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 703 毫秒
1.
近年来发现,K+通道与乳腺癌细胞的增殖和转化密切相关,但机制尚不清楚。本研究室前期报道了K+通道阻断剂4-氨基吡啶(4-aminopyridine,4-AP)能够抑制人乳腺上皮细胞的增殖,本文则进一步检测几种电压门控K+通道(voltage-gatedK+channel,Kv)在人乳腺上皮细胞系MCF10A中的表达,运用全细胞膜片钳技术,初步研究了该细胞K+通道的特性,观察K+通道阻断剂对细胞增殖以及信号通路蛋白活性的影响。结果显示,MCF10A细胞均有Kv1.1、Kv1.2、Kv1.3和Kv1.5基因mRNA的表达,其中Kv1.5表达量明显高于乳腺癌细胞MCF7。全细胞膜片钳钳制细胞于-60mV,给予持续时间800ms、范围从-60mV到+60mV的去极化刺激电压,步幅为10mV,然后给予持续150ms的-60mV的刺激,刺激频率为1Hz,可记录到一种跨膜电流,该电流具有电压依赖、外向整流的特性,并且能被Kv通道阻断剂4-AP阻断,证实该细胞膜存在Kv通道。此外,4-AP阻断K+通道10min后,与增殖相关的有丝分裂原活化蛋白激酶(mitogen-activated protein kinases,MAPK)信号通路ERK1/2蛋白活性增强而p38蛋白活性减弱;5mmol/L4-AP处理细胞48h后,MCF10A的生长抑制率为25.29%。以上结果提示,在人乳腺上皮细胞系MCF10A细胞膜上存在不同亚型的Kv通道,该通道可被4-AP阻断,并且4-AP能够抑制MCF10A细胞的增殖,其机制可能与细胞增殖信号通路不同成员的活性调节有关。  相似文献   

2.
HIV-1Tat蛋白抑制DNA修复和增强细胞辐射敏感性   总被引:1,自引:0,他引:1  
近年来临床研究发现,艾滋病合并肿瘤患者放疗后产生的正常组织和皮肤毒性反应明显高于普通肿瘤患者.本研究将探讨HIV-1Tat蛋白是否影响细胞对电离辐射敏感性及机理. 两个表达Tat蛋白的细胞系TT2和TE671-Tat均来源于人的横纹肌肉瘤细胞(TE671)并已转染了不同来源的tat基因.使用细胞辐射后克隆形成率检测辐射敏感性,RT-PCR和Western 印迹检测基因表达,彗星电泳和γ-H2AX位点检测DNA双链断裂和修复. TT2和TE671-Tat细胞的辐射敏感性与转染空载体及对照细胞相比明显增加.彗星电泳和γ-H2AX位点检测表明,在表达Tat蛋白的细胞中,辐射诱导DNA双链断裂的修复水平明显降低.通过RT-PCR和Western 印迹检测进一步证实,表达Tat蛋白的细胞中DNA修复蛋白DNA-PKcs的表达被抑制. HIV-1Tat蛋白抑制DNA-PKcs的表达,降低DNA双链断裂的修复,使细胞的电离辐射敏感性增高.本研究为了解AIDS合并肿瘤患者对放射治疗敏感性变化提供了重要信息.  相似文献   

3.
目的:构建原钙黏蛋白7b(PCDH7b)与绿色荧光蛋白(GFP)的融合基因表达载体pQCXIP-PCDH7b-EGFP,并检测PCDH7b-GFP融合蛋白在细胞中的表达定位。方法:以MDA-MB-436细胞基因组DNA为模板,PCR扩增人PCDH7b基因,克隆入逆转录病毒载体pQCXIP-EGFP-N1构建pQCXIP-PCDH7b-EGFP,病毒包装后感染人乳腺上皮细胞系MCF10A,免疫荧光染色检测其与膜蛋白E钙黏蛋白的相对表达定位。结果:构建获得逆转录病毒载体pQCXIP-PCDH7b-EGFP,融合蛋白PCDH7b-GFP定位于细胞浆和细胞膜,与E钙黏蛋白有共定位。结论:PCDH7b-GFP融合蛋白的表达,为后续活细胞成像分析PCDH7b在细胞内的动态分布奠定了基础。  相似文献   

