首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 218 毫秒
1.
Ras信号通过细胞外信号调节激酶(extracellular signal regulated kinase,ERK1/2)的磷酸化调节通路活性,Ras突变或者异常激活都会导致肿瘤发生。为了研究在MCF-7乳腺癌细胞中,激活Ras信号通路后组蛋白H4K16位发生乙酰化修饰状态(H4K16ac)水平对癌细胞增殖、迁移能力的影响,研究采用Western blot技术检测不同转染组的ERK1/2、p-ERK1/2、H4和H4K16ac的表达水平;免疫组织化学技术检测乳腺癌组织和癌旁组织中p-ERK1/2水平和H4K16ac水平;分别构建突变质粒激活细胞Ras信号通路和模拟细胞内H4K16ac;CCK-8技术检测细胞的增殖能力,Transwell技术检测细胞的迁移能力。在对照组,激活Ras信号组,模拟H4K16ac组中,p-ERK1/2相对表达水平分别为0.285±0.017、0.407±0.026、0.373±0.028;H4K16ac相对表达水平分别为0.331±0.013、0.082±0.005、0.082±0.007。在乳腺癌组织和癌旁组织中p-ERK1/2表达分别为0.064±0.001、0.051±0.001;H4K16ac表达分别为0.028±0.003、0.063±0.005。激活Ras信号通路后癌细胞的增殖和迁移能力分别增加34.8%和103.1%(p0.05),与激活Ras信号通路的癌细胞相比,模拟H4K16ac的细胞的增殖和迁移能力分别降低25.1%和42.7%(p0.05)。结果表明在MCF-7乳腺癌细胞中激活Ras信号通路后p-ERK1/2水平上升,导致H4K16ac降低。细胞中H4K16ac上升会抑制Ras信号通路活性,从而抑制乳腺癌细胞的增殖和迁移。H4K16ac在癌细胞的增殖和向周围组织转移过程中发挥重要作用。  相似文献   

2.
Notch1信号通路在调节细胞的增殖、分化和凋亡中起重要作用。Notch1信号通路的异常激活可促进乳腺癌的发生、发展,但调控此过程的表观遗传机制尚有待于阐明。本研究发现,与相应的癌旁组织相比,乳腺癌组织中Notch1活性片段NIC1呈高表达,而组蛋白H4第16位赖氨酸残基乙酰化(H4K16ac)水平较低。敲低MCF-7细胞中Notch1可导致H4K16ac水平升高,且MCF-7细胞的增殖和迁移能力下降。本研究结果提示表观遗传修饰H4K16ac参与乳腺癌细胞Notch1致瘤信号传递,为乳腺癌发生、发展的机制研究提供了新思路。  相似文献   

3.
为了研究WSTF调控Ras相关乳腺癌细胞特性的机制。研究采用Western Blot检测WSTF磷酸化和组蛋白修饰水平;经GST pull-down验证WSTF与PCAF、MOF之间的相互作用,采用乙酰化酶活性实验验证PCAF和MOF酶活性;经Ch IP、Real-time PCR检测基因表达,并采用体内成瘤实验验证细胞成瘤能力。研究发现WSTF与PCAF、MOF之间存在相互作用。Ras通路激活后,WSTF磷酸化水平上调,使其与PCAF的结合增强的同时与MOF的结合减弱。这一变化引起PCAF的乙酰化酶活性上调而MOF的酶活性下调,导致H3K9ac上调和H4K16ac下调。组蛋白修饰的变化引起通路下游肿瘤相关基因表达发生改变,增强细胞成瘤能力。综上,WSTF蛋白通过调节与PCAF和MOF的相互作用,影响下游H3K9ac和H4K16ac水平及肿瘤相关基因表达,调控乳腺癌细胞成瘤能力。  相似文献   

