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 156 毫秒
1.
细胞因子信号传导抑制因子3(SOCS3)是细胞因子信号传导抑制因子蛋白质家族(SOCS)的一员。SOCS3是一种重要的细胞内蛋白质,在体内负调控细胞因子介导的信号通路,参与机体免疫、生长、造血、新陈代谢及肿瘤增殖等各种关键过程。近年的研究发现,SOCS3参与疼痛的调控,在神经病理性疼痛、炎性疼痛等多种类型疼痛及吗啡耐受中发生表达的变化。在坐骨神经慢性压迫损伤(CCI)模型中,磷酸二聚化的STAT3转移到细胞核内诱导脊髓背角SOCS3表达增加,在完全弗氏佐剂(CFA)炎性疼痛大鼠中,下丘脑室旁核(PVN)内SOCS3在急性期蛋白质表达水平增加、其慢性期表达下降,在骨癌疼痛大鼠腰2~5背根神经节(DRG)中SOCS3蛋白质水平显著下降。鞘内注射SOCS3慢病毒载体、阿司匹林触发的脂蛋白A4(ATL)和芍药苷,或通过抑制非编码RNA表达降低非编码RNA对SOCS3的抑制作用,能够增加SOCS3表达发挥镇痛作用。SOCS3通过抑制Janus激酶/信号转导子和转录激活子3(JAK/STAT3)信号通路及下游基因的表达,阻碍白细胞介素-1(IL-1)、IL-6和肿瘤坏死因子α(TNF-α)等多种炎...  相似文献   

2.
为了揭示干扰素刺激基因(interferon-stimulated gene, ISG)表达的改变对乙型肝炎病毒感染治疗效果的影响,本研究检测了干扰素刺激基因STAT1、MX和SOCS3在慢性乙型肝炎患者的外周血单核细胞(peripheral blood mononuclear cell, PBMC)和肝脏样品中的表达情况。结果显示,采用聚乙二醇干扰素(Peg-IFN)治疗后,Peg-IFN应答者的PBMC和肝组织中的STAT1和MX表达水平显著升高,而非应答者的SOCS3表达显著升高。在应答者的活组织检查中,Peg-IFN治疗24 h后细胞核中磷酸化STAT1的染色比例显著增加,而非应答者在治疗前肝细胞核染色比例较高,治疗后染色比例显著减少。此外,治疗前非应答者的肝SOCS3表达水平显著高于应答者,并且随着IFN的治疗SOCS3表达继续增加。本研究表明,STAT1和MX是Peg-IFN抗病毒免疫应答的正向调节因子,而SOCS3 (JAK/STAT途径的负调节因子)激活干扰素刺激基因的负调控,并抑制Peg-IFN的免疫应答。  相似文献   

3.
Ⅲ型干扰素(typeⅢinterferon, IFN-Ⅲ;亦称为IFN-λ)是2003年被发现的一种新型IFN,由IFN-λ1(IL-29)、IFN-λ2(IL-28A)、IFN-λ3(IL-28B)和IFN-λ4等细胞因子组成。IFN-λ通过与细胞膜上特异性受体IFN-λ受体1(IL-28RA)和IL-10R2结合,激活Janus激酶/信号转导与转录激活子等信号通路进行信号转导,从而发挥其生物学作用。IFN-λ是一种极具临床应用前景的细胞因子,与Ⅰ型IFN相比,在慢性丙型肝炎病毒(hepatitis C virus, HCV)感染中,能更好地改善病毒学应答以及提高病毒清除率,且不良反应更少。现就IFN-λ在消化道、肝脏以及呼吸道感染中的抗病毒作用作一概述。  相似文献   

4.
细胞信号转导异常可揭示人类疾病发生的本质, 一些病毒的致病机制即源于其蛋白所致宿主细胞内信号转导的紊乱. 丙型肝炎病毒(HCV)感染是引起人类严重肝脏疾病的主要病因, 但致病机制尚未明确. HCV包膜蛋白2(E2蛋白)能介导病毒吸附并结合至靶细胞表面, 此乃HCV感染的前提及首发事件. 推测HCV E2蛋白经与其受体(人CD81)的相互作用而将病毒感染信号传递至宿主细胞内, 使细胞增殖和分化异常, 从而导致感染细胞发生早期病变. 为进一步验证此致病机制, 研究了HCV E2蛋白等诸因素对差异表达人CD81的U937, Molt-4细胞内MAPK/ERK途径的影响, 结果表明, HCV E2蛋白可特异性激活细胞内MAPK/ERK途径, 而HCV E2单抗、CD81单抗、 慢性HCV感染患者血清或MAPK/ERK途径上游MEK1的抑制剂(PD98059)均不同程度地减弱或抑制HCV E2蛋白对MAPK/ERK的激活. 此外, PD98059尚可抑制HCV E2蛋白对MAPK/ERK途径下游转录因子Elk-1的活化. 研究认为, HCV E2蛋白经其相应受体引发的宿主细胞跨膜信号转导异常很可能是HCV的致病机制之一.  相似文献   

5.
6.
7.
已知丙型肝炎病毒(hepatitis C virus,HCV)可通过其蛋白酶NS3/4A切割线粒体抗病毒信号蛋白(mitochondrial antiviral signaling protein,MAVS)来逃逸天然免疫识别,但尚不清楚其切割动力学及切割在抑制干扰素中的作用。本研究旨在细胞模型中探讨HCV感染过程中病毒复制建立及病毒NS3/4A切割MAVS的动态过程,探究NS3/4A切割MAVS对病毒逃逸宿主天然免疫建立感染的贡献。首先构建基于绿色荧光蛋白(green fluorescent protein,GFP)的MAVS切割报告系统(GFP-NLS-MAVS-TM462),用 HCV Jc1-Gluc 感染Huh7.5/GFP-NLS-MAVS-TM462细胞。结果显示,病毒复制早期MAVS切割效率较低;NS3/4A高效切割MAVS发生于HCV复制晚期,且其切割效率与NS3蛋白水平相关。利用带有GFP ypet的HCV报告病毒Jc1-378-1感染Huh7.5/RFP-NLS-MAVS-TM462细胞,在单细胞水平观察HCV感染阳性细胞中MAVS被切割情况,发现HCV复制细胞中仅部分细胞MAVS被切割。进一步研究发现,不同基因型NS3/4A切割MAVS的效率仅与NS3表达水平相关。以上结果提示,HCV蛋白酶NS3/4A切割MAVS依赖NS3/4A蛋白在病毒复制过程中的累积,对在病毒复制早期逃逸宿主天然免疫建立感染可能无显著贡献。  相似文献   

8.
MicroRNA(miRNA)是一类长度约为22核苷酸的非编码小RNA,能够在转录后水平上对基因的表达进行调节,进而影响细胞活动。病毒依赖宿主细胞进行复制。宿主可以通过免疫系统抵御病毒的入侵,病毒会发展不同的策略用于逃避宿主的免疫。目前,一些病毒已经被证明可以编码miRNA来重塑细胞环境。疱疹病毒是一类双链DNA病毒,该类病毒能够在宿主体内保持长久的潜伏状态。疱疹病毒家族的很多成员都能够表达miRNA。越来越多实验结果表明,疱疹病毒的miRNA对于调节病毒潜伏以及抑制宿主的抗病毒免疫反应方面发挥着重要的作用。该文讨论了疱疹病毒所编码的miRNA的产生过程和功能,希望有助于了解疱疹病毒的潜伏感染以及病毒与宿主的相互作用。  相似文献   

9.
侯敏  刘新  张文艳 《病毒学报》2017,33(2):284-292
SOCS(Suppressor of cytokine signaling),是一种细胞因子信号通路抑制蛋白,目前认为该蛋白家族可以调节LIF(Leukemia inhibitory factor)、G-CSF(Granulocyte colony-stimulating factor)、IL-6(Interleukin-6)、IL-10(Interleukin-10)、IFN-λ(interferon-λ)等30多种细胞因子,而这些因子是机体抵抗入侵的外来病原体的主要免疫防御反应。病毒在感染宿主的过程中通过劫持宿主中的SOCS蛋白,从而对细胞中的JAK/STAT、NF-κB等与抗病毒因子调控相关的信号通路以及对T细胞的分化的调控调节病毒感染。近年来,大量的文献证实SOCS蛋白的变化与病毒感染的程度以及愈后的器官损伤具有紧密的联系,使得SOCS蛋白作为抗病毒靶点的研究尤为重要。本文主要讨论SOCS蛋白通过调控JAK-STAT、NF-κB等信号通路,在病毒感染过程中发挥的作用和作用机制。  相似文献   

10.
11.
Hepatitis C virus (HCV) infection involves a variety of viral and host factors, which leads to the dysregulation of number of relevant genes including long noncoding RNAs (LncRNAs). LncRNA urothelial carcinoma-associated 1 (UCA1) has been reported to be upregulated in HCV-infected individuals. In a bid to elucidate on the contribution of UCA1 on HCV replication, we infected Huh7.5 cells with cell culture-derived HCV and found that UCA1 expression was elevated in time- and dose-dependent manners. Functionally, UCA1 knockdown by siRNA upregulated interferon (IFN) responses, thereby increasing the expression of interferon-stimulating genes (ISGs), and subsequently suppressing HCV replication. Bioinformatics analysis and experimental results indicated that, functioning as competitive endogenous RNA, UCA1 could sponge microRNA (miR)-145-5p, which targeted suppressor of cytokine signaling 7 (SOCS7) mRNA and subsequently mediated SOCS7 silencing. Moreover, SOCS7 protein exerted an inhibitory effect on IFN responses, thereby facilitating HCV replication. Taken together, at first, our findings demonstrate that UCA1 can counteract the expression of miR-145-5p, thereby upregulating the level of SOCS7, and in turn leading to the suppression of antiviral response in Huh7.5 cells.  相似文献   

12.
Influenza A virus (IAV) has evolved multiple mechanisms to compromise type I interferon (IFN) responses. The antiviral function of IFN is mainly exerted by activating the JAK/STAT signalling and subsequently inducing IFN‐stimulated gene (ISG) production. However, the mechanism by which IAV combat the type I IFN signalling pathway is not fully elucidated. In this study, we explored the roles of human microRNAs modulated by IAV infection in type I IFN responses. We demonstrated that microRNA‐30 (miR‐30) family members were downregulated by IAV infection. Our data showed that the forced expression of miR‐30 family members inhibited IAV proliferation, while miR‐30 family member inhibitors promoted IAV proliferation. Mechanistically, we found that miR‐30 family members targeted and reduced SOCS1 and SOCS3 expression, and thus relieved their inhibiting effects on IFN/JAK/STAT signalling pathway. In addition, miR‐30 family members inhibited the expression of NEDD4, a negative regulator of IFITM3, which is important for host defence against influenza viruses. Our findings suggest that IAV utilises a novel strategy to restrain host type I IFN‐mediated antiviral immune responses by decreasing the expression of miR‐30 family members, and add a new way to understand the mechanism of immune escape caused by influenza viruses.  相似文献   

