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 125 毫秒
1.
抗菌肽P7抑制大肠杆菌的非膜作用机制北大核心CSCD   总被引:1,自引:0,他引:1  
陈旋  李莉蓉 《微生物学报》2016,56(11):1737-1745
【目的】研究抗菌肽P7抑制大肠杆菌的非膜作用机制。【方法】P7与溴化乙锭竞争结合大肠杆菌基因组DNA的荧光光谱,分析P7与DNA的结合方式;流式细胞术分析P7与大肠杆菌基因组DNA结合对细菌细胞周期的影响;采用磁珠富集和PCR扩增相结合的方法分析P7特异结合的DNA序列;通过实时荧光定量PCR分析P7对大肠杆菌DNA复制和SOS损伤修复基因表达的影响;核酸染料的荧光分析研究P7对大肠杆菌DNA和RNA合成的影响。【结果】P7以嵌插的方式作用于大肠杆菌基因组DNA碱基对并形成肽-DNA复合物,使溴化乙锭-DNA复合体系的荧光强度减弱。P7可以显著增加大肠杆菌细胞周期中S期细胞数目,抑制大肠杆菌DNA复制。P7特异性结合rnh A使该基因表达水平显著下调2.24倍。同时,在肽的影响下参与大肠杆菌DNA复制相关的ssb、dna G、lig B和rnh A基因的表达水平显著下调(P<0.05),DNA损伤修复的rec A和rec N基因显著上调(P<0.05)。P7可降低大肠杆菌DNA和RNA的合成。【结论】P7特异性地结合rnh A序列引起大肠杆菌DNA的损伤并抑制大肠杆菌的DNA复制。在P7的影响下,参与大肠杆菌DNA复制相关的基因的表达水平下调,DNA损伤修复基因显著上调,同时抑制大肠杆菌DNA和RNA的合成。  相似文献   

2.
从DNA修复机理看细胞癌变的发生机制   总被引:3,自引:0,他引:3  
DNA损伤是引起基因突变,导致细胞恶性转化的重要原因.DNA损伤的修复过程非常复杂,是与细胞周期调节、DNA复制和DNA转录等生命活动紧密相连的.首先DNA修复需要细胞周期停滞,避免DNA损伤进入子代细胞.其次,参与DNA转录的某些基因产物参与DNA损伤的识别,有利于转录链的优先修复.最后,DNA修复系统NER、MMR参与损伤修复.上述DNA修复过程任何环节的异常,都将造成DNA修复功能减弱,导致某些功能基因突变,从而导致细胞的恶性转化.  相似文献   

3.
乳腺癌易感蛋白1在DNA损伤修复中的作用   总被引:1,自引:0,他引:1  
人类乳腺癌易感基因1(breast cancer susceptibility gene 1,BRCA1)首先是在乳腺癌家族中发现的,是具有遗传倾向的乳腺癌和卵巢癌易感基因,其基因的突变与家族性乳腺癌及卵巢癌的发生有密切联系。BRCA1是一种抑癌基因,其基因产物可以参与维持基因组稳定性的多条细胞信号通路,例如DNA损伤诱导的细胞周期调控、DNA损伤修复、基因转录调节、细胞凋亡、泛素化等重要的细胞活动。本文就近几年来BRCA1在DNA损伤修复中的作用的研究进展作一综述,包括DNA损伤诱导的细胞周期检查点的激活和DNA损伤修复两方面。  相似文献   

4.
目的探讨细胞周期素G1和G2在膀胱移行细胞癌(尿路上皮癌)中的表达及临床意义。方法收集武汉大学人民医院病理科2000-2006年有完整临床和病理资料的膀胱移行细胞癌存档蜡块50例和5例癌旁组织,采用免疫组织化学S-P法检测50例膀胱移行细胞癌和5例癌旁组织中细胞周期素G1和G2的表达水平。采用HPIAS-1000高清晰度彩色病理图文报告管理系统,对细胞周期素G1和G2的表达进行定量分析,并用SPSS11.5软件对各组免疫组织化学反应阳性颗粒的平均光密度、阳性面积率做单因素方差分析和SNK(q)检验。结果细胞周期素G1在膀胱移行细胞癌中呈高表达,癌旁组织中呈低表达。而细胞周期素G2在膀胱移行细胞癌中呈低表达,癌旁组织中呈高表达。膀胱移行细胞癌与癌旁组织相比,差异有显著性(P〈0.05)。随着移行细胞癌组织中细胞周期素G1表达的增高,周期素G2的表达却显著下降,两者呈显著负相关(P〈0.01)。结论细胞周期素G1和G2在膀胱移行细胞癌与癌旁组织中对细胞周期调控和/或DNA修复起了重要作用,并参与了诱导细胞凋亡的过程。  相似文献   

5.
目的:利用生物信息学方法从鼻咽癌组织基因芯片中筛选差异表达基因,并分析它们之间的互相作用。方法:利用R语言程序包等相关软件分析鼻咽癌组织和正常组织中差异表达的基因,并对其进行在线GO分析、KEGG富集分析及蛋白互作分析。结果:从25例鼻咽癌组织样本和3例正常组织对照样本中共分析出1103个差异表达基因,包括600个上调基因和503个下调基因(P0.05)。GO分析结果显示,差异表达基因在生物过程中显著富集,包括细胞分裂、DNA复制、G1/S有丝分裂细胞周期的转变和有丝分裂核分裂;在分子功能中,包括与蛋白质结合、与ATP结合、蛋白同二聚化活性、与DNA复制起点结合以及与受损的DNA结合;在细胞组分中,包括细胞质、核质、胞外体和船体中部。KEGG通路分析显示,差异表达基因在细胞周期、DNA复制、癌症途径、p53信号通路、错配修复、小细胞肺癌、卵母细胞减数分裂、氨基糖和核苷酸糖代谢、基部切除修复和人T淋巴细胞病毒(HTLV-Ⅰ)感染等信号通路中显著富集。在蛋白互作分析网络中筛选出10个节点度最高核心基因CDC45、MCM10、MCM3、MCM5、MCM2、CDC7、CDT1、CDC6、SMC2和NCAPG。结论:通过鼻咽癌组织和正常组织的对比筛选,发现多个基因表达发生变化,为相关临床研究提供了重要的信息基础。  相似文献   

6.
7.
宁顺斌  王玲 《生命科学》1998,10(4):167-171
抑癌基因p53蛋白产物是一种多功能的转录调控因子,其C-末端含DNA结合区,N-末端含转录激活区。它能通过磷酸化或构象变化来激活其DNA结合活性,从而与某些基因的启动子区结合而激活它们的转录,而对不具有与其结合位点基因的转录起抑制作用。p53蛋白还能作为一种DNA复制因子,通过与某些基因的复制起始区结合而对它们的复制或修复进行调控。对基因转录或复制的调控都最终反映在对细胞周期的调控上。  相似文献   

