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.
<正>类风湿关节炎(rheumatoid arthritis, RA)是一种全身性慢性自身免疫性疾病,其病理特征以滑膜炎、血管翳生成以及关节软骨破坏为主[1]。RA发病中,免疫细胞分泌的细胞因子通过特定受体介导细胞间通讯以促进RA的发生发展[2]。  相似文献   

2.
<正>肾结石是泌尿外科多发性疾病,可造成肾功不全、肾萎缩和脓胸等并发症,给患者带来严重的经济负担[1-2]。草酸钙(calcium oxalate,Ca Ox)肾结石的形成涉及活性氧(reactive oxygen species,ROS)产生增多、免疫炎症反应和肾小管上皮损伤等病理过程[3]。既往研究证实,巨噬细胞的不同极化状态能够影响Ca Ox肾结石形成[4]。  相似文献   

3.
系统性红斑狼疮(SLE)是一种自身免疫性疾病,其发病机制极为复杂,随着研究的深入,炎症小体在狼疮性肾炎(LN)的发病中作用也逐渐受到重视。其中,核苷酸结合寡聚化结构域样受体蛋白3(NLRP3)炎症小体是目前研究最详尽的一类炎症体。我们对近年来NLRP3 炎症小体在LN 中作用的相关研究进行归纳总结,发现NLRP3 炎症小体不仅在LN 的发病中起着重要的作用,并通过循环免疫细胞和固有细胞参与肾脏损伤的过程。最后介绍了两种NLRP3 炎症小体的特异性抑制剂β-羟基丁酸酯和MCC950,为LN 治疗提供了新的策略。  相似文献   

4.
目的探讨核苷酸结合寡聚化结构域样受体蛋白3(NLRP3)炎症小体在神经病理性疼痛大鼠中脑导水管周围灰质腹外侧区(vlPAG)的表达水平及其作用。方法选取SPF级SD大鼠20只,采用随机数字表法分为假手术组(Sham组)与慢性坐骨神经压迫性损伤组(CCI组),每组10只。于手术前后测定2组机械痛阈与热痛阈;Western blot检测2组NLRP3炎症小体各组分蛋白表达水平;RT-PCR检测2组NLRP3炎症小体各组分基因mRNA表达水平;酶联免疫吸附实验(ELISA)检测2组炎症因子白介素-lβ(IL-1β)及白介素-18(IL-18)表达水平;免疫荧光检测2组中NLRP3炎症小体在vlPAG的分布情况,并与小胶质细胞标志物IBA1进行共定位分析。结果与Sham组相比,CCI组术后3 d、7 d、14 d、21 d机械痛阈与热痛阈均明显降低(P<0.001);与Sham组相比,CCI组核心蛋白NLRP3、效应蛋白Caspase-1 p10及前体蛋白pro-Caspase-1表达水平均明显升高(P<0.001);与Sham组相比,CCI组NLRP3(P<0.001)、Caspase-1(P<0.001)及IL-1β(P<0.01)mRNA表达水平均明显升高,炎症因子IL-1β(P<0.001)与IL-18(P<0.01)表达水平也明显升高;与Sham组相比,CCI组在vlPAG的NLRP3阳性细胞数增加,且与IBA1存在共表达。结论神经病理性疼痛大鼠vlPAG中NLRP3炎症小体被激活且表达水平增高,提示NLRP3炎症小体激活水平与神经病理性疼痛大鼠痛阈下降相关。  相似文献   

5.
<正>阿尔茨海默症(Alzheimer′sdisease,AD)为一种致病机制尚不明确的神经退行性疾病,其中β-淀粉样蛋白的积累、磷酸tau蛋白的神经原纤维缠结的形成以及有害的神经炎症是AD进展的主要标志性特征[1-3]。全世界现约有5000万AD患者,预计到2050年将增加到1.52亿,AD治疗药物有着巨大的临床规模需求[4]。1991年威廉·弗雷二世提出了一种用于治疗大脑神经系统疾病的鼻腔药物递送方法。鼻腔药物递送方法已经应用于包括帕金森病、疼痛、癌症等诸多领域[5]。  相似文献   

6.
<正>表观遗传学是指DNA序列不变的情况下,基于非基因序列改变所致的基因表达水平的变化[1],如DNA甲基化[2]、RNA甲基化[3]、组蛋白修饰[4]、染色质构象变化[5]等。表观遗传学在真核生物中的变化主要是调控细胞增殖[6-7]、分化[8]、代谢[3,9]、周期循环[10]以及免疫调控[11]等生物学过程,这一过程中通过某些调控分子量的变化或发生结构修饰变化等,进而靶向调节下游靶基因的生物学效应。  相似文献   

