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 431 毫秒
1.
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   

2.
肝细胞生长因子(HGF)是一种具有多重功能的细胞调控因子。HGF与其受体Met酪氨酸激酶(c-Met)的结合可激发多种生物学反应,从而调节细胞的增殖、分化、形态发生和侵袭运动等。有多种因素参与了HGF/c-Met信号传导的调控,从而防止信号的过度放大,其中Cbl1、Rab、泛素化激酶和HGF/c-Met的内吞等发挥了重要的作用。因此,对HGF/c-Met内吞过程的研究,了解内吞对于HGF/c-Met的信号传导及其调控的影响,探讨HGF/c-Met信号传导通路的调控机理和相互作用模式,可进一步阐明HGF/c-Met信号传导的调控机制,从而验证肝细胞中内吞作用直接调节HGF/c-Met信号通路的作用机制。  相似文献   

3.
目的:探讨肝细胞生长因子(HGF)和其受体c-Met在鼻咽癌(NPC)中的表达及其意义.方法:采用免疫组织化学S-P法检测55例NPC,15例慢性鼻咽炎症中HGF/c-Met蛋白的表达,并结合临床病理进行分析.结果:HGF、c-Met在慢性鼻咽粘膜炎症中,主要表达于柱状上皮细胞胞膜或细胞浆;在NPC中HGF主要表达于肿瘤间质、癌细胞有少量表达;c-Met表迭于癌细胞,间质不表达.HGF、c-Met蛋白在NPC中的阳性表达率分别为90.9%(39/55)、70.9%(50/55),与对照组相比,差异有显著性(P<0.05);HGF,c-Met的阳性表达与临床分期呈正相关(P<0.05),而与年龄、性别、组织分型无相关性;有淋巴结转移的c-Met蛋白阳性表达率分别显著高于无淋巴结转移,差异有显著性(P<0.01).结论:HGF/c-Met过度表达可能在鼻咽癌癌细胞的侵袭转移过程中起调节作用,c-Met基因的异常表达与NPC侵袭转移生物学行为密切相关,c-Met对于判断NPC预后具有一定价值.  相似文献   

4.
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   

5.
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   

6.
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   

7.
目的:探讨肝细胞生长因子(HGF)和其受体c—Met在鼻咽癌(NPC)中的表达及其意义。方法:采用免疫组织化学S-P法检测55例NPC,15例慢性鼻咽炎症中HGF/c—Met蛋白的表达,并结合临床病理进行分析。结果:HGF、C—Met在慢性鼻咽粘膜炎症中,主要表达于柱状上皮细胞胞膜或细胞浆;在NPC中HGF主要表达于肿瘤间质、癌细胞有少量表达;c-Met表达于癌细胞,间质不袁达。HGF、C—Met蛋白在NPC中的阳性表达率分别为90.9%(39/55)、70.9%(50/55),与对照组相比,差异有显著性(P〈0.05);HGF,c-Met的阳性表达与临床分期呈正相关(P〈0.05),而与年龄、性别、组织分型无相关性;有淋巴结转移的c-Met蛋白阳性表达率分别显著高于无淋巴结转移,差异有显著性(P〈0.01)。结论:HGF/c—Met过度表达可能在鼻咽癌癌细胞的侵袭转移过程中起调节作用,c-Met基因的异常表达与NPC侵袭转移生物学行为密切相关,c-Met对于判断NPC预后具有一定价值。  相似文献   

8.
c-Met 为一种由原癌基因MET 编码的跨膜受体酪氨酸激酶,是肝细胞生长因子的受体,与多种癌症的不良预后有关。近年来,c-Met抑制剂作为一种对抗癌症的有效策略备受关注。结合汤森路透数据库资源——Thomson Reuters Integrity 和Cortellis for Competitive Intelligence,对c-Met 在疾病中的作用、相关药物研究进展、适应证、研发机构、交易、专利、文献等情报进行了数据层面的分析。  相似文献   

