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 109 毫秒
1.
细胞焦亡是由gasdermin家族蛋白D(gasdermin D, GSDMD)介导的一种新型程序性细胞死亡方式,表现为早期细胞凋亡样染色质凝结和DNA断裂,之后形成细胞膜孔,细胞肿胀,膜破裂,导致细胞内容物和促炎介质的释放。细胞焦亡途径主要包括依赖含半胱氨酸的天冬氨酸蛋白水解酶1(cysteinyl aspartate specific proteinase-1, caspase-1)的经典途径和依赖caspase-4/caspase-5/caspase-11的非经典途径。细胞焦亡的主要特点是Nod样受体蛋白3(nod-like receptor protein 3, NLRP3)炎性小体活化,caspase-1激活,细胞膜孔形成,白细胞介素(interleukin, IL)-1β和IL-18释放,从而放大炎症级联反应。NLRP3炎性小体活化与高血压、糖尿病、高脂血症及肥胖等心血管危险因素有关,是心血管炎症的重要触发和内源性调节因子。本文主要就细胞焦亡在心血管疾病中的相关研究进展进行综述。  相似文献   

2.
细胞焦亡是一种与炎性应答有关的细胞程序性死亡方式,与高血脂、高血糖、痛风和动脉粥样硬化等多种代谢性疾病密切相关。通过caspase-1依赖或非依赖的机制调节的细胞焦亡都参与代谢性疾病的发展。在caspase-1依赖的细胞焦亡中,多种代谢性疾病有关的危险信号激活Nod样受体蛋白3炎症小体,导致细胞焦亡、白介素-1β水平增加,进而激活局部及全身炎症反应,是代谢性疾病发生发展的重要原因之一。革兰氏阴性菌释放的脂多糖能直接激活caspase-4/5/11,导致caspase-1非依赖的细胞焦亡。抑制细胞炎症小体–焦亡通路未来可能成为改善代谢性疾病的有效治疗策略之一,然而,由于细胞焦亡调节代谢稳态的机制仍不清楚,因此还需要进一步研究。  相似文献   

3.
细胞焦亡(pyroptosis)是一种高度促炎性的细胞程序性死亡,最早是在受细菌感染或者细菌毒素处理后的巨噬细胞中观察到的,很长一段时间被误认为是一种巨噬细胞特异的、依赖于能够切割白介素1β的促炎性蛋白酶caspase-1的细胞死亡.后续的研究发现,胞浆内模式识别受体识别病原体来源的模式分子或者机体本身来源的危险信号分子形成炎症小体(inflammasomes),招募和激活caspase-1导致细胞焦亡;鼠的caspase-11和人的caspase-4/5直接作为模式识别受体识别细菌脂多糖类脂A组装的炎症小体也导致细胞焦亡,这一发现颠覆了传统炎症小体的概念.与caspase-1不同, caspase-11/4/5不能切割白介素且引起的细胞焦亡在非单核细胞中也普遍存在.最新的研究发现, caspase-1以及caspase-11/4/5都能切割共同的底物gasdermin D(GSDMD)导致裂解性细胞死亡.GSDMD属于一类具有膜打孔活性的gasdermin家族蛋白成员,细胞焦亡也被重新定义为gasdermin介导的程序性坏死样细胞死亡,开创了细胞焦亡研究的新领域.本文回顾了细胞焦亡研究的历史以及细胞焦亡概念的进化过程,总结了caspase-1和caspase-11/4/5上游目前已知的天然免疫通路,讨论了关于细胞焦亡的研究进展尤其是GSDMD以及其他gasdermin家族细胞焦亡执行蛋白的功能和作用机制,以及细胞焦亡和相关蛋白在对抗感染以及人的自身炎症性疾病过程中的作用.  相似文献   

4.
细胞焦亡是一种程序性细胞死亡,参与了多种疾病的发生发展,而炎症反应在细胞焦亡中的作用是目前的研究热点。炎症小体是炎症反应的重要组成部分,其中黑色素瘤缺乏因子2 (absent in melanoma 2,AIM2)炎症小体的激活是诱发由含半胱氨酸的天冬氨酸蛋白酶1 (caspase-1)介导的细胞焦亡的重要因素。靶向AIM2炎症小体激活与细胞焦亡可作为临床相关疾病治疗的有效策略,本文综述了AIM2炎症小体介导的细胞焦亡的研究进展。  相似文献   

