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 349 毫秒
1.
Toll样受体2(Toll-like receptor 2,TLR2)是由胞内段、跨膜段和胞外段组成的单次跨膜受体,也是固有免疫中重要的模式识别受体(pattern recognition receptor,PRR)。可溶性Toll样受体2(soluble Tolllike receptor 2,s TLR2)是TLR2胞外段脱落生成的可溶性蛋白。s TLR2作为TLR2的特异性调节分子,充当诱饵受体与配体结合,抑制TLR2信号,进而减轻炎症,减少过度免疫造成的损伤。近年来,s TLR2在越来越广泛的体液中被检测出来,且在很多疾病中出现明显异常的表达,成为这些疾病潜在的诊断指标或治疗工具。本文简要综述了s TLR2的生成、结构、功能及其与疾病的关系等。  相似文献   

2.
金丹  吕凤林 《病毒学报》2005,21(2):155-159
1984年,果蝇的Toll样蛋白被发现。13年后,人们发现了第一个与果蝇Toll蛋白同源的人Toll样受体(toll like receptor,TLR)蛋白(hTLR4)。迄今,已有10种TLR蛋白(即hTLR1~10)被发现,它们代表了一类保守的受体家族,分别识别不同的抗原。其中TLR3能专一性地识别双链RNA(double stranded RNA,dsRNA),通过依赖MyD88的信号通路及不依赖MyD88的信号通路,  相似文献   

3.
Toll样受体4与肝损伤的研究进展   总被引:1,自引:0,他引:1  
Wang JQ  Li J  Zou YH 《生理科学进展》2007,38(4):365-368
Toll样受体4((toll like receptor4,TLR4)是内毒素(LPS)的关键受体,为Toll蛋白家族中的一个成员,是联系固有免疫和适应性免疫的纽带。TLR4主要表达于髓源性细胞,其启动的胞内信号转导在肝损伤的发生和发展过程中发挥重要作用。这一信号转导途径主要通过NF-κB、p38、JNK等的激活,使细胞产生炎症转录因子,介导肝脏炎症。TLR4与氧化应激的相互作用,使得肝脏对TLR4的配体及细胞因子的敏感性增加,从而加重肝脏损伤。随着TLR4在肝损伤中的作用进一步阐明,其在肝脏疾病中的治疗作用将会产生广阔的应用前景。  相似文献   

4.
髓样分化蛋白-2在天然免疫中的作用   总被引:1,自引:0,他引:1  
Xu FL  Li L 《生理科学进展》2004,35(2):139-142
Toll样受体 (Toll likereceptor ,TLR)家族作为模式识别受体 ,在天然免疫中具有重要作用。髓样分化蛋白 2 (myeloiddifferentialprotein 2 ,MD 2 )可能含有两个相对独立的功能结构域 ,既能与Toll样受体家族中的TLR4、TLR2结合 ,也能与多种配体结合 (包括lipopolysaccharide ,LPS)。这种特殊的结构可能与其三方面的主要功能有关 :(1)MD 2与TLR4结合 ,赋予TLR4对各种配体 (包括LPS)的反应性 ;(2 )MD 2与TLR2结合 ,赋予TLR2对LPS的反应性 ,并增强TLR2对细菌及其胞壁成分的反应性 ;(3)MD 2能促进TLR4和TLR2的表达 ,并且与TLR4在细胞内的分布密切相关。这表明MD 2可以通过两种方式直接或间接调控TLRs的功能 :与TLR2 /TLR4结合 ,或调控TLR2 /TLR4的表达与分布。因而MD 2不仅仅是TLR4的辅助分子 ,而且还是天然免疫中的调控分子 ,可能在感染、炎症、免疫等病理生理过程中具有更广泛的生物学功能  相似文献   

5.
肽聚糖(peptidoglycan, PGN)是细菌细胞壁的主要结构,与体内多种受体信号分子如核苷酸结合寡聚化结构域蛋白1/2(nucleotide-binding oligomerization domain protein 1/2, NOD1/2)、Toll样受体2(toll like receptor 2, TLR2)和PGN识别蛋白(peptidoglycan recognition proteins, PGRP)等结合后,参与人体如发烧、睡眠和骨生成等生理功能。研究发现,PGN作为病原微生物细菌的结构成分,在临床血流感染和自身免疫疾病诊断中也发挥着重要作用。现就PGN在人体内的代谢、功能及疾病诊断应用价值作一概述。  相似文献   

