NLRP3 inhibitor

NLRP3 抑制剂
  • 文章类型: Journal Article
    已在各种人类疾病中观察到NLRP3炎性体的异常激活。用小分子抑制剂靶向NLRP3蛋白显示出作为疾病干预的有效策略的巨大潜力。在这里,设计并合成了一系列新型联苯磺酰胺NLRP3炎性体抑制剂。代表性化合物H28被鉴定为有效且特异性的NLRP3炎性体抑制剂,IC50值为0.57μM。初步机制研究表明,化合物H28表现出与NLRP3蛋白(KD:1.15μM)的直接结合,有效抑制NLRP3炎性体的组装和激活。在小鼠急性腹膜炎模型中的结果表明,H28有效抑制NLRP3炎性体通路,证明了它们的抗炎特性。我们的发现强烈支持H28作为治疗NLRP3相关疾病的潜在先导化合物的进一步发展。
    The aberrant activation of NLRP3 inflammasome has been observed in various human diseases. Targeting the NLRP3 protein with small molecule inhibitors shows immense potential as an effective strategy for disease intervention. Herein, a series of novel biphenyl-sulfonamide NLRP3 inflammasome inhibitors were designed and synthesized. The representative compound H28 was identified as potent and specific NLRP3 inflammasome inhibitor with IC50 values of 0.57 μM. Preliminary mechanistic studies have revealed that compound H28 exhibits direct binding to the NLRP3 protein (KD: 1.15 μM), effectively inhibiting the assembly and activation of the NLRP3 inflammasome. The results in a mouse acute peritonitis model revealed that H28 effectively inhibit the NLRP3 inflammasome pathway, demonstrating their anti-inflammatory properties. Our findings strongly support the further development of H28 as potential lead compound for treating NLRP3-related diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    NLRP3是一种细胞内传感蛋白,可检测各种危险信号和环境危害。其活化导致保护性促炎反应,其设计为通过NLRP3炎性体的形成损害病原体和修复组织损伤。NLRP3炎性体的组装导致促炎细胞因子IL-1β和IL-18的胱天蛋白酶1依赖性分泌释放,并导致gasdermind介导的细胞凋亡。在这里,我们描述了一种高亲和力的新型吲唑系列的发现,通过筛选DNA编码的文库和直接从筛选鉴定的有效先导化合物3(BAL-0028,IC50=25nM),NLRP3激活的可逆抑制剂。SPR研究显示化合物3与NLRP3的NACHT结构域紧密结合(KD范围104-123nM)。化合物3与NLRP3NACHT结构域的相互作用的CADD分析提出了与ADP和MCC950的结合位点不同的结合位点,并且包括特异性位点相互作用。我们预计,化合物3(BAL-0028)和其他成员的这种新型的吲哚类中性抑制剂将表现出显著不同的物理,生物化学,和与先前鉴定的NLRP3抑制剂相比的生物学特性。
    NLRP3 is an intracellular sensor protein that detects a broad range of danger signals and environmental insults. Its activation results in a protective pro-inflammatory response designed to impair pathogens and repair tissue damage via the formation of the NLRP3 inflammasome. Assembly of the NLRP3 inflammasome leads to caspase 1-dependent secretory release of the pro-inflammatory cytokines IL-1β and IL-18 as well as to gasdermin d-mediated pyroptotic cell death. Herein, we describe the discovery of a novel indazole series of high affinity, reversible inhibitors of NLRP3 activation through screening of DNA-encoded libraries and the potent lead compound 3 (BAL-0028, IC50 = 25 nM) that was identified directly from the screen. SPR studies showed that compound 3 binds tightly (KD range 104-123 nM) to the NACHT domain of NLRP3. A CADD analysis of the interaction of compound 3 with the NLRP3 NACHT domain proposes a binding site that is distinct from those of ADP and MCC950 and includes specific site interactions. We anticipate that compound 3 (BAL-0028) and other members of this novel indazole class of neutral inhibitors will demonstrate significantly different physical, biochemical, and biological properties compared to NLRP3 inhibitors previously identified.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    有机合成方法的进展可以促进后期铅化合物的改性和新型活性化合物的发现。有机合成领域的分子编辑技术,包括外围和骨骼编辑,促进快速获得先导化合物的分子多样性。CH键活化的外围编辑逐渐用于铅优化,以提供新型活性支架和化学空间开发。开发具有高抗炎效力的冬凌草甲素衍生物,新型冬凌草甲素磺酰胺是通过基于可见光光催化外周编辑的支架跳跃策略设计和合成的。所有新化合物都显示出可测量的IL-1β抑制和THP-1细胞中的低细胞毒性。对接研究表明,最佳活性化合物ZM640通过两个氢键相互作用被容纳在NLRP3的结合位点。这些初步结果证实了α,冬凌草甲素的β-不饱和羰基对于NLRP3抑制作用不是必需的。这种新的冬凌草甲素支架具有作为一类有前途的NLRP3抑制剂进一步开发的潜力。