5.
【目的】牛病毒性腹泻病毒(bovine viral diarrhea virus, BVDV)是引起牛病毒性腹泻-黏膜病的关键病毒。BVDV的结构蛋白Erns可在病毒感染的初期削弱宿主的免疫防御,引发牛群炎症反应。核苷酸寡聚化结构域样受体(nucleotide-binding oligomerization domain, NOD)热蛋白结构域相关蛋白3 (NLRP3)炎症小体是NOD样受体(NOD-like receptor, NLRs)家族重要成员,调控炎症性疾病的发生发展,同时激活的NLRP3炎症小体能够引起宿主细胞焦亡,进而诱发级联放大的炎症反应。但BVDV Erns蛋白在BVDV感染诱发炎症反应的分子机制尚不清楚。【方法】为进一步探索Erns蛋白对BVDV感染激活NLRP3炎症小体诱发细胞焦亡的影响,构建了BVDV Erns蛋白的真核表达质粒pCMV-HA-Erns,过表达BVDV Erns蛋白,检测BVDV感染细胞中NLRP3炎症小体组分[半胱氨酸蛋白酶(caspase-1)、凋亡相关斑点样蛋白(apoptosis-associated speck-like protein, ASC)和NLRP3]、IL-1β的mRNA转录水平和蛋白表达水平,以及细胞死亡调节蛋白(gasdermin D, GSDMD)的基因表达和蛋白剪切情况,并通过扫描电镜观察牛睾丸(bovine testis, BT)细胞膜成孔及BT细胞内容物释放情况,以分析Erns蛋白诱导BT细胞产生细胞焦亡。【结果】Erns蛋白能够显著引起NLRP3炎症小体活化进而激活caspase-1,活化的caspase-1一方面切割GSDMD,形成有活性的GSDMD-N端并在BT细胞膜形成孔洞,释放内容物,诱导BT细胞发生细胞焦亡;另一方面活化的caspase-1切割pro-IL-1β,形成有活性的IL-1β,并释放到BT细胞外,引起BT细胞上清中IL-1β水平上升。【结论】系统解析了BVDV Erns蛋白激活NLRP3炎症小体介导细胞焦亡的产生,对疫苗及治疗药物的研制具有重要指导意义。  相似文献   

6.
脊髓损伤的治疗与康复一直是医学领域的重大难题,尤其是在改善损伤的神经功能方面进展甚微。继发性损伤是造成脊髓损伤后神经功能障碍的主要原因,炎症反应是继发性损伤阶段最重要的病理过程。急性期通过抑制神经炎症来减轻继发性损伤被认为可减轻神经功能损害而达到神经保护作用。炎性小体是一类蛋白质复合体,由模式识别受体中的NLRs家族和PHYIN家族的受体蛋白质作为主要框架组装并命名,常见的炎性小体包括NLRP1、NLRP3、NLRC4(IPAF)、AIM2等。在感染或受到损伤刺激时,炎性小体在细胞质内组装,并激活促炎症蛋白酶胱天蛋白酶1(caspase-1),活化的胱天蛋白酶1一方面促进促炎症细胞因子IL-1β和IL-18的前体成熟和分泌,另一方面介导细胞焦亡。细胞焦亡以细胞肿胀破裂并释放细胞内容物为特征,是在炎症和应激的病理条件下诱导的程序性细胞死亡方式。促炎症细胞因子和焦亡释放的胞内物质都可作为促炎信号引发炎症反应。近期发现,炎性小体通过诱导促炎因子释放以及介导细胞焦亡等途径, 参与激活脊髓损伤后的炎症级联反应,加重继发性神经炎症。靶向抑制炎性小体的激活可减轻炎症反应,促进神经细胞存活,达到神经保护作用。因此,炎性小体有望成为脊髓损伤治疗的新靶点。本文拟从炎性小体的结构及其在脊髓损伤中的作用、激活机制和治疗前景进行综述,以期为后续研究提供思路。  相似文献   