6.
<正>白细胞介素1受体相关激酶1旁路引发和直接连接Toll样受体至快速核苷酸结合寡聚化结构域样受体含热蛋白结构域3炎性体激活致病性感染和组织损伤引发炎性体的组装,这种胞质蛋白复合物激活半胱天冬酶-1(caspase-1),促进白细胞介素1β前体(pro-IL-1β)和pro-IL-18的切割成熟,还能调节caspase-1依赖的程序性细胞死亡(pyroptosis)。众所周知,微生物识别Toll样受体(TLR)主要是诱导激活caspase-1,但已知的TLR功能仅限于上调炎性体的作用。感染致命微生物后,TLR和核苷酸结合寡聚化结构域样受体(NLR)有可能同时  相似文献   

7.
易世杰  赵礼金 《蛇志》2013,25(2):183-187
Toll样受体(toll-like receptors,TLRs)因其积极的研究成果而成为近年来广受关注的一种病原体识别受体,TLRs分布相对比较广泛,不但在小肠上皮、呼吸上皮细胞表达,同时也在血管内皮细胞、树突状细胞[1]、大鼠脾及心肌细胞[2]等细胞中表达.研究证实,它属于模式识别受体(pattern recognition receptors,PRRs),病原相关分子模式(pathogen-associated molecule pattern,PAMPs)可被其辨别,然后引发一系列的信号转导,TLRs 是备受关注的一种PRRs.Toll样受体4(toll-like receptor 4,TLR4)是TLRs家族中极为重要的成员,是天然免疫系统识别病原微生物的主要受体,在天然免疫反应中扮演着关键性作用.细菌脂多糖(lipopolysaccharide,LPS)作为一类受体,主要作用是介导信号跨膜转导,尤其对革兰氏阴性菌所引起的感染性炎症起着极为关键的作用.由于近年来对TLR4介导的信号转导及TLR4与疾病的关系研究成为热点,本文就TLR4的信号转导、TLR4与LPS的关系及TLR4信号通路调节进行综述如下.  相似文献   

8.
HMGB1在缺血-再灌注损伤中的作用机制研究进展   总被引:1,自引:0,他引:1  
高迁移率族蛋白B1(high mobility group protein B1,HMGB1)是一类核内非组蛋白,人HMGB1含有215个氨基酸残基,定位于人染色体13q12,它在核内主要参与维持核小体的结构、识别和结合DNA、调控DNA的复制转录等.HMGB1主要通过两种途径释放到细胞外,与Toll样受体、晚期糖基化终产物受体等受体相互作用激活核因子κB、有丝分裂原激活蛋白激酶p38信号通路,释放炎症介质参与缺血-再灌注损伤的发生发展.对HMGB1作用的生物学基础的深入研究,将提供治疗缺血-再灌注损伤的新靶点.  相似文献   

9.
动脉粥样硬化是一种慢性免疫炎症性疾病,它与自身的先天性免疫和适应性免疫密切相关。Toll样受体(Toll-like receptors,TLR)作为激活非特异性免疫的重要受体蛋白,可以识别病原微生物,激活免疫反应。Toll样受体9是TLR家族中的重要一员,是先天免疫系统中识别细菌和病毒Cp G DNA的重要受体,其与动脉粥样硬化(atherosclerosis,AS)的发生发展紧密相关。研究发现,TLR9与动脉粥样硬化的发生、发展(内皮受损和泡沫化细胞形成)密切相关,但也有研究发现TLR9在AS进程中具有潜在的保护效应。本文对Toll样受体9与动脉粥硬化疾病之间关系做一个简要的阐述,简明的总结了TLR9与树突细胞及自噬之间的联系,并为其作为靶点治疗动脉粥样硬化提供新的思路。  相似文献   

10.
Toll样受体介导的信号转导通路在对抗外来病原体的天然免疫应答中起重要作用。Toll样受体是一个天然模板识别受体家族,能识别固有性模板(微生物和哺乳动物所共有的病原相联的分子模板PAMPs)。Toll样受体通过巨噬细胞和其他免疫细胞来识别,其中TLR4识别内毒素、TLR2识别肽聚糖、TLR9识别细菌DNA、TLR5识别鞭毛蛋白、TLR3识别双链RNA等。本探讨了多种Toll受体家族成员在动物体内识别机理及功能,概述了其应用研究进展。  相似文献   