    The progress of organicsyntheticmethod can promote late-stage lead compound modification and novel active compound discovery. Molecular editing technology in the field of organic synthesis, including peripheral and skeletal editing, facilitates rapid access to molecular diversity of a lead compound. Peripheral editing of CH bond activation is gradually used in lead optimization to afford novel active scaffolds and chemical space exploitation. To develop oridonin derivatives with high anti-inflammatory potency, novel oridonin sulfamides had been designed and synthesized by a scaffoldhopping strategy based on a visible-light photocatalysis peripheral editing. All novel compounds revealed measurable inhibition of IL-1β and low cytotoxicity in THP-1 cells. The docking study indicated that the best active compound ZM640 was accommodated in thebinding site of NLRP3 with two hydrogen bond interaction. These preliminary results confirm that α, β-unsaturated carbonyl of oridonin is not essential for NLRP3 inhibitory effect. This new oridonin scaffold has its potential to be further developed as a promising class of NLRP3 inhibitors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    阿尔茨海默病(AD)仍然是一种无法治愈的神经退行性疾病,对人类构成威胁。大脑中的免疫信号,特别是含有3的NLR家族pyrin结构域(NLRP3),目前针对AD治疗。基于NLRP3的NACHT结构域的晶体结构及其著名的抑制剂MCC950,我们设计并合成了19种磺酰脲类化合物,并评估了它们抑制caspase-1和白介素-1β(IL-1β)的能力。其中,选择9个用于测量其caspase-1的IC50和细胞毒性分析。最后,选择三种化合物来评估它们对小鼠IL-1β的抑制作用。结果表明,在较低的给药浓度下,与MCC950相比,化合物5m具有更好的降低脑中IL-1β水平的能力,表明5m具有穿透血脑屏障(BBB)并在体外和体内抑制炎症的潜力。化合物5m在NLRP3上的对接研究揭示了类似于MCC950的结合模式。这些发现表明化合物5m有望作为AD治疗的NLRP3抑制剂。
    Alzheimer\'s disease (AD) remains an incurable neurodegenerative condition that poses a threat to humanity. Immune signaling in the brain, particularly the NLR family pyrin domain containing 3 (NLRP3), is currently targeted for AD treatment. Based on the crystal structure of the NACHT domain of NLRP3 and its renowned inhibitor MCC950, we designed and synthesized nineteen sulfonylurea compounds and evaluated their capacity to inhibit caspase-1 and interleukin-1β (IL-1β). Of these, nine were selected for measuring their IC50 for caspase-1 and cytotoxicity analysis. Finally, three compounds were chosen to assess their inhibitory effect on IL-1β in mice. The results showed that compound 5m had a superior ability to reduce IL-1β levels in the brain compared to MCC950 at a lower dosing concentration, indicating that 5m has the potential to penetrate the blood-brain barrier (BBB) and inhibit inflammation both in vitro and in vivo. Docking studies of compound 5m on NLRP3 revealed a binding mode similar to MCC950. These findings suggest that compound 5m holds promise as an NLRP3 inhibitor for AD treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    神经炎症是卒中相关脑损伤的关键病理生理特征。局部先天免疫应答通过激活炎性体触发中风后的神经炎症。核苷酸结合寡聚化结构域富含亮氨酸的重复序列和含pyrin结构域的蛋白3(NLRP3)炎性体与中风病理生物学密切相关。中风后,已经提出了几种刺激来触发NLRP3炎性体的组装。最近的研究提高了人们的理解,并揭示了一些调节NLRP3炎症小体介导的神经炎症的新参与者。本文讨论了NLRP3组装的最新进展,并强调了卒中诱导的线粒体功能障碍是调节NLRP3激活的主要检查点。NLRP3炎性体激活导致caspase-1依赖性成熟和IL-1β释放,IL-18和gasderminD。此外,在卒中实验模型中,NLRP3炎性体激活和下游信号传导的遗传或药理学抑制已被证明可减轻脑梗死并改善神经系统预后.靶向NLRP3炎性体的几种药物样小分子正处于不同的发展阶段,作为各种炎症状态的新疗法。包括中风。了解这些分子如何干扰NLRP3炎性体组装对于更好地优化和/或开发新的NLRP3抑制剂至关重要。在这次审查中,我们总结了NLRP3炎性体的组装,并讨论了在了解NLRP3炎性体介导的卒中后神经炎症的上游调节因子方面的最新进展.此外,我们严格研究了NLRP3炎性体介导的信号传导在卒中病理生理学中的作用,以及针对NLRP3炎性体相关信号传导用于卒中治疗的治疗模式的发展.