4.
沉默DNA-PKcs对细胞信号转导相关基因转录的影响   总被引:2,自引:0,他引:2  
利用RNA干扰技术构建DNA-PKcs表达抑制细胞模型,探讨DNA-PKcs对HeLa细胞信号转导相关基因表达的调控作用.通过观察细胞对辐射及顺铂的敏感性,鉴定细胞表型变化.用寡核苷酸芯片检测细胞信号转导相关基因的转录谱,并用RT-PCR方法和SEAP检测系统进一步验证基因的表达变化.所筛选出的DNA-PKcs表达抑制细胞对辐射及顺铂的敏感性升高,15个与细胞信号转导相关的基因表达升高,其中7个是与干扰素信号转导反应相关的基因.8个表达下降,包括有细胞增殖分化相关基因,如NFAT.RT-PCR检测结果与芯片结果相一致,利用SEAT报告系统检测,进一步证实NFAT转录活性下调.实验结果表明,DNA-PKcs除了参与DNA修复外,还调控细胞信号转导相关基因的表达,而且大多与细胞增殖分化相关.  相似文献   

5.
大鼠高血压相关基因表达蛋白抑制血管平滑肌细胞增殖   总被引:8,自引:0,他引:8  
大鼠高血压相关基因 ( r HRG- 1 )编码一新细胞内信号传递蛋白 .体外转染 r HRG- 1表达蛋白发现 r HRG- 1表达蛋白能抑制自发性高血压大鼠血管平滑肌细胞内 Raf蛋白 ( Raf- 1 )和丝裂素活化蛋白激酶 ( MAPK)活性 ,抑制抗细胞凋亡基因 ( bcl- 2 )和增殖细胞核抗原 ( PCNA)基因 m RNA表达 ,同时还抑制该细胞 DNA的合成 .r HRG- 1是一正常血压大鼠血管平滑肌细胞内高度表达的基因 ,由此推测在自发性高血压大鼠血管平滑肌细胞内转染 r HRG- 1表达蛋白抑制其细胞 DNA合成的作用可能是抑制细胞内 Raf- 1活性与 MAPK活性及抑制 PCNA和 bcl- 2基因表达的结果  相似文献   

6.
Rad9是一种重要的细胞周期监控点调控蛋白.越来越多的证据显示,Rad9也可与多种DNA损伤修复通路中的蛋白质相互作用,并调节其功能,在DNA损伤修复中发挥重要作用.非同源末端连接修复是DNA双链断裂的一条重要修复途径.Ku70、Ku80和DNA依赖的蛋白激酶催化亚基(DNA-PKcs)共同组成DNA依赖的蛋白激酶复合物(DNA-PK),在非同源末端修复连接中起重要作用.本研究中,检测到Rad9与Ku70有直接的物理相互作用和功能相互作用.我们在不同的细胞模型中发现,Rad9基因敲除、Rad9蛋白去除或Rad9表达降低会导致非同源末端连接效率明显下降.已有的研究表明,DNA损伤可导致细胞中Ku70与染色质结合增加及DNA-PKcs激酶活性增强.我们的结果显示,与野生小鼠细胞相比,Rad9基因敲除的小鼠细胞中, DNA损伤诱导的上述效应均减弱.综上所述,我们的研究首次报道了Rad9与非同源末端连接修复蛋白Ku70间有相互作用,并提示Rad9可通过调节Ku70/Ku80/DNA-PKcs复合物功能参与非同源末端连接修复.  相似文献   

7.
该研究探讨了无赖氨酸激酶4(with no lysine kinase 4, WNK4)对于小鼠气管上皮细胞液体转运中的调节作用。在原代培养的小鼠气管上皮细胞中,应用siRNA特异性沉默WNK4基因,半定量PCR和Western blot实验验证沉默效率;随后应用尤斯灌流装置和Western blot实验记录该激酶的低表达对小鼠气管上皮细胞的短路电流及钠离子通道α-亚基蛋白表达水平的影响。半定量PCR和Western blot结果显示,该研究选用的siRNA序列可以沉默WNK4的表达。尤斯灌流和Western blot结果显示,沉默该激酶后,小鼠气管上皮细胞的阿米洛利敏感性电流和钠离子通道α-亚基蛋白表达明显增加。该研究表明,降低WNK4基因表达能增加小鼠气管上皮细胞的上皮钠离子通道α-亚基蛋白表达,促进钠离子转运,此过程可能参与相关水肿性肺疾患的修复。  相似文献   

