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.
肝细胞癌(hepatocellular carcinoma,HCC)是常见的恶性肿瘤之一,由于其转移能力强、易复发以及对放化疗抵抗,导致其预后很差。研究表明,HGF/c-Met介导的信号传导在肝癌的侵袭和转移中起着重要作用,c-Met的激活能引发"侵入性生长"程序,使细胞运动性、侵袭性增强,减少细胞的凋亡。因此,更加深入了解与肝癌细胞发生发展相关的信号通路,以及探寻治疗肝癌的分子靶点将是未来研究的重点领域。本文综述了HGF/c-Met信号通路概况、c-Met的活化和调节、该通路与肝癌恶性行为的关系以及基于c-Met的肝癌靶向治疗。  相似文献   

2.
受体酪氨酸激酶c-Met即肝细胞生长因子HGF受体。HGF/c-Met信号通路在肿瘤形成、生长和转移过程中被频繁激活,因此, c-Met 已成为抗癌药物研究中一个重要靶标。重点介绍近年来基于c-Met通路的抗癌药物研究进展。  相似文献   

3.
受体酪氨酸激酶 c-Met 是抗肿瘤治疗的一个重要靶点,c-Met/HGF 通路在肿瘤的发生、发展、转移及血管再生中发挥重要作用。 综述 c-Met 及与配体 HGF 的复合物结构特征、c-Met/HGF 通路的生物学作用以及靶向 c-Met 抗肿瘤小分子抑制剂的研究进展。  相似文献   

4.
蛋白质修饰对Wnt信号通路的调控   总被引:1,自引:0,他引:1  
Wnt信号通路与细胞的生长发育和分化等密切相关,是细胞中重要的信号转导途径,在 多种癌症中,都有该通路的异常改变.Wnt信号通路主要是通过一系列蛋白将Wnt信号传导至β连环蛋白(β-catenin,β-cat),使后者入核并与转录因子T细胞因子/淋巴细胞增 强因子(T cell factor / lymphoid enhancer factor,TCF/LEF)结合,从而促进下游基因的转录,进而调控细胞的多种生理过程.在该通路中,涉及轴蛋白(Axin)、结肠腺瘤样息 肉病蛋白(adenomatous polyposis coli,APC)、糖原合酶激酶3β (glycogen synthase kinase-3β, GSK-3β)、β连环蛋白和酪蛋白激酶I (casein kinase I,CKI)等众多调节因子,这些因子能发生多种化学修饰,如磷酸化、泛素化(ubiquitylation)、苏素化 (small ubiquitin related moditier,SUMO)和乙酰化等,从而影响β连环蛋白、T细胞因子的稳定性、细胞定位以及活性,最终起到调节Wnt信号通路的作用.  相似文献   

5.
促分裂原激活的蛋白激酶(MAPK)信号传导通路的研究进展   总被引:12,自引:0,他引:12  
牟金叶  陈晓光 《生命科学》2002,14(4):208-211,203
MAPK信号传导通路在真核生物细胞的生化和分化、细胞周期调节和细胞凋亡过程中发挥着重要的作用。生物化学研究和分子生物学鉴定表明:在酵母和哺乳动物细胞中MAPK信号传导通路都有一个保守的三组分激活模件,该模件内的激酶引发了一系列的磷酸化级联反应。了解MAPK信号传导通路的组成部分、调控方式和作用机制,有助于对因信号传导通路的调节失控而引起的疾病进行预防和治疗。  相似文献   

6.
脑信号蛋白(semaphorin)是分泌的或膜相关糖蛋白,其通过与相应的受体结合后刺激激酶、调节RhoGTP酶,通过调节R-Ras调控整合素、细胞骨架,从而调控细胞运动。脑信号蛋白信号系统也调节肿瘤细胞的运动,调控肿瘤血管生成,并和肝细胞生长因子HGF/Met相偶联,控制肿瘤的侵袭转移。  相似文献   

7.
肝细胞生长因子(HGF)是一种多功能细胞因子,其生物学活性由c-Met蛋白所介导,通过多种途径调节细胞的生长,发挥重要的生理效应.Met活化失调在许多肿瘤的发生及侵袭中都有重要的作用.目前,对HGF/c-Met系统的研究已取得了很大进展.c-Met抑制剂成为抗肿瘤药物的研究热点,其中NK4能显著抑制肿瘤的生长、增殖和转移,本文对NK4在抗肿瘤方面的作用做一综述.  相似文献   

8.
丝裂原激活蛋白激酶(mitogen-activated protein kinases,MAPKs)信号通路是生物体内重要的信号传导通路,其主要参与调控细胞的增殖、生长、分化、凋亡和炎症反应等多种生理病理过程。MAPKs信号通路在多种心血管疾病的病理过程中起着重要调控作用。动脉粥样硬化(athrosclerosis,AS)所致的各种急重症严重危害人类的健康,发病率呈逐年上升的趋势,但是动脉粥样硬化发生发展的分子机制尚不完全清楚。近年来,MAPKs信号通路在动脉粥样硬化(athrosclerosis,AS)的发生发展中的作用已成为是研究的热点。  相似文献   

9.
肝细胞生长因子(hepatocyte growth factor, HGF)是一种多功能的细胞因子,其生物学活性由c-Met蛋白所介导.HGF/c-Met信号通路在肿瘤生成、侵袭、转移以及肿瘤新生血管生成方面起重要促进作用. 因此, HGF/c-Met信号转导通路可以作为抗肿瘤药物设计的靶点.其中,HGF-α链N端447个氨基酸组成的NK4蛋白是HGF的特异性拮抗剂,它不仅通过抑制HGF/c-Met系统的信号转导发挥抗肿瘤效应;而且可以通过拮抗HGF和其它血管生成因子如成纤维细胞生长因子(fibroblast growth factors, FGF)、血管内皮生长因子(vascular endothelial growth factor, VEGF)的活性,进而抑制肿瘤新生血管生成,最终导致肿瘤细胞的凋亡.NK4的这种双重抗肿瘤功能使其成为一类很有前景的新型抗肿瘤药物.本文就NK4对肿瘤的抑制作用及其机制的研究进展进行综述.  相似文献   

10.
cJun氨基末端激酶(JNK)家族是促分裂原活化蛋白激酶(MAPK)超家族成员之一,MAPK信号通路是多级蛋白激酶的级联反应,包括三个关键的激酶:MAPK、MAPK的激酶(MAPKK)和MAPK激酶的激酶(MAPKKK).JNK信号通路中有许多支架蛋白,如:JIP、JAMP、POSH等,能够与JNK及JNK信号通路中相关成员结合成复合物,调节它们的活性和细胞内定位,JNK信号通路可被细胞因子、生长因子、应激等多种因素激活,大量实验提示JNK活化在细胞增殖、细胞凋亡、应激反应以及多种人类疾病的发生与发展中起着重要的作用.JNK信号通路与其他信号通路间也有着相互作用.现对JNK活化机制的研究进展进行综述.  相似文献   

11.
Redox signaling is emerging as an essential mechanism in the regulation of biological activities of the cell. The HGF/c-Met signaling pathway has been implicated as a key regulator of the cellular redox homeostasis and oxidative stress. We previously demonstrated that genetic deletion of c-Met in hepatocytes disrupts redox homeostasis by a mechanism involving NADPH oxidase. Here, we were focused to address the mechanism of NADPH oxidase regulation by HGF/c-Met signaling in primary mouse hepatocytes and its relevance. HGF induced a biphasic mechanism of NADPH oxidase regulation. The first phase employed the rapid increase in production of ROS as signaling effectors to activate the Nrf2-mediated protective response resulting in up-regulation of the antioxidant proteins, such as NAD(P)H quinone oxidoreductase and γ-glutamylcysteine synthetase. The second phase operated under a prolonged HGF exposure, caused a suppression of the NADPH oxidase components, including NOX2, NOX4, p22 and p67, and was able to abrogate the TGFβ-induced ROS production and improve cell viability. In conclusion, HGF/c-Met induces a Nrf2-mediated protective response by a double mechanism driven by NADPH oxidase.  相似文献   

12.
Perineural invasion (PNI) is a pathologic feature of pancreatic cancer and is associated with poor outcomes, metastasis, and recurrence in pancreatic cancer patients. However, the molecular mechanism of PNI remains unclear. The present study aimed to investigate the mechanism that HGF/c-Met pathway facilitates the PNI of pancreatic cancer. In this study, we confirmed that c-Met expression was correlated with PNI in pancreatic cancer tissues. Activating the HGF/c-Met signaling pathway potentiated the expression of nerve growth factor (NGF) to recruit nerves and promote the PNI. Activating the HGF/c-Met signaling pathway also enhanced the migration and invasion ability of cancer cells to facilitate cancer cells invading nerves. Mechanistically, HGF/c-Met signaling pathway can active the mTOR/NGF axis to promote the PNI of pancreatic cancer. Additionally, we found that knocking down c-Met expression inhibited cancer cell migration along the nerve, reduced the damage of the sciatic nerve caused by cancer cells and protected the function of the sciatic nerve in vivo. Taken together, our findings suggest a supportive mechanism of the HGF/c-Met signaling pathway in promoting PNI by activating the mTOR/NGF axis in pancreatic cancer. Blocking the HGF/c-Met signaling pathway may be an effective target for the treatment of PNI.Subject terms: Pancreatic cancer, Cancer microenvironment  相似文献   

13.
There is growing evidence that the signal pathway between hepatocyte growth factor (HGF) and its receptor c-Met plays an important role in the development of lung cancer, although the specificity of such role is to be clarified. It seems clear that the HGF/c-Met signal contributes to the metastasis of cancer cells to the lung by stimulating the hyperproduction and overactivation of cytokines and enzymes, e.g. HGF, vascular endothelial growth factor and matrix metalloproteases. The HGF/c-Met signal may act as the candidate responsible for the development of epidermal growth factor receptor (EGFR) kinase inhibitor resistance. Experimental evidence showed that the combination of both EGFR and c-Met inhibitors had synergetic or additive therapeutic effects on lung cancer. Although the mechanism of interaction between HGF/c-Met and transforming growth factor-a/EGFR remains unclear, the cross-talk and balance between those two signal pathways are critical and necessary in the development of new therapies for lung cancer.  相似文献   

14.
15.
Small cell lung cancer (SCLC) is an aggressive illness with early metastases. There are several receptor tyrosine kinases (RTKs) overexpressed in SCLC, including c-Met. c-Met contains an external semaphorin-like domain, a cytoplasmic juxtamembrane domain, tyrosine kinase domain and multiple tyrosines that bind to adapter molecules. We have previously reported that c-Met is abundantly expressed in the NCI-H69 SCLC cell line and now have determined the downstream effects of stimulating c-Met via its ligand hepatocyte growth factor (HGF). Utilizing unique phospho-specific antibodies generated against various tyrosines of c-Met, we show that Y1003 (binding site for c-Cb1 and a negative regulatory site), Y1313 (binding site for PI3K), Y1230/Y1234/Y1235 (autophosphorylation site), Y1349 (binding site for Grb2), Y1365 (important in cell morphogenesis) are phosphorylated in response to HGF (40 ng/ml, 7.5 min) in H69 cells. Since multiple biological and biochemical effects are transduced through the PI3K pathway, we determine the role of PI3K in the c-Met/HGF stimulation pathway. We initially determined that by inhibiting PI3K with LY294002 (50μM over 72 hours), there was at least a 55% decrease in viability of H69 cells. Since H69 SCLC cells form clusters in cell culture, we determined the effects of HGF and LY294002 on cell motility of the clusters by time-lapse video microscopy. In response to HGF, SCLC moved much faster and formed more clusters, and this was inhibited by LY294002. Finally, we determined the downstream signal transduction of HGF stimulation of c-Met with and without inhibition of c-Met (with geldanamycin, an anisamycin antibiotic that inhibits c-Met in SCLC) or PI3K (with LY294002). We show that association of c-Met with PI3K and GAB2 is diminished by inhibiting c-Met. In summary, activation of the c-Met pathway targets the PI3K pathway in SCLC and this may be an important therapeutic target.  相似文献   

16.
17.
The activated c-Met receptor has potent effects on normal tissues and tumors. c-Met levels are regulated by hepatocyte growth factor (HGF); however, it is unknown if they can be regulated by gastrointestinal (GI) hormones. c-Met is found in many GI tissues/tumors that possess GI hormone receptors. We studied the effect of GI hormones on c-Met in rat pancreatic acini, which possess both receptors. CCK-8, carbachol, and bombesin, but not VIP/secretin, decreased c-Met. CCK-8 caused rapid and potent c-Met down-regulation and abolished HGF-induced c-Met and Gab1 tyrosine phosphorylation, while stimulating c-Met serine phosphorylation. The effect of cholecystokinin (CCK) was also seen in intact acini using immunofluorescence, in a biotinylated fraction representing membrane proteins, in single acinar cells, in Panc-1 tumor cells, and in vivo in rats injected with CCK. CCK-8 did not decrease cell viability or overall responsiveness. GF109203X, thapsigargin, or their combination partially reversed the effect of CCK-8. In contrast to HGF-induced c-Met down-regulation, the effect of CCK was decreased by a lysosome inhibitor (concanamycin) but not the proteasome inhibitor lactacystin. Inhibitors of clathrin-mediated endocytosis blocked the effect of CCK. HGF but not CCK-8 caused c-Met ubiquitination. These results show CCK and other GI hormones can cause rapid c-Met down-regulation, which occurs by a novel mechanism. These results could be important for c-Met regulation in normal as well as in neoplastic tissue in the GI tract.  相似文献   

18.
T cell dependent humoral immune responses are initiated by the activation of naive B cells in the T cell areas of the secondary lymphoid tissues. This primary B cell activation leads to migration of germinal center (GC) cell precursors into B cell follicles where they engage follicular dendritic cells (FDC) and T cells, and differentiate into memory B cells or plasma cells. Both B cell homing to the GC and interaction with FDC critically depend on integrin-mediated adhesion. We have recently indentified the c-met-encoded receptor tyrosine kinase and its ligand, the growth and motility factor hepatocyte growth factor/scatter factor (HGF/SF), as a novel paracrine signalling pathway regulating B cell adhesion (van der Voort et al., 1997, J. Exp. Med. 185, 2121–2131). The c-Met protein is expressed on B cells localized in the dark zone of the GC (centroblasts) and is induced by CD40 plus BCR ligation. Stimulation of c-Met with HGF/SF. which is produced at high levels by tonsillar stromal cells and FDC, leads to receptor phosphorylation and to enhanced integrin-mediated adhesion of B cells to both VCAM-l and fibronectin. Interestingly, these responses to HGF/SF are promoted by heparan-sulfate proteoglycan forms of CD44 (CD44-HS). Like c-Met, CD44-HS is induced on B cells by CD40 ligation. It efficiently binds HGF/SF and strongly promotes signalling through c-Met. We conclude that integrin regulation during antigen specific B cell differentiation involves cross-talk between the HGF/SF-c-Met pathway and CD44-HS.  相似文献   

19.
The receptor tyrosine kinase c-Met and its ligand, hepatocyte growth factor/scatter factor (HGF/SF), modulate signaling cascades implicated in cellular proliferation, survival, migration, invasion, and angiogenesis. Therefore, dysregulation of HGF/c-Met signaling can compromise the cellular capacity to moderate these activities and can lead to tumorigenesis, metastasis, and therapeutic resistance in various human malignancies. To facilitate studies investigating HGF/c-Met receptor coupling or c-Met signaling events in real time and in living cells and animals, here we describe a genetically engineered reporter where bioluminescence can be used as a surrogate for c-Met tyrosine kinase activity. c-Met kinase activity in cultured cells and tumor xenografts was monitored quantitatively and dynamically in response to the activation or inhibition of the HGF/c-Met signaling pathway. Treatment of tumor-bearing animals with a c-Met inhibitor and the HGF neutralizing antibody stimulated the reporter’s bioluminescence activity in a dose-dependent manner and led to a regression of U-87 MG tumor xenografts. Results obtained from these studies provide unique insights into the pharmacokinetics and pharmacodynamics of agents that modulate c-Met activity and validate c-Met as a target for human glioblastoma therapy.  相似文献   

20.
Sublethal irradiation promotes invasiveness of neuroblastoma cells   总被引:1,自引:0,他引:1  
Neuroblastoma is the most frequent extracranial solid tumour of childhood. Despite multiple clinical efforts, clinical outcome has remained poor. Neuroblastoma is considered to be radiosensitive, but some clinical studies including the German trial NB90 failed to show a clinical benefit of radiation therapy. The mechanisms underlying this apparent discrepancy are still unclear. We have therefore investigated the effects of radiation on neuroblastoma cell behaviour in vitro. We show that sublethal doses of irradiation up-regulated the expression of the hepatocyte growth factor (HGF) and its receptor c-Met in some neuroblastoma cell lines. The increase in HGF/c-Met expression was correlated with enhanced invasiveness and activation of proteases degrading the extracellular matrix. Thus, irradiation at sublethal doses may promote the metastatic dissemination of neuroblastoma cells through activating the HGF/c-Met pathway and triggering matrix degradation.  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