AIM To investigate the role of suppressor of cytokine signaling 1(SOCS1)in regulating MET-mediated invasive potential of hepatocellular carcinoma(HCC)cells.METHODSStable derivatives of mouse(Hepa1-6)and human(hep3B,He...AIM To investigate the role of suppressor of cytokine signaling 1(SOCS1)in regulating MET-mediated invasive potential of hepatocellular carcinoma(HCC)cells.METHODSStable derivatives of mouse(Hepa1-6)and human(hep3B,Hep G2)HCC cell lines expressing SOCS1or control vector were evaluated for their ability to migrate towards hepatocyte growth factor(HGF)in the transwell migration assay,invade extracellular matrix in response to HGF stimulation in a 3-D invasion assay by confocal microscopy,and to undergo anchorageindependent proliferation in semisolid agar.Following intravenous and intrasplenic inoculation into NOD.scid.gamma mice,the ability of Hepa cells to form othotopic tumors was evaluated.Following HGF stimulation of Hepa and Hep3B cells,expression of proteins implicated in epithelial-to-mesenchymal transition was evaluated by western blot and qR T-PCR.RESULTS SOCS1 expression in mouse and human HCC cells inhibited HGF-induced migration through matrigel.In the 3-D invasion assay,HGF stimulation induced invasion of HCC cells across type-Ⅰcollagen matrix,and SOCS1expression significantly reduced the depth of invasion.SOCS1 expression also reduced the number and size of colonies formed by anchorage-independent growth in semisolid agar.Following intravenous inoculation,control Hepa cell formed large tumor nodules that obliterated the liver whereas the SOCS1-expressing Hepa cells formed significantly smaller nodules.Tumors formed by SOCS1-expressing cells showed reduced phosphorylation of STAT3 and ERK that was accompanied by reduced levels of MET protein expression.HGF stimulated Hepa cells expressing SOCS1 showed increased expression of E-cadherin and decreased expression of EGR1,SNAI1and ZEB1.Comparable results were obtained with Hep3B cells.SOCS1 expressing HCC cells also showed reduced levels of EGR1 and SNAI1 transcripts.CONCLUSION Our findings indicate that loss of SOCS1-dependent control over epithelial-to-mesenchymal transition may contribute to MET-mediated migration,invasion and metastatic growth of HCC.展开更多
High mobility group box 1 protein (HMGB1) is a highly conserved, ubiquitous protein in the nuclei and cytoplasm of nearly all cell types. HMGB1 is secreted into the extracellular milieu and acts as a proinflammatory...High mobility group box 1 protein (HMGB1) is a highly conserved, ubiquitous protein in the nuclei and cytoplasm of nearly all cell types. HMGB1 is secreted into the extracellular milieu and acts as a proinflammatory cytokine. In this article we reviewed briefly the cellular immune response mediated by HMGB1 in inflammation and sepsis. This systemic review is mainly based on our own work and other related reports HMGB1 can actively affect the immune functions of many types of cells including T lymphocytes, regulatory T cells (Tregs), dendritic cells (DCs), macrophages, and natural killer cells (NK cells). Various cellular responses can be mediated by HMGB1 which binds to cell-surface receptors [e.g., the receptor for advanced glycation end products (RAGE), Toll-like receptor (TLR)2, and TLR4]. Anti-HMGB1 treatment, such as anti-HMGB1 polyclonal or monoclonal antibodies, inhibitors (e.g., ethyl pyruvate) and antagonists (e.g., A box), can protect against sepsis lethality and give a wider window for the treatment opportunity. HMGB1 is an attractive target for the development of new therapeutic strategies in the treatment of patients with septic complications.展开更多
Background:Hepatocellular carcinoma(HCC)is one of the most prevalent human cancers with high mortality.Long non-coding RNA heart and neural crest derivatives expressed 2 anti-sense 1(HAND2-AS1)is down-regulated in sev...Background:Hepatocellular carcinoma(HCC)is one of the most prevalent human cancers with high mortality.Long non-coding RNA heart and neural crest derivatives expressed 2 anti-sense 1(HAND2-AS1)is down-regulated in several cancers including HCC,yet the precise mechanisms how HAND2-AS1 regulates cell survival in HCC remains poorly understood.Methods:The expression levels of HAND2-AS1 and miR-300 were measured using quantitative real-time PCR.The protein levels of suppressor of cytokine signaling 5(SOCS5),Bcl-2,Bax and cleaved caspase-3 were determined by Western blot.Cell viability and cell proliferation were assessed using cell counting kit-8 and clone formation assay,respectively.Cell apoptosis was detected using flow cytometry.The interactions between HAND2-AS1 and miR-300,miR-300 and SOCS5 were validated using luciferase reporter assay.Results:HAND2-AS1 was down-regulated in HCC tissues and cell lines,and the expression level of HAND2-AS1 was positively correlated to patient survival.HAND2-AS1 over-expression reduced viability and proliferation in HCC cells.Elevated HAND2-AS1 level induced apoptosis in HCC cells,accompanied with increased Bax and cleaved caspase-3 levels and decreased Bcl-2 level.We also validated that HAND2-AS1 acted as a sponge of miR-300,and there was a negative correlation between expression levels of HAND2-AS1 and miR-300 in HCC tissues.Furthermore,we found that SOCS5 was a downstream target of miR-300.In addition,miR-300 mimics abolished HAND2-AS1-mediated inhibition of cell viability and proliferation.miR-300 mimics also reversed the HAND2-AS1-induced apoptosis in HCC cells.Conclusion:lncRNA HAND2-AS1 inhibits proliferation in HCC through regulating miR-300/SOCS5 axis.展开更多
目的探讨染色体区域稳定蛋白1(chromosomal region maintenance 1,CRM1)与细胞因子信号传导抑制因子1(suppressor of cytokine signaling 1,SOCS1)表达与胃癌演进、临床病理特征及预后的关系。方法采用免疫组化检测低级别上皮内瘤变胃组...目的探讨染色体区域稳定蛋白1(chromosomal region maintenance 1,CRM1)与细胞因子信号传导抑制因子1(suppressor of cytokine signaling 1,SOCS1)表达与胃癌演进、临床病理特征及预后的关系。方法采用免疫组化检测低级别上皮内瘤变胃组织27例、高级别上皮内瘤变胃组织26例,以及进展期胃癌组织67例及对应癌旁组织67例中CRM1及SOCS1蛋白表达水平,分析两者在不同病变胃黏膜中的变化以及与胃癌临床病理特征的关系,通过在线分析工具Kaplan-Meier Plotter、单因素及多因素Cox分析影响胃癌患者预后的相关因素。结果胃癌组织中CRM1表达高于癌旁组织,SOCS1表达低于癌旁组织及瘤变组织(P<0.05);CRM1在胃癌组织高表达与浸润深度及淋巴结转移有关(P<0.05);生存分析显示高表达CRM1提示预后不良(P<0.05),SOCS1表达水平与预后无关(P>0.05);在胃癌组织中,CRM1与SOCS1表达无显著相关性(R=-0.025,P=0.842)。结论CRM1和SOCS1参与胃癌发生,CRM1高表达可能参与胃癌侵袭及转移等恶性进展。展开更多
基金Supported by the Cancer Research Society,Montreal,Canada,No.16195
文摘AIM To investigate the role of suppressor of cytokine signaling 1(SOCS1)in regulating MET-mediated invasive potential of hepatocellular carcinoma(HCC)cells.METHODSStable derivatives of mouse(Hepa1-6)and human(hep3B,Hep G2)HCC cell lines expressing SOCS1or control vector were evaluated for their ability to migrate towards hepatocyte growth factor(HGF)in the transwell migration assay,invade extracellular matrix in response to HGF stimulation in a 3-D invasion assay by confocal microscopy,and to undergo anchorageindependent proliferation in semisolid agar.Following intravenous and intrasplenic inoculation into NOD.scid.gamma mice,the ability of Hepa cells to form othotopic tumors was evaluated.Following HGF stimulation of Hepa and Hep3B cells,expression of proteins implicated in epithelial-to-mesenchymal transition was evaluated by western blot and qR T-PCR.RESULTS SOCS1 expression in mouse and human HCC cells inhibited HGF-induced migration through matrigel.In the 3-D invasion assay,HGF stimulation induced invasion of HCC cells across type-Ⅰcollagen matrix,and SOCS1expression significantly reduced the depth of invasion.SOCS1 expression also reduced the number and size of colonies formed by anchorage-independent growth in semisolid agar.Following intravenous inoculation,control Hepa cell formed large tumor nodules that obliterated the liver whereas the SOCS1-expressing Hepa cells formed significantly smaller nodules.Tumors formed by SOCS1-expressing cells showed reduced phosphorylation of STAT3 and ERK that was accompanied by reduced levels of MET protein expression.HGF stimulated Hepa cells expressing SOCS1 showed increased expression of E-cadherin and decreased expression of EGR1,SNAI1and ZEB1.Comparable results were obtained with Hep3B cells.SOCS1 expressing HCC cells also showed reduced levels of EGR1 and SNAI1 transcripts.CONCLUSION Our findings indicate that loss of SOCS1-dependent control over epithelial-to-mesenchymal transition may contribute to MET-mediated migration,invasion and metastatic growth of HCC.