8.
Rad9是一种重要的细胞周期监控点调控蛋白.越来越多的证据显示,Rad9也可与多种DNA损伤修复通路中的蛋白质相互作用,并调节其功能,在DNA损伤修复中发挥重要作用.非同源末端连接修复是DNA双链断裂的一条重要修复途径.Ku70、Ku80和DNA依赖的蛋白激酶催化亚基(DNA-PKcs)共同组成DNA依赖的蛋白激酶复合物(DNA-PK),在非同源末端修复连接中起重要作用.本研究中,检测到Rad9与Ku70有直接的物理相互作用和功能相互作用.我们在不同的细胞模型中发现,Rad9基因敲除、Rad9蛋白去除或Rad9表达降低会导致非同源末端连接效率明显下降.已有的研究表明,DNA损伤可导致细胞中Ku70与染色质结合增加及DNA-PKcs激酶活性增强.我们的结果显示,与野生小鼠细胞相比,Rad9基因敲除的小鼠细胞中, DNA损伤诱导的上述效应均减弱.综上所述,我们的研究首次报道了Rad9与非同源末端连接修复蛋白Ku70间有相互作用,并提示Rad9可通过调节Ku70/Ku80/DNA-PKcs复合物功能参与非同源末端连接修复.  相似文献   

9.
c-Myc蛋白与DNA-PKcs作用位点的鉴定   总被引:1,自引:0,他引:1  
DNA-PK复合物由Ku蛋白和DNA依赖蛋白激酶催化亚基(DNA-PKcs)组成,DNA-PKcs属于PI3K相关激酶家族成员.我们前期工作发现,DNA-Kcs沉默后,c-Myc的稳定性下降,且二者存在相互作用.为进一步确定c-Myc蛋白与DNA-PKcs相互作用位点,本研究利用原核表达系统活动了c-Myc及其截短体蛋白,利用GST pull-down技术结合Western印迹法,发现c-Myc蛋白294~370位氨基酸与DNA-PKcs存在相互作用.在细胞内表达GFP-c-Myc各截短体蛋白,发现294~370位氨基酸是c-Myc蛋白降解必需的.利用免疫荧光技术,发现DNA-PKcs与c-Myc蛋白有相同的细胞亚定位,进一步表明两者在生物学功能上具有相关性.有文献报道294~370位氨基酸是乙酰转移酶p300的底物,此位点的乙酰化导致c-Myc的降解.本实验结果提示,c-Myc蛋白的294~370位氨基酸与DNA-PKcs结合,可能阻止了乙酰转移酶p300的结合,从而达到提高c-Myc蛋白稳定性的作用.  相似文献   

10.
DNA双链断裂修复与重症联合免疫缺陷   总被引:1,自引:0,他引:1  
Wang KY  Zhao YH  Li WG 《生理科学进展》2008,39(2):182-184
DNA双链断裂(double-strand breaks, DSBs)是细胞DNA损伤的主要类型,它的修复通过同源重组(HR)和非同源末端连接(NHEJ)两种机制实现.NHEJ是人和哺乳动物细胞DSBs修复的重要通路,主要由DNA依赖性蛋白激酶(DNA-PK)、X射线修复交叉互补蛋白4(XRCC4)、DNA连接酶Ⅳ、Artemis、XLF/Cernunnos和其它DNA损伤修复辅助因子组成.本文重点介绍了NHEJ机制主要成分的特性及其功能,以及这些组分的基因发生突变或缺失所引起的DSBs修复缺陷与辐射敏感性重症联合免疫缺陷(radiosensitive severe combined immunodeficiencies, RS-SCIDs).  相似文献   