7.
<正>上皮钠通道(epithelial sodium channel,ENa C)是一种非电压依赖性的离子通道,是上皮细胞主要的Na+转运通道之一,属于ENa C/退化蛋白(degenerin)超家族[1-2]。ENa C对阿米洛利敏感,主要分布于人体肺部[3]、肾脏[4]、结肠、生殖道[5]及中枢神经系统[6]等多种组织的上皮细胞,能够控制Na+进入上皮细胞,对稳定细胞内Na+浓度及维持细胞渗透平衡具有重要作用[7]。细胞内外液体渗透平衡是维持细胞正常生理功能的基础,这种平衡取决于细胞内外的相对渗透压和膜的渗透性[8],而人体细胞内液和细胞外液的渗  相似文献   

8.
嘌呤能2X7受体(purinergic 2X7 receptor, P2X7R)是一种离子通道型受体, 可引起核苷酸结合寡聚化结构域样受体3(nucleotide-binding oligomerization domain-like receptor protein 3, NLRP3)的激活, 进而影响炎症细胞因子如IL-1β、IL-18等的释放从而参与多种炎症性疾病。近年来, P2X7R/NLRP3信号通路已成为炎症性疾病研究较多的通路之一, 已有部分P2X7R和NLRP3炎性小体的拮抗剂进入早期的临床治疗。本文就P2X7R和NLRP3炎性小体的相关进展进行综述, 为进一步验证P2X7R/NLRP3的激活导致IL-1β等炎症细胞因子在肿瘤和炎症性疾病中释放增加提供参考, 为研究P2X7R/NLRP3作为肿瘤和炎症性疾病的重要病理机制和潜在的治疗靶点提供新的思路。  相似文献   

9.
NLRP3炎性小体是一种胞内多蛋白复合体,主要由NOD样受体家族成员NLRP3、接头蛋白ASC以及前体半胱天冬酶1(pro-caspase-1)组成,该炎性小体可以通过激活caspase-1促进促炎因子白细胞介素-1 beta(IL-1β)和IL-18的分泌以及细胞焦亡(pyroptosis)的形成.NLRP3炎性小体的激活在很多自身免疫性及自身炎症性疾病中扮演着重要的角色,因此,深入探究NLRP3炎性小体激活的调控机制可为NLRP3炎性小体相关疾病的治疗提供更多新的思路.  相似文献   

10.
<正>慢性阻塞性肺疾病(chronic obstructive pulmonary disease, COPD)是一种呼吸道慢性炎症性疾病,高发病率和死亡率造成了巨大社会和经济负担,2019年WHO公布COPD已成为全球第三大死亡原因[1]。但COPD的发病机制尚不明了。随着脂质代谢研究的深入,在COPD中肺泡巨噬细胞(alveolar macrophage, AM)内的脂质代谢变化也备受关注。Fujii等[2]在研究中发现了AM中的脂质组成以COPD疾病等级依赖性的方式发生变化,  相似文献   

11.
Inflammasomes in innate immune cells mediate the induction of inflammation by sensing microbes and pathogen‐associated/damage‐associated molecular patterns. Inflammasomes are also known to be involved in the development of some human and animal autoimmune diseases. The Nod‐like receptor family pyrin domain containing 3 (NLRP3) inflammasome is currently the most fully characterized inflammasome, although a limited number of studies have demonstrated its role in demyelinating autoimmune diseases in the central nervous system of humans and animals. Currently, the development of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), is known to be induced by the NLRP3 inflammasome through enhanced recruitment of inflammatory immune cells in the central nervous system. On the other hand, interferon‐β (IFNβ), a first‐line drug to treat MS, inhibits NLRP3 inflammasome activation, and ameliorates EAE. The NLRP3 inflammasome is indeed a factor capable of inducing EAE, but it is dispensable when EAE is induced by aggressive disease induction regimens. In such NLRP3 inflammasome‐independent EAE, IFN‐β treatment is generally not effective. This might therefore be one mechanism that leads to occasional failures of IFN‐β treatment in EAE, and possibly, in MS as well. In the current review, we discuss inflammasomes and autoimmunity; in particular, the impact of the NLRP3 inflammasome on MS/EAE, and on IFN‐β therapy.  相似文献   