8.
高原鼢鼠肝脏组织细胞周期相关基因的进化和表达   总被引:1,自引:0,他引:1  
高原鼢鼠Myospalax baileyi是一种世居青藏高原的地下鼠,对严重的低氧环境有很强的适应性。低氧诱导细胞周期G1、G2期阻滞。为了探讨高原鼢鼠适应低氧环境的分子机制,应用生物信息学方法对p53下游细胞周期基因p21、CyclinD1、CyclinE、CDK6、CDK2、14-3-3-σ、Gadd45α、B99和CyclinB1的序列和编码的氨基酸序列进行了进化分析,并以SD大鼠Rattus norvegicus为对照,研究了这些基因在不同海拔(3300 m、2260 m)条件下的表达模式。结果表明:(1)高原鼢鼠细胞周期相关基因的序列与以色列鼹鼠Nannospalax galili同源性最高,达到90%以上;p21、CyclinD1、CyclinE和CyclinB1编码蛋白与以色列鼹鼠存在明显的趋同进化位点;SIFT评估发现,p21和CyclinB1氨基酸序列分别在第27号位点和第105号位点的变异对细胞周期调控功能有显著影响;(2)与低海拔条件相比,在高海拔条件下,高原鼢鼠肝脏组织中与G1期相关的基因p21表达水平显著上升,p21下游基因CyclinD1、CyclinE、CDK6和CDK2表达水平显著下降,而在SD大鼠中没有显著变化;与G2期相关的基因Gadd45α、B99、14-3-3-δ和CyclinB1在高原鼢鼠和SD大鼠中随海拔变化不发生明显变化。在不同海拔条件下,高原鼢鼠肝脏组织中的上述细胞周期相关基因的表达水平均极显著高于SD大鼠(P<0.01)。以上结果提示,高原鼢鼠经过长期的低氧适应,通过上调p21基因的表达抑制下游CyclinD1、CyclinE、CDK6和CDK2基因的表达,导致细胞周期G1期阻滞,从而提供充足的时间进行DNA修复,保证了DNA复制的准确性;同时高原鼢鼠肝脏组织中细胞周期的调控不仅与细胞周期基因的表达水平有关,而且可能与细胞周期因子p21的第27号位点和CyclinB1的第105号位点的变异有关。  相似文献   

9.
侯芷晗  陶敏  白晓彦 《生命的化学》2023,(11):1670-1680
牛痘相关激酶1(vaccinia-related kinase 1,VRK1)是一种核染色质丝苏氨酸蛋白激酶,在癌症中不发生基因突变,但在很多类型的肿瘤中表达上调并与不良预后相关。在细胞核内,VRK1可以磷酸化几种转录因子、组蛋白和涉及DNA损伤反应途径的蛋白质,还可以参与转录过程中组蛋白的乙酰化修饰,调节细胞周期、有丝分裂等过程促进细胞增殖,并且在DNA损伤修复中发挥至关重要的作用。在DNA损伤修复反应中,VRK1调控组蛋白乙酰化,介导DNA损伤反应的触发,进一步参与非同源末端连接DNA修复途径,还可以调控p53相关的DNA损伤修复过程。基于VRK1的以上生物学功能,癌组织中VRK1的高表达可以促进肿瘤细胞增殖、转移以及参与肿瘤细胞DNA修复过程。在癌症靶向治疗研究中,VRK1可以作为癌症合成致死性策略的选择靶点用于多种癌症的防治。  相似文献   

10.
细胞周期的失控与疾病   总被引:5,自引:0,他引:5  
细胞周期的运行失控可导致各种疾病。细胞周期的超常快速进行或在DNA复制不或有损伤时继续推进,将导致癌变。多种癌瘤常伴随细胞周期蛋白的过量表达;相反地、衰老细胞中细胞周期蛋白表达常减少。生长因子、癌基因及抑癌基因的失常,最终均归结到细胞周期的失控。  相似文献   

11.
Coupling of human circadian and cell cycles by the timeless protein   总被引:9,自引:0,他引:9       下载免费PDF全文
The Timeless protein is essential for circadian rhythm in Drosophila. The Timeless orthologue in mice is essential for viability and appears to be required for the maintenance of a robust circadian rhythm as well. We have found that the human Timeless protein interacts with both the circadian clock protein cryptochrome 2 and with the cell cycle checkpoint proteins Chk1 and the ATR-ATRIP complex and plays an important role in the DNA damage checkpoint response. Down-regulation of Timeless in human cells seriously compromises replication and intra-S checkpoints, indicating an intimate connection between the circadian cycle and the DNA damage checkpoints that is in part mediated by the Timeless protein.  相似文献   

12.
A variety of telomere protection programs are utilized to preserve telomere structure. However, the complex nature of telomere maintenance remains elusive. The Timeless protein associates with the replication fork and is thought to support efficient progression of the replication fork through natural impediments, including replication fork block sites. However, the mechanism by which Timeless regulates such genomic regions is not understood. Here, we report the role of Timeless in telomere length maintenance. We demonstrate that Timeless depletion leads to telomere shortening in human cells. This length maintenance is independent of telomerase, and Timeless depletion causes increased levels of DNA damage, leading to telomere aberrations. We also show that Timeless is associated with Shelterin components TRF1 and TRF2. Timeless depletion slows telomere replication in vitro, and Timeless-depleted cells fail to maintain TRF1-mediated accumulation of replisome components at telomeric regions. Furthermore, telomere replication undergoes a dramatic delay in Timeless-depleted cells. These results suggest that Timeless functions together with TRF1 to prevent fork collapse at telomere repeat DNA and ensure stable maintenance of telomere length and integrity.  相似文献   

13.
A variety of telomere protection programs are utilized to preserve telomere structure. However, the complex nature of telomere maintenance remains elusive. The Timeless protein associates with the replication fork and is thought to support efficient progression of the replication fork through natural impediments, including replication fork block sites. However, the mechanism by which Timeless regulates such genomic regions is not understood. Here, we report the role of Timeless in telomere length maintenance. We demonstrate that Timeless depletion leads to telomere shortening in human cells. This length maintenance is independent of telomerase, and Timeless depletion causes increased levels of DNA damage, leading to telomere aberrations. We also show that Timeless is associated with Shelterin components TRF1 and TRF2. Timeless depletion slows telomere replication in vitro, and Timeless-depleted cells fail to maintain TRF1-mediated accumulation of replisome components at telomeric regions. Furthermore, telomere replication undergoes a dramatic delay in Timeless-depleted cells. These results suggest that Timeless functions together with TRF1 to prevent fork collapse at telomere repeat DNA and ensure stable maintenance of telomere length and integrity.  相似文献   

14.
Tipin was originally isolated as a protein interacting with Timeless/Tim1/Tim (Tim), which is known to be involved in both circadian rhythm and cell cycle checkpoint regulation. The endogenous Tim and Tipin proteins in human cells, interacting through the N-terminal segment of each molecule, form a complex throughout the cell cycle. Tipin and Tim are expressed in the interphase nuclei mostly at constant levels during the cell cycle, and small fractions are recovered in the chromatin-enriched fractions during S phase. Depletion of endogenous Tipin results in reduced growth rate, and this may be due in part to inefficient progression of S phase and DNA synthesis. Knockdown of Tipin induces radioresistant DNA synthesis and inhibits phosphorylation of Chk1 kinase caused by replication stress, as was observed with that of Tim. Knockdown of Tipin or Tim results in reduced protein level and relocation to the cytoplasm of the respective binding partner, suggesting that the complex formation may be required for stabilization and nuclear accumulation of both proteins. Furthermore, both Tipin and Tim may facilitate the accumulation of Claspin in the nuclei under replication stress, whereas nuclear localization of Tipin and Tim is unaffected by Claspin. Our results indicate that mammalian Tipin is a checkpoint mediator that cooperates with Tim and may regulate the nuclear relocation of Claspin in response to replication checkpoint.  相似文献   

15.
Circadian clock genes have become a hot topic in cancer research in recent years, and more and more studies are showing that clock genes are involved in regulating cell proliferation cycle and apoptosis of malignant tumors, neuroendocrine and immune function, and other processes. Lung cancer is a malignant tumor with increasing incidence worldwide. The pathogenesis of lung cancer is extremely complicated and includes genetic factors, living environment, and smoking, and the occurrence of lung cancer is related to the regulation of many oncogenes and tumor suppressor genes. But there are few studies on clock genes in lung cancer. Studies on clock genes may help to better understand the mechanism of lung cancer development for an improved treatment. The expressions of all 14 kinds of clock genes in adenocarcinoma (ADC) and squamous cell carcinoma (SCC), two main kinds of non-small-cell lung cancer (NSCLC), were studied based on integration and analysis of data from The Cancer Genome Atlas (TCGA) to show the association between clock gene expression and prognosis of cancer patients. Analysis of TCGA data indicated that overexpression of Cry2, BMAL1, and RORA with underexpression of Timeless and NPAS2 was associated with a favorable prognosis of ADC, and the expression of NPAS2 was associated with the time of patient survival. Additionally, the expression of Cry2 was related to TNM stage. In SCC, high expression of DEC1 was correlated with poor overall survival in patients and the expression of Timeless was associated with the time of patient survival. In NSCLC, circadian clock genes constitute cancer circadian rhythm by interacting with each other, showing that asynchrony with normal tissues, which collectively controlling the occurrence and development of NSCLC.  相似文献   

16.
17.

Background

The role of CTGF varies in different types of cancer. The purpose of this study is to investigate the involvement of CTGF in tumor progression and prognosis of human nasopharyngeal carcinoma (NPC).

Experimental design

CTGF expression levels were examined in NPC tissues and cells, nasopharynx (NP) tissues, and NP69 cells. The effects and molecular mechanisms of CTGF expression on cell proliferation, migration, invasion, and cell cycle were also explored.

Results

NPC cells exhibited decreased mRNA expression of CTGF compared to immortalized human nasopharyngeal epithelial cell line NP69. Similarly, CTGF was observed to be downregulated in NPC compared to normal tissues at mRNA and protein levels. Furthermore, reduced CTGF was negatively associated with the progression of NPC. Knocking down CTGF expression enhanced the colony formation, cell migration, invasion, and G1/S cell cycle transition. Mechanistic analysis revealed that CTGF suppression activated FAK/PI3K/AKT and its downstream signals regulating the cell cycle, epithelial-mesenchymal transition (EMT) and MMPs. Finally, DNA methylation microarray revealed a lack of hypermethylation at the CTGF promoter, suggesting other mechanisms are associated with suppression of CTGF in NPC.

Conclusion

Our study demonstrates that reduced expression of CTGF promoted cell proliferation, migration, invasion and cell cycle progression through FAK/PI3K/AKT, EMT and MMP pathways in NPC.  相似文献   

18.
19.
20.
The Timeless-Tipin complex and Claspin are mediators of the ATR-dependent activation of Chk1 in the intra-S checkpoint response to stalled DNA replication forks. Tim-Tipin and Claspin also contribute to sister chromatid cohesion (SCC) in various organisms, likely through a replication-coupled process. Some models of the establishment of SCC posit that interactions between cohesin rings and replisomes could result in physiological replication stress requiring fork stabilization. The contributions of Timeless, Tipin, Claspin, Chk1 and ATR to SCC were investigated in genetically stable, human diploid fibroblast cell lines. Whereas Timeless, Tipin and Claspin showed similar contributions to UVC-induced activation of Chk1, siRNA-mediated knockdown of Timeless induced a 100-fold increase in sister chromatid discohesion, whereas the inductive effects of knocking down Tipin, Claspin and ATR were 4–20-fold. Knockdown of Chk1 did not significantly affect SCC. Consistent findings were obtained in two independently derived human diploid fibroblast lines and support a conclusion that SCC in human cells is strongly dependent on Timeless but independent of Chk1. Furthermore, the 10-fold difference in discohesion observed when depleting Timeless versus Tipin indicates that Timeless has a function in SCC that is independent of the Tim-Tipin complex, even though the abundance of Timeless is reduced when Tipin is targeted for depletion. A better understanding of how Timeless, Tipin and Claspin promote SCC will elucidate non-checkpoint functions of these proteins at DNA replication forks and inform models of the establishment of SCC.  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