首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 171 毫秒
1.
人类生存环境中的有害物质、机体正常代谢产生的氧化自由基、端粒缩短或端粒酶活性改变、原癌基因激活或抑癌基因失活等均可造成DNA损伤。通过启动DNA损伤修复反应,激活p53/p21或p16/Rb信号转导途径可以引发细胞周期阻滞,为修复破损的DNA赢得时间,避免不完整的DNA信息继续传递下去。过度的细胞周期阻滞将引起不可逆的细胞增殖停滞并最终引起细胞衰老,而当损伤的DNA没有完全修复就无限制的进入细胞周期时,将会诱发肿瘤的形成。肿瘤和衰老的发生机制是相互对立、相互交织的,而DNA损伤修复反应是联系二者的纽带。  相似文献   

2.
郝一 《生物技术通讯》2011,22(2):264-268
哺乳动物细胞在遭受应激损伤因素刺激时会启动一系列信号传导通路,从而引发细胞周期阻滞、DNA修复或细胞凋亡等效应,这些机制的异常与肿瘤的发生发展密切相关。GADD45α作为生长阻滞及DNA损伤诱导基因编码家族的一员,参与维持基因组稳定性、调控细胞周期行进、DNA损伤修复、细胞衰老及细胞凋亡等多种生物学过程,在肿瘤发生发展和肿瘤抑制反应中具有重要作用。我们简要综述了GADD45α参与维持基因组稳定性并发挥肿瘤抑制效应的分子机制。  相似文献   

3.
何艳  刘静 《生命科学》2010,(5):411-415
细胞衰老是细胞脱离细胞周期并不可逆地丧失增殖能力后进入的一种相对稳定的状态,虽然基本代谢过程仍然能够维持,但丧失合成DNA及增殖能力。细胞衰老具有复制衰老、癌基因诱导的衰老及加速衰老等类型。衰老细胞具有细胞体积大而扁平、细胞停止分裂及SA-β-gal反应阳性等明显特性,复制衰老还具有端粒缩短到无法维持染色体结构完整性的特征。目前已知,p53-p21和p16-pRB在细胞衰老过程中起着重要的调控作用,细胞衰老对肿瘤的形成起着天然的屏障作用。通过抑制端粒酶活性来诱导肿瘤细胞衰老和通过胞外刺激或化学治疗药物诱导肿瘤细胞发生衰老样生长停滞,已成为抗肿瘤研究的新思路。  相似文献   

4.
衰老是细胞的重要生命现象之一,衰老假说之一认为细胞中残留DNA损伤的积累可加速细胞的衰老.因此,细胞内DNA损伤监测及修复系统的正常运行与细胞衰老调控密切相关,DNA损伤监测及修复相关酶如PARP、DNA-PK、ATM、p53等在细胞衰老中的调控作用日益受到广泛关注.研究这些蛋白质分子间的相互作用及其在细胞衰老过程中的调控功能,有利于揭示DNA损伤应激、损伤修复调控与细胞衰老之间的内在联系,为抗衰老研究及从衰老角度治疗肿瘤提供新的思路.  相似文献   

5.
细胞衰老是指细胞生长永久阻滞于细胞周期的G1期,出现形态、生化及表观遗传的变化特性.细胞衰老由端粒缩短、DNA损伤、缺氧或癌基因失调等因素引起,它是抵抗肿瘤发生的主要壁垒.原癌基因c-myc编码转录因子,可调控很多基因,进而影响细胞周期演进、衰老、凋亡、代谢等生物学过程.c-Myc蛋白与细胞衰老密切相关,它可影响hTERT、p16、p53、Bmi-1和p27等衰老相关基因转录.c-Myc不仅可抑制复制性衰老,也能抑制癌基因诱发的衰老.c-Myc抑制ras诱导的细胞衰老取决于CDK2.c-Myc失活不仅能够诱导非恶性细胞(如人成纤维细胞)衰老,而且在许多肿瘤细胞中也可诱导衰老.然而,与ras基因类似,在特定条件下,c-Myc也可诱导细胞衰老,并可促进维氏综合症(Werner syndrome,WRN)缺失细胞的衰老.  相似文献   