12.
The aptly named inflammasomes are powerful signaling complexes that sense inflammatory signals under a myriad of conditions, including those from infections and endogenous sources. The inflammasomes promote inflammation by maturation and release of the pro‐inflammatory cytokines, IL‐1β and IL‐18. Several inflammasomes have been identified so far, but this review focuses mainly on the NLRP3 inflammasome. By still ill‐defined activation mechanisms, a sensor molecule, NLRP3 (NACHT, LRR and PYD domains‐containing protein 3), responds to danger signals and rapidly recruits ASC (apoptosis‐associated speck‐like protein containing a CARD) and pro‐caspase‐1 to form a large oligomeric signaling platform—the inflammasome. Involvement of the NLRP3 inflammasome in infections, metabolic disorders, autoinflammation, and autoimmunity, underscores its position as a central player in sensing microbial and damage signals and coordinating pro‐inflammatory immune responses. Indeed, evidence in patients with multiple sclerosis (MS) suggests inflammasome activation occurs during disease. Experiments with the mouse model of MS, experimental autoimmune encephalomyelitis (EAE), specifically describe the NLRP3 inflammasome as critical and necessary to disease development. This review discusses recent studies in EAE and MS which describe associations of inflammasome activation with promotion of T cell pathogenicity, infiltration of cells into the central nervous system (CNS) and direct neurodegeneration during EAE and MS.  相似文献   

13.
Aging is the greatest risk factor for the development of chronic diseases such as arthritis, type 2 diabetes, cardiovascular disease, kidney disease, Alzheimer's disease, macular degeneration, frailty, and certain forms of cancers. It is widely regarded that chronic inflammation may be a common link in all these age-related diseases. This raises the question, can one alter the course of aging and potentially slow the development of all chronic diseases by manipulating the mechanisms that cause age-related inflammation? Emerging evidence suggests that pro-inflammatory cytokines interleukin-1 (IL-1) and IL-18 show an age-dependent regulation implicating inflammasome-mediated caspase-1 activation in the aging process. The Nod-like receptor (NLR) family of innate immune cell sensors, such as the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome controls the caspase-1 activation in myeloid-lineage cells in several organs during aging. The NLRP3 inflammasome is especially relevant to aging as it can get activated in response to structurally diverse damage-associated molecular patterns (DAMPs) such as extracellular ATP, excess glucose, ceramides, amyloids, urate, and cholesterol crystals, all of which increase with age. Interestingly, reduction in NLRP3-mediated inflammation prevents age-related insulin resistance, bone loss, cognitive decline, and frailty. NLRP3 is a major driver of age-related inflammation and therefore dietary or pharmacological approaches to lower aberrant inflammasome activation holds promise in reducing multiple chronic diseases of age and may enhance healthspan.  相似文献   

14.
The NLRP3 inflammasome plays a crucial role in innate immune-mediated inflammation and contributes to the pathogenesis of multiple autoinflammatory, metabolic and neurodegenerative diseases, but medications targeting the NLRP3 inflammasome are not available for clinical use. RRx-001 is a well-tolerated anticancer agent currently being investigated in phase III clinical trials, but its effects on inflammatory diseases are not known. Here, we show that RRx-001 is a highly selective and potent NLRP3 inhibitor that has strong beneficial effects on NLRP3-driven inflammatory diseases. RRx-001 inhibits the activation of the canonical, noncanonical, and alternative NLRP3 inflammasomes but not the AIM2, NLRC4 or Pyrin inflammasomes. Mechanistically, RRx-001 covalently binds to cysteine 409 of NLRP3 via its bromoacetyl group and therefore blocks the NLRP3-NEK7 interaction, which is critical for the assembly and activation of the NLRP3 inflammasome. More importantly, RRx-001 treatment attenuates the symptoms of lipopolysaccharide (LPS)-induced systemic inflammation, dextran sulfate sodium (DSS)-induced colitis and experimental autoimmune encephalomyelitis (EAE) in mice. Thus, our study identifies RRx-001 as a new potential therapeutic agent for NLRP3-driven diseases.  相似文献   

15.
急性肺损伤/急性呼吸窘迫综合征(ALI/ARDS)是由多种非心源性肺内外因素引起的急性进行性呼吸衰竭,发病核心为过度放大或失控的炎症反应,目前没有特效的治疗药物.NOD样受体蛋白3(nucleotide-binding domain (NOD)-like receptor protein 3,NLRP3)炎症小体是细胞受到刺激时形成的多蛋白复合体,活化后导致细胞焦亡及IL-1β、IL-18等产生,在多种感染性、炎症性疾病中起重要作用.引起肺损伤的多种因素均可导致NLRP3炎症小体形成、活化,有研究提示,与ALI/ARDS中的过度炎症反应有关.因而深入研究NLRP3炎症小体在ALI/ARDS中的作用,对于进一步阐明ALI/ARDS的发病机制有重要意义,甚至有望成为治疗ALI/ARDS的新靶点.  相似文献   