11.
12.
13.
The nuclear protein high mobility group box protein 1 (HMGB1) promotes inflammation upon extracellular release. HMGB1 induces proinflammatory cytokine production in macrophages via Toll-like receptor (TLR)-4 signaling in a redox-dependent fashion. Independent of its redox state and endogenous cytokine-inducing ability, HMGB1 can form highly immunostimulatory complexes by interaction with certain proinflammatory mediators. Such complexes have the ability to enhance the induced immune response up to 100-fold, compared with induction by the ligand alone. To clarify the mechanisms for these strong synergistic effects, we studied receptor requirements. Interleukin (IL)-6 production was assessed in supernatants from cultured peritoneal macrophages from mice each deficient in one of the HMGB1 receptors (receptor for advanced glycation end products [RAGE], TLR2 or TLR4) or from wild-type controls. The cultures were stimulated with the TLR4 ligand lipopolysaccaride (LPS), the TLR2 ligand Pam3CysSerLys4 (Pam3CSK4), noninflammatory HMGB1 or each TLR ligand in complex with noninflammatory HMGB1. The activity of the HMGB1-TLR ligand complexes relied on engagement of the same receptor as for the noncomplexed TLR ligand, since HMGB1-LPS complexes used TLR4 and HMGB1-Pam3CSK4 complexes used TLR2. Deletion of any of the intracellular adaptor molecules used by TLR2 (myeloid differentiation factor-88 [MyD88], TIR domain–containing adaptor protein [TIRAP]) or TLR4 (MyD88, TIRAP, TIR domain–containing adaptor-inducing interferon-β [TRIF], TRIF-related adaptor molecule [TRAM]) had similar effects on HMGB1 complex activation compared with noncomplexed LPS or Pam3CSK4. This result implies that the enhancing effects of HMGB1-partner molecule complexes are not regulated by the induction of additional signaling cascades. Elucidating HMGB1 receptor usage in processes where HMGB1 acts alone or in complex with other molecules is essential for the understanding of basic HMGB1 biology and for designing HMGB1-targeted therapies.  相似文献   

14.
15.
Phagocytosis of apoptotic cells by macrophages, known as efferocytosis, is a critical process in the resolution of inflammation. High mobility group box 1 (HMGB1) protein was first described as a nuclear nonhistone DNA-binding protein, but is now known to be secreted by activated cells during inflammatory processes, where it participates in diminishing efferocytosis. Although HMGB1 is known to undergo modification when secreted, the effect of such modifications on the inhibitory actions of HMGB1 during efferocytosis have not been reported. In the present studies, we found that HMGB1 secreted by Toll-like receptor 4 (TLR4) stimulated cells is highly poly(ADP-ribosyl)ated (PARylated). Gene deletion of poly(ADP)-ribose polymerase (PARP)-1 or pharmacological inhibition of PARP-1 decreased the release of HMGB1 from the nucleus to the extracellular milieu after TLR4 engagement. Preincubation of macrophages or apoptotic cells with HMGB1 diminished efferocytosis through mechanisms involving binding of HMGB1 to phosphatidylserine on apoptotic cells and to the receptor for advanced glycation end products (RAGE) on macrophages. Preincubation of either macrophages or apoptotic cells with PARylated HMGB1 inhibited efferocytosis to a greater degree than exposure to unmodified HMGB1, and PARylated HMGB1 demonstrated higher affinity for phosphatidylserine and RAGE than unmodified HMGB1. PARylated HMGB1 had a greater inhibitory effect on Ras-related C3 botulinum toxin substrate 1 (Rac-1) activation in macrophages during the uptake of apoptotic cells than unmodified HMGB1. The present results, showing that PARylation of HMGB1 enhances its ability to inhibit efferocytosis, provide a novel mechanism by which PARP-1 may promote inflammation.  相似文献   

16.
Toll-like receptor 4 (TLR4) is ubiquitously expressed on parenchymal and immune cells of the liver and is the most studied TLR responsible for the activation of proinflammatory signaling cascades in liver ischemia and reperfusion (I/R). Since pharmacological inhibition of TLR4 during the sterile inflammatory response of I/R has not been studied, we sought to determine whether eritoran, a TLR4 antagonist trialed in sepsis, could block hepatic TLR4-mediated inflammation and end organ damage. When C57BL/6 mice were pretreated with eritoran and subjected to warm liver I/R, there was significantly less hepatocellular injury compared to control counterparts. Additionally, we found that eritoran is protective in liver I/R through inhibition of high-mobility group box protein B1 (HMGB1)-mediated inflammatory signaling. When eritoran was administered in conjunction with recombinant HMGB1 during liver I/R, there was significantly less injury, suggesting that eritoran blocks the HMGB1–TLR4 interaction. Not only does eritoran attenuate TLR4-dependent HMGB1 release in vivo, but this TLR4 antagonist also dampened HMGB1’s release from hypoxic hepatocytes in vitro and thereby weakened HMGB1’s activation of innate immune cells. HMGB1 signaling through TLR4 makes an important contribution to the inflammatory response seen after liver I/R. This study demonstrates that novel blockade of HMGB1 by the TLR4 antagonist eritoran leads to the amelioration of liver injury.  相似文献   

17.
TLR9(Toll-likereceptor9)是一种微生物病原相关分子结构模式识别受体,TLR9能够识别CpG—ODN(胞嘧啶磷酸鸟甘-寡聚脱氧核苷酸),使病原相关受体在先天性免疫细胞上表达,并激活下游炎性通路。研究表明,TLR9在先天性免疫反应中产生了重要作用,如脓毒血症、自身免疫性疾病、刀豆体球蛋白A介导肝炎性肝脏损伤、炎性泡沫细胞形成、缺血再灌注损伤等,并且与多种致病因子相关联,如肝x受体、甲酰多肽受体、线粒体DNA等。  相似文献   