6.
细胞衰老是指细胞生长永久阻滞于细胞周期的G1期,出现形态、生化及表观遗传的变化特性.细胞衰老由端粒缩短、DNA损伤、缺氧或癌基因失调等因素引起,它是抵抗肿瘤发生的主要壁垒.原癌基因c-myc编码转录因子,可调控很多基因,进而影响细胞周期演进、衰老、凋亡、代谢等生物学过程.c-Myc蛋白与细胞衰老密切相关,它可影响hTERT、p16、p53、Bmi-1和p27等衰老相关基因转录.c-Myc不仅可抑制复制性衰老,也能抑制癌基因诱发的衰老.c-Myc抑制ras诱导的细胞衰老取决于CDK2.c-Myc失活不仅能够诱导非恶性细胞(如人成纤维细胞)衰老,而且在许多肿瘤细胞中也可诱导衰老.然而,与ras基因类似,在特定条件下,c-Myc也可诱导细胞衰老,并可促进维氏综合症(Werner syndrome,WRN)缺失细胞的衰老.  相似文献   

7.
DNA双链断裂损伤反应及它的医学意义   总被引:2,自引:0,他引:2  
DNA损伤应激反应是维持基因组稳定性的基石.细胞在长期进化中形成了由损伤监视、周期调控、损伤修复、凋亡诱导等在内的自稳平衡机制.一方面,借助感应、识别并启动精细而复杂的修复机制修复损伤;另一方面,通过DNA损伤应激活化的细胞周期检查点机制,延迟或阻断细胞周期进程,为损伤修复提供时间,使细胞能安全进入新一轮细胞周期;损伤无法修复时则诱导细胞凋亡.DNA双链断裂(double strand breaks,DSBs)是真核基因组后果最严重的损伤类型之一,其修复不利,同肿瘤等人类疾病的发生发展密切相关.新进展揭示:DSBs损伤反应信号分子ATM-Chk2-p53、H2AX等的组成性活化,是肿瘤形成早期所激活的细胞内可诱导的抗癌屏障,其信号网络的精确、精细调控在基因组稳定性维持中发挥重要作用.此外,HIV病毒整合进入宿主细胞基因组的过程也依赖于宿主细胞中ATM介导的DSBs损伤反应信号转导;ATM特异性的小分子抑制剂在抗HIV感染中显示重要的功能意义.文中重点讨论调控DSBs损伤应激反应信号网络的主要研究进展,及其在肿瘤发生、发展及抗HIV感染中的新医学意义.  相似文献   

8.
林德玲  罗瑛  宋宜 《遗传》2014,(4):309-315
DNA损伤发生时,细胞会激活一系列复杂的信号网络来调控细胞周期检查,完成DNA损伤修复或当损伤超过修复能力时诱导凋亡,这一信号网络被称为DNA损伤反应(DNA damage response,DDR)。以往DDR信号网络的研究主要集中于基因转录调控和蛋白共价修饰对功能分子的稳定性和活性调控。近年来,mRNA稳定性调控和mRNA翻译调控等基因转录后调控机制在DDR中的重要作用引起研究者越来越多的关注。研究证明:多种microRNAs和RNA结合蛋白(RNA-binding proteins,RBPs)在转录后水平调控诸多重要功能蛋白的表达,在DDR信号网络中起着不可或缺的作用。文章针对DDR反应中转录后调控的研究进展以及参与其中的microRNAs和RBPs进行阐述和讨论。  相似文献   

9.
DNA和RNA肿瘤病毒可以通过整合基因、病毒自身携带的癌基因及病毒的致癌蛋白 细胞内源怀癌基因或抑癌基因相互作用,顺式或反式激活癌基因,致使基因过度表达,产生细胞转化和组织癌变。本文阐述病毒基因及其产物对细胞癌基因的顺式激作用与反式激活作用,进而深入了解病毒诱导癌基因转录的分子机制。  相似文献   

10.
生物有机体基因组DNA经常会受到内源或外源因素的影响而导致结构发生变化,产生损伤;在长期进化过程中,有机体也相应形成了一系列应对与修复损伤DNA,并维持染色体基因组正常结构功能的机制。其中DNA损伤检验点(DNA damage checkpoint)就是在感应DNA损伤的基础上,对损伤感应信号进行转导,或引起细胞周期的暂停,从而使细胞有足够的时间对损伤DNA进行修复,或最终导致细胞发生凋亡。DNA损伤检验点信号转导途径是一个高度保守的信号感应过程,整个途径大致可以分为损伤感应、信号传递及信号效应3个组成部分。其中3-磷脂酰肌醇激酶家族类成员ATM(ataxia-telangiectasia mutated)和ATR(ataxia-telangiectasia and Rad3-related)活性的增加构成整个途径活化的第一步。它们通过激活下游的效应激酶,Chk2/Chk1,通过协同作用许多其他调控细胞周期、DNA复制、DNA损伤修复及细胞凋亡等过程的蛋白质因子来实现细胞对DNA损伤的高度协调反应。近十几年,随着此领域研究的不断深入,人们逐步揭示了DNA损伤检验点途径发生过程中,各种核心组分通过与不同调节因子、效应因子及DNA损伤修复蛋白间的复杂相互作用,以实现监测感应异常DNA结构并实施相应反应的机制;其中,检验点衔接因子(mediators)及染色质结构,尤其是核小体组蛋白的共价修饰在调控ATM/ATR活性,促进ATM/ATR与底物间的相互作用以及介导DNA损伤位点周围染色质区域上多蛋白复合物在时间与空间上的动态形成发挥着重要的作用。同时,人们也开始发现DNA损伤检验点途径与DNA损伤修复、基因组稳定性以及肿瘤发生等过程之间某些内在的联系。该反应途径在通过协调细胞针对DNA损伤做出各种反应的基础上,直接或间接地参与或调控DNA损伤修复过程,并与DNA损伤修复途径协同作用最终保证染色体基凶组结构的完整性,而检验点途径的改变,则会引起基因组不稳定的发生,包括从突变频率的提高到大范围的染色体重排,以及染色体数量的畸变。如:突变发生在肿瘤形成早期,会大大增加肿瘤发生的几率。文章将对DNA损伤检验点途径机制及其对DNA损伤修复、基因组稳定性影响的最新进展进行综述。  相似文献   

11.
The mechanisms of tumor suppression must be linked to the oncogenic threats that may affect a normal cell. An important cancer causing mechanism is the accidental activation of genes that stimulate cell proliferation (oncogenes) by a variety of endogenous or environmental mutagens. This event has been experimentally modelled by enforcing the expression of oncogenes in primary cells. The astonishing outcome of these manipulations is that oncogenes trigger antiproliferative responses preventing progression to malignant transformation. These responses bring to an end proliferation due to cell death or a permanent cell cycle arrest called senescence. Here we review evidence indicating that oncogene induced senescence (OIS) involves activation of p53 via the DNA damage response (DDR). These results imply mechanisms of DNA damage in cells expressing oncogenes, that may be secondary to reactive oxygen species and/or some form of “oncogenic stress” that affect normal DNA replication. Interestingly, DNA damage signals persist in cells that escape from senescence. The implications of these signals for tumorigenesis are also discussed. Given that DNA damage signals have now been observed in cells treated with any stimuli known to induce senescence, the process can be redefined as a metabolically viable but permanent cell cycle arrest with persistent DNA damage signaling.  相似文献   

12.
Primary mouse embryonic fibroblasts (MEFs) are a popular tool for molecular and cell biology studies. However, when MEFs are grown in vitro under standard tissue culture conditions, they proliferate only for a limited number of population doublings (PD) and eventually undergo cellular senescence. Presently, the molecular mechanisms halting cell cycle progression and establishing cellular senescence under these conditions are unclear. Here, we show that a robust DNA damage response (DDR) is activated when MEFs undergo replicative cellular senescence. Senescent cells accumulate senescence-associated DDR foci (SDFs) containing the activated form of ATM, its phosphorylated substrates and γH2AX. In senescent MEFs, DDR markers do not preferentially accumulate at telomeres, the end of linear chromosomes. It has been observed that proliferation of MEFs is extended if they are cultured at low oxygen tension (3% O2). We observed that under these conditions, DDR is not observed and senescence is not established. Importantly, inactivation of ATM in senescent MEFs allows escape from senescence and progression through the S-phase. Therefore, MEFs undergoing cellular senescence arrest their proliferation due to the activation of a DNA damage checkpoint mediated by ATM kinase. Finally, we observed that spontaneously immortalized proliferating MEFs display markers of an activated DDR, indicating the presence of chromosomal DNA damage in these established cell lines.  相似文献   