9.
慢性乙肝病毒感染是亚洲国家肝癌的常见病因,乙肝病毒所表达的乙肝病毒X蛋白(Hepatitis B virus X protein,HBx)是肝癌发生发展的重要推动因子。核因子-κB(Nuclear factorκB,NF-κB)是模式识别受体下游重要的转录因子,肝细胞生长因子(Hepatocyte growth factor,HGF)/c-Met途径能够促进NF-κB活化,并通过活化的NF-κB来调节肝癌细胞的迁移、侵袭等生物学环节。但HBx是否直接通过NF-κB通路来调节肝癌细胞的迁移、侵袭尚未明确。为探究HBx通过NF-κB通路调节肝癌细胞迁移、侵袭的作用,本研究采用培养肝癌HepG2细胞并分组,空白对照组用不含药物及质粒的DMEM处理,空白质粒组转染1.2μg的pcDNA3.1空白质粒,HBx质粒组转染不同浓度的HBx表达质粒pcDNA3.1-HBx,HBx+PDTC组转染1.2μg的HBx表达质粒pcDNA3.1-HBx并用含有50μmol/L PDTC的DMEM进行处理;皮下注射HepG2细胞建立移植瘤小鼠模型,称量移植瘤的质量。检测细胞迁移及侵袭活力、细胞及移植瘤中NF-κB通路分子、迁移基因、侵袭基因的表达。结果显示,与空白对照组、空白质粒组比较,HBx质粒组细胞中NF-κB、HGF、c-Met、N-cadherin、Vimentin、MMP2、MMP9的蛋白表达水平及相对愈合面积、侵袭数目均明显增多(P0.05);与HBx质粒组比较,HBx+PDTC组细胞中NF-κB、HGF、c-Met、N-cadherin、Vimentin、MMP2、MMP9的蛋白表达水平及相对愈合面积、侵袭数目均明显减少(P0.05);与空白对照组、空白质粒组比较,HBx质粒组移植瘤的质量及移植瘤中NF-κB、HGF、c-Met的蛋白表达水平明显增加(P0.05)。本研究得出结论,HBx能够促进肝癌细胞的迁移、侵袭,且该作用与激活NF-κB通路有关。  相似文献   

10.
肝细胞生长因子在正常大鼠腰段脊髓和背根神经节的表达   总被引:1,自引:0,他引:1  
目的:观察肝细胞生长因子(hepatocyte growth factor,HGF)在大鼠脊髓和背根神经节(dorsal root ganglion,DRG)的表达。方法:取健康成年6只SD大鼠运用免疫组织化学染色技术检测HGF在腰段脊髓、背根神经节内的表达和分布。结果:在L4-6段脊髓,HGF免疫阳性产物可见于各板层神经元,尤以脊髓前角运动神经元明显;在DRG中,HGF免疫阳性物质可见于以大、中型为主的神经元的胞浆及突起中。结论:脊髓和背根神经节内的HGF通过与受体c-Met结合可能在神经再生及突触可塑性方面起一定作用。  相似文献   

11.
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   

12.
The growth factor/receptor pair HGF/c-Met exerts control on proliferation, morphogenesis and motility, and through overexpression and mutation is implicated in cancer. Here we have investigated the relationship between receptor signalling and traffic, and its control by specific PKC isotypes. It is shown that c-Met signalling to the ERK cascade occurs within endosomal compartments and that it is in this compartment that PKCepsilon specifically exerts its control on the pathway with the consequent accumulation of ERK in focal complexes. These events are clearly separated from the subsequent microtubule-dependent sorting of c-Met to its perinuclear destination, which is shown to be under the control of PKCalpha. Thus while it is shown that traffic to endosomes is essential for HGF/c-Met to trigger an ERK response, the subsequent traffic and signalling of c-Met controlled by these two PKC isotypes are unconnected events. The dynamic properties conferred by the PKCepsilon control are shown to be essential for a normal HGF-dependent migratory response. Thus PKCs are shown to control both receptor traffic and signal traffic to relay HGF/c-Met responses.  相似文献   

13.
《MABS-AUSTIN》2013,5(6):710-723
The c-Met proto-oncogene is a multifunctional receptor tyrosine kinase that is stimulated by its ligand, hepatocyte growth factor (HGF), to induce cell growth, motility and morphogenesis. Dysregulation of c-Met function, through mutational activation or overexpression, has been observed in many types of cancer and is thought to contribute to tumor growth and metastasis by affecting mitogenesis, invasion, and angiogenesis. We identified human monoclonal antibodies that bind to the extracellular domain of c-Met and inhibit tumor growth by interfering with ligand-dependent c-Met activation. We identified antibodies representing four independent epitope classes that inhibited both ligand binding and ligand-dependent activation of c-Met in A549 cells. In cells, the antibodies antagonized c-Met function by blocking receptor activation and by subsequently inducing downregulation of the receptor, translating to phenotypic effects in soft agar growth and tubular morphogenesis assays. Further characterization of the antibodies in vivo revealed significant inhibition of c-Met activity (≥ 80% lasting for 72–96 h) in excised tumors corresponded to tumor growth inhibition in multiple xenograft tumor models. Several of the antibodies identified inhibited the growth of tumors engineered to overexpress human HGF and human c-Met (S114 NIH 3T3) when grown subcutaneously in athymic mice. Furthermore, lead candidate antibody CE-355621 inhibited the growth of U87MG human glioblastoma and GTL-16 gastric xenografts by up to 98%. The findings support published pre-clinical and clinical data indicating that targeting c-Met with human monoclonal antibodies is a promising therapeutic approach for the treatment of cancer.  相似文献   