7.
细胞焦亡是一种促炎症性和溶解性的细胞模式死亡,它是依赖半胱天冬酶-1(caspase-1)的激活所引起的一系列反应,最终作用于细胞,产生一种介于凋亡和坏死之间的细胞死亡。机体通过触发细胞焦亡而起到保护机体免受外界感染的作用,其中炎症小体激活半胱天冬酶-1(caspase-1)对于细胞焦亡的诱导起着重要的作用。本文主要介绍细胞焦亡及其被诱发的分子机制新进展,及与临床贴近的最新进展。  相似文献   

8.
沙门菌主要通过食物传播,严重威胁了人类健康。肠道上皮细胞作为抵抗沙门菌入侵的重要屏障,可通过多种方式抵抗沙门菌的定植与入侵。同时,肠道固有层巨噬细胞可特异性识别正常菌群与沙门菌,激活炎性小体并分泌白细胞介素(interleukin,IL)-1β等炎症因子诱导炎症反应清除沙门菌。Caspase家族属于半胱氨酸蛋白酶,它们被激活后可执行各种细胞功能。Caspase-1是炎性小体的重要组成部分,可切割消皮素D(gasdermin D)诱导细胞焦亡,引发炎症反应。研究发现,Caspase-8同样参与炎性小体复合物的形成,但其功能尚不明确。新近研究发现,在沙门菌感染所诱导的细胞焦亡被抑制时,Caspase-8在炎性小体中被强烈激活,并在肠道上皮细胞和巨噬细胞中调控细胞死亡与炎症反应,以限制沙门菌感染。因此,Caspase-8在沙门菌感染期间也是调节宿主抗感染免疫的关键分子,研究其调控宿主细胞死亡以及炎症因子释放的机制对深入了解沙门菌感染与宿主抗感染免疫应答之间的关系具有重要意义。  相似文献   

9.
细胞焦亡(pyroptosis)是近年来发现的一种区别于细胞凋亡的促炎程序性死亡方式。焦亡途径包括半胱天冬酶(caspase)-1介导的经典焦亡途径和Caspase-4/5/11介导的非经典焦亡途径。细胞焦亡涉及多种炎性小体的激活,如核苷酸结合寡聚化结构域样受体蛋白3(NLR pyrin domain containing 3,NLRP3)、核苷酸结合寡聚化结构域样受体蛋白C4 (NLR containing a caspase recruitment domain 4,NLRC4)以及黑色素瘤缺乏因子2 (absent in melanoma 2,AIM2)等。Gasdermin-D(GSDMD)是参与细胞焦亡的关键切割蛋白,最终导致膜蛋白通道开放、膜孔形成、白细胞介素(interleukins,ILs)释放,从而扩大炎症反应。细胞焦亡介导许多疾病如感染性疾病、神经系统疾病、心血管疾病、代谢性疾病以及炎症免疫性疾病等。本文综述了细胞焦亡机制及与疾病关系的研究进展。  相似文献   

10.
炎性小体是先天性免疫系统的受体和传感器,在许多疾病的发生和进展中起着关键的病理作用。近期研究表明,NOD样受体家族核苷酸结合寡聚化结构域样受体3 (NOD-like receptor thermal protein domain associated protein 3, NLRP3)炎性小体参与了对公共健康具有高度影响的疾病的发生,如肌肉骨骼系统疾病。肌肉骨骼系统疾病是主要由工作和周围环境引起或加重的肌肉、关节、骨骼等运动系统疾病,以及相关神经、循环系统损伤的疾病。NLRP3小体的激活可以诱导炎症及引发焦亡,造成机体进一步损伤。因此,以NLRP3炎性小体为切入点,开展对肌肉骨骼系统疾病的预防和治疗具有重要意义。研究炎症性疾病中NLRP3炎性小体活动的机制及作用已然成为新的研究方向。本文对NLRP3炎性小体的激活途径及机制进行了概述,并分析了NLRP3炎性小体在肌少症、骨质疏松症和关节炎等肌肉骨骼系统疾病中的作用,以期为肌肉骨骼系统疾病的治疗提供理论依据。  相似文献   