13.
The DNA damage response (DDR) is activated upon DNA damage generation to promote DNA repair and inhibit cell cycle progression in the presence of a lesion. Cellular senescence is a permanent cell cycle arrest characterized by persistent DDR activation. However, some reports suggest that DDR activation is a feature only of early cellular senescence that is then lost with time. This challenges the hypothesis that cellular senescence is caused by persistent DDR activation. To address this issue, we studied DDR activation dynamics in senescent cells. Here we show that normal human fibroblasts retain DDR markers months after replicative senescence establishment. Consistently, human fibroblasts from healthy aged donors display markers of DDR activation even three years in culture after entry into replicative cellular senescence. However, by extending our analyses to different human cell strains, we also observed an apparent DDR loss with time following entry into cellular senescence. This though correlates with the inability of these cell strains to survive in culture upon replicative or irradiation-induced cellular senescence. We propose a model to reconcile these results. Cell strains not suffering the prolonged in vitro culture stress retain robust DDR activation that persists for years, indicating that under physiological conditions persistent DDR is causally involved in senescence establishment and maintenance. However, cell strains unable to maintain cell viability in vitro, due to their inability to cope with prolonged cell culture-associated stress, show an only-apparent reduction in DDR foci which is in fact due to selective loss of the most damaged cells.  相似文献   

14.
Nibrin plays an important role in the DNA damage response (DDR) and DNA repair. DDR is a crucial signaling pathway in apoptosis and senescence. To verify whether truncated nibrin (p70), causing Nijmegen Breakage Syndrome (NBS), is involved in DDR and cell fate upon DNA damage, we used two (S4 and S3R) spontaneously immortalized T cell lines from NBS patients, with the founding mutation and a control cell line (L5). S4 and S3R cells have the same level of p70 nibrin, however p70 from S4 cells was able to form more complexes with ATM and BRCA1. Doxorubicin-induced DDR followed by cell senescence could only be observed in L5 and S4 cells, but not in the S3R ones. Furthermore the S3R cells only underwent cell death, but not senescence after doxorubicin treatment. In contrary to doxorubicin treatment, cells from all three cell lines were able to activate the DDR pathway after being exposed to γ-radiation. Downregulation of nibrin in normal human vascular smooth muscle cells (VSMCs) did not prevent the activation of DDR and induction of senescence. Our results indicate that a substantially reduced level of nibrin or its truncated p70 form is sufficient to induce DNA-damage dependent senescence in VSMCs and S4 cells, respectively. In doxorubicin-treated S3R cells DDR activation was severely impaired, thus preventing the induction of senescence.  相似文献   

15.
DNA lesions trigger the DNA damage response (DDR) machinery, which protects genomic integrity and sustains cellular survival. Increasing data underline the significance of the integrity of the DDR pathway in chemotherapy response. According to a recent work, persistent exposure of A549 lung carcinoma cells to doxorubicin induces an initial DDR‐dependent checkpoint response, followed by a later DDR‐independent, but p27Kip1‐dependent one. Prompted by the above report and to better understand the involvement of the DDR signaling after chemotherapeutic stress, we examined the potential role of the canonical DDR pathway in A549 cells treated with doxorubicin. Exposure of A549 cells, prior to doxorubicin treatment, to ATM, ATR and DNA‐PKcs inhibitors either alone or in various combinations, revealed that the earlier documented two‐step response was DDR‐dependent in both steps. Notably, inhibition of both ATM and ATR or selective inhibition of ATM or DNA‐PKcs resulted in cell‐cycle re‐entry despite the increased levels of p27Kip1 at all time points analyzed. We further investigated the regulation of p27Kip1 protein levels in the particular setting. Our results showed that the protein status of p27Kip1 is mainly determined by p38‐MAPK, whereas the role of SKP2 is less significant in the doxoroubicin‐treated A549 cells. Cumulatively, we provide evidence that the DNA damage signaling is responsible for the prolonged cell cycle arrest observed after persistent chemotherapy‐induced genotoxic stress. In conclusion, precise identification of the molecular mechanisms that are activated during the chemotherapeutic cycles could potentially increase the sensitization to the therapy applied.  相似文献   