11.
Akt phosphorylation has previously been described to be involved in mediating DNA damage repair through the nonhomologous end-joining (NHEJ) repair pathway. Yet the mechanism how Akt stimulates DNA-protein kinase catalytic subunit (DNA-PKcs)-dependent DNA double-strand break (DNA-DSB) repair has not been described so far. In the present study, we investigated the mechanism by which Akt can interact with DNA-PKcs and promote its function during the NHEJ repair process. The results obtained indicate a prominent role of Akt, especially Akt1 in the regulation of NHEJ mechanism for DNA-DSB repair. As shown by pull-down assay of DNA-PKcs, Akt1 through its C-terminal domain interacts with DNA-PKcs. After exposure of cells to ionizing radiation (IR), Akt1 and DNA-PKcs form a functional complex in a first initiating step of DNA-DSB repair. Thereafter, Akt plays a pivotal role in the recruitment of AKT1/DNA-PKcs complex to DNA duplex ends marked by Ku dimers. Moreover, in the formed complex, Akt1 promotes DNA-PKcs kinase activity, which is the necessary step for progression of DNA-DSB repair. Akt1-dependent DNA-PKcs kinase activity stimulates autophosphorylation of DNA-PKcs at S2056 that is needed for efficient DNA-DSB repair and the release of DNA-PKcs from the damage site. Thus, targeting of Akt results in radiosensitization of DNA-PKcs and Ku80 expressing, but not of cells deficient for, either of these proteins. The data showed indicate for the first time that Akt through an immediate complex formation with DNA-PKcs can stimulate the accumulation of DNA-PKcs at DNA-DSBs and promote DNA-PKcs activity for efficient NHEJ DNA-DSB repair.  相似文献   

12.
Non-homologous end joining (NHEJ) is one of the primary pathways for the repair of ionizing radiation (IR)-induced DNA double-strand breaks (DSBs) in mammalian cells. Proteins required for NHEJ include the catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs), Ku, XRCC4 and DNA ligase IV. Current models predict that DNA-PKcs, Ku, XRCC4 and DNA ligase IV assemble at DSBs and that the protein kinase activity of DNA-PKcs is essential for NHEJ-mediated repair of DSBs in vivo. We previously identified a cluster of autophosphorylation sites between amino acids 2609 and 2647 of DNA-PKcs. Cells expressing DNA-PKcs in which these autophosphorylation sites have been mutated to alanine are highly radiosensitive and defective in their ability to repair DSBs in the context of extrachromosomal assays. Here, we show that cells expressing DNA-PKcs with mutated autophosphorylation sites are also defective in the repair of IR-induced DSBs in the context of chromatin. Purified DNA-PKcs proteins containing serine/threonine to alanine or aspartate mutations at this cluster of autophosphorylation sites were indistinguishable from wild-type (wt) protein with respect to protein kinase activity. However, mutant DNA-PKcs proteins were defective relative to wt DNA-PKcs with respect to their ability to support T4 DNA ligase-mediated intermolecular ligation of DNA ends. We propose that autophosphorylation of DNA-PKcs at this cluster of sites is important for remodeling of DNA-PK complexes at DNA ends prior to DNA end joining.  相似文献   

13.
Interstitial telomeric sequences (ITSs) in hamster cells are hot spots for spontaneous and induced chromosome aberrations (CAs). Most data on ITS instability to date have been obtained in DNA repair-proficient cells. The classical non-homologous end joining repair pathway (C-NHEJ), which is the principal double strand break (DSB) repair mechanism in mammalian cells, is thought to restore the morphologically correct chromosome structure. The production of CAs thus involves DNA-PKcs-independent repair pathways. In our current study, we investigated the participation of DNA-PKcs from the C-NHEJ pathway in the repair of spontaneous or radiation-induced DSBs in ITSs using wild-type and DNA-PKcs mutant Chinese hamster ovary cells. Our data demonstrate that DNA-PKcs stabilizes spontaneous DSBs within ITSs from the chromosome 9 long arm, leading to the formation of terminal deletions. In addition, we show that DNA-PKcs-dependent C-NHEJ is employed following radiation-induced DSBs in other ITSs and restores morphologically correct chromosomes, whereas DNA-PKcs independent mechanisms co-exist in DNA-PKcs proficient cells leading to an excess of CAs within ITSs.  相似文献   

14.
DNA damage initiates signaling events through kinase cascades that result in cell cycle checkpoint control and DNA repair. However, it is not yet clear how the signaling pathways relay to DNA damage repair. Using the repeat region of checkpoint protein MDC1 (mediator of DNA damage checkpoint protein 1), we identified DNA-PKcs/Ku as MDC1-associated proteins. Here, we show that MDC1 directly interacts with the Ku/DNA-PKcs complex. Down-regulation of MDC1 resulted in defective phospho-DNA-PKcs foci formation and DNA-PKcs autophosphorylation, suggesting that MDC1 regulates autophosphorylation of DNA-PKcs following DNA damage. Furthermore, DNA-PK-dependent DNA damage repair is defective in cells depleted of MDC1. Taken together, these results suggest that the MDC1 repeat region is involved in protein-protein interaction with DNA-PKcs/Ku, and MDC1 regulates DNA damage repair by influencing DNA-PK autophosphorylation. Therefore, MDC1 acts not only as a mediator of DNA damage checkpoint but also as a mediator of DNA damage repair.  相似文献   