16.
17.
Neuroinflammation is considered as a detrimental factor in neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), etc. Nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3), the most well-studied inflammasome, is abundantly expressed in microglia and has gained considerable attention. Misfolded proteins are characterized as the common hallmarks of neurodegenerative diseases due to not only their induced neuronal toxicity but also their effects in over-activating microglia and the NLRP3 inflammasome. The activated NLRP3 inflammasome aggravates the pathology and accelerates the progression of neurodegenerative diseases. Emerging evidence indicates that microglial autophagy plays an important role in the maintenance of brain homeostasis and the negative regulation of NLRP3 inflammasome-mediated neuroinflammation. The excessive activation of NLRP3 inflammasome impairs microglial autophagy and further aggravates the pathogenesis of neurodegenerative diseases. In this review article, we summarize and discuss the NLRP3 inflammasome and its specific inhibitors in microglia. The crucial role of microglial autophagy and its inducers in the removal of misfolded proteins, the clearance of damaged mitochondria and reactive oxygen species (ROS), and the degradation of the NLRP3 inflammasome or its components in neurodegenerative diseases are summarized. Understanding the underlying mechanisms behind the sex differences in NLRP3 inflammasome-mediated neurodegenerative diseases will help researchers to develop more targeted therapies and increase our diagnostic and prognostic abilities. In addition, the superiority of the combined use of microglial autophagy inducers with the specific inhibitors of the NLRP3 inflammasome in the inhibition of NLRP3 inflammasome-mediated neuroinflammation requires further preclinical and clinical validations in the future.  相似文献   

18.
Inflammasomes are multi-protein signaling complexes that trigger the activation of inflammatory caspases and the maturation of interleukin-1β. Among various inflammasome complexes, the NLRP3 inflammasome is best characterized and has been linked with various human autoinflammatory and autoimmune diseases. Thus, the NLRP3 inflammasome may be a promising target for anti-inflammatory therapies. In this review, we summarize the current understanding of the mechanisms by which the NLRP3 inflammasome is activated in the cytosol. We also describe the binding partners of NLRP3 inflammasome complexes activating or inhibiting the inflammasome assembly. Our knowledge of the mechanisms regulating NLRP3 inflammasome signaling and how these influence inflammatory responses offers further insight into potential therapeutic strategies to treat inflammatory diseases associated with dysregulation of the NLRP3 inflammasome.  相似文献   

19.
Aging-related diseases such as cancer, cardiovascular diseases, diabetes, and neurodegenerative diseases are often accompanied by fibrosis. The NLRP3 inflammasome triggers the inflammatory response and subsequently promotes fibrosis through pathogen-associated molecular patterns (PAMPs). In this review, we first introduce the general background and specific mechanism of NLRP3 in fibrosis. Second, we investigate the role of NLRP3 in fibrosis in different organs/tissues. Third, we discuss the relationship between NLRP3 and fibrosis during aging. In summary, this review describes the latest progress on the roles of NLRP3 in fibrosis and aging and reveals the possibility of NLRP3 as an antifibrotic and anti-aging treatment target.  相似文献   

20.
Aberrant inflammasome activation contributes to the pathogenesis of various human diseases, including atherosclerosis, gout, and metabolic disorders. Elucidation of the underlying mechanism involved in the negative regulation of the inflammasome is important for developing new therapeutic targets for these diseases. Here, we showed that Raf kinase inhibitor protein (RKIP) negatively regulates the activation of the NLRP1, NLRP3, and NLRC4 inflammasomes. RKIP deficiency enhanced caspase-1 activation and IL-1β secretion via NLRP1, NLRP3, and NLRC4 inflammasome activation in primary macrophages. The overexpression of RKIP in THP-1 cells inhibited NLRP1, NLRP3, and NLRC4 inflammasome activation. RKIP-deficient mice showed increased sensitivity to Alum-induced peritonitis and Salmonella typhimurium-induced inflammation, indicating that RKIP inhibits NLRP3 and NLRC4 inflammasome activation in vivo. Mechanistically, RKIP directly binds to apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) and competes with NLRP1, NLRP3, or NLRC4 to interact with ASC, thus interrupting inflammasome assembly and activation. The depletion of RKIP aggravated inflammasome-related diseases such as monosodium urate (MSU)-induced gouty arthritis and high-fat diet (HFD)-induced metabolic disorders. Furthermore, the expression of RKIP was substantially downregulated in patients with gouty arthritis or type 2 diabetes (T2D) compared to healthy controls. Collectively, our findings suggest that RKIP negatively regulates NLRP1, NLRP3, and NLRC4 inflammasome activation and is a potential therapeutic target for the treatment of inflammasome-related diseases.  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