基金supported,in part,by grants from the National Natural Science Foundation(Nos.81130035,30901561,30971192,81071545)the National Basic Research Program of China(No.2012CB518102)the China Postdoctoral Science Foundation(Nos.20100480347,201104125)
文摘High mobility group box 1 protein (HMGB1) is a highly conserved, ubiquitous protein in the nuclei and cytoplasm of nearly all cell types. HMGB1 is secreted into the extracellular milieu and acts as a proinflammatory cytokine. In this article we reviewed briefly the cellular immune response mediated by HMGB1 in inflammation and sepsis. This systemic review is mainly based on our own work and other related reports HMGB1 can actively affect the immune functions of many types of cells including T lymphocytes, regulatory T cells (Tregs), dendritic cells (DCs), macrophages, and natural killer cells (NK cells). Various cellular responses can be mediated by HMGB1 which binds to cell-surface receptors [e.g., the receptor for advanced glycation end products (RAGE), Toll-like receptor (TLR)2, and TLR4]. Anti-HMGB1 treatment, such as anti-HMGB1 polyclonal or monoclonal antibodies, inhibitors (e.g., ethyl pyruvate) and antagonists (e.g., A box), can protect against sepsis lethality and give a wider window for the treatment opportunity. HMGB1 is an attractive target for the development of new therapeutic strategies in the treatment of patients with septic complications.
文摘Background:Hepatocellular carcinoma(HCC)is one of the most prevalent human cancers with high mortality.Long non-coding RNA heart and neural crest derivatives expressed 2 anti-sense 1(HAND2-AS1)is down-regulated in several cancers including HCC,yet the precise mechanisms how HAND2-AS1 regulates cell survival in HCC remains poorly understood.Methods:The expression levels of HAND2-AS1 and miR-300 were measured using quantitative real-time PCR.The protein levels of suppressor of cytokine signaling 5(SOCS5),Bcl-2,Bax and cleaved caspase-3 were determined by Western blot.Cell viability and cell proliferation were assessed using cell counting kit-8 and clone formation assay,respectively.Cell apoptosis was detected using flow cytometry.The interactions between HAND2-AS1 and miR-300,miR-300 and SOCS5 were validated using luciferase reporter assay.Results:HAND2-AS1 was down-regulated in HCC tissues and cell lines,and the expression level of HAND2-AS1 was positively correlated to patient survival.HAND2-AS1 over-expression reduced viability and proliferation in HCC cells.Elevated HAND2-AS1 level induced apoptosis in HCC cells,accompanied with increased Bax and cleaved caspase-3 levels and decreased Bcl-2 level.We also validated that HAND2-AS1 acted as a sponge of miR-300,and there was a negative correlation between expression levels of HAND2-AS1 and miR-300 in HCC tissues.Furthermore,we found that SOCS5 was a downstream target of miR-300.In addition,miR-300 mimics abolished HAND2-AS1-mediated inhibition of cell viability and proliferation.miR-300 mimics also reversed the HAND2-AS1-induced apoptosis in HCC cells.Conclusion:lncRNA HAND2-AS1 inhibits proliferation in HCC through regulating miR-300/SOCS5 axis.
文摘目的探讨染色体区域稳定蛋白1(chromosomal region maintenance 1,CRM1)与细胞因子信号传导抑制因子1(suppressor of cytokine signaling 1,SOCS1)表达与胃癌演进、临床病理特征及预后的关系。方法采用免疫组化检测低级别上皮内瘤变胃组织27例、高级别上皮内瘤变胃组织26例,以及进展期胃癌组织67例及对应癌旁组织67例中CRM1及SOCS1蛋白表达水平,分析两者在不同病变胃黏膜中的变化以及与胃癌临床病理特征的关系,通过在线分析工具Kaplan-Meier Plotter、单因素及多因素Cox分析影响胃癌患者预后的相关因素。结果胃癌组织中CRM1表达高于癌旁组织,SOCS1表达低于癌旁组织及瘤变组织(P<0.05);CRM1在胃癌组织高表达与浸润深度及淋巴结转移有关(P<0.05);生存分析显示高表达CRM1提示预后不良(P<0.05),SOCS1表达水平与预后无关(P>0.05);在胃癌组织中,CRM1与SOCS1表达无显著相关性(R=-0.025,P=0.842)。结论CRM1和SOCS1参与胃癌发生,CRM1高表达可能参与胃癌侵袭及转移等恶性进展。