11.
Pyroptosis is a form of necrotic and inflammatory programmed cell death, which could be characterized by cell swelling, pore formation on plasma membranes, and release of proinflammatory cytokines (IL-1β and IL-18). The process of pyroptosis presents as dual effects: protecting multicellular organisms from microbial infection and endogenous dangers; leading to pathological inflammation if overactivated. Two pathways have been found to trigger pyroptosis: caspase-1 mediated inflammasome pathway with the involvement of NLRP1-, NLRP3-, NLRC4-, AIM2-, pyrin-inflammasome (canonical inflammasome pathway) and caspase-4/5/11-mediated inflammasome pathway (noncanonical inflammasome pathway). Gasdermin D (GSDMD) has been proved to be a substrate of inflammatory caspases (caspase-1/4/5/11), and the cleaved N-terminal domain of GSDMD oligomerizes to form cytotoxic pores on the plasma membrane. Here, we mainly reviewed the up to date mechanisms of pyroptosis, and began with the inflammasomes as the activator of caspase-1/caspase-11, 4, and 5. We further discussed these inflammasomes functions in diseases, including infectious diseases, sepsis, inflammatory autoimmune diseases, and neuroinflammatory diseases.  相似文献   

12.
NOD-like receptor (NLR) proteins (Nlrps) are cytosolic sensors responsible for detection of pathogen and danger-associated molecular patterns through unknown mechanisms. Their activation in response to a wide range of intracellular danger signals leads to formation of the inflammasome, caspase-1 activation, rapid programmed cell death (pyroptosis) and maturation of IL-1β and IL-18. Anthrax lethal toxin (LT) induces the caspase-1-dependent pyroptosis of mouse and rat macrophages isolated from certain inbred rodent strains through activation of the NOD-like receptor (NLR) Nlrp1 inflammasome. Here we show that LT cleaves rat Nlrp1 and this cleavage is required for toxin-induced inflammasome activation, IL-1 β release, and macrophage pyroptosis. These results identify both a previously unrecognized mechanism of activation of an NLR and a new, physiologically relevant protein substrate of LT.  相似文献   

13.
Inflammasomes are protein complexes assembled upon recognition of infection or cell damage signals, and serve as platforms for clustering and activation of procaspase-1. Oligomerisation of initiating proteins such as AIM2 (absent in melanoma-2) and NLRP3 (NOD-like receptor family, pyrin domain-containing-3) recruits procaspase-1 via the inflammasome adapter molecule ASC (apoptosis-associated speck-like protein containing a CARD). Active caspase-1 is responsible for rapid lytic cell death termed pyroptosis. Here we show that AIM2 and NLRP3 inflammasomes activate caspase-8 and -1, leading to both apoptotic and pyroptotic cell death. The AIM2 inflammasome is activated by cytosolic DNA. The balance between pyroptosis and apoptosis depended upon the amount of DNA, with apoptosis seen at lower transfected DNA concentrations. Pyroptosis had a higher threshold for activation, and dominated at high DNA concentrations because it happens more rapidly. Gene knockdown showed caspase-8 to be the apical caspase in the AIM2- and NLRP3-dependent apoptotic pathways, with little or no requirement for caspase-9. Procaspase-8 localised to ASC inflammasome ‘specks'' in cells, and bound directly to the pyrin domain of ASC. Thus caspase-8 is an integral part of the inflammasome, and this extends the relevance of the inflammasome to cell types that do not express caspase-1.  相似文献   

14.
The inflammasome is a signalling platform leading to caspase-1 activation. Caspase-1 causes pyroptosis, a necrotic-like cell death. AIM2 is an inflammasome sensor for cytosolic DNA. The adaptor molecule ASC mediates AIM2-dependent caspase-1 activation. To date, no function besides caspase-1 activation has been ascribed to the AIM2/ASC complex. Here, by comparing the effect of gene inactivation at different levels of the inflammasome pathway, we uncovered a novel cell death pathway activated in an AIM2/ASC-dependent manner. Francisella tularensis, the agent of tularaemia, triggers AIM2/ASC-dependent caspase-3-mediated apoptosis in caspase-1-deficient macrophages. We further show that AIM2 engagement leads to ASC-dependent, caspase-1-independent activation of caspase-8 and caspase-9 and that caspase-1-independent death is reverted upon caspase-8 inhibition. Caspase-8 interacts with ASC and active caspase-8 specifically colocalizes with the AIM2/ASC speck thus identifying the AIM2/ASC complex as a novel caspase-8 activation platform. Furthermore, we demonstrate that caspase-1-independent apoptosis requires the activation of caspase-9 and of the intrinsic pathway in a typical type II cell manner. Finally, we identify the AIM2/ASC-dependent caspase-1-independent pathway as an innate immune mechanism able to restrict bacterial replication in vitro and control IFN-γ levels in vivo in Casp1(KO) mice. This work underscores the crosstalk between inflammasome components and the apoptotic machinery and highlights the versatility of the pathway, which can switch from pyroptosis to apoptosis.  相似文献   

15.
Infection of macrophages by bacterial pathogens can trigger Toll-like receptor (TLR) activation as well as Nod-like receptors (NLRs) leading to inflammasome formation and cell death dependent on caspase-1 (pyroptosis). Complicating the study of inflammasome activation is priming. Here, we develop a priming-free NLRC4 inflammasome activation system to address the necessity and role of priming in pyroptotic cell death and damage-associated molecular pattern (DAMP) release. We find pyroptosis is not dependent on priming and when priming is re-introduced pyroptosis is unaffected. Cells undergoing unprimed pyroptosis appear to be independent of mitochondrial involvement and do not produce inflammatory cytokines, nitrous oxide (NO), or reactive oxygen species (ROS). Nevertheless, they undergo an explosive cell death releasing a chemotactic isoform of the DAMP high mobility group protein box 1 (HMGB1). Importantly, priming through surface TLRs but not endosomal TLRs during pyroptosis leads to the release of a new TLR4-agonist cysteine redox isoform of HMGB1. These results show that pyroptosis is dominant to priming signals and indicates that metabolic changes triggered by priming can affect how cell death is perceived by the immune system.  相似文献   

16.

Background

Nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome is associated with metabolic disorder and cell death, which are important triggers in diabetic cardiomyopathy (DCM). We aimed to explore whether NLRP3 inflammasome activation contributes to DCM and the mechanism involved.

Methods

Type 2 diabetic rat model was induced by high fat diet and low dose streptozotocin. The characteristics of type 2 DCM were evaluated by metabolic tests, echocardiography and histopathology. Gene silencing therapy was used to investigate the role of NLRP3 in the pathogenesis of DCM. High glucose treated H9c2 cardiomyocytes were used to determine the mechanism by which NLRP3 modulated the DCM. The cell death in vitro was detected by TUNEL and EthD-III staining. TXNIP-siRNA and pharmacological inhibitors of ROS and NF-kB were used to explore the mechanism of NLRP3 inflammasome activation.

Results

Diabetic rats showed severe metabolic disorder, cardiac inflammation, cell death, disorganized ultrastructure, fibrosis and excessive activation of NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), pro-caspase-1, activated caspase-1 and mature interleukin-1β (IL-1β). Evidence for pyroptosis was found in vivo, and the caspase-1 dependent pyroptosis was found in vitro. Silencing of NLRP3 in vivo did not attenuate systemic metabolic disturbances. However, NLRP3 gene silencing therapy ameliorated cardiac inflammation, pyroptosis, fibrosis and cardiac function. Silencing of NLRP3 in H9c2 cardiomyocytes suppressed pyroptosis under high glucose. ROS inhibition markedly decreased nuclear factor-kB (NF-kB) phosphorylation, thioredoxin interacting/inhibiting protein (TXNIP), NLRP3 inflammasome, and mature IL-1β in high glucose treated H9c2 cells. Inhibition of NF-kB reduced the activation of NLRP3 inflammasome. TXNIP-siRNA decreased the activation of caspase-1 and IL-1β.

