Abstract

Inflammasomes are multimeric protein complexes that induce the cleavage and release of bioactive IL-1β and cause a lytic form of cell death, termed pyroptosis. Due to its diverse triggers, ranging from infectious pathogens and host danger molecules to environmental irritants, the NOD-like receptor protein 3 (NLRP3) inflammasome remains the most widely studied inflammasome to date. Despite intense scrutiny, a universal mechanism for its activation remains elusive, although, recent research has focused on mitochondrial dysfunction or potassium (K+) efflux as key events. In this review, we give a general overview of NLRP3 inflammasome activation and explore the recently emerging noncanonical and alternative pathways to NLRP3 activation. We highlight the role of the NLRP3 inflammasome in the pathogenesis of metabolic disease that is associated with mitochondrial and oxidative stress. Finally, we interrogate the mechanisms proposed to trigger NLRP3 inflammasome assembly and activation. A greater understanding of how NLRP3 inflammasome activation is triggered may reveal new therapeutic targets for the treatment of inflammatory disease.

Review of the ambiguities surrounding mitochondrial damage-mediated activation of NLRP3, including new models for how cell death may activate NLRP3 to influence inflammation.

1 INTRODUCTION

Inflammasomes are cytosolic multimeric protein complexes that assemble upon exposure to pathogen and host-derived danger signals to modulate the host immune response. Assembly of the inflammasome triggers autoproteolytic cleavage and activation of the inflammatory cysteine aspartic protease, caspase-1. Active caspase-1 subsequently induces the cleavage and maturation of the proinflammatory cytokines, pro-IL-1β and pro-IL-18, and cleaves gasdermin D (GSDMD) to cause pore formation and induce cellular lysis, or pyroptotic cell death.1,2,3 Since the discovery of the nucleotide-binding oligomerization domain (NOD)-like receptor 1 (NLRP1) inflammasome by the laboratory of Jürg Tschopp in 2002,4 several cytosolic pattern recognition receptors (PRRs) have now been shown to assemble functional inflammasomes, or trigger caspase-1 activation, upon sensing pathogen- or host damage-associated molecular patterns (PAMPS and DAMPS).5 These include members of the NLR family, such as NLRP3, NLRC4 (IPAF), and the less well studied human NLRP2, NLRP7 and mouse NLRP6, NLRP9b, and NLRP12 sensor proteins,6,7,8,9,10,11 the HIN-200 absent in melanoma-like receptors, AIM2 and IFI-16,12,13 and the tripartite motif (TRIM) family receptor, Pyrin.14 To date, the majority of inflammasome sensors exhibit selectivity for physiological triggers. NLRP1 is activated by anthrax lethal toxin secreted by Bacillus anthracis,15,16 while AIM2 senses cytosolic bacterial, viral or host dsDNA,12,17,18 and Pyrin is triggered by Rho GTPase inactivating bacterial toxins/effectors, such as Clostridium difficile toxin A/B.14 In the case of NLRC4, it does not directly sense Gram-negative bacteria (e.g., Salmonella Typhimurium) but instead utilizes NLR family of apoptosis inhibitory protein (NAIP) co-receptors that specifically sense flagellin and components of the type III secretion system (T3SS).19,20,21,22 It is important to note that in humans there is a sole NAIP co-receptor, while in mice NAIP5 and NAIP6 sense flagellin, and NAIP1 and NAIP2 recognize needle and inner rod proteins of the T3SS, respectively.19,20,21,22 In contrast to the selective nature of most inflammasome sensors, NLRP3 recognizes a diverse range of pathogen and danger molecules, including bacterial toxins (e.g., pore-forming toxin nigericin), host metabolites (e.g., uric acid, cholesterol crystals) and environmental irritants (e.g., asbestos, silica), which has implicated the NLRP3 inflammasome in the pathogenesis of numerous infectious and inflammatory diseases.23

Upon sensing a danger signal, inflammasome sensors recruit the bipartite adaptor, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), via common pyrin domain (PYD) interactions, or in the case of CARD containing receptors (i.e., NLRC4, NLRP1) associate via CARD-CARD interactions. This facilitates ASC polymerization through homotypic PYD-PYD interactions to form long helical prion-like filaments, which are condensed into a discrete ASC speck, in a CARD-dependent manner.24,25,26,27 In turn, ASC recruits pro-caspase-1 via its N-terminal CARD domain (CARD-CARD interaction) to promote proximity-induced caspase-1 dimerization and autoproteolysis to release the large (p20) and small (p10) subunits. Until recently, it was widely assumed that the labile caspase-1 tetramer comprising two p20 and two p10 subunits (p20/10) was the active form. Undeniably, superphysiological levels of recombinant p20/10 caspase-1 is catalytically active.28,29,30 However, recent work by the laboratory of Kate Schroder has indicated that, in fact, the inflammasome-associated full-length caspase-1 (p46), as well as the transient p33/p10 species, are the bioactive forms.30 Instead, the p20/10 tetramers are the result of p33/10 self-cleavage to terminate caspase-1 signaling.30 In support of this notion, the CARD-containing NLRC4 and NLRP1b inflammasome sensors can interact directly with caspase-1 to drive pyroptosis (and limited IL-1β activation) independent of caspase-1 proteolysis.31,32 Moreover, dimerized caspase-1 has been shown to induce the activation and secretion of IL-1β in the absence of proteolysis and cell death.33 Hence, it remains of interest to reassess the cell-specific kinetics of activation/inactivation of caspase-1 by different inflammasome complexes.

The primary functions of active caspase-1 is to induce the cleavage of pro-IL-1β and -IL-18 to their bioactive forms, and cleave GSDMD (at Asp 276 in mice and Asp275 in humans) to release an active effector N-terminal GSDMD (GSDMD-NT) p30 fragment that causes pyroptotic cell death.2,3,34 GSDMD-NT binds and inserts into lipid membranes, and oligomerizes to form transmembrane pores (∼10–20 nm) that causes a loss of cell membrane integrity and the lytic release of cytoplasmic contents (e.g., HMGB1, IL-1α).35,36,37,38 GSDMD pores also facilitate the release of mature IL-1β and IL-18, and this has recently been suggested to occur immediately prior to pyroptotic cell death.39,40 It is worth mentioning that caspase-1 and GSDMD are not essential for cell death to all inflammasome triggers.3,34,41,42,43 In fact, in certain cases NLRP3 itself is dispensable, such as upon exposure to lysosomal-disrupting agents, crystalline and particulate matter.44,45,46 Likewise, inflammasome-associated IL-1β secretion can occur in a GSDMD-independent manner, via caspase-8-mediated apoptosis or secondary necrosis/pyroptosis, and can even occur largely in the absence of cell death.41,47,48 Furthermore, caspase-8 is recruited to several inflammasomes to drive caspase-1-independent IL-1β activation and/or apoptotic cell death.43,49,50 Remarkably, it has now also become apparent that there is bidirectional crosstalk between apoptotic and pyroptotic cell death pathways. For example, under certain conditions, caspase-1 can cleave and activate caspase-3 and -7 to drive apoptosis.51,52 Conversely, caspase-3 and -7 cleave and inactivate GSDMD (at Asp87) to limit pyroptosis.52

Despite our improved knowledge of inflammasome assembly and downstream signaling, the upstream events that trigger activation of the most widely studied inflammasome, the NLRP3 inflammasome, remain contentious. Mitochondrial damage and oxidative stress are widely touted to be the universal mechanism to trigger NLRP3, yet, there is considerable evidence that potassium ion (K+) efflux is essential in most cases. In this review, we give an overview of NLRP3 inflammasome activity in health and disease, particularly highlighting the recent link with mitochondrial damage and oxidative stress in metabolic disease. We discuss the ambiguities surrounding mechanisms of NLRP3 activation, such as ion flux, mitochondrial damage, and reactive oxygen species (ROS) generation.

2 MODES OF NLRP3 INFLAMMASOME ACTIVATION

2.1 Canonical activation of the NLRP3 inflammasome

The NLRP3 sensor (also known as NALP3 and Cryopyrin, encoded by the NLRP3 or CIAS gene) has been extensively studied due to its involvement in many disease pathologies and the diverse nature of its triggers, which include host DAMPS, such as extracellular ATP, uric acid, and cholesterol crystals, PAMPs (e.g., bacterial mRNA and DNA, DNA/RNA viruses, fungi, and protozoa) and environmental irritants (e.g., silica, asbestos, aluminum hydroxide).23 NLRP3 comprises a C-terminal leucine-rich repeat (LRR) that may act as the sensor domain, a NACHT (or NOD) domain that enables ATP-dependent oligomerization and activation, and a PYD domain at the N-terminus to recruit the adaptor protein ASC.