16.
Cells respond to genotoxic stress by activating the DNA damage response (DDR). When injury is severe or irreparable, cells induce apoptosis or cellular senescence to prevent transmission of the lesions to the daughter cells upon cell division. Resistance to apoptosis is a hallmark of cancer that challenges the efficacy of cancer therapy. In this work, the effects of ionizing radiation on apoptosis-resistant E1A + E1B transformed cells were investigated to ascertain whether the activation of cellular senescence could provide an alternative tumor suppressor mechanism. We show that irradiated cells arrest cell cycle at G2/M phase and resume DNA replication in the absence of cell division followed by formation of giant polyploid cells. Permanent activation of DDR signaling due to impaired DNA repair results in the induction of cellular senescence in E1A + E1B cells. However, irradiated cells bypass senescence and restore the population by dividing cells, which have near normal size and ploidy and do not express senescence markers. Reversion of senescence and appearance of proliferating cells were associated with downregulation of mTOR, activation of autophagy, mitigation of DDR signaling, and expression of stem cell markers.  相似文献   

17.
18.
DNA damage has been thought to be directly associated with the neoplastic progression by enabling mutations in tumor suppressor genes and activating/and amplifying oncogenes ultimately resulting in genomic instability. DNA damage causes activation of the DNA damage response (DDR) that is an important cellular mechanism for maintaining genomic integrity in the face of genotoxic stress. While the cellular response to genotoxic stress has been extensively studied in cancer models, less is known about the cellular response to oncogenic stress in the premalignant context. In the present study, by using breast tissues samples from women at different risk levels for invasive breast cancer (normal, proliferative breast disease and ductal carcinoma in situ) we found that DNA damage is inversely correlated with risk of invasive breast cancer. Similarly, in MCF10A based in vitro model system where we recapitulated high DNA damage conditions as seen in patient samples by stably cloning in cyclin E, we found that high levels of oncogene induced DNA damage, by triggering inhibition of a major proliferative pathway (AKT), inhibits cell growth and causes cells to die through autophagy. These data suggest that AKT-mTOR pathway is a novel component of oncogene induced DNA damage response in immortalized ‘normal-like’ breast cells and its suppression may contribute to growth arrest and arrest of the breast tumorigenesis.  相似文献   

19.
DNA lesions, constantly produced by endogenous and exogenous sources, activate the DNA damage response (DDR), which involves detection, signaling and repair of the damage. Autophagy, a lysosome-dependent degradation pathway that is activated by stressful situations such as starvation and oxidative stress, regulates cell fate after DNA damage and also has a pivotal role in the maintenance of nuclear and mitochondrial genomic integrity. Here, we review important evidence regarding the role played by autophagy in preventing genomic instability and tumorigenesis, as well as in micronuclei degradation. Several pathways governing autophagy activation after DNA injury and the influence of autophagy upon the processing of genomic lesions are also discussed herein. In this line, the mechanisms by which several proteins participate in both DDR and autophagy, and the importance of this crosstalk in cancer and neurodegeneration will be presented in an integrated fashion. At last, we present a hypothetical model of the role played by autophagy in dictating cell fate after genotoxic stress.  相似文献   

20.
We have previously demonstrated that the human papillomavirus (HPV) genome replicates effectively in U2OS cells after transfection using electroporation. The transient extrachromosomal replication, stable maintenance, and late amplification of the viral genome could be studied for high- and low-risk mucosal and cutaneous papillomaviruses. Recent findings indicate that the cellular DNA damage response (DDR) is activated during the HPV life cycle and that the viral replication protein E1 might play a role in this process. We used a U2OS cell-based system to study E1-dependent DDR activation and the involvement of these pathways in viral transient replication. We demonstrated that the E1 protein could cause double-strand DNA breaks in the host genome by directly interacting with DNA. This activity leads to the induction of an ATM-dependent signaling cascade and cell cycle arrest in the S and G2 phases. However, the transient replication of HPV genomes in U2OS cells induces the ATR-dependent pathway, as shown by the accumulation of γH2AX, ATR-interacting protein (ATRIP), and topoisomerase IIβ-binding protein 1 (TopBP1) in viral replication centers. Viral oncogenes do not play a role in this activation, which is induced only through DNA replication or by replication proteins E1 and E2. The ATR pathway in viral replication centers is likely activated through DNA replication stress and might play an important role in engaging cellular DNA repair/recombination machinery for effective replication of the viral genome upon active amplification.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号