14.
The c-Met proto-oncogene is a multifunctional receptor tyrosine kinase that is stimulated by its ligand, hepatocyte growth factor (HGF), to induce cell growth, motility and morphogenesis. Dysregulation of c-Met function, through mutational activation or overexpression, has been observed in many types of cancer and is thought to contribute to tumor growth and metastasis by affecting mitogenesis, invasion, and angiogenesis. We identified human monoclonal antibodies that bind to the extracellular domain of c-Met and inhibit tumor growth by interfering with ligand-dependent c-Met activation. We identified antibodies representing four independent epitope classes that inhibited both ligand binding and ligand-dependent activation of c-Met in A549 cells. In cells, the antibodies antagonized c-Met function by blocking receptor activation and by subsequently inducing downregulation of the receptor, translating to phenotypic effects in soft agar growth and tubular morphogenesis assays. Further characterization of the antibodies in vivo revealed significant inhibition of c-Met activity (≥ 80% lasting for 72–96 h) in excised tumors corresponded to tumor growth inhibition in multiple xenograft tumor models. Several of the antibodies identified inhibited the growth of tumors engineered to overexpress human HGF and human c-Met (S114 NIH 3T3) when grown subcutaneously in athymic mice. Furthermore, lead candidate antibody CE-355621 inhibited the growth of U87MG human glioblastoma and GTL-16 gastric xenografts by up to 98%. The findings support published pre-clinical and clinical data indicating that targeting c-Met with human monoclonal antibodies is a promising therapeutic approach for the treatment of cancer.  相似文献   

15.
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   

16.
c-Met, the high affinity receptor for hepatocyte growth factor (HGF), is one of the most frequently activated tyrosine kinases in many human cancers and a target for cancer therapy. However, inhibitory targeting of c-Met with antibodies has proven difficult, because most antibodies have intrinsic agonist activity. Therefore, the strategy for reducing the agonism is critical for successful development of cancer therapies based on anti-c-Met antibodies. Here we developed a mechanism-based assay method for rapid screening of anti-c-Met antibodies, involving the determination of Akt phosphorylation and c-Met degradation for agonism and efficacy, respectively. Using the method, we identified an antibody, F46, that binds to human c-Met with high affinity (Kd = 2.56 nM) and specificity, and induces the degradation of c-Met in multiple cancer cells (including MKN45, a gastric cancer cell line) with minimal activation of c-Met signaling. F46 induced c-Met internalization in both HGF-dependent and HGF-independent cells, suggesting that the degradation of c-Met results from antibody-mediated receptor internalization. Furthermore, F46 competed with HGF for binding to c-Met, resulting in the inhibition of both HGF-mediated invasion and angiogenesis. Consistently, F46 inhibited the proliferation of MKN45 cells, in which c-Met is constitutively activated in an HGF-independent manner. Xenograft analysis revealed that F46 markedly inhibits the growth of subcutaneously implanted gastric and lung tumors. These results indicate that F46, identified by a novel mechanism-based assay, induces c-Met degradation with minimal agonism, implicating a potential role of F46 in therapy of human cancers.  相似文献   

17.
The biological behaviors of hepatocellular carcinoma (HCC) are complex mainly due to heterogeneity of progressive genetic and epigenetic mutations as well as tumor environment. Hepatocyte growth factor (HGF)/c-Met signaling pathway is regarded to be a prototypical example for stromal-epithelial interactions during developmental morphogenesis, wound healing, organ regeneration and cancer progression. And p53 plays as an important regulator of Met-dependent cell motility and invasion. Present study showed that 2 HCC cell lines, Hep3B and HepG2, displayed different invasive capacity when treated with HGF which was secreted by hepatic stellate cells (HSCs). We found that HGF promoted Hep3B cells invasion and migration as well as epithelial-mesenchymal transition (EMT) occurrence because Hep3B was p53 deficient, which leaded to the c-Met over-expression. Then we found that HGF/c-Met promoted Hep3B cells invasion and migration by upregulating Snail expression. In conclusion, HGF/c-Met signaling is enhanced by loss of p53 expression, resulting in increased ability of invasion and migration by upregulating the expression of Snail.  相似文献   

18.
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   

19.
Recent studies have demonstrated that the transplantation of bone marrow cells following diabetes induced by streptozotocin can support the recovery of pancreatic β-cell mass and a partial reversal of hyperglycemia. To address this issue, we examined whether the c-Met/hepatocyte growth factor (HGF) signaling pathway was involved in the recovery of β-cell injury after bone marrow transplantation (BMT). In this model, donor-derived bone marrow cells were positive for HGF immunoreactivity in the recipient spleen, liver, lung, and pancreas as well as in the host hepatocytes. Indeed, plasma HGF levels were maintained at a high value. The frequency of c-Met expression and its proliferative activity and differentiative response in the pancreatic ductal cells in the BMT group were greater than those in the PBS-treated group, resulting in an elevated number of endogenous insulin-producing cells. The induction of the c-Met/HGF signaling pathway following BMT promotes pancreatic regeneration in diabetic rats.  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