Activation of the NLRP3 inflammasome in innate immune cells is generally thought of as a 2-step process (Fig. 1). In the first step (signal 1), microbial engagement of TLRs, or sterile priming (i.e., TNF receptor 1 [TNFR1] signaling), activates NF-κB-mediated transcription of NLRP3 and other proinflammatory genes, including pro-IL-1β. The second signal, such as a DAMP or PAMP, then triggers assembly and activation of the NLRP3 inflammasome via an ill-defined manner that may involve one or more events, including K+ efflux, mitochondrial damage and ROS generation, calcium (Ca2+) influx, and lysosomal rupture (see Section 4, Figs. 1 and 4). Notable exceptions to this 2-step model, include the spontaneous NLRP3 activation observed upon LPS stimulation of murine bone marrow dendritic cells (BMDCs) and human monocytes in vitro, as well as the P2X7 receptor (P2X7R)-independent activation of NLRP3 in mice upon LPS challenge (discussed further in Sections 2.2.1 and 2.2.2).53,54,55

Modes of canonical NLRP3 inflammasome activation. Activation of the canonical NLRP3 inflammasome requires a priming signal, signal 1, via engagement of pattern recognition receptors (PRRs) that induce NF-κβ-mediated transcriptional up-regulation of inflammasome machinery, namely NLRP3, pro-IL-1β and pro-IL-18. Signal 2, an NLRP3 activation signal, is triggered by a variety of DAMPS (e.g., cholesterol crystals, ATP) and PAMPS (e.g., pore-forming toxins, viral RNA). PAMPS and DAMPS trigger formation of the multimeric inflammasome complex comprising NLRP3, ASC and pro-caspase-1 via an ill-defined mechanism that may involve K+ efflux and/or Ca2+ influx, lysosomal rupture, ROS production and mitochondrial dysfunction (see Fig.). NEK7-NLRP3 interactions are essential to promote NLRP3 oligomerization and the subsequent recruitment of ASC via homotypic pyrin domain interactions. ASC oligomerization leads to amyloid-like fibril structures that can interact with capase-1 via common CARD domains and promote proximity-induced caspase-1 activation. Active caspase-1 (putative p33/10 and p46 caspase-1 subunits [dotted line box]), in turn, cleaves substrates pro-IL-1β and pro-IL-18 to their bioactive forms. Caspase-1 also cleaves GSDMD to release the N-terminal fragment that can translocate to the plasma membrane and form oligomeric pores to induce a lytic form of cell death, termed pyroptosis, and facilitate IL-1β release
Figure 1

Modes of canonical NLRP3 inflammasome activation. Activation of the canonical NLRP3 inflammasome requires a priming signal, signal 1, via engagement of pattern recognition receptors (PRRs) that induce NF-κβ-mediated transcriptional up-regulation of inflammasome machinery, namely NLRP3, pro-IL-1β and pro-IL-18. Signal 2, an NLRP3 activation signal, is triggered by a variety of DAMPS (e.g., cholesterol crystals, ATP) and PAMPS (e.g., pore-forming toxins, viral RNA). PAMPS and DAMPS trigger formation of the multimeric inflammasome complex comprising NLRP3, ASC and pro-caspase-1 via an ill-defined mechanism that may involve K+ efflux and/or Ca2+ influx, lysosomal rupture, ROS production and mitochondrial dysfunction (see Fig.). NEK7-NLRP3 interactions are essential to promote NLRP3 oligomerization and the subsequent recruitment of ASC via homotypic pyrin domain interactions. ASC oligomerization leads to amyloid-like fibril structures that can interact with capase-1 via common CARD domains and promote proximity-induced caspase-1 activation. Active caspase-1 (putative p33/10 and p46 caspase-1 subunits [dotted line box]), in turn, cleaves substrates pro-IL-1β and pro-IL-18 to their bioactive forms. Caspase-1 also cleaves GSDMD to release the N-terminal fragment that can translocate to the plasma membrane and form oligomeric pores to induce a lytic form of cell death, termed pyroptosis, and facilitate IL-1β release

In recent work by the laboratories of Bruce Beutler, Gabriel Núñez, and Veit Hornung, the serine threonine kinase NIMA-related kinase 7 (NEK7) was identified as a critical positive regulator of NLRP3 inflammasome assembly downstream of K+ efflux and ROS.56,57,58 NEK7 was shown to interact via its catalytic domain with the LRR and NACHT domain of NLRP3 to facilitate inflammasome activation in a manner independent of its kinase activity.56,57 Notably, this interaction can reportedly be disrupted by intrinsic apoptosis regulator cytochrome c to presumably limit pyroptotic cell death.59 Similarly, the anticancer ent-kaurene diterpenoid, Oridonin, which is the major constituent of Rabdosia rubescens, was recently suggested to covalently bind cysteine 279 of the NACHT domain to block NEK7-NLRP3 interactions.60

It should be acknowledged that in addition to transcriptional regulation (i.e., signal 1), TLR signaling also induces post-translational regulation of NLRP3 activation to expedite an inflammatory response.61,62,63,64,65 Brief exposure of macrophages to TLR agonists that signal via MyD88 (10–20 min), and NLRP3 activators nigericin and ATP, or Listeria monocytogenes, induces rapid IL-1 receptor associated kinase-1 (IRAK1)/IRAK4- and mitochondrial ROS (mtROS)-dependent inflammasome assembly and activation.64,65 Further to this, Juliana et al. earlier showed that both LPS and ATP are capable of inducing the deubiquitylation of NLRP3 to regulate inflammasome activity, although surprisingly, only LPS required mtROS to trigger deubiquitinase (DUB) activity.63 The DUB, BRCC3 (BRCC36 in humans), was recently revealed to deubiquitylate NLRP3 to induce inflammasome activation.66,67 A number of additional ubiquitin modification events have now been proposed to regulate NLRP3 inflammasome assembly and activation. For example, the E3 ubiquitin ligase HOIL-1, a linear ubiquitination chain assembly complex (LUBAC) component, was suggested to ubiquitylate ASC to promote NLRP3 inflammasome assembly; however, related complex member SHARPIN was found to only be required for inflammasome priming.68,69 In contrast, the DUB A20 has been shown to limit NLRP3 activation by deubiquitylating inflammasome-associated IL-1β.70 Several E3 ubiquitin ligases, including F-box L2 (FBXL2) and TRIM31, as well as membrane-associated RING finger protein 7 (MARCH7), have also been reported to promote K48-linked polyubiquitylation of NLRP3 to promote its proteasomal or autophagolysomal degradation.71,72,73 It has now become increasingly clear that multiple post-translational modification events fine-tune NLRP3 inflammasome assembly and activation, including ubiquitylation, phosphorylation, acetylation, and nitrosylation; however, most results require further validation (reviewed in Poudel et al.).74

2.2 Noncanonical and alternative pathways to NLRP3 activation

2.2.1 Noncanonical caspase-11-mediated activation of the NLRP3 inflammasome

In 2011, the laboratory of Vishva Dixit identified that the murine inflammatory caspase, caspase-11, acts as the cytosolic sensor of intracellular LPS derived from Gram-negative bacteria to promote noncanonical activation of NLRP375 (Fig. 2). In this pioneering work, Dixit and colleagues confirmed reports by the laboratory of Junying Yuan that the widely used caspase-1-deficient mouse lines derived in 129 embryonic stem cells lacked caspase-11 expression.76 This effect was caused by a mutation in the 129 caspase-11 locus, which due to its close proximity to caspase-1, could not be segregated upon backcrossing onto the C57BL/6 background.75 Subsequent examination of caspase-11-deficient bone marrow-derived macrophages (BMDMs) revealed that caspase-11 was essential for NLRP3 inflammasome-mediated IL-1β and IL-18 activation in response to cholera toxin B, Escherichia coli, Citrobacter rodentium, and Vibrio cholerae, but not canonical activators, such as ATP and MSU.75 Importantly, while caspase-11 activation triggered the NLRP3-caspase-1 inflammasome to activate IL-1β and IL-18, caspase-11 alone was sufficient to trigger pyroptotic cell death and lethal sepsis.75 Caspase-11 has now been shown to be critical for inducing lethal sepsis in response to LPS,75,77 and protecting mice from Gram-negative bacterial infections78,79,80,81 and acute dextran sulfate sodium-induced colitis.82,83

The noncanonical caspase-11-dependent pathway to NLRP3 inflammasome activation. Activation of the noncanonical caspase-11 NLRP3 inflammasome requires TLR- and type I IFN-induced transcription of pro-caspase-11, guanylate binding proteins (GBP), as well as NLRP3 machinery. Caspase-11 is activated within the cytosol by LPS derived from Gram-negative bacteria (e.g., Burkholderia, Legionella) that escape phagocytic vacuoles in a GBP-dependent manner, or accesses the cytosol via outer membrane vesicles (OMVs). The cytosolic hydrophobic hexa-acetylated lipid A moiety of LPS directly binds the CARD domain of caspase-11 to facilitate caspase-11 oligomerization and activation. Active caspase-11 induces the cleavage and release of the N-terminal domain of GSDMD to form pores in the cell membrane and cause pyroptotic cell death. Active caspase-11 also reportedly cleaves the pannexin-1 hemichannel to facilitate the release of ATP to perpetuate pyroptosis via P2X7R signalling, and activates NLRP3 via K+ efflux
Figure 2

The noncanonical caspase-11-dependent pathway to NLRP3 inflammasome activation. Activation of the noncanonical caspase-11 NLRP3 inflammasome requires TLR- and type I IFN-induced transcription of pro-caspase-11, guanylate binding proteins (GBP), as well as NLRP3 machinery. Caspase-11 is activated within the cytosol by LPS derived from Gram-negative bacteria (e.g., Burkholderia, Legionella) that escape phagocytic vacuoles in a GBP-dependent manner, or accesses the cytosol via outer membrane vesicles (OMVs). The cytosolic hydrophobic hexa-acetylated lipid A moiety of LPS directly binds the CARD domain of caspase-11 to facilitate caspase-11 oligomerization and activation. Active caspase-11 induces the cleavage and release of the N-terminal domain of GSDMD to form pores in the cell membrane and cause pyroptotic cell death. Active caspase-11 also reportedly cleaves the pannexin-1 hemichannel to facilitate the release of ATP to perpetuate pyroptosis via P2X7R signalling, and activates NLRP3 via K+ efflux

Several recent studies have shed light on how murine caspase-11, and human homologs caspase-4 and -5, are activated by the hexa-acetylated lipid A moiety of LPS to trigger NLRP3 inflammasome activity and cause pyroptosis in innate immune cells.75,84,85,86,87 The laboratory of Feng Shao proposed that caspase-11, -4, and -5 specifically bind the lipid A moiety of LPS via their CARD domains to induce caspase oligomerization and activation.84 It is important to note, that caspase-11 can be activated via transfection, electroporation or cholera toxin A delivery of LPS to the intracellular compartment.75,77,87 In activated dendritic cells, and possibly retinal epithelial cells, caspase-11 can also reportedly be triggered by oxidized phospholipids (OxPAPC) released from dying cells.88,89 Moreover, high doses of OxPAPC components have been shown to trigger both noncanonical and canonical NLRP3 activation in macrophages.90,91 Perplexingly, a recent study disputed these findings and concluded that OxPAPC binds caspase-11 to actually limit NLRP3 activation by Gram-negative bacteria.92 More recently, it has become evident that outer membrane vesicles derived from Gram-negative bacteria (e.g., Pseudomonas aeruginosa, Neisseria meningitides) are also sensed by specific inflammatory caspases to direct innate immune cell responses.93,94

Upon activation, caspase-11, akin to caspase-1 in the canonical pathway, induces pyroptotic cell death by directly cleaving and activating GSDMD2,3,34 (Fig. 2). However, it has additionally been shown that active casapase-11 can contribute to pyroptosis, in a GSDMD-independent manner, via the cleavage of the pannexin-1 hemichannel to allow ATP release and engagement of the purinergic P2X7R.95 Similar to most canonical triggers of NLRP3 activation, noncanonical caspase-11-mediated activation of NLRP3 is dependent on K+ efflux,95,96 and it has been suggested that caspase-11 mediated opening of the pannexin-1 channel allows K+ efflux.95 Remarkably, despite efforts at dissecting apart these pathways in myeloid cells in vitro, it now appears that caspase-11-induced GSDMD activity is not required in the hematopoietic compartment for lethal endotoxic shock in vivo.97,98 In fact, in response to E. coli endotoxin and live bacteria, the hematopoietic system initiates inflammatory events via type I IFN and TNF production, which then in turn promotes pro-pyroptotic caspase-11 and pro-apoptotic caspase-8 collaborative activity in target tissue cells (e.g., gut epithelial cell death) to amplify injury.98

Despite improved knowledge on how caspase-11 activates NLRP3 and GSDMD to drive inflammatory events, the upstream events required for activation remain ill defined. A number of studies have identified that type I IFN signaling, induced via TLR3/4 and Toll/IL-1R domain-containing adaptor-inducing IFN-β (TRIF), can amplify the expression of caspase-11.77,78,87,100 Recently, the complement-related carboxypeptidase B1 was implicated in up-regulation of caspase-4, -5, and -11 downstream of type I IFN.101 Notably, type I IFNs (and type II IFNs) also influence caspase-11 activation at another level, via the induction of IFN-inducible GTPases, including guanylate binding proteins (GBPs) and IFN-inducible protein IRGB10.102,103,104 It has been shown that GBPs and IRGB10 can liberate bacterial ligands from bacteria-containing vacuoles, and possibly outer membrane vesicles, to allow sensing by caspase-11 (Fig. 2).103,104 Interestingly, a vacuole-independent role for GBPs in the rapid noncanonical and canonical activation of NLRP3 has recently been reported upon Chlamydia muridarum infection of macrophages.105 Moreover, IRGB10 has not only been shown to permit caspase-11-mediated NLRP3 inflammasome activation to Francisella novicida, but also lyses the bacterial membrane, releasing sufficient levels of DNA to activate the AIM2 inflammasome.106 Hence, IFN-inducible GTPases appear to play a vital role in coordinating inflammasome responses to Gram-negative bacteria, and it is now emerging that bacteria have evolved effector mechanism to evade these antibacterial defense mechanisms.107

2.2.2 Alternative and cell death-induced pathways to NLRP3 inflammasome activation

Apoptosis is an evolutionarily conserved programmed cell death pathway to remove superfluous or damaged cells, promote pathogen clearance and maintain immune tolerance. Intrinsic “mitochondria-dependent” apoptosis is triggered by a range of cellular stressors, including growth factor withdrawal and ultra violet irradiation. Regulated by the balance of pro- and anti-apoptotic BCL-2 family members, the irreversible step culminating in cell death is activation of BAX and BAK oligomeric pores in the mitochondrial membrane that allows the release of mitochondrial contents, including cytochrome c. Subsequently, cytochrome c orchestrates formation of an apoptosome complex (comprising cytochrome c, apoptosis protease activating factor-1 [APAF-1] and pro-caspase-9) that induces energy-dependent autoactivation of caspase-9 to cleave effector caspases, such as caspase-3 and -7, to induce apoptosis (Fig. 3B).108 In contrast, death receptor signaling, via members of the TNFR superfamily, such as TNFR1, and members of the TLR family (i.e. TLR3 and TLR4), activates a death or “ripoptosome” complex comprising Fas-associated protein with death domain (FADD), receptor interacting protein kinase 1 (RIPK1) and pro-caspase-8, which leads to a caspase cascade and cellular demise (Fig. 3B). If caspase-8 activity is compromised, TLR and TNFR1 signaling can also induce a lytic form of cell death, called necroptosis. Necroptosis requires the kinase activity of RIPK1 and RIPK3, and the pseudokinase, Mixed lineage kinase domain-like (MLKL). Phosphorylation of MLKL by RIPK3 induces a conformational change that exposes the N-terminal four-helix bundle (4HB) killing domain and promotes its oligomerization and migration to the plasma membrane, where it inserts into the membrane to cause lytic cell death109 (Fig. 3B).

Alternative and cell death-induced pathways to NLRP3 inflammasome activation. A. The alternative NLRP3 inflammasome is activated in human monocytes by LPS. LPS signaling via TRIF recruits RIPK1, FADD and caspase-8 to trigger NLRP3 activation in a K+ efflux-independent manner. Unique to this pathway is the lack of requirement for ASC speck formation and pyroptosis. B. Multiple programmed cell death pathways trigger NLRP3 activation via K+ efflux. In the presence of LPS priming: (i) IAP loss induces formation of a RIPK1-RIPK3-FADD-caspase-8 complex, whereby activation of extrinsic apoptotic caspase-8 can activate IL-1β directly or trigger NLRP3 inflammasome activation via K+ efflux. (ii) When IAPs are absent and/or caspase-8 levels are compromised, a RIPK1/3 kinase- and MLKL-dependent necroptotic cell death pathway triggers NLRP3 activation in a K+ efflux-dependent manner. (iii) Specific activation of intrinsic effector molecules BAX and BAK, upon loss of pro-survival proteins MCL-1 and BCL-XL, triggers IAP loss to promote caspase-8 activity. BAX and BAK signaling activates effector caspases, caspase-3 and caspase-7, which cause K+ efflux to activate NLRP3 and are also required for activation of caspase-8 to cleave and activate IL-1β directly. Of note, pyroptotic effector GSDMD is dispensable for cell death and IL-1β secretion in all modes of cell-death mediated NLRP3 inflammasome activation
Figure 3

Alternative and cell death-induced pathways to NLRP3 inflammasome activation. A. The alternative NLRP3 inflammasome is activated in human monocytes by LPS. LPS signaling via TRIF recruits RIPK1, FADD and caspase-8 to trigger NLRP3 activation in a K+ efflux-independent manner. Unique to this pathway is the lack of requirement for ASC speck formation and pyroptosis. B. Multiple programmed cell death pathways trigger NLRP3 activation via K+ efflux. In the presence of LPS priming: (i) IAP loss induces formation of a RIPK1-RIPK3-FADD-caspase-8 complex, whereby activation of extrinsic apoptotic caspase-8 can activate IL-1β directly or trigger NLRP3 inflammasome activation via K+ efflux. (ii) When IAPs are absent and/or caspase-8 levels are compromised, a RIPK1/3 kinase- and MLKL-dependent necroptotic cell death pathway triggers NLRP3 activation in a K+ efflux-dependent manner. (iii) Specific activation of intrinsic effector molecules BAX and BAK, upon loss of pro-survival proteins MCL-1 and BCL-XL, triggers IAP loss to promote caspase-8 activity. BAX and BAK signaling activates effector caspases, caspase-3 and caspase-7, which cause K+ efflux to activate NLRP3 and are also required for activation of caspase-8 to cleave and activate IL-1β directly. Of note, pyroptotic effector GSDMD is dispensable for cell death and IL-1β secretion in all modes of cell-death mediated NLRP3 inflammasome activation

There is growing literature that there is significant crosstalk between modes of cell death and inflammatory signaling pathways, particularly the NLRP3 inflammasome.110 Caspase-8 has long been acknowledged to exhibit some overlap in substrate specificity with caspase-1. In 2008, the laboratory of Rudi Beyaert demonstrated that recombinant caspase-8, but not caspase-3, could cleave IL-1β at the same site as caspase-1.111 TLR3 and TLR4 ligation and protein synthesis inhibition were also shown to promote caspase-8 mediated cleavage of IL-1β.29,111 In fact, TLR-TRIF signaling alone was sufficient to trigger the formation of a signaling complex comprising FADD, RIPK1, RIPK3, and caspase-8 to induce both caspase-8 and caspase-1-mediated activation of IL-1β in BMDCs.54 Many subsequent studies have demonstrated that in TLR- and TNF-primed myeloid cells, a diverse range of cell stressors can activate caspase-8 to trigger NLRP3, or directly cleave pro-IL-1β and pro-IL-18 in myeloid cells (reviewed in Feltham et al.).110 For example, ligation of the death receptor Fas in macrophages induces a FADD-caspase-8 complex to activate IL-1β directly, in a RIPK3-independent manner.112 Whilst, treatment of LPS-primed BMDCs with pro-apoptotic chemotherapeutics, such as doxorubicin or staurosporine, induces RIPK3-dependent IL-1β production, dominantly via direct caspase-8 proteolysis.113 How diverse stressors trigger capsase-8-mediated IL-1β activity remains unclear; however, it is likely to involve changes in the level of cell death inhibitors.113,114 In line with this notion, chemical or genetic loss of all 3 E3 ubiquitin ligase inhibitor of apoptosis (IAP) proteins, namely cellular IAP1 (cIAP1) and cIAP2 and X-linked IAP (XIAP), in TLR- or TNF-primed BMDMs, BMDCs, and neutrophils induced RIPK1-RIPK3-caspase-8-mediated apoptosis (RIPK1-capase-8 in neutrophils), proteolysis of IL-1β and NLRP3 activation29,115,116,117,118 (Fig. 3B). It is important to note, that XIAP loss is essential for TLR-mediated activation of this inflammatory pathway in innate immune cells.115,116,119 Moreover, it has recently been shown that TLR-MyD88 and TNF-TNFR2-mediated proteasomal degradation of cIAP1, and its adaptor TRAF2, is required to induce caspase-8-driven NLRP3 inflammasome and IL-1β activation in the absence of XIAP.119 Intriguingly, loss of the NF-κB inhibitor and DUB, A20, also promotes TLR4-induced, RIPK3-dependent NLRP3 activation, presumably via caspase-8 activation.70 Whereas, loss of cellular FADD-like IL-1β-converting enzyme-inhibitor protein (c-FLIP), an endogenous capase-8 inhibitor, exaggerates caspase-8-mediated IL-1β secretion in response to a mimetic of the natural IAP inhibitor, second mitochondria-derived activator of caspases (SMAC).120

A number of recent studies have proposed that caspase-8 can associate directly with the NLRP3 inflammasome to drive IL-1β activation and apoptotic cell death.41,50,70,121 Work performed in the laboratory of Thirumala-Devi Kanneganti suggested that the FADD-RIPK3-caspase-8 ripoptosome complex associates with the NLRP3 inflammasome to promote both canonical and noncanonical caspase-11-mediated (e.g., C. rodentium) inflammasome activity.121 An alternative model proposed that upon Yersinia infection a RIPK1-FADD-caspase-8 complex can directly cleave caspase-1, in the absence of NLRP3 and NLRC4.122 Complicating the interpretation of these findings is the fact that caspase-8 plays a fundamental role in the transcription of cytokines (e.g. pro-IL-1β) and NLRP3 itself, as evidenced by defective TLR3/4 and Gram-negative bacterial inflammatory cytokine responses in Ripk3−/−Caspase-8−/−, Ripk3−/−FADD−/−, and Mlkl−/−FADD−/− mice.121,123,124,125 Is it worth noting that there are conflicting reports as to whether defective canonical NF-κB activation is responsible for reduced transcriptional responses in Ripk3−/−Caspase-8−/− macrophages.121,123,124 In addition, other groups have reported relatively normal canonical NLRP3 inflammasome activation upon sufficient TLR-priming in Ripk3−/−Caspase-8−/− myeloid cells (as indicated by ASC oligomerization and caspase-1 cleavage), suggesting that these findings warrant closer examination.116,122,123 Nevertheless, a recent study by Olaf Groß and colleagues established that similar to other inflammasomes, caspase-8 (as well as FADD, RIPK1 and c-FLIP) can indeed be recruited to the canonical NLRP3 inflammasome to activate IL-1β and induce secondary pyroptosis, although, this phenomenon was only pronounced in the absence of caspase-1 protease, or GSDMD, activity.41 Intriguingly, reminiscent of the noncanonical caspase-11 inflammasome pathway, a few studies have now proposed that caspase-8 can act upstream of the NLRP3 inflammasome (Fig. 3A, B).53,116 Chemical loss of the IAP proteins in LPS-primed BMDMs was first reported to induce RIPK1-RIPK3-caspase-8-dependent (and RIPK kinase independent) activation of NLRP3 via ROS and K+ efflux29,116,126 (Fig. 3B). However, divergent from the noncanonical pathway, GSDMD did not play a major role in cell death or IL-1β secretion.119 The requirement for upstream caspase-8 activity was established by James Vince and colleagues upon making the observation that NLRP3-associated caspase-1 cleavage events were blocked in unprimed, NLRP3-sufficient, Ripk3−/−Caspase-8−/− macrophages upon IAP inhibition.116 The laboratory of Veit Hornung later coined the term the “alternative NLRP3 inflammasome” to describe a similar TLR-TRIF-RIPK1-FADD-caspase-8-mediated route to NLRP3 inflammasome activation in human BlaER1 monocytic cells, which was surprisingly not dependent on RIPK3, ASC oligomerization or K+ efflux, and occurred in the absence of cell death53 (Fig. 3A).

Work centralized around unravelling how caspase-8 loss triggers lethal necroptotic signaling also uncovered that the RIPK1-RIPK3-MLKL “necrosome” complex can trigger NLRP3 inflammasome activation. The laboratory of David Wallach discovered that deletion of caspase-8 in dendritic cells rendered mice hypersensitive to LPS-induced lethality associated with RIPK3-driven NLRP3 inflammasome activity.127 In caspase-8-deficient BMDCs, TLR2/4 ligation led to spontaneous RIPK1/3 kinase, MLKL and mitochondrial phosphatase PGAM5-dependent NLRP3 inflammasome assembly, and IL-1β and IL-18 activation, but surprisingly not necrotic or pyroptotic cell death.127 Of note, a recent study revealed that PGAM5 is dispensable for necroptotic signaling, and therefore most likely PGAM5 is redundant in this mode of NLRP3 activation.128 Other groups, however, have reaffirmed that TLR ligation triggers RIPK3 and MLKL-mediated NLRP3 activation in caspase-8-deficient BMDMs and human BlaER1 monocytes, as well as in RIPK1-deficient monocyte/macrophages.53,116,129 It has also become apparent that the RIPK3-MLKL-driven NLRP3 inflammasome pathway can be further amplified in the absence of caspase-8, via the chemical or genetic loss of the IAP proteins, particularly XIAP115,116,126 (Fig. 3B). Of note, these necroptotic cell death signaling pathways to NLRP3 inflammasome activation were dependent on the kinase activities of RIPK1 and/or RIPK3, as well as the common event, K+ efflux.115,116,126

Controversially, mitochondrial dysfunction and apoptosis have been suggested to be the universal trigger for canonical NLRP3 activation (see Section 4.2). Until recently, whether direct activation of mitochondria-dependent apoptosis could drive NLRP3 inflammasome activation in innate immune cells remained unclear. Work by the laboratory of Thomas Naderer recently identified that upon infection of macrophages, Legionella inhibits host cell protein synthesis, and accordingly, reduces pro-survival MCL-1 levels, thereby rendering infected cells exquisitely sensitive to BAX/BAK-dependent apoptotic cell death upon BCL-XL inhibition.130 Remarkably, two subsequent studies have now discovered that in LPS-primed macrophages, activation of BAX and BAK signaling can trigger a novel K+ efflux-dependent pathway to NLRP3 inflammasome and IL-1β activation (Fig. 3B; Fig. #4, 11, 130,131). BAX/BAK activation, upon chemical/genetic BCL-XL and MCL-1 loss, or treatment with myxobacteria-derived cyclic peptides, vioprolides (targets BCL-2 and MCL-1), was shown to cause IAP degradation to promote caspase-8-mediated activation of IL-1β.131,132 However, wholly unexpectedly in the paper by Vince et al., caspase-8 did not act upstream of NLRP3 activation, as posited by previous studies.53,116 Instead, intrinsic apoptotic activation of effector caspases, caspase-3 and -7, triggered NLRP3 activation via K+ efflux, which was presumably caused by plasma membrane damage.131 Remarkably, caspase-3 and caspase-7 activity was also found to act upstream of caspase-8 activation.131 Furthermore, cellular demise and IL-1β secretion proceeded largely in the absence of caspase-1 and caspase-3, and their respective pyroptotic cell death effectors GSDMD and GSDME.2,3,131,133,134 Given the potential for three programmed cell death pathways to converge on NLRP3 activation, one could collectively consider them alternative cell death-induced inflammasomes.

Proposed models for NLRP3 inflammasome activation via mitochondrial dysfunction. Canonical stimuli are proposed to trigger NLRP3 inflammasome assembly and activation by damaging the mitochondria and inducing mitochondrial ROS (mtROS) production, releasing oxidized mitochondrial DNA (mtDNA) and promoting the mitochondrial localization of NLRP3 and ASC for activation. (1) Mitochondrial autophagy, mitophagy, limits NLRP3 activation by eliminating damaged or stressed mitochondria from the cell. (2-6) NLRP3 activation can be mediated by VDAC1/2-mediated mtROS production and (3) BAX/BAK activation, which can both be repressed by pro-survival protein BCL-2. (4, 6) ER stress induces C/EPB homologous protein (CHOP)-mediated transcriptional control of IP3R-dependent Ca2+ efflux from the ER into the cytosol. This induces transport of Ca2+ ions into the mitochondria that generates mtROS. (5) mtROS induces deubiquitylation of NLRP3 to promote inflammasome activation, and (6) may facilitate mtDNA release to activate NLRP3 by inducing mitochondrial pore transition (MPT) pore opening. (7) TLR4-IRF1 mediated transcriptional up-regulation of CMPK2 can generate short strands of mtDNA that exit the mitochondria in an oxidized form to bind and activate the NLRP3 inflammasome. (8) Depletion of mitochondrial NAD+ upon Sirtuin 2 inhibition may lead to hyper-acetylation of α-tubulin and induce Dynein-mediated transport of ASC on the mitochondria to NLRP3 on the ER, and facilitate NLRP3 activation. (9, 10) Cardiolipin and the mitochondrial antiviral signaling protein (MAVS) can reportedly tether NLRP3 to the mitochondria for activation in a mtROS-dependent manner. (11) Specific activation of BAX/BAK triggered by loss of BCL-XL and MCL-1 activity can trigger NLRP3 activation, via activation of downstream effector caspases, caspase-3 and -7, in a K+ efflux-dependent manner (see Fig.)
Figure 4

Proposed models for NLRP3 inflammasome activation via mitochondrial dysfunction. Canonical stimuli are proposed to trigger NLRP3 inflammasome assembly and activation by damaging the mitochondria and inducing mitochondrial ROS (mtROS) production, releasing oxidized mitochondrial DNA (mtDNA) and promoting the mitochondrial localization of NLRP3 and ASC for activation. (1) Mitochondrial autophagy, mitophagy, limits NLRP3 activation by eliminating damaged or stressed mitochondria from the cell. (2-6) NLRP3 activation can be mediated by VDAC1/2-mediated mtROS production and (3) BAX/BAK activation, which can both be repressed by pro-survival protein BCL-2. (4, 6) ER stress induces C/EPB homologous protein (CHOP)-mediated transcriptional control of IP3R-dependent Ca2+ efflux from the ER into the cytosol. This induces transport of Ca2+ ions into the mitochondria that generates mtROS. (5) mtROS induces deubiquitylation of NLRP3 to promote inflammasome activation, and (6) may facilitate mtDNA release to activate NLRP3 by inducing mitochondrial pore transition (MPT) pore opening. (7) TLR4-IRF1 mediated transcriptional up-regulation of CMPK2 can generate short strands of mtDNA that exit the mitochondria in an oxidized form to bind and activate the NLRP3 inflammasome. (8) Depletion of mitochondrial NAD+ upon Sirtuin 2 inhibition may lead to hyper-acetylation of α-tubulin and induce Dynein-mediated transport of ASC on the mitochondria to NLRP3 on the ER, and facilitate NLRP3 activation. (9, 10) Cardiolipin and the mitochondrial antiviral signaling protein (MAVS) can reportedly tether NLRP3 to the mitochondria for activation in a mtROS-dependent manner. (11) Specific activation of BAX/BAK triggered by loss of BCL-XL and MCL-1 activity can trigger NLRP3 activation, via activation of downstream effector caspases, caspase-3 and -7, in a K+ efflux-dependent manner (see Fig.)

3 NLRP3 IN HEALTH AND DISEASE

3.1 Cryopyrin associated periodic syndromes and inflammatory disease

Historically, NLRP3 was first linked to the development of Familial cold autoinflammatory syndrome (FCAS) and Muckle-Wells syndrome (MWS), which are both characterized by dysregulated systemic inflammation leading to intermittent episodes of rash, arthralgia, fever, and conjunctivitis.135 Subsequently, a spectrum of typically dominantly inherited autoinflammatory disease syndromes have been described in patients harboring “gain-of-function” mutations in NLRP3 that precipitate in IL-1β activation. These include the mild to moderate features observed in FCAS and MWS, through to the severe and often fatal disease manifestations exhibited in chronic infantile neurologic, cutaneous, articular (CINCA) syndrome, also known as neonatal-onset multisystem inflammatory disease (NOMID).136,137 In 2004, the laboratory of Jürg Tschopp established that these NLRP3 mutations induced spontaneous inflammasome assembly and caused pathogenic IL-1β secretion.6 These syndromes are now collectively known as cryopyrin-associated periodic syndromes (CAPS). Most CAPS patients (∼60-75%) are heterozygous germline carriers of missense mutations in the NLRP3 gene located on chromosome 1 (1q44), with 209 different nucleotide variants listed in the infevers database and ∼90 mutations described to date.137 However, in CINCA/NOMID somatic mutations during fetal development account for ∼40% of the affected paediatric patients. It is also now emerging that somatic NLRP3 mosaicism may account for CAPS symptoms in previously documented “mutation negative” patients.138 Importantly, most of the mutations in NLRP3 disrupt exon 3, which encodes the NACHT domain that is required to control NLRP3 oligomerization. Generation of knock-in mice harboring CAPS patient point mutations has confirmed the requirement for elevated myeloid cell-specific NLRP3 inflammasome-mediated IL-1β activation to drive disease pathologies.139 Furthermore, the rapid remission of CAPS symptoms in 97% of patients following anti-IL-1β monoclonal antibody (i.e., Canakinumab) therapy has confirmed the pathologic role for IL-1β in these syndromes.140

In addition to these autoactivating NLRP3 mutations associated with CAPS, single nucleotide polymorphisms in NLRP3 have been associated with increased caspase-1 activity in common diseases, such as Crohn's disease, rheumatoid arthritis, ankylosing spondylitis, and celiac disease.141 The importance of NLRP3 inflammasome-mediated activation of IL-1β and IL-18 in the pathogenesis of common diseases has largely been validated in preclinical models, including models of Alzheimer's disease,142 acute graft-versus-host-disease,143 multiple sclerosis,144 liver inflammation and fibrosis,145 gout,146 obesity and type 2 diabetes,147 and cardiovascular disease (e.g., atherosclerosis).148 Moreover, the results from the phase I clinical trial of Canakinumab (Canakinumab Anti-inflammatory Thrombosis Outcomes Study [CANTOS] clinical trial) in cardiovascular indications, suggest that inhibiting NLRP3 inflammasome or IL-1β activity holds promise for cardiovascular and metabolic diseases.149,150 This potential has led to a growing interest in developing compounds that target inflammasome machinery. In particular, agents that target NLRP3 itself, such as CP-456,733 (also known as MCC950), have been developed and tested successfully in a range of preclinical disease models (2018). Impeding the progression of NLRP3 antagonists to the clinic is the lack of knowledge surrounding mechanisms of activation, protein structure, and drug targeting.

3.2 Mitochondrial dysfunction-induced NLRP3 activation in metabolic disorders

NLRP3 inflammasome activation is commonly linked with metabolic disorders that exhibit well-defined host-derived triggers. These include uric acid crystals in gout,146 islet amyloid polypeptide (IAPP) in type 2 diabetes,152 crystalline cholesterol generated from oxidized LDL uptake in atherosclerosis,148 and fibrillar peptide amyloid-beta in Alzheimer's disease.153 These crystalline and amyloid NLRP3 agonists activate the canonical NLRP3 pathway largely via “frustrated” phagocytosis and lysosomal rupture (see Section 4.3.2). In this section, we discuss the emerging concept that oxidative stress and mitochondrial dysfunction are key mechanisms triggering NLRP3 activation in aging and obesity-induced metabolic syndrome.

3.2.1 Mitochondrial perturbation in metabolic disease and aging

One common denominator in the development of metabolic disease and degenerative aging is mitochondrial dysfunction. This is not surprising, because mitochondria are the central organelle responsible for many key metabolic processes, of which, the most important is the stepwise conversion of glucose or fats into the cellular energy source ATP. In the case of glucose, pyruvate generated via glycolysis is next converted into acetyl-coenzyme A (acetyl-coA). Acetyl-coA is further metabolized within the mitochondria matrix via the Krebs cycle (also known as the citric acid cycle or tricarboxylic acid [TCA] cycle) to yield NADH and FADH2, which then undergo oxidative phosphorylation via the electron transport chain to generate ATP.154 Mitochondrial fatty acid β-oxidation is another critical pathway to yield energy upon uptake of free fatty acids liberated via lipolysis. Fatty acid oxidation of long chain acyl-CoA esters, which form the backbone for phospholipid, cholesterol, and triacylglycerol synthesis, generates acetyl-coA that is utilized by the Krebs cycle and oxidative phosphorylation to generate ATP.154 Interestingly, a side product of oxidative phosphorylation is the production of mtROS that can cause cellular and mitochondrial DNA (mtDNA) damage.154 Both mtROS and oxidized mtDNA have been proposed to directly activate the NLRP3 inflammasome (see Section 4.2).

The role of mitochondrial dysfunction in aging is well established; by the fact that aging mitochondria lose their integrity, exhibit morphologic abnormalities, and decreased oxidative phosphorylation, as well as a general decrease in mitochondrial mass and mtDNA copy-numbers (2013). Similarly, mitochondria isolated from the skeletal muscle and adipose tissue of obese and/or insulin-resistant patients display decreased oxidative phosphorylation capacity, lipid metabolism, and reduced mitochondrial size that is associated with impaired glycolysis rates.156,157 It is now becoming abundantly clear that these mitochondrial defects associated with metabolic stress may contribute to NLRP3 activation in metabolic disease. For example, reduced activity of key glycolytic enzyme, hexokinase, is not only associated with defective mitochondrial bioenergetics in metabolic disease,158 but can also activate the NLRP3 inflammasome (see Section 4.1).159 Moreover, and as discussed below, defective glucose metabolism is also linked to oxidative stress and thioredoxin-interacting protein (TXNIP)-mediated NLRP3 activation,160,161 albeit controversially (see Section 4.2.2). Accumulation of free fatty acids, particularly saturated fatty acid palmitate, in nonadipose tissues (e.g., muscle, pancreatic β-cells and cardiomyocytes) induces mitochondria-dependent apoptotic cell death and lipotoxicity, which is now associated with the activation of NLRP3 via mtROS production162,163 (see Section 3.2.2). Finally, reduced levels of mitochondrial mitofusin proteins (mediate mitochondrial fusion) and increased levels of dynamin-related protein 1 (DRP1; mediates mitochondrial fission) are associated with metabolic disease progression and NLRP3 activity.164,165 For instance, increased DRP1 activity and mitochondrial fission is linked with NLRP3 activation in high glucose situations (see Section 4.2.4).165

3.2.2 NLRP3 activation in degenerative aging

Loss of metabolic control upon aging is linked to increased canonical NLRP3 inflammasome activity in myeloid cells and chronic low-grade systemic sterile inflammation or “inflammaging.” For example, in aging mice NLRP3 activity promotes a decline in glycemic control, and induces thymic involution and T cell senescence, bone loss, as well as hippocampal astrogliosis that is associated with cognitive decline and dementia.166,167 How NLRP3 is triggered during the aging process remains uncertain, although as discussed above, there is accumulating evidence that defective mitochondrial biogenesis, and an accumulation of DAMPs with age and sedentary lifestyles (e.g., free fatty acids, cholesterol, ceramide) drives NLRP3 activation. With advanced age, elevated TNF levels have also been linked to increased transcriptional up-regulation of NLRP3 for activation. In point of fact, neutralization of TNF in aging mice dampened NLRP3 expression and caspase-1 cleavage in adipose tissues (and liver), improved glucose metabolism, and attenuated insulin resistance.168 Interestingly, one recent study also suggested that the macrophage somatotropic (growth hormone and insulin-like growth factor-1) axis up-regulates NLRP3 expression and activity in aging visceral adipose tissue to drive chronic inflammation and metabolic dysfunction.169 In addition to driving chronic IL-1β activity in aging, NLRP3 activation also maintains macrophage numbers in visceral adipose tissue and blunts lipolysis by increasing the secretion of growth differentiation factor 3 and monamine oxidase A, which lowers the amount of bioavailable lipolysis-inducing norepinephrine.170 This reduced capacity for lipolysis is associated with the common signs of aging, including increased adiposity, lower exercise capacity, reduced thermoregulation during cold stress, and reduced ability to survive starvation. NLRP3 inflammasome activity may also globally contribute to degenerative aging, and most likely tumorigenesis, as downstream IL-1 signaling promotes cell senescence and associated activation of the proinflammatory senescence-associated secretory phenotype.171

3.2.3 Fatty acids and NLRP3 activation in obesity-induced metabolic disease

Nutrient overload through the consumption of saturated fats, cholesterol, and highly processed carbohydrates (i.e., a Western lifestyle diet) induces chronic low-grade inflammation that causes metabolic syndrome. A weight of evidence gathered from obese patients and preclinical models suggests that elevated NLRP3 and IL-1β activity drives this inflammation and contributes to insulin resistance.172 In fact, NLRP3-deficient mice have been reported to exhibit significant protection from high fat diet-induced metabolic syndrome, characterized by reduced sterile inflammation (i.e., a decrease in the frequency of effector T cell infiltrate in visceral adipose tissue), reduced adipocyte hypertrophy and liver steatosis, and preservation of pancreatic β-cell mass that results in improved insulin sensitivity and glucose tolerance.147 A number of studies have now largely confirmed this pathogenic role for NLRP3 and ASC in obesity-related disorders, such as insulin resistance and Type 2 diabetes, nonalcoholic steatohepatitis (NASH) and cardiovascular disease (e.g., atherosclerosis).172,173,174,175,176

Accumulating evidence suggests that gut dysbiosis contributes to NLRP3 inflammasome activation in metabolic syndrome via the release of microbiome-derived LPS to prime the inflammasome.177 In point of fact, morbidly obese people exhibit chronically elevated levels of LPS,178 and mice lacking TLR4 are protected from high fat diet-induced inflammation and disease manifestations associated with metabolic syndrome (e.g., NASH, atherosclerosis).179,180,181,182 Dietary saturated fatty acids can also signal via PRRs, such as TLR4 and the endocytic receptor CD36, to induce transcription of inflammatory cytokines and NLRP3 itself.183,184 Moreover, saturated fatty acids, such as palmitate and its breakdown product ceramide, have been shown to trigger activation of the NLRP3 inflammasome in macrophages and dendritic cells.147,185,186

Exactly how saturated fatty acids trigger NLRP3 remains unclear. Palmitate was shown to inactivate AMP-activated protein kinase (AMPK) and inhibit autophagic clearance of damaged mitochondria, thereby triggering mtROS-induced activation of NLRP3, presumably via oxidized mtDNA release.186 An alternative model proposed that palmitate-induced lipotoxicity induces ER stress responses and the resultant activation of inositol-required enzyme 1 (IRE1) triggers NLRP3 activation. Unexpectedly, this IRE1 pathway induced by saturated fatty acids did not require the unfolded protein response, nor did it require the mobilization of Ca2+ or downstream target TXNIP (see Sections 3.2.3 and 4.2.2).187,188 Instead, IRE1 activation was associated with mtROS-induced NLRP3 activation.188 Remarkably, one recent report suggested that akin to cholesterol crystals and amyloid-β fibrils generated upon CD36-medated phagocytosis,184 saturated fatty acid uptake may induce crystalline structures to cause lysosomal rupture and activate NLRP3 in macrophages.189

Omega-3 polyunsaturated fatty acids are reported to counterbalance saturated fatty acid-induced metabolic dysfunction by altering the gut microbiota and increasing the production of short chain fatty acids (e.g., butyrate). It has been shown that omega-3 fatty acids can block canonical NLRP3 activation and protect from high fat diet-induced metabolic dysfunction, although this effect may be partly due to effects on inflammasome priming.190,191 The laboratory of Vishwa Deep Dixit also recently revealed that the ketone body β-hydroxybutyrate, which is generated by short chain fatty acid metabolism in the liver during periods of energy deficit, can act as a negative metabolic regulator of NLRP3 inflammasome activation to both canonical triggers and lipotoxic fatty acids by blocking K+ efflux.192,193 Moreover, administration of β-hydroxybutyrate, or a ketogenic-diet, have also been shown to suppress inflammasome activity and inflammation in preclinical models of MSU-induced gouty peritonitis and CAPS.192,193 How β-hydroxybutyrate exerts its activities and the levels required to combat chronic inflammation still needs to be resolved.

3.2.4 Hyperglycemia, oxidative stress and TXNIP in NLRP3 activation and metabolic disease

TXNIP is an inhibitor of thioredoxin, a major thiol-reducing and antioxidant protein. Over the last few years TXNIP has garnered attention as an important regulator of glucose and lipid metabolism. Evidence for a connection between TXNIP and metabolic disease was revealed through a genome wide study that reported high expression of TXNIP in pre-diabetic and diabetic patient muscle biopsies.194 Subsequently, specific TXNIP variants have been associated with hyperglycemia and high triglyceride levels in diabetes patients.195 Correlating with this data, high glucose induces TXNIP expression in numerous cell types associated with metabolic disease (e.g., adipocytes, skeletal myocytes, and β-cells), and conversely, insulin can repress TXNIP expression.160,194,196 Moreover, TXNIP promotes glucose uptake by insulin responsive cells,194 and its overexpression promotes β-cell loss (a hallmark of diabetes) via mitochondrial apoptosis upon exposure to high glucose, while its deficiency prevents cell death,197,198 Fitting with a key role in glucose metabolism, mice lacking TXNIP are hypoglycemic, hypoinsulinemic and also exhibit a central defect in glucose release both to a normal diet and upon high fat diet challenge.199 Intriguingly, palmitate has been shown to suppress glucose-mediated TXNIP expression and attenuate ROS generation in β-cells, but not to alter cell death.198,200 Collectively, these results suggest that fatty acids and insulin act to counterbalance TXNIP-dependent β-cell oxidative stress caused by hyperglycemia, and highlights the fact that palmitate utilizes a TXNIP-independent pathway to induce β-cell apoptosis.201

The correlation between oxidative stress, TXNIP activity and NLRP3 was first made following the discovery that TXNIP binds to NLRP3, using a yeast two-hybrid screen.160 Subsequently, ER stressors (e.g., thapsigargin) and/or high glucose have been suggested to trigger NLRP3 inflammasome activation in islets, adipose tissue/adipocytes, macrophage and monocytes in a TXNIP-dependent manner160,196,202 (see Section 4.2.2 for ambiguities surrounding TXNIP involvement in NLRP3 activation). It is worth noting that there are discrepancies between mouse and human as to whether β-cells have a functional NLRP3 inflammasome.203,204 One recent study reported that mouse β-cells only express low levels of NLRP3 machinery, and consequently observed that high glucose failed to activate caspase-1 or IL-1β in WT and NLRP3 autoactivating mutant β-cells.203 Therefore, it will be important to clarify the exact mechanisms and cell-types by which metabolic stressors trigger NLRP3 activity. This will be vital as the ROS-TXNIP-NLRP3 pathway is implicated in the pathology of a growing number of metabolic disease models, such as type 1 and 2 diabetes,205 arterial damage206 and conversion of NASH to fatty liver disease.207

4 MECHANISMS FOR NLRP3 ACTIVATION

Based on the diverse nature of canonical NLRP3 activators, it is widely assumed that there must be a universal mechanism to trigger NLRP3 activation. Currently several common molecular and cellular events have been proposed to induce the activation and assembly of the NLRP3 inflammasome (Figs. 1 and 4), including K+ efflux, Ca2+ mobilization, lysosomal rupture, ROS production, and mitochondrial dysfunction. Of these, K+ efflux is widely believed to be the unifying event for NLRP3 inflammasome activation,45,95 with a few notable exceptions.53,208 Nevertheless, mitochondrial dysfunction has recently emerged as a major, if not controversial, contender. It therefore remains of great debate as to whether there is a single universal event responsible for NLRP3 inflammasome activation. In this section, we will discuss in detail the strengths and weaknesses surrounding the most widely documented mechanisms—K+ efflux and mitochondrial dysfunction—and give a brief overview on the ambiguities surrounding other potential triggers.

4.1 The universal event: potassium (K+) efflux

K+ efflux is the most common upstream event required for NLRP3 inflammasome activation.45 Extracellular ATP binding the purinergic P2X7R, and pore-forming toxins like potassium ionophore nigericin, were the first stimuli demonstrated to lower intracellular K+ levels and activate NLRP3.209,210 Subsequently, the laboratory of Gabriel Núñez has established that almost all known stimuli trigger NLRP3 in a K+ efflux-dependent manner.45 In fact, exposure of LPS-primed macrophages to high extracellular potassium (range 30–60 mM) inhibits NLRP3 activation to ATP, pore-forming toxins, and lysosomal disruptors (i.e., crystalline/particulates and Leu-Leu-O-methyl ester [LL-OMe]). Fittingly, lowering extracellular K+ levels promotes spontaneous NLRP3 activation.211,212 In line with this unifying model for NLRP3 activation, K+ efflux is also required for noncanonical activation of the NLRP3 inflammasome,95 and for most alternative and cell death-induced inflammasome pathways126 (Figs. 2 and 3).

Despite it being clear that perturbations in membrane integrity lead to K+ efflux and activation of NLRP3, how K+ efflux occurs to most NLRP3 triggers remains unresolved. Most studies have focused on pore-forming toxins and H+/K+ ionophores, such as nigericin, and the ATP-triggered P2X7R cation channel. In the case of extracellular ATP binding P2X7R, a larger nonselective pannexin-1 pore was originally implicated in the activation of caspase-1 activation and IL-1β release.213 However, later studies indicated that the pannexin-1 hemichannel was not essential for increased permeability to pore-forming toxins or cell regulatory volume decreases.214,215 In fact, examination of pannexin-1-deficient macrophages suggested that pannexin-1 is dispensable for both DAMP uptake and NLRP3 activity.216 It should be noted, that whilst pannexin-1 is not obligatory for extracellular ATP-induced NLRP3 activation, caspase-11 has also been proposed to cleave and activate pannexin-1 hemichannels to permit the extracellular release of ATP to trigger P2X7R-mediated events (see Section 2.2.1 and Fig.).95 Intriguingly, a K+ channel inhibitor screen recently performed by the laboratory of Asrar Malik, uncovered that a quinine-sensitive two pore TWIK2 potassium efflux channel, may be required for ATP/P2X7R-mediated activation of NLRP3.217 As these studies focused on ATP/P2X7R-mediated events, it remains to be seen how K+ efflux is triggered by an ever-growing number of canonical triggers.

Another conundrum in the K+ efflux model is the mechanism for NLRP3 activation. The work of Núñez and colleagues has clearly demonstrated that K+ efflux acts upstream of NLRP3 activation. In macrophages harboring the autoactivating NLRP3R258W mutation, K+ efflux was not required for NLRP3 activation and high extracellular potassium failed to inhibit inflammasome activity.45 NEK7 has been shown to bind the LRR domain of NLRP3 in a K+ efflux-dependent manner to trigger NLRP3 inflammasome oligomerization and activation.57,58 The fact inflammasome activation in NLRP3R258W mutant macrophages critically required NEK7 but not K+ efflux, may argue for a role for K+ efflux in inducing a conformational change to promote NEK7-NLRP3 interactions.45,57 Despite the clear connect between K+ efflux and NLRP3 activation, it remains to be seen if reduced intracellular K+ levels directly trigger NLRP3 activation, or if other homeostatic changes are required. The laboratory of Rongbin Zhou recently proposed that nigericin and ATP activate chloride intracellular channels (CLICs) downstream of K+ efflux and mtROS, and thereby promote NEK7 interactions with NLRP3.218 However, in direct contradiction, Núñez and colleagues observed K+ efflux and Na+ influx in response to nigericin but no change in intracellular Cl levels.45 Zhou and colleagues postulated that these discrepancies may be explained by the transient nature of Cl ion flux in response to activators in their hands. As other groups have also implicated CLICs in the priming and activation of NLRP3,219 it may be worth revisiting the role of ion flux homeostasis in NLRP3 inflammasome activation.

At face value, K+ efflux appears to be the unifying event to trigger NLRP3 inflammasome activation; yet recent reports support the possibility that K+ efflux-independent NLRP3 triggers exist. For example, alternative NLRP3 inflammasome activation via a RIPK1-caspase-8 complex in human monocytes occurs in the absence of cell death (and thus plasma membrane damage) and does not require K+ efflux.53 Likewise, cytosolic Gram-positive bacterial peptidoglycan-N-acetylglucosamine was shown to bind and inhibit the glycolytic enzyme hexokinase resulting in its dissociation from the mitochondrial outer membrane and activation of NLRP3 in a K+ efflux-independent manner. How hexokinase release from the mitochondria activates NLRP3 remains unclear, but Wolf et al. eluded to the possibility that disruption of hexokinase interactions with voltage-dependent anion channels (VDACs) may facilitate mtDNA release.159 Importantly, blockade of glycolysis also reportedly triggered NLRP3 activation by causing mitochondria damage and mtROS production.220 Finally, the laboratory of Olaf Groß recently reported that TLR7 agonist imiquimod, and related molecule CL097, also target the mitochondria, specifically the ROS-producing quinone oxidoreductases NQO2 and mitochondrial Complex I, to trigger NLRP3 activation in a manner independent of K+ efflux but dependent on NEK7.208

4.2 An emerging culprit: Mitochondrial dysfunction

Mitochondria play an essential role in the generation of energy in the form of ATP. However, as discussed above, mitochondria also critically regulate intrinsic “mitochondria-dependent” apoptosis. In addition, mitochondria have also recently been implicated in innate antiviral immune responses. For example, upon sensing cytoplasmic viral RNA, the retinoic acid inducible gene-I-like (RIG-I) receptors, RIG-I and MDA5, signal via the outer mitochondrial membrane adaptor protein mitochondria antiviral signaling (MAVS) to activate NF-κB and IFN regulator factor 3 (IRF3) transcription factors and induce type I IFN production.221 Likewise, mitochondrial apoptotic signaling causes the release of mtDNA, which in the absence of intrinsic apoptotic caspase activity, triggers the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) to drive IFN responses.222,223 Finally, there is an emerging dogma that mitochondrial dysfunction is the universal trigger for NLRP3 activation, via events including ROS and mtDNA release, apoptosis, and mitochondrial localization (Fig. 4).

4.2.1 BCL-2 levels

Mitochondria were first implicated in inflammasome activation in 2007 through work performed in the laboratory of John Reed, which showed that the BCL-2 pro-survival family members, BCL-2 and BCL-XL could bind and suppress NLRP1 inflammasome activation.224 Consistent with this finding, a vaccinia virus homologue of BCL-2, F1L, was later found to also bind and inhibit NLRP1 and promote virulence.225 Possible caveats to the work by Reed and colleagues, is the growing evidence that the bacterial peptidoglycan muramyl dipeptide (MDP) used in this study may not be a ligand of NLRP1.226 This is best exemplified by the fact that macrophages derived form NLRP1b-deficient mice (129 genetic background) respond normally to MDP-conjugated to adjuvant titanium oxide (TiO2) and L18-MDP (that has improved cellular uptake).227 It should be noted that TiO2 is a bona fide NLRP3 trigger.227,228 Key experiments were also performed in the presence of the now recognized NLRP3 trigger, ATP.210 As such, reports of enhanced or diminished inflammasome-associated IL-1β activity in macrophages deficient or overexpressing BCL-2, respectively, is more suggestive of a role for BCL-2 in limiting activation of the NLRP3 inflammasome.161,229 However, more recently, mice overexpressing BCL-2 in the haematopoietic system were shown to exhibit comparable IL-1β responses upon MSU crystal injection, compared to littermate control mice.230 Likewise, BMDMs derived from two independent BCL-2 transgenic mouse lines exhibited normal NLRP3 inflammasome responses to a range of canonical triggers.123

4.2.2 Autophagy, mitophagy, and ROS

Autophagy is a conserved cellular process for the delivery and targeted lysosomal degradation of cytosolic constituents, such as proteins and damaged organelles. In 2008, the laboratory of Shizuo Akira first made the connection between autophagy and NLRP3 inflammasome regulation by demonstrating that loss of autophagy-related protein ATG16L1 in mice rendered them sensitive to spontaneous IL-1β activation upon TLR ligation, and exaggerated canonical NLRP3 responses.231 Subsequently, chemical inhibition of autophagy, and deficiency in a range of autophagy related proteins, including microtubule light chain 3 beta (LC3B), Beclin1, ATG5, and ATG7, have been shown to trigger NLRP3 activation.231,232,233 Moreover, mice deficient in LC3B were highly susceptible to the cecal-ligation and puncture-induced sepsis model and produced excessive levels of NLRP3 inflammasome-associated IL-1β.232

Autophagy has been proposed to limit inflammasome activation and IL-1 family cytokine production and secretion in several ways. One obvious way autophagy could limit NLRP3 activation is by delivering inflammasome machinery into the lysosomes for degradation. Arguing against this theory, is the fact that early studies showed no significant differences in NLRP3, ASC, caspase-1 and pro-IL-1β levels in macrophages lacking autophagy machinery, despite increased NLRP3 activity.231,232 In work performed using chemical inhibitors and activators of autophagy, a role for autophagy in the degradation of pro-IL-1β was counter-proposed.234 Subsequently, TRIM20/MEFV has emerged as an autophagic receptor to promote the degradation of inflammasome machinery, including NLRP3, caspase-1 and NLRP1. In fact, mutations in MEFV, which are associated with Familial Mediterranean Fever, diminish recruitment of autophagy machinery to NLRP3 and promote inflammasome activity.235 Collectively, this work suggests that autophagy acts to remove inflammasome machinery and limit inflammation. Remarkably, it is now emerging that inflammasomes themselves may coordinate their own autophagic destruction via caspase-1 activation.236,237,238

ROS have long been associated with NLRP3 inflammasome activation. Initial studies using RNA interference against NADPH oxidase and ROS scavengers suggested that cellular ROS was the trigger for NLRP3.211,239 However, later studies using peripheral blood mononuclear cells from chronic granulomatous disease patients that exhibit defective NOX1-4 activity, and mice genetically deficient in NADPH oxidase subunits, suggested that NADPH oxidase was not required for NLRP3 activation to canonical activators or upon autophagy inhibition.234,240,241 In 2011, two independent laboratories proposed that stressed mitochondria were the source of ROS essential for NLRP3 activation, and that autophagic clearance of damaged mitochondria (mitophagy) was required to prevent mtROS production and excessive NLRP3 activation161,232 (Fig. 4). Zhou et al. postulated that canonical NLRP3 inflammasome activation was dependent on VDAC1/2 facilitating Ca2+ uptake into the mitochondria to generate mtROS. Increased mtROS subsequently disrupted TXNIP/thioredoxin interactions and allowed TXNIP to bind and recruit NLRP3 and ASC to the mitochondria-associated ER membranes (ER-MAMs) for activation (Fig. 4).161 In contrast, Nakahira et al. suggested that LPS and ATP-induced mtROS triggered mitochondrial permeability transition (MPT) pore opening to cause mtDNA release and inflammasome activation (Fig. 4, 6).232 Notably, in this scenario, NLRP3 was required for mtDNA release, suggesting that mtDNA release acted to amplify inflammasome activity. Subsequently, other groups reported similar models that centralized around mitochondrial damage inducing mtROS and mtDNA release (see Section 4.2.3 and Fig.).229,242 Based on these models, there has been great interest in connecting mitochondria, oxidative stress, and NLRP3 inflammasome activation with disease, as discussed above (see Section 3.2).

Stemming from the work of Zhou and colleagues,161 it is now widely assumed that mtROS is the common event triggering activation of NLRP3. Nevertheless, several groups have now failed to reproduce these findings and thereby weakened the mitochondrial dysfunction model. For instance, the spontaneous NLRP3 activation observed upon inhibition of the mitochondrial respiratory chain complex with rotenone is not reproducible.45,63,243 Similarly, it was inferred that ROS-dependent recruitment of TXNIP to the mitochondria was required for NLRP3 binding and activation; however, a study by the laboratory of Luke O'Neill observed no defect in NLRP3 activation in TXNIP-deficient macrophages exposed to IAPP, MSU, silica, and ATP.152 Intriguingly, a recent study may point to an indirect role for TXNIP in NLRP3 activation, as TXNIP represses LPS-induced generation of nitric oxide that is a putative negative regulator of NLRP3 activation.74,244 Finally, the data supporting the concept that BCL-2 suppresses VDAC1/2 to limit mtROS-induced NLRP3 inflammasome activation has been directly contradicted by another recent study that showed normal canonical NLRP3 activation in BCL-2 transgenic macrophages.123

The laboratory of Tiffany Horng suggested that NLRP3 activators universally trigger Ca2+ mobilization from the ER stores to damage mitochondria, induce ROS, and causes a loss of mitochondrial membrane potential to release mtDNA (Fig. 4, 6). Complicating this model, is the fact that Ca2+ influx has been demonstrated downstream of ROS production induced by crystal and liposome NLRP3 activators.245 It also appears that mtROS is not obligatory for NLRP3 activation to all stimuli, including some pore-forming toxins, influenza, encephalomyocarditis and measles viruses, and even the widely used trigger ATP.45,246,247,248 Moreover, the ROS scavengers and inducers of mtROS utilized to model NLRP3 activation may have off-target effects. For example, Veit Hornung and colleagues documented that ROS scavengers, diphyliodonium (DPI) and N-acetylcysteine (NAC), can blunt LPS-induced NLRP3 inflammasome priming rather than block activation.240 Nevertheless, others have shown that addition of ROS scavengers after TLR priming can still inhibit NLRP3 activity.29,245,249 Intriguingly, mtROS has now been suggested to contribute to nontranscriptional priming of NLRP3 by promoting deubiquitylation of NLRP3 upon LPS stimulation63 (Fig. 4).

The concept that clearance of damaged ROS-producing mitochondria occurs to limit NLRP3 inflammasome activation is largely based on data obtained upon the genetic loss of autophagy machinery or chemical inhibition of autophagy161,231,232 (Fig. 4). Chemicals that induce mitochondrial pore transition (MPT), such as the mitochondrial un-coupler carbonyl cyanide m-chlorophenylhydrazone (CCCP), can activate NLRP3 but also trigger the accumulation of mitochondrial serine/threonine kinase Pink1 to recruit the E3 ubiquitin ligase Parkin (encoded by Park2) and activate mitophagy.161,250 Challenging the belief that mitophagy restricts inflammasome activation, a study by Allam et al. discerned no defect in NLRP3 inflammasome activation in Park2-deficient macrophages to canonical activators ATP, MSU, and alum, as well as apoptotic stimuli, despite the accumulation of mitochondria.123 In contrast, in a more recent study, Parkin deficiency was associated with exaggerated responses to NLRP3 activators.251 The reason for these discrepancies remains unclear, although, Parkin has recently been ascribed nonmitophagic functions that might affect NLRP3 activation, including the K63 ubiquitylation of RIPK1 to promote activation of NF-κB.252

4.2.3 Apoptosis and mtDNA release

Apoptotic cell death is classically viewed as an immunologically silent process; however, as discussed above, targeted activation of apoptotic caspases, caspase-8 and caspase-3/7, can contribute to activation of the NLRP3 inflammasome and IL-1β in innate immune cells115,116,123,131,132 (see Fig. 3B;  Fig. #11). In 2012, the laboratory of Moshe Arditi put forward mitochondrial apoptosis as the unifying model for how diverse stimuli trigger NLRP3 (Fig. 4). It was proposed that NLRP3 activators, such as ATP and nigericin, trigger mitochondrial apoptosis to induce the release of oxidized mtDNA that binds and activates NLRP3. Fitting with this idea, apoptosis-inducing drugs, such as the protein translation inhibitor cycloheximide, can trigger NLRP3 activation.229 Discrediting this model, macrophages deficient in essential intrinsic apoptotic effectors BAX and BAK, or alternative effector BOK, or initiator caspase, caspase-9, responded normally to both canonical NLRP3 activators (e.g., alum, Nigericin, and ATP), as well as apoptotic stimuli (e.g., cycloheximide, staurosporine, and ultraviolet irradiation).123,131 Indeed, a growing consensus is that apoptosis-inducing cancer chemotherapeutics promote TLR-induced caspase-8-mediated activation of IL-1β and/or NLRP3 activation.29,50,113

Augustine Choi and colleagues proposed that MPT enables mtDNA release to activate NLRP3232 (Fig. 4). Inconsistent with the notion, NLRP3 activity was an upstream prerequisite for mtDNA release. Evidence for the role of the MPT in NLRP3 activation was also gathered using cyclosporine A, a potent inhibitor of MPT component cyclophilin D.232 A more recent study, however, showed genetically that cyclophilin D is not essential for canonical NLRP3 activation. Furthermore, cyclosporine A still inhibited NLRP3 activation in the absence of cyclophilin D, suggesting that cyclosporine A targets another unknown protein to block NLRP3 activation.123

Recently, the laboratory of Michael Karin reproduced the core finding that mtDNA can activate NLRP3 using transcription factor A, mitochondria (TFAM) knockout macrophages that exhibit reduced mtDNA content.253 Extending these findings, Karin and colleagues suggested that TLR engagement was required to induce the synthesis of new mtDNA, via IRF1-mediated up-regulation of the mitochondrial deoxyribonuleotide kinase, UMP-CMPK2; Fig. #7). Correspondingly, reduced LPS-induced mtDNA synthesis in IRF1- or UMP-CPMK2-deficient BMDMs precipitated in defective oxidized mtDNA release and IL-1β responses to canonical NLRP3 triggers.253 Confusing these matters, however, is a previous report that suggested moderate mtDNA stress caused by TFAM loss in murine embryonic fibroblasts and macrophages leads to reduced total mitochondrial DNA levels and increased cytosolic mtDNA accumulation to trigger cGAS-STING signaling.254 Furthermore, in direct contradiction to this TLR-IRF1-UMP-CMK2 pathway to mtDNA up-regulation, Si Ming Man et al. previously demonstrated that IRF1 is not required for either canonical or noncanonical NLRP3 activation but is obligatory for AIM2 activation.255 A final caveat to the mtDNA model, is that a related study found no defect in ATP-induced NLRP3 responses upon mtDNA depletion with ethidium bromide.256

4.2.4 Mitochondrial localization and homeostasis

Since the original report that mtROS stimulated the recruitment of NLRP3 from the ER to the ER-MAMs to trigger activation,160 several mitochondrial proteins have been linked to this process. For example, mitochondrial damage inflicted by NLRP3 activators is suggested to deplete co-enzyme NAD+ stores. Reduced NAD+ levels, in turn, inactivates the α-tubulin deacetylase sirtuin 2 and promotes acetylated-α-microtubule, dynein-dependent transport of perinuclear mitochondria to the ER-MAMs to bring ASC into the proximity of NLRP3 and drive inflammasome assembly (Fig. 4).257 However, incompatible with this model is the fact that (i) prior to NLRP3 activation ASC is predominantly found in the nucleus and rapidly redistributes to the cytosol upon activation,258 (ii) other reports suggest that NLRP3 is localized to the mitochondria,161 and (iii) the lack of requirement for either mtROS and K+ efflux.257

MAVS was also proposed by the laboratory of Ronald Germain to tether NLRP3 to the mitochondria (Fig. 4) and facilitate activation to soluble (e.g., ATP and nigericin), but not particulate/crystalline matter (e.g., alum, cholesterol crystals, or MSU crystals).259 Challenging this study, several groups have now identified that while MAVS, and interacting protein mitofusin 2, are required for NLRP3 activation to RNA viruses, they do not impact canonical NLRP3 activation.123,247,260,261 Inner mitochondrial membrane phospholipid, cardiolipin, also allegedly binds NLRP3 to localize it to the mitochondria for activation (Fig. 4), as knockdown of cardiolipin synthase attenuated NLRP3 activation.262 Nevertheless, it was recently suggested that mtROS generation at the inflammasome priming stage facilitates both NLRP3 and caspase-1 association with cardiolipin on the outer mitochondria membrane, and thus cardiolipin may act as scaffolding for NLRP3 inflammasome assembly, rather than its activation.263 Notably, if the mitochondria acts as the inflammatory platform and trigger, it remains to be reconciled why in many studies the active inflammasome appears to be cytosolic, as evidenced by a lack of co-localization of ASC specks with organelle markers.264

It is also noteworthy that the mitochondrial GTPase DRP1 has been suggested to be required for NLRP3 activation downstream of RIPK1-RIPK3 upon exposure to dsRNA or RNA viruses, although a latter study failed to reproduce these findings.265,266 It has also recently been proposed that the mitochondrial phosphatase PGAM5, which also maintains mitochondrial homeostasis and triggers mitophagy, is required for mtROS-induced NLRP3 activation in macrophages to not only vesicular stomatosis virus infection but also soluble and particulate triggers.128 It therefore remains plausible that mitochondrial dysfunction may trigger NLRP3 activity under certain circumstances.

4.3 THE MINOR PLAYERS

4.3.1 Calcium mobilization

Intracellular Ca2+ was first implicated in ATP- and Nigericin-induced NLRP3 activation in a study utilizing the Ca2+ chelator BAPTA-AM and ER-Ca2+ ATPase thapsigargin.267 Several studies have now reported that NLRP3 can be triggered by Ca2+ mobilization from the ER stores upon activation of G protein coupled calcium sensing receptor (CaSR) signaling, and Ca2+ influx induced via store operated-Ca2+ entry (SOCE) channels or cationic transient receptor potential (TRP) channels.215,242,245,268,269 Ca2+ mobilization from the ER stores was initially postulated to be a common event to trigger NLRP3 via CaSR, phospholipase C (PLC) and 1,4,5-triphosphate signaling. Indeed, inhibition or knockdown of CaSR, PLC, or the ER Ca2+ release channel, 1,4,5-triphosphate receptor (IP3R), inhibited intracellular Ca2+ levels and ATP-mediated NLRP3 activation. Conversely, high extracellular Ca2+ and direct activation of PLC triggered spontaneous IL-1β activation in the absence of NLRP3 activators.268 A subsequent report suggested that the release of Ca2+ by necrotic cells engaged both the CaSR and related G protein coupled calcium sensing receptor (GPRC6A) to mobilize ER Ca2+ stores and trigger NLRP3.269 A further model proposed that cell swelling followed by regulatory volume decrease triggered TRP vanilloid 2 (TRPV2)-mediated Ca2+ influx and TGF-β-activated kinase 1 (TAK1) activation to activate NLRP3.215 However, this TAK1-dependent pathway has largely been discounted, as genetