt tissues during aging, cellular senescence promotes tumorigenesis. Therefore, antagonistic pleiotropy may explain, if not in whole the apparently paradoxical effects of cellular senescence, though this still remains an open question.

  • PDF

Melatonin Rescues Mesenchymal Stem Cells from Senescence Induced by the Uremic Toxin p-Cresol via Inhibiting mTOR-Dependent Autophagy

  • Yun, Seung Pil;Han, Yong-Seok;Lee, Jun Hee;Kim, Sang Min;Lee, Sang Hun
    • Biomolecules & Therapeutics
    • /
    • v.26 no.4
    • /
    • pp.389-398
    • /
    • 2018
  • p-Cresol, found at high concentrations in the serum of chronic kidney failure patients, is known to cause cell senescence and other complications in different parts of the body. p-Cresol is thought to mediate cytotoxic effects through the induction of autophagy response. However, toxic effects of p-cresol on mesenchymal stem cells have not been elucidated. Thus, we aimed to investigate whether p-cresol induces senescence of mesenchymal stem cells, and whether melatonin can ameliorate abnormal autophagy response caused by p-cresol. We found that p-cresol concentration-dependently reduced proliferation of mesenchymal stem cells. Pretreatment with melatonin prevented pro-senescence effects of p-cresol on mesenchymal stem cells. We found that by inducing phosphorylation of Akt and activating the Akt signaling pathway, melatonin enhanced catalase activity and thereby inhibited the accumulation of reactive oxygen species induced by p-cresol in mesenchymal stem cells, ultimately preventing abnormal activation of autophagy. Furthermore, preincubation with melatonin counteracted other pro-senescence changes caused by p-cresol, such as the increase in total 5'-AMP-activated protein kinase expression and decrease in the level of phosphorylated mechanistic target of rapamycin. Ultimately, we discovered that melatonin restored the expression of senescence marker protein 30, which is normally suppressed because of the induction of the autophagy pathway in chronic kidney failure patients by p-cresol. Our findings suggest that stem cell senescence in patients with chronic kidney failure could be potentially rescued by the administration of melatonin, which grants this hormone a novel therapeutic role.

Tenovin-1 Induces Senescence and Decreases Wound-Healing Activity in Cultured Rat Primary Astrocytes

  • Bang, Minji;Ryu, Onjeon;Kim, Do Gyeong;Mabunga, Darine Froy;Cho, Kyu Suk;Kim, Yujeong;Han, Seol-Heui;Kwon, Kyoung Ja;Shin, Chan Young
    • Biomolecules & Therapeutics
    • /
    • v.27 no.3
    • /
    • pp.283-289
    • /
    • 2019
  • Brain aging induces neuropsychological changes, such as decreased memory capacity, language ability, and attention; and is also associated with neurodegenerative diseases. However, most of the studies on brain aging are focused on neurons, while senescence in astrocytes has received less attention. Astrocytes constitute the majority of cell types in the brain and perform various functions in the brain such as supporting brain structures, regulating blood-brain barrier permeability, transmitter uptake and regulation, and immunity modulation. Recent studies have shown that SIRT1 and SIRT2 play certain roles in cellular senescence in peripheral systems. Both SIRT1 and SIRT2 inhibitors delay tumor growth in vivo without significant general toxicity. In this study, we investigated the role of tenovin-1, an inhibitor of SIRT1 and SIRT2, on rat primary astrocytes where we observed senescence and other functional changes. Cellular senescence usually is characterized by irreversible cell cycle arrest and induces senescence- associated ${\beta}$-galactosidase (SA-${\beta}$-gal) activity. Tenovin-1-treated astrocytes showed increased SA-${\beta}$-gal-positive cell number, senescence-associated secretory phenotypes, including IL-6 and IL-$1{\beta}$, and cell cycle-related proteins like phospho-histone H3 and CDK2. Along with the molecular changes, tenovin-1 impaired the wound-healing activity of cultured primary astrocytes. These data suggest that tenovin-1 can induce cellular senescence in astrocytes possibly by inhibiting SIRT1 and SIRT2, which may play particular roles in brain aging and neurodegenerative conditions.

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
    • /
    • v.49 no.11
    • /
    • 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
    • /
    • v.48 no.10
    • /
    • 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.