    Neuroinflammation is a key pathophysiological feature of stroke-associated brain injury. A local innate immune response triggers neuroinflammation following a stroke via activating inflammasomes. The nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome has been heavily implicated in stroke pathobiology. Following a stroke, several stimuli have been suggested to trigger the assembly of the NLRP3 inflammasome. Recent studies have advanced the understanding and revealed several new players regulating NLRP3 inflammasome-mediated neuroinflammation. This article discussed recent advancements in NLRP3 assembly and highlighted stroke-induced mitochondrial dysfunction as a major checkpoint to regulating NLRP3 activation. The NLRP3 inflammasome activation leads to caspase-1-dependent maturation and release of IL-1β, IL-18, and gasdermin D. In addition, genetic or pharmacological inhibition of the NLRP3 inflammasome activation and downstream signaling has been shown to attenuate brain infarction and improve the neurological outcome in experimental models of stroke. Several drug-like small molecules targeting the NLRP3 inflammasome are in different phases of development as novel therapeutics for various inflammatory conditions, including stroke. Understanding how these molecules interfere with NLRP3 inflammasome assembly is paramount for their better optimization and/or development of newer NLRP3 inhibitors. In this review, we summarized the assembly of the NLRP3 inflammasome and discussed the recent advances in understanding the upstream regulators of NLRP3 inflammasome-mediated neuroinflammation following stroke. Additionally, we critically examined the role of the NLRP3 inflammasome-mediated signaling in stroke pathophysiology and the development of therapeutic modalities to target the NLRP3 inflammasome-related signaling for stroke treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Randomized Controlled Trial
    背景:NLRP3炎性体驱动促炎细胞因子(包括白介素(IL)-1β和IL-18)的释放,是溃疡性结肠炎(UC)的潜在靶标。Selnoflast(RO7486967)是一种口服活性物质,强力,选择性和可逆的小分子NLRP3抑制剂。我们做了一个随机的,安慰剂对照1b期研究,以评估安全性,耐受性,塞诺司特的药代动力学(PK)和药效学(PD)。
    方法:将19例先前诊断为UC且目前患有活动性中度至重度疾病的成年人以2:1的比例随机分配至selnoflast或安慰剂治疗7天。选择450mgQD(每日一次)的剂量以在血浆和结肠组织中实现90%IL-1β抑制。连续的血,分析乙状结肠活检和粪便样本的各种PD标志物。还评估了安全性和PK。
    结果:Selnoflast耐受性良好。口服后血浆浓度迅速增加,给药后1小时达到Tmax。在整个给药间隔内,平均血浆浓度保持在IL-1βIC90水平以上(第1天和第5天的平均Ctrough:2.55μg/mL和2.66μg/mL,分别)。在稳定状态下,乙状结肠中的给药后氟烷酮浓度(5-20μg/g)高于IC90。在用脂多糖(LPS)离体刺激后,全血中的IL-1β的产生减少(在selnoflast臂中)。未观察到血浆IL-18水平的变化。在乙状结肠组织中IL-1相关基因签名的表达没有有意义的差异,粪便生物标志物的表达无差异。
    结论:Selnoflast是安全且耐受性良好的。Selnofflast450mgQD实现了血浆和组织暴露,预测在给药间隔内维持IL-1βIC90。然而,PD生物标志物结果显示治疗组之间没有显著差异,提示在UC中没有重大的治疗效果。这项研究的局限性在于其样本量小和对组织中IL-1β的影响的间接评估。
    背景:ISRCTN16847938。
    The NLRP3 inflammasome drives release of pro-inflammatory cytokines including interleukin (IL)-1β and IL-18 and is a potential target for ulcerative colitis (UC). Selnoflast (RO7486967) is an orally active, potent, selective and reversible small molecule NLRP3 inhibitor. We conducted a randomized, placebo-controlled Phase 1b study to assess the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of selnoflast.