Conclusion

NLRP3 inflammasome contributed to the development of DCM. NF-κB and TXNIP mediated the ROS-induced caspase-1 and IL-1β activation, which are the effectors of NLRP3 inflammasome. NLRP3 gene silencing may exert a protective effect on DCM.  相似文献   

17.
Mycobacterium tuberculosis (Mtb) has evolved to evade host innate immunity by interfering with macrophage functions. Interleukin-1β (IL-1β) is secreted by macrophages after the activation of the inflammasome complex and is crucial for host defense against Mtb infections. We have previously shown that Mtb is able to inhibit activation of the AIM2 inflammasome and subsequent pyroptosis. Here we show that Mtb is also able to inhibit host cell NLRP3 inflammasome activation and pyroptosis. We identified the serine/threonine kinase PknF as one protein of Mtb involved in the NLRP3 inflammasome inhibition, since the pknF deletion mutant of Mtb induces increased production of IL-1β in bone marrow-derived macrophages (BMDMs). The increased production of IL-1β was dependent on NLRP3, the adaptor protein ASC and the protease caspase-1, as revealed by studies performed in gene-deficient BMDMs. Additionally, infection of BMDMs with the pknF deletion mutant resulted in increased pyroptosis, while the IL-6 production remained unchanged compared to Mtb-infected cells, suggesting that the mutant did not affect the priming step of inflammasome activation. In contrast, the activation step was affected since potassium efflux, chloride efflux and the generation of reactive oxygen species played a significant role in inflammasome activation and subsequent pyroptosis mediated by the Mtb pknF mutant strain. In conclusion, we reveal here that the serine/threonine kinase PknF of Mtb plays an important role in innate immune evasion through inhibition of the NLRP3 inflammasome.  相似文献   

18.
Listeria monocytogenes (LM) infection induces pyroptosis, a form of regulated necrosis, in host macrophages via inflammasome activation. Here, we examined the role of Mint3 in macrophages, which promotes glycolysis via hypoxia-inducible factor-1 activation, during the initiation of pyroptosis following LM infection. Our results showed that Mint3-deficient mice were more resistant to lethal listeriosis than wild-type (WT) mice. Additionally, the mutant mice showed higher levels of IL-1β/IL-18 in the peritoneal fluid during LM infection than WT mice. Moreover, ablation of Mint3 markedly increased the activation of caspase-1, maturation of gasdermin D, and pyroptosis in macrophages infected with LM in vitro, suggesting that Mint3 depletion promotes pyroptosis. Further analyses revealed that Mint3 depletion upregulates inflammasome assembly preceding pyroptosis via glycolysis reduction and reactive oxygen species production. Pharmacological inhibition of glycolysis conferred resistance to listeriosis in a Mint3-dependent manner. Moreover, Mint3-deficient mice treated with the caspase-1 inhibitor VX-765 were as susceptible to LM infection as WT mice. Taken together, these results suggest that Mint3 depletion promotes pyroptosis in host macrophages, thereby preventing the spread of LM infection. Mint3 may serve as a target for treating severe listeriosis by inducing pyroptosis in LM-infected macrophages.Subject terms: Cell death, Infection  相似文献   

19.
Acute inflammation in response to both exogenous and endogenous danger signals can lead to the assembly of cytoplasmic inflammasomes that stimulate the activation of caspase-1. Subsequently, caspase-1 facilitates the maturation and release of cytokines and also, under some circumstances, the induction of cell death by pyroptosis. Using a mouse line lacking expression of NLRP1, we show that assembly of this inflammasome in cells is triggered by a toxin from anthrax and that it initiates caspase-1 activation and release of IL-1β. Furthermore, NLRP1 inflammasome activation also leads to cell death, which escalates over 3 d following exposure to the toxin and culminates in acute lung injury and death of the mice. We show that these events are not dependent on production of IL-1β by the inflammasome but are dependent on caspase-1 expression. In contrast, muramyl dipeptide-mediated inflammasome formation is not dependent on NLRP1 but NLRP3. Taken together, our findings show that assembly of the NLRP1 inflammasome is sufficient to initiate pyroptosis, which subsequently leads to a self-amplifying cascade of cell injury within the lung from which the lung cannot recover, eventually resulting in catastrophic consequences for the organism.  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