15.
The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) is an enormous, 470-kDa protein serine/threonine kinase that has homology with members of the phosphatidylinositol (PI) 3-kinase superfamily. This protein contributes to the repair of DNA double-strand breaks (DSBs) by assembling broken ends of DNA molecules in combination with the DNA-binding factors Ku70 and Ku80. It may also serve as a molecular scaffold for recruiting DNA repair factors to DNA strand breaks. This study attempts to better define the role of protein kinase activity in the repair of DNA DSBs. We constructed a contiguous 14-kb human DNA-PKcs cDNA and demonstrated that it can complement the DNA DSB repair defects of two mutant cell lines known to be deficient in DNA-PKcs (M059J and V3). We then created deletion and site-directed mutations within the conserved PI 3-kinase domain of the DNA-PKcs gene to test the importance of protein kinase activity for DSB rejoining. These DNA-PKcs mutant constructs are able to express the protein but fail to complement the DNA DSB or V(D)J recombination defects of DNA-PKcs mutant cells. These results indicate that the protein kinase activity of DNA-PKcs is essential for the rejoining of DNA DSBs in mammalian cells. We have also determined a model structure for the DNA-PKcs kinase domain based on comparisons to the crystallographic structure of a cyclic AMP-dependent protein kinase. This structure gives some insight into which amino acid residues are crucial for the kinase activity in DNA-PKcs.  相似文献   

16.
Cells exposed to UV irradiation are predominantly arrested at S-phase as well as at the G(1)/S boundary while repair occurs. It is not known how UV irradiation induces S-phase arrest and yet permits DNA repair; however, UV-induced inhibition of replication is efficiently reversed by the addition of replication protein A (RPA), suggesting a role for RPA in this regulatory event. Here, we show evidence that DNA-dependent protein kinase (DNA-PK), plays a role in UV-induced replication arrest. DNA synthesis of M059K (DNA-PK catalytic subunit-positive (DNA-PKcs(+))), as measured by [(3)H]thymidine incorporation, was significantly arrested by 4 h following UV irradiation, whereas M059J (DNA-PKcs(-)) cells were much less affected. Similar results were obtained with the in vitro replication reactions where immediate replication arrest occurred in DNA-PKcs(+) cells following UV irradiation, and only a gradual decrease in replication activity was observed in DNA-PKcs(-) cells. Reversal of replication arrest was observed at 8 h following UV irradiation in DNA-PKcs(+) cells but not in DNA-PKcs(-) cells. Reversal of UV-induced replication arrest was also observed in vitro by the addition of a DNA-PK inhibitor, wortmannin, or by immunodepletion of DNA-PKcs, supporting a positive role for DNA-PK in damage-induced replication arrest. The RPA-containing fraction from UV-irradiated DNA-PKcs(+) cells poorly supported DNA replication, whereas the replication activity of the RPA-containing fraction from DNA-PKcs(-) cells was not affected by UV, suggesting that DNA-PKcs may be involved in UV-induced replication arrest through modulation of RPA activity. Together, our results strongly suggest a role for DNA-PK in S-phase (replication) arrest in response to UV irradiation.  相似文献   

17.
Although the roles of DNA-dependent protein kinase catalytic subunits (DNA-PKcs) in the non-homologous end joining (NHEJ) of DNA repair are well-recognized, the biological mechanisms and regulators by DNA-PKcs besides DNA repair, have not been clearly described. Here, we show that active DNA-PKcs caused by ionizing radiation, phosphorylated Snail1 at serine (Ser) 100, led to increased Snail1 stability. Furthermore, phosphorylated Snail1 at Ser100 reciprocally inhibited the kinase activity of DNA-PKcs, resulting in an inhibition of DNA repair activity. Moreover, Snail1 phosphorylation by DNA-PKcs was involved in genomic instability and aggressive tumor characteristics. Our results describe novel cellular mechanisms that affect genomic instability, sensitivity to DNA-damaging agents, and the migration of tumor cells by reciprocal regulation between DNA-PKcs and Snail1.  相似文献   