    Nineteen adults with previous diagnosis of UC and current active moderate to severe disease were randomized 2:1 to selnoflast or placebo for 7 days. A dose of 450 mg QD (once daily) was selected to achieve 90% IL-1β inhibition in plasma and colon tissue. Consecutive blood, sigmoid colon biopsies and stool samples were analyzed for a variety of PD markers. Safety and PK were also evaluated.
    Selnoflast was well-tolerated. Plasma concentrations increased rapidly after oral administration, reaching Tmax 1 h post-dose. Mean plasma concentrations stayed above the IL-1β IC90 level throughout the dosing interval (mean Ctrough on Day 1 and Day 5: 2.55 μg/mL and 2.66 μg/mL, respectively). At steady state, post-dose selnoflast concentrations in sigmoid colon (5-20 μg/g) were above the IC90 . Production of IL-1β was reduced in whole blood following ex vivo stimulation with lipopolysaccharide (LPS) (in the selnoflast arm). No changes were observed in plasma IL-18 levels. There were no meaningful differences in the expression of an IL-1-related gene signature in sigmoid colon tissue, and no differences in the expression of stool biomarkers.
    Selnoflast was safe and well-tolerated. Selnoflast 450 mg QD achieved plasma and tissue exposure predicted to maintain IL-1β IC90 over the dosing interval. However, PD biomarker results showed no robust differences between treatment arms, suggesting no major therapeutic effects are to be expected in UC. The limitations of this study are its small sample size and indirect assessment of the effect on IL-1β in tissue.
    ISRCTN16847938.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    脓毒症诱发的心肌功能障碍(SIMD)是世界范围内严重影响健康的问题之一。目前,SIMD的机制仍未明确阐明。NOD样受体蛋白3(NLRP3)炎症小体被认为通过调节多个生物学过程参与SIMD的病理生理学。NLRP3炎性体及其相关信号通路可能影响炎症的调节,自噬,凋亡,以及SIMD中的焦亡。NLRP3炎性体的一些分子特异性抑制剂(例如,褪黑激素,乌司他丁,Irisin,硝呋嗪,和人参皂苷Rg1等。)已经开发出来了,在SIMD的细胞或动物模型中显示出有希望的抗炎作用。这些实验结果表明,NLRP3炎性体可能是SIMD治疗的有希望的治疗靶标。然而,用于治疗SIMD的NLRP3抑制剂的临床转化仍需要强有力的体内和临床前试验.
    Sepsis-induced myocardial dysfunction (SIMD) is one of the serious health-affecting problems worldwide. At present, the mechanisms of SIMD are still not clearly elucidated. The NOD-like receptor protein 3 (NLRP3) inflammasome has been assumed to be involved in the pathophysiology of SIMD by regulating multiple biological processes. NLRP3 inflammasome and its related signaling pathways might affect the regulation of inflammation, autophagy, apoptosis, and pyroptosis in SIMD. A few molecular specific inhibitors of NLRP3 inflammasome (e.g., Melatonin, Ulinastatin, Irisin, Nifuroxazide, and Ginsenoside Rg1, etc.) have been developed, which showed a promising anti-inflammatory effect in a cellular or animal model of SIMD. These experimental findings indicated that NLRP3 inflammasome could be a promising therapeutic target for SIMD treatment. However, the clinical translation of NLRP3 inhibitors for treating SIMD still requires robust in vivo and preclinical trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Preprint
    尽管年龄相关性黄斑变性(AMD)中脉络膜新生血管(CNV)的发病机制在很大程度上是未知的,炎症小体可能有助于CNV的发生和发展。为了了解NLRP3炎性体在CNV中的作用,我们使用Ccr2RFPCx3cr1GFP双报告小鼠来表征激光诱导的布鲁赫膜破裂后Ccr2RFP阳性单核细胞和Cx3cr1GFP阳性小胶质细胞向CNV病变的迁移。MCC950用作NLRP3抑制剂。免疫染色用于确认NLRP3炎性体在LCNV病变中的定位。使用共聚焦显微镜对LCNV体积进行成像和定量。ELISA和qRT-PCR通过监测LCNV小鼠脉络膜组织中IL-1β蛋白和mRNA的表达来确认NLRP3的激活。此外,使用NLRP3(-/-)LCNV小鼠来研究NLRP3炎性体是否有助于LCNV病变的发展。我们观察到RFP阳性单核细胞衍生的巨噬细胞和GFP阳性小胶质细胞衍生的巨噬细胞,除了其他细胞类型,在激光损伤后第7天定位在LCNV病变中。此外,NLRP3炎性体与LCNV病变相关。抑制NLRP3炎性体,使用MCC950,导致Ccr2RFP阳性巨噬细胞增加,Cx3cr1GFP阳性小胶质细胞,和其他细胞导致总病变大小增加。NLRP3(-/-)LCNV小鼠,显示与年龄匹配的对照组相比,病变大小显着增加。抑制NLRP3,导致脉络膜组织中IL-1βmRNA和蛋白表达降低,提示病变大小增加可能与IL-1β没有直接关系。