18.
IntroductionHigh mobility group box-1 (HMGB1), a typical damage-associated molecular pattern (DAMP) protein, is associated with inflammatory conditions and tissue damage. Our recent study found that circulating HMGB1 levels could reflect the disease activity of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). The current study aimed to investigate whether HMGB1 participated in ANCA-induced neutrophil activation, which is one of the most important pathogenic aspects in the development of AAV.MethodsThe various effects of HMGB1 in ANCA-induced neutrophil activation were measured. Antagonists for relevant receptors and signaling molecules were employed.ResultsANCA antigens translocation on neutrophils primed with HMGB1 was significantly higher than non-primed neutrophils. The levels of respiratory burst and degranulation increased significantly in HMGB1-primed neutrophils activated with ANCA-positive IgG, as compared with non-primed neutrophils. Furthermore, blocking Toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE), rather than TLR2, resulted in a significant decrease in HMGB1-induced ANCA antigens translocation, respiratory burst and degranulation. Similar effects were also found when blocking MyD88 and NF-κB.ConclusionsHMGB1 could prime neutrophils by increasing ANCA antigens translocation, and the primed neutrophils could be further induced by ANCA, resulting in the respiratory burst and degranulation. This process is TLR4- and RAGE-dependent through the MyD88/NF-κB pathway.

Electronic supplementary material

The online version of this article (doi:10.1186/s13075-015-0587-4) contains supplementary material, which is available to authorized users.  相似文献   

19.
Survival rates for patients with pulmonary hypertension (PH) remain low, and our understanding of the mechanisms involved are incomplete. Here we show in a mouse model of chronic hypoxia (CH)-induced PH that the nuclear protein and damage-associate molecular pattern molecule (DAMP) high mobility group box 1 (HMGB1) contributes to PH via a Toll-like receptor 4 (TLR4)-dependent mechanism. We demonstrate extranuclear HMGB1 in pulmonary vascular lesions and increased serum HMGB1 in patients with idiopathic pulmonary arterial hypertension. The increase in circulating HMGB1 correlated with mean pulmonary artery pressure. In mice, we similarly detected the translocation and release of HMGB1 after exposure to CH. HMGB1-neutralizing antibody attenuated the development of CH-induced PH, as assessed by measurement of right ventricular systolic pressure, right ventricular hypertrophy, pulmonary vascular remodeling and endothelial activation and inflammation. Genetic deletion of the pattern recognition receptor TLR4, but not the receptor for advanced glycation end products, likewise attenuated CH-induced PH. Finally, daily treatment of mice with recombinant human HMGB1 exacerbated CH-induced PH in wild-type (WT) but not Tlr4−/− mice. These data demonstrate that HMGB1-mediated activation of TLR4 promotes experimental PH and identify HMGB1 and/or TLR4 as potential therapeutic targets for the treatment of PH.  相似文献   

20.
Toll-like receptor (TLR) family plays a key role in innate immunity and various inflammatory responses. TLR4, one of the well-characterized pattern-recognition receptors, can be activated by endogenous damage-associated molecular pattern molecules such as high mobility group box 1 (HMGB1) to sustain sterile inflammation. Evidence suggested that blockade of TLR4 signaling may confer protection against abdominal aortic aneurysm (AAA). Herein we aimed to obtain further insight into the mechanism by which TLR4 might promote aneurysm formation. Characterization of the CaCl2-induced AAA model in mice revealed that upregulation of TLR4 expression, localized predominantly to vascular smooth muscle cells (VSMCs), was followed by a late decline during a 28-day period of AAA development. In vitro, TLR4 expression was increased in VSMCs treated with HMGB1. Knockdown of TLR4 by siRNA attenuated HMGB1-enhanced production of proinflammatory cytokines, specifically interleukin-6 and monocyte chemoattractant protein-1 (MCP-1), and matrix-degrading matrix metalloproteinase (MMP)-2 from VSMCs. In vivo, two different strains of TLR4-deficient (C57BL/10ScNJ and C3H/HeJ) mice were resistant to CaCl2-induced AAA formation compared to their respective controls (C57BL/10ScSnJ and C3H/HeN). Knockout of TLR4 reduced interleukin-6 and MCP-1 levels and HMGB1 expression, attenuated macrophage accumulation, and eventually suppressed MMP production, elastin destruction and VSMC loss. Finally, human AAA exhibited higher TLR4 expression that was localized to VSMCs. These data suggest that TLR4 signaling contributes to AAA formation by promoting a proinflammatory status of VSMCs and by inducing proteinase release from VSMCs during aneurysm initiation and development.  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