18.
DNA double-strand breaks (DSBs) are repaired by nonhomologous end-joining (NHEJ) and homologous recombination (HR). The NHEJ/HR decision is under complex regulation and involves DNA-dependent protein kinase (DNA-PKcs). HR is elevated in DNA-PKcs null cells, but suppressed by DNA-PKcs kinase inhibitors, suggesting that kinase-inactive DNA-PKcs (DNA-PKcs-KR) would suppress HR. Here we use a direct repeat assay to monitor HR repair of DSBs induced by I-SceI nuclease. Surprisingly, DSB-induced HR in DNA-PKcs-KR cells was 2- to 3-fold above the elevated HR level of DNA-PKcs null cells, and ~4- to 7-fold above cells expressing wild-type DNA-PKcs. The hyperrecombination in DNA-PKcs-KR cells compared to DNA-PKcs null cells was also apparent as increased resistance to DNA crosslinks induced by mitomycin C. ATM phosphorylates many HR proteins, and ATM is expressed at a low level in cells lacking DNA-PKcs, but restored to wild-type level in cells expressing DNA-PKcs-KR. Several clusters of phosphorylation sites in DNA-PKcs, including the T2609 cluster, which is phosphorylated by DNA-PKcs and ATM, regulate access of repair factors to broken ends. Our results indicate that ATM-dependent phosphorylation of DNA-PKcs-KR contributes to the hyperrecombination phenotype. Interestingly, DNA-PKcs null cells showed more persistent ionizing radiation-induced RAD51 foci (but lower HR levels) compared to DNA-PKcs-KR cells, consistent with HR completion requiring RAD51 turnover. ATM may promote RAD51 turnover, suggesting a second (not mutually exclusive) mechanism by which restored ATM contributes to hyperrecombination in DNA-PKcs-KR cells. We propose a model in which DNA-PKcs and ATM coordinately regulate DSB repair by NHEJ and HR.  相似文献   

19.
DNA polymerase beta (Polbeta) has been implicated in base excision repair. Polbeta knockout mice exhibit apoptosis in postmitotic neuronal cells and die at birth. Also, mice deficient in nonhomologous end-joining (NHEJ), a major pathway for DNA double-strand break repair, cause massive neuronal apoptosis. Severe combined immunodeficiency (SCID) mice have a mutation in the gene encoding DNA-dependent protein kinase catalytic subunit (DNA-PKcs), the component of NHEJ, and exhibit defective lymphogenesis. To study the interaction between Polbeta and DNA-PKcs, we generated mice doubly deficient in Polbeta and DNA-PKcs. Polbeta(-/-)DNA-PKcs(scid/scid) embryos displayed greater developmental delay, more extensive neuronal apoptosis, and earlier lethality than Polbeta(-/-) and DNA-PKcs(scid/scid) embryos. Furthermore, to study the involvement of p53 in the phenotype, we generated Polbeta(-/-)DNA-PKcs(scid/scid)p53(-/-) triple-mutant mice. The mutants did not exhibit apoptosis but were lethal with defective neurulation at midgestation. These results suggest a genetic interaction between Polbeta and DNA-PKcs in embryogenesis and neurogenesis.  相似文献   

20.
DNA-dependent protein kinase (DNA-PK), consisting of Ku and DNA-PKcs subunits, is the key component of the non-homologous end-joining (NHEJ) pathway of DNA double strand break (DSB) repair. Although the kinase activity of DNA-PKcs is essential for NHEJ, thus far, no in vivo substrate has been conclusively identified except for an autophosphorylation site on DNA-PKcs itself (threonine 2609). Here we report the ionizing radiation (IR)-induced autophosphorylation of DNA-PKcs at a novel site, serine 2056, the phosphorylation of which is required for the repair of DSBs by NHEJ. Interestingly, IR-induced DNA-PKcs autophosphorylation is regulated in a cell cycle-dependent manner with attenuated phosphorylation in the S phase. In contrast, DNA replication-associated DSBs resulted in DNA-PKcs autophosphorylation and localization to DNA damage sites. These results indicate that although IR-induced DNA-PKcs phosphorylation is attenuated in the S phase, DNA-PKcs is preferentially activated by the physiologically relevant DNA replication-associated DSBs at the sites of DNA synthesis.  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