    Though the pathogenesis of choroidal neovascularization (CNV) is largely unknown in age-related macular degeneration (AMD), inflammasomes may contribute to CNV development and progression. To understand the role NLRP3 inflammasomes in CNV, we used Ccr2RFPCx3cr1GFP dual-reporter mice to characterize migration of Ccr2RFP positive monocytes and Cx3cr1GFP positive microglial cells into CNV lesions after laser-induced rupture of Bruch\'s membrane. MCC950 was used as NLRP3 inhibitor. Immunostaining was used to confirm localization of NLRP3 inflammasomes in the LCNV lesions. Confocal microscopy was used to image and quantify LCNV volumes. ELISA and qRT-PCR were used to confirm the activation of NLRP3 by monitoring the expression of IL-1β protein and mRNA in choroidal tissues from LCNV mice. In addition, NLRP3 (-/-) LCNV mice were used to investigate whether NLRP3 inflammasomes contribute to the development of LCNV lesions. We observed that RFP positive monocyte-derived macrophages and GFP positive microglia-derived macrophages, in addition to other cell types, were localized in LCNV lesions at day 7 post-laser injury. In addition, NLRP3 inflammasomes are associated with LCNV lesions. Inhibition of NLRP3 inflammasomes, using MCC950, caused an increased Ccr2RFP positive macrophages, Cx3cr1GFP positive microglia, and other cells resulting in an increase in total lesion size. NLRP3 (-/-) LCNV mice, showed significantly increased lesion size compared to age-matched controls. Inhibition of NLRP3, resulted in decreased IL-1β mRNA and protein expression in the choroidal tissues, suggesting that increased lesion size may not be directly related to IL-1β.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 尽管对更新的需求不断增加,更安全,更有效,和更实惠的疗法来治疗多种疾病和病症,药物开发需要太长时间,成本太高,并且太不确定,无法在不授予排他性权利或进入壁垒以激励和维持对其的投资的情况下进行。这些进入壁垒采取保护知识产权的专利和法规规定的营销排他性条款的形式。这篇综述的重点是新化学实体(NCEs)的监管和专利排他性的基本来龙去脉。指的是以前从未批准过的具有全新活性成分的药物,符合资格,并使用RRx-001,一种小分子航空航天衍生的NCE开发用于治疗癌症,辐射毒性,和NLR家族pyrin结构域含3(NLRP3)炎性体的疾病,作为一个“现实世界”的例子。这旨在作为“101型”的底漆;其目的是帮助原始药物的开发人员浏览专利的迷宫,其他知识产权法规,以及主要市场的法定排他性,以便他们能够适当利用它们。
    Despite an ever-increasing need for newer, safer, more effective, and more affordable therapies to treat a multitude of diseases and conditions, drug development takes too long, costs too much, and is too uncertain to be undertaken without the conferment of exclusionary rights or entry barriers to motivate and sustain investment in it. These entry barriers take the form of patents that protect intellectual property and marketing exclusivity provisions that are provided by statute. This review focuses on the basic ins and outs of regulatory and patent exclusivities for which new chemical entities (NCEs), referring to never-before approved drugs with an entirely new active ingredient, are eligible and uses RRx-001, a small molecule aerospace-derived NCE in development for the treatment of cancer, radiation toxicity, and diseases of the NLR family pyrin domain containing 3 (NLRP3) inflammasome, as a \"real world\" example. This is intended as a \'101-type\' of primer; its aim is to help developers of original pharmaceuticals navigate the maze of patents, other IP regulations, and statutory exclusivities in major markets so that they can make proper use of them.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:Cryopyrin相关周期性综合征(CAPS)是一种由NLRP3功能获得突变引起的遗传性自身炎症性疾病。尽管CAPS患者经常患有感音神经性听力损失,目前尚不清楚NLRP3中CAPS相关突变是否与听力损失进展相关.
    方法:我们产生了小鼠,在耳蜗驻留的CX3CR1巨噬细胞中条件表达了CAPS相关的NLRP3突变体(D301N),并检查了CAPS小鼠在局部和全身炎症环境中对炎症介导的听力损失的易感性。
    结果:中耳腔注射脂多糖(LPS)后,NLRP3突变小鼠表现出严重的耳蜗炎症,炎症体激活和听力损失。然而,这种中耳注射模型在对照小鼠中引起相当大的听力损失,并且不可避免地引起不依赖炎症的听力损失,这可能是由于注射程序引起的耳组织损伤。随后,我们优化了全身LPS注射模型,在NLRP3突变小鼠中引起明显的听力损失,但在对照小鼠中没有。反复低剂量注射LPS引起的外周炎症导致血液迷宫屏障破坏,巨噬细胞以NLRP3依赖性方式渗入耳蜗和耳蜗炎症体激活。有趣的是,耳蜗浸润巨噬细胞和常驻巨噬细胞均导致CAPS小鼠外周炎症介导的听力损失.此外,NLRP3特异性抑制剂,MCC950和白介素-1受体拮抗剂可显着减轻NLRP3突变小鼠的全身性LPS诱导的听力损失和炎症表型。
    结论:我们的研究结果表明,在我们的CAPS小鼠模型中,CAPS相关的NLRP3突变对于外周炎症诱导的听力损失至关重要,NLRP3特异性抑制剂可用于治疗炎症介导的感觉神经性听力损失。
    背景:韩国国家研究基金会由韩国政府和延世大学医学院团队科学奖资助。
    BACKGROUND: Cryopyrin-associated periodic syndrome (CAPS) is an inherited autoinflammatory disease caused by a gain-of-function mutation in NLRP3. Although CAPS patients frequently suffer from sensorineural hearing loss, it remains unclear whether CAPS-associated mutation in NLRP3 is associated with the progression of hearing loss.
    METHODS: We generated a mice with conditional expression of CAPS-associated NLRP3 mutant (D301N) in cochlea-resident CX3CR1 macrophages and examined the susceptibility of CAPS mice to inflammation-mediated hearing loss in a local and systemic inflammation context.
    RESULTS: Upon lipopolysaccharide (LPS) injection into middle ear cavity, NLRP3 mutant mice exhibited severe cochlear inflammation, inflammasome activation and hearing loss. However, this middle ear injection model induced a considerable hearing loss in control mice and inevitably caused an inflammation-independent hearing loss possibly due to ear tissue damages by injection procedure. Subsequently, we optimized a systemic LPS injection model, which induced a significant hearing loss in NLRP3 mutant mice but not in control mice. Peripheral inflammation induced by a repetitive low dose of LPS injection caused a blood-labyrinth barrier disruption, macrophage infiltration into cochlea and cochlear inflammasome activation in an NLRP3-dependent manner. Interestingly, both cochlea-infiltrating and -resident macrophages contribute to peripheral inflammation-mediated hearing loss of CAPS mice. Furthermore, NLRP3-specific inhibitor, MCC950, as well as an interleukin-1 receptor antagonist significantly alleviated systemic LPS-induced hearing loss and inflammatory phenotypes in NLRP3 mutant mice.
    CONCLUSIONS: Our findings reveal that CAPS-associated NLRP3 mutation is critical for peripheral inflammation-induced hearing loss in our CAPS mice model, and an NLRP3-specific inhibitor can be used to treat inflammation-mediated sensorineural hearing loss.
    BACKGROUND: National Research Foundation of Korea Grant funded by the Korean Government and the Team Science Award of Yonsei University College of Medicine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号