首页 | 本学科首页   官方微博 | 高级检索  
检索        


AIM2 and NLRC4 inflammasomes contribute with ASC to acute brain injury independently of NLRP3
Authors:Adam Denes  Graham Coutts  Nikolett Lénárt  Sheena M Cruickshank  Pablo Pelegrin  Joanne Skinner  Nancy Rothwell  Stuart M Allan  David Brough
Abstract:Inflammation that contributes to acute cerebrovascular disease is driven by the proinflammatory cytokine interleukin-1 and is known to exacerbate resulting injury. The activity of interleukin-1 is regulated by multimolecular protein complexes called inflammasomes. There are multiple potential inflammasomes activated in diverse diseases, yet the nature of the inflammasomes involved in brain injury is currently unknown. Here, using a rodent model of stroke, we show that the NLRC4 (NLR family, CARD domain containing 4) and AIM2 (absent in melanoma 2) inflammasomes contribute to brain injury. We also show that acute ischemic brain injury is regulated by mechanisms that require ASC (apoptosis-associated speck-like protein containing a CARD), a common adaptor protein for several inflammasomes, and that the NLRP3 (NLR family, pyrin domain containing 3) inflammasome is not involved in this process. These discoveries identify the NLRC4 and AIM2 inflammasomes as potential therapeutic targets for stroke and provide new insights into how the inflammatory response is regulated after an acute injury to the brain.Proinflammatory cytokines of the interleukin-1 (IL-1) family are critical regulators of host responses to infection and orchestrate damaging inflammatory responses that occur during disease (1). One of the main mediators of damaging sterile inflammation is IL-1β, which is implicated in the etiology of many major diseases, including acute cerebrovascular disease (2). Acute cerebrovascular disease presents as a range of conditions, including devastating injuries such as subarachnoid hemorrhage (SAH) and ischemic stroke, which account for up to 10% of mortality worldwide and are the leading cause of morbidity (2). Treatments for acute stroke are limited to thrombolysis for up to 10% of all strokes, antiplatelet agents, and stroke unit care. Thus, treatment of acute cerebrovascular disease remains an area of unmet clinical need. Understanding the mechanisms regulating production of IL-1β during ischemic brain injury may lead to the identification of new therapeutic targets.IL-1β is produced by many cells, most commonly those of macrophage lineage, as a pro–IL-1β precursor. Pro–IL-1β is expressed in response to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) that bind to pattern recognition receptors (PRRs) to up-regulate proinflammatory gene expression (3, 4). PAMPs are motifs carried by pathogens, such as bacterial endotoxin (or LPS), and DAMPs are commonly endogenous molecules released by necrosis. Pro–IL-1β is inactive and remains intracellular until a further PAMP or DAMP stimulation activates cytosolic PRRs, often of the nucleotide-binding domain and leucine-rich repeat containing receptor (NLR) family, to form large multiprotein complexes called inflammasomes (5). These complexes consist of the PRR, procaspase-1, and, depending upon the PRR, an adaptor protein called ASC, that interact via CARD and pyrin homology-binding domains (5). When the PRR senses PAMPs or DAMPs, it recruits ASC, which in turn recruits caspase-1, causing its activation. Caspase-1 then processes pro–IL-1β to a mature form that is rapidly secreted from the cell (5). The activation of caspase-1 can also cause cell death (6).A number of inflammasome-forming PRRs have been identified, including NLR family, pyrin domain containing 1 (NLRP1); NLRP3; NLRP6; NLRP7; NLRP12; NLR family, CARD domain containing 4 (NLRC4); AIM 2 (absent in melanoma 2); IFI16; and RIG-I (5). Of these inflammasomes identified to date, the best characterized, and most strongly associated with sterile inflammation, is formed by NLRP3 (7). Indeed, there are now several studies suggesting that NLRP3 inflammasomes contribute to ischemic brain injury (8, 9). However, the picture is more complicated. NLRP1 inflammasomes have been implicated in several models of brain injury (6, 10, 11), as have AIM2 inflammasomes, which are suggested to mediate pyroptotic neuronal cell death (12). There is also evidence supporting a role for caspase-1 in brain injury (13), with a selective caspase-1 inhibitor, VRT-018858, a nonpeptide, active metabolite of the prodrug pralnacasan, showing marked protection in a rat model of stroke (14). However, data for the related caspase-1 inhibitor VRT-043198 suggest that it is also an effective inhibitor of caspase-4 (15), a human ortholog of caspase-11. Caspase-11 is also implicated in ischemic brain injury (16, 17), and given that we now also know that the original caspase-1−/− mouse is also deficient in caspase-11 (18), it is clear that caspase-11 could have a role in ischemic brain injury. Our aim here was to elucidate which inflammasomes contribute to ischemic brain injury, using mice in which specific inflammasome components are deleted (−/−).
Keywords:inflammation  inflammasome  cerebral ischemia  brain injury  cell death
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