13.
Effective recognition of viral infection and subsequent triggering of antiviral innate immune responses are essential for the host antiviral defense, which is tightly regulated by multiple regulators, including microRNAs. Our previous study showed that a panel of microRNAs, including miR-155, was markedly upregulated in macrophages upon vesicular stomatitis virus infection; however, the biological function of miR-155 during viral infection remains unknown. In this paper, we show that RNA virus infection induces miR-155 expression in macrophages via TLR/MyD88-independent but retinoic acid-inducible gene I/JNK/NF-κB-dependent pathway. And the inducible miR-155 feedback promotes type I IFN signaling, thus suppressing viral replication. Furthermore, suppressor of cytokine signaling 1 (SOCS1), a canonical negative regulator of type I IFN signaling, is targeted by miR-155 in macrophages, and SOCS1 knockdown mediates the enhancing effect of miR-155 on type I IFN-mediated antiviral response. Therefore, we demonstrate that inducible miR-155 feedback positively regulates host antiviral innate immune response by promoting type I IFN signaling via targeting SOCS1.  相似文献   

14.
15.
16.
17.
18.
19.
Although the use of IFN-alpha in combination with ribavirin has improved the treatment efficacy of chronic hepatitis C virus (HCV) infection, 20-50% of patients still fail to eradicate the virus depending on the HCV genotype. Recently, overexpression of HCV core protein has been shown to inhibit IFN signaling and induce SOCS-3 expression. Aim of this study was to examine the putative role of SOCS proteins in IFN resistance. By Western blot analysis, a 4-fold induction of STAT-1/3 phosphorylation by IFN-alpha was observed in mock-transfected HepG2 clones. In contrast, IFN-induced STAT-1/3 phosphorylation was considerably downregulated by SOCS-1/3 overexpression. In mock-transfected cells, IFN-alpha induced 2',5'-OAS and myxovirus resistance A (MxA) promoter activity 40- to 80-fold and 10- to 35-fold, respectively, and this effect was abrogated in SOCS-1/3 overexpressing cells. As detected by Northern blot technique, IFN-alpha potently induced 2',5'-OAS and MxA mRNA expression in the control clones. Overexpression of SOCS-1 completely abolished both 2',5'-OAS and MxA mRNA expression, whereas SOCS-3 mainly inhibited 2',5'-OAS mRNA expression. Our results demonstrate that SOCS-1 and SOCS-3 proteins inhibit IFN-alpha-induced activation of the Jak-STAT pathway and expression of the antiviral proteins 2',5'-OAS and MxA. These data suggest a potential role of SOCS proteins in IFN resistance during antiviral treatment.  相似文献   

20.

Background

Epstein-Barr virus is recognized to cause lymphoproliferative disorders and is also associated with cancer. Evidence suggests that monocytes are likely to be involved in EBV pathogenesis, especially due to a number of cellular functions altered in EBV-infected monocytes, a process that may affect efficient host defense. Because type I interferons (IFNs) are crucial mediators of host defense against viruses, we investigated the effect of EBV infection on the IFNα pathway in primary human monocytes.

Methodology/Principal Findings

Infection of monocytes with EBV induced IFNα secretion but inhibited the positive feedback loop for the amplification of IFNα. We showed that EBV infection induced the expression of suppressor of cytokine signaling 3 (SOCS3) and, to a lesser extent, SOCS1, two proteins known to interfere with the amplification of IFNα secretion mediated by the JAK/STAT signal transduction pathway. EBV infection correlated with a blockage in the activation of JAK/STAT pathway members and affected the level of phosphorylated IFN regulatory factor 7 (IRF7). Depletion of SOCS3, but not SOCS1, by small interfering RNA (siRNA) abrogated the inhibitory effect of EBV on JAK/STAT pathway activation and significantly restored IFNα secretion. Finally, transfection of monocytes with the viral protein Zta caused the upregulation of SOCS3, an event that could not be recapitulated with mutated Zta.

Conclusions/Significance

We propose that EBV protein Zta activates SOCS3 protein as an immune escape mechanism that both suppresses optimal IFNα secretion by human monocytes and favors a state of type I IFN irresponsiveness in these cells. This immunomodulatory effect is important to better understand the aspects of the immune response to EBV.  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