4.
PI3K/AKT信号通路调控Myogenin和MCK基因的表达   总被引:1,自引:0,他引:1  
李晶  张云生  李宁  胡晓湘  石国庆  刘守仁  柳楠 《遗传》2013,35(5):637-642
骨骼肌分化过程受多个信号通路调控, PI3K/AKT信号通路是其中最重要的信号转导通路之一。PI3K/AKT信号通路可以调控骨骼肌分化, 但在染色质水平上的调控机制还不是很清楚。文章以小鼠成肌细胞(C2C12)为研究材料, 采用免疫印迹、染色质免疫共沉淀(Chromatin immunoprecipitation, ChIP)、定量PCR (Q-PCR)的方法研究PI3K/AKT信号通路调控Myogenin和MCK基因的表达。研究发现, C2C12细胞分化过程中添加PI3K/AKT信号通路激活剂处理24 h, Myogenin和MCK蛋白表达水平显著升高, 组蛋白H3K27me3去甲基化酶UTX的表达也升高, H3K27me3在Myogenin基因启动子区和MCK基因启动子及增强子区的富集与对照组相比显著降低。用PI3K/AKT信号通路抑制剂处理, 结果相反。因此, PI3K/AKT信号通路可能通过调控组蛋白去甲基化酶UTX的表达活性改变靶基因的H3K27me3的富集进而调控骨骼肌分化。  相似文献   

5.
目的:探讨长链非编码RNA SNHG3对人乳腺癌细胞MCF-7增殖、迁移与侵袭的影响。方法:构建SNHG3过表达质粒,实验分别设置阴性对照组(pcDNA-3.1+)与SNHG3基因过表达组(pcDNA-3.1+/SNHG3)。将MCF-7细胞转染对照组质粒和SNHG3过表达质粒,采用实时定量PCR 方法检测 SNHG3 mRNA 转录水平,Western blot 检测MMP9及EMT相关蛋白质水平;集落形成实验检测MCF-7细胞增殖能力;划痕愈合实验检测MCF-7细胞横向迁移能力; Transwell 小室实验检测MCF-7细胞纵向迁移能力及侵袭能力。结果:过表达SNHG3后,MCF-7细胞中SNHG3的mRNA水平显著增高(P<0.001);MCF-7细胞的体外增殖能力明显增加(P<0.01),迁移(P<0.01)与侵袭能力(P<0.001)也显著增强,实时定量PCR, Western blot 结果显示SNHG3可激活EMT相关通路。结论:过表达SNHG3可能通过激活EMT通路促进乳腺癌MCF-7细胞的增殖,迁移与侵袭。  相似文献   

6.
EBP50通过抑制LIMK/cofilin信号通路和PI3K/Akt/m TOR/MMP信号通路抑制乳腺癌细胞的侵袭和转移。然而,EBP50是否可以通过其他机制抑制乳腺癌细胞的侵袭和转移尚未可知。本研究发现,EBP50能通过Wnt3a/β-catenin信号通路影响乳腺癌细胞的侵袭和转移。Western印迹结果显示,EBP50在低转移细胞株MCF-7和正常乳腺细胞株MCF-10A中高表达,而在高转移细胞株MDA-MB-231低表达。采用RNAi技术将小RNA质粒瞬时转染乳腺癌细胞株MCF-7,同时将质粒pc DNA3.1-EBP50转入乳腺癌细胞株MDA-MB-231。Western印迹结果显示,Si EBP50/MCF-7细胞组的EBP50表达水平明显下调,MDA231/EBP50细胞组的EBP50表达水平明显上调。Transwell侵袭实验和划痕实验结果显示,用Wnt3a刺激后,Si EBP50/MCF-7细胞组体外迁移和侵袭能力明显增强,MDA231/EBP50细胞组体外迁移和侵袭能力明显减弱。Western印迹结果显示,与未用Wnt3a或同时用Wnt3a和抑制剂Dkk1刺激的相比,Si EBP50/MCF-7细胞组上皮-钙粘蛋白(Ecadherin)下调,波形蛋白(vimentin)上调,细胞核中β-联蛋白(β-catenin)的表达水平升高,而MDA231/EBP50细胞组上皮-钙粘蛋白上调,波形蛋白下调,细胞核中β-联蛋白表达下降。上述结果表明,EBP50通过Wnt3a/β-catenin信号通路影响β-联蛋白的转核,抑制EMT的发生,进而抑制乳腺癌细胞的侵袭和转移。  相似文献   

7.
目的:探讨磷脂酰肌醇-3-激酶/丝苏氨酸蛋白激酶(phosphatidylinositol 3 kinase/serine-threonine kinase,PI3K/AKT)信号通路与乳腺癌多药耐药和侵袭转移的相关性。方法:以乳腺癌细胞系MCF-7为母本,持续低浓度加药诱导建立阿霉素(Adriamycin,ADR)耐药系MCF-7/ADR’。细胞免疫荧光检测两细胞系中磷酸化AKT(phosphorylated AKT,P-AKT)、P-糖蛋白(P-Glycoprotein,P-gp)、基质金属蛋白酶2(matrix metalloproteinase-2,MMP-2)的表达。PI3K抑制剂LY294002作用两系前后,Western Blot检测P-AKT、MMP-2、P-gp的表达改变及qRT-PCR检测MMP-2、MDR1的表达改变。结果:P-AKT、P-gp(MDR1)、MMP-2在MCF-7中为低表达或不表达,MCF-7/ADR’中为高表达。LY294002作用两系后,P-AKT、P-gp(MDR1)、MMP-2在MCF-7/ADR’中的表达明显减低(P<0.05),MCF-7无明显改变。结论:抑制PI3K/AKT信号通路可有效降低MCF-7/ADR’耐药和侵袭转移能力,PI3K/AKT通路是调控乳腺癌多药耐药和侵袭转移的重要信号通路之一。  相似文献   

8.
【目的】果蝇Ras信号通路在细胞增殖与生长过程中发挥着重要的作用。Myc基因是bHLH转录因子家族基因,可调控细胞生长、竞争和再生增殖等生理过程。本研究旨在明确Ras信号通路与Myc的关系,探索Ras信号调控核内复制细胞生长的作用机制。【方法】生物信息学分析转基因家蚕Bombyx mori后部丝腺Myc基因的转录水平,并通过qPCR验证;在黑腹果蝇Drosophila melanogaster Kc细胞中,分别转染pAc5.1-HisB-Ras~(V12)-V5或pAc5.1-HisB-Raf-Flag过表达Ras~(V12)或Raf后,通过qPCR和Western blot技术分别检测Myc基因在mRNA和蛋白水平的相对表达量;在黑腹果蝇幼虫脂肪体和唾液腺中,结合黑腹果蝇遗传工具和分子生物学手段,验证Ras信号通路对Myc基因的调控作用。【结果】家蚕后部丝腺过表达Ras1~(CA)上调Myc转录水平。激活Ras信号使得黑腹果蝇Kc细胞内Myc在转录水平和蛋白水平上的表达量上调;黑腹果蝇幼虫唾液腺和脂肪体中,游走期Myc基因的表达量高于取食期;过表达Myc或激活Ras信号可以促进细胞核内周期进程;激活Ras信号促进Myc表达。【结论】Ras信号通路激活Myc表达,促进细胞核内周期进程,促进器官发育。  相似文献   

9.
本研旨在探讨Janus激酶3 (JAK3)对乳腺癌细胞迁移能力的影响以及其作用机制。利用si RNA (si JAK3)沉默乳腺癌MCF-7细胞JAK3的表达,用划痕实验检测细胞迁移能力,用荧光钙成像技术检测钙池操纵性钙通道(store-operatedcalcium channel,SOCC)活性,用Westernblot和RT-PCR检测钙池操纵性钙内流(store-operatedcalciumentry,SOCE)过程中的重要分子Orai1和STIM1表达水平。结果显示,SOCC抑制剂2-APB对MCF-7细胞的迁移能力有抑制作用。si JAK3转染可显著抑制MCF-7细胞的迁移能力,降低SOCC活性,下调Orai1和STIM1的m RNA和蛋白表达水平。在JAK3沉默细胞中过表达Orai1或STIM1,可使MCF-7细胞迁移力恢复。上述结果提示,JAK3通过影响SOCC促进乳腺癌细胞的迁移。  相似文献   

10.
本研旨在探讨Janus激酶3 (JAK3)对乳腺癌细胞迁移能力的影响以及其作用机制。利用si RNA (si JAK3)沉默乳腺癌MCF-7细胞JAK3的表达,用划痕实验检测细胞迁移能力,用荧光钙成像技术检测钙池操纵性钙通道(store-operatedcalcium channel,SOCC)活性,用Westernblot和RT-PCR检测钙池操纵性钙内流(store-operatedcalciumentry,SOCE)过程中的重要分子Orai1和STIM1表达水平。结果显示,SOCC抑制剂2-APB对MCF-7细胞的迁移能力有抑制作用。si JAK3转染可显著抑制MCF-7细胞的迁移能力,降低SOCC活性,下调Orai1和STIM1的m RNA和蛋白表达水平。在JAK3沉默细胞中过表达Orai1或STIM1,可使MCF-7细胞迁移力恢复。上述结果提示,JAK3通过影响SOCC促进乳腺癌细胞的迁移。  相似文献   

11.
12.
13.
14.
The PI3K/Akt/mTOR signaling pathway plays a key regulatory function in cell survival, proliferation, migration, metabolism and apoptosis. Aberrant activation of the PI3K/Akt/mTOR pathway is found in many types of cancer and thus plays a major role in breast cancer cell proliferation. In our previous studies, benzo[b]furan derivatives were evaluated for their anticancer activity and the lead compounds identified were 26 and 36. These observations prompted us to investigate the molecular mechanism and apoptotic pathway of these lead molecules against breast cancer cells. Benzo[b]furan derivatives (26 and 36) were evaluated for their antiproliferative activity against human breast cancer cell lines MCF-7 and MDA MB-231. These compounds (26 and 36) have shown potent efficiency against breast cancer cells (MCF-7) with IC50 values 0.057 and 0.051 μM respectively. Cell cycle analysis revealed that these compounds induced cell cycle arrest at G2/M phase in MCF-7 cells. Western blot analysis revealed that these compounds inhibit the PI3K/Akt/mTOR signaling pathway and induced mitochondrial mediated apoptosis in human breast cancer cells (MCF-7).  相似文献   

15.
We designed to investigate the effects of down-regulating the tumor susceptibility gene 101 (TSG101) on the proliferation and apoptosis of the human breast cancer MCF-7 cell line, and the role of the MAPK/ERK signal pathway in this process. The siRNA against TSG101 was transfected into the breast cancer MCF-7 cell line using Lipofectamine 2000. After TSG101 knockdown, the proliferation of MCF-7 cells was measured by the MTT assay. The cell cycle distribution and apoptosis were examined by using flow cytometry while cell migration was measured using a transwell assay. The protein level of p-ERK was further assessed by immunofluorescence and western blotting. Our results are as following, the MCF-7 cells transfected with TSG101 siRNA proliferated significantly slower and exhibited significantly increased rates of apoptosis compared to the control cells. In the TSG101 siRNA transfected cells, the percentage of cells in the G?/G? and S phase of the cell cycle was significantly higher and lower, respectively, compared to the control cells. Moreover, the migration ability of TSG101 siRNA transfected cells was lower than the control groups. Lastly, the level of p-ERK protein in TSG101 siRNA transfected cells was significantly decreased compared with the control cells. In conclusion, TSG101 knockdown in breast cancer cells induces apoptosis and inhibits proliferation. The TSG101 depleted cells are arrested at the G?/S transition of the cell cycle. The migration of breast cancer cells is also impaired by TSG101 siRNA. TSG101 may play a biological role through modulation of the MAPK/ERK signaling pathway in breast cancer.  相似文献   

16.
Urokinase-type plasminogen activator (uPA) activates the mitogen activated protein (MAP) kinases, extracellular signal-regulated kinase (ERK) 1 and 2, in diverse cell types. In this study, we demonstrate that uPA stimulates migration of MCF-7 breast cancer cells, HT 1080 fibrosarcoma cells, and uPAR-overexpressing MCF-7 cells by a mechanism that depends on uPA receptor (uPAR)-ligation and ERK activation. Ras and MAP kinase kinase (MEK) were necessary and sufficient for uPA-induced ERK activation and stimulation of cellular migration, as demonstrated in experiments with dominant-negative and constitutively active mutants of these signaling proteins. Myosin light chain kinase (MLCK) was also required for uPA-stimulated cellular migration, as determined in experiments with three separate MLCK inhibitors. When MCF-7 cells were treated with uPA, MLCK was phosphorylated by a MEK-dependent pathway and apparently activated, since serine-phosphorylation of myosin II regulatory light chain (RLC) was also increased. Despite the transient nature of ERK phosphorylation, MLCK remained phosphorylated for at least 6 h. The uPA-induced increase in MCF-7 cell migration was observed selectively on vitronectin-coated surfaces and was mediated by a beta1-integrin (probably alphaVbeta1) and alphaVbeta5. When MCF-7 cells were transfected to express alphaVbeta3 and treated with uPA, ERK was still phosphorylated; however, the cells did not demonstrate increased migration. Neutralizing the function of alphaVbeta3, with blocking antibody, restored the ability of uPA to promote cellular migration. Thus, we have demonstrated that uPA promotes cellular migration, in an integrin-selective manner, by initiating a uPAR-dependent signaling cascade in which Ras, MEK, ERK, and MLCK serve as essential downstream effectors.  相似文献   

17.
Recent studies indicate the importance of spatiotemporal regulation in the diversity and specificity of intracellular signaling. Here, we show that Ras-PI3K signaling plays an important role in the local regulation of phosphatidylinositol metabolism in the endosome through live-cell imaging by using a bimolecular fluorescence complementation technique, in which molecular interaction is indicated by fluorescence emission. Using several possible combinations of Ras and the Ras-binding domain, we identified an optimal set of probe molecules that yielded the most significant increase in fluorescence intensity between the active and inactive forms of Ras. This combination revealed that, among the Ras effectors tested, phosphatidylinositol 3-kinase (PI3K) was specifically implicated in signaling in the endosome. We also found that full length PI3K was recruited to the endosome in EGF- and Ras-dependent manners, which appears to be essential for the activation of PI3K in this compartment. Taken together, these findings demonstrate that the spatiotemporal regulation of Ras-PI3K signaling may dictate the activation of PI3K and subsequent downstream signaling in the endosome.  相似文献   

18.
Nucleosome destabilization by histone variants and modifications has been implicated in the epigenetic regulation of gene expression, with the histone variant H2A.Z and acetylation of H3K56 (H3K56ac) being two examples. Here we find that deletion of SWR1, the major subunit of the SWR1 complex depositing H2A.Z into chromatin in exchange for H2A, promotes epigenetic white-opaque switching in Candida albicans. We demonstrate through nucleosome mapping that SWR1 is required for proper nucleosome positioning on the promoter of WOR1, the master regulator of switching, and that its effects differ in white and opaque cells. Furthermore, we find that H2A.Z is enriched adjacent to nucleosome-free regions at the WOR1 promoter in white cells, suggesting a role in the stabilization of a repressive chromatin state. Deletion of YNG2, a subunit of the NuA4 H4 histone acetyltransferase (HAT) that targets SWR1 activity through histone acetylation, produces a switching phenotype similar to that of swr1, and both may act downstream of the GlcNAc signaling pathway. We further uncovered a genetic interaction between swr1 and elevated H3K56ac with the discovery that the swr1 deletion mutant is highly sensitive to nicotinamide. Our results suggest that the interaction of H2A.Z and H3K56ac regulates epigenetic switching at the nucleosome level, as well as having global effects.  相似文献   

19.
Ras GTPase-activating protein (RasGAP) is hypothesized to be an effector of oncogenic Ras stimulating numerous downstream cellular signaling cascades involved in survival, proliferation and motility. In this study, we identified calpain small subunit-1 (Capns1) as a new RasGAP-SH3 domain binding partner, using yeast two-hybrid screening. The interaction was confirmed by co-immunoprecipitation assay and was found specific to cells expressing oncogenic K-Ras. We used confocal microscopy to analyze our stably transfected cell model producing mutant Ras (PC3Ras(V12)). Staining for RasGAP-SH3/Capns1 co-localization was two-fold stronger in the protrusions of Ras(V12) cells than in PC3 cells. RasGAP or Capns1 knockdown in PC3Ras(V12) cells induced a two- to three-fold increase in apoptosis. Capns1 gene silencing reduced the speed and increased the persistence of movement in PC3Ras(V12) cells. In contrast, RasGAP knockdown in PC3Ras(V12) cells increased cell migration. Knockdown of both proteins altered the speed and directionality of cell motility. Our findings suggest that RasGAP and Capns1 interaction in oncogenic Ras cells is involved in regulating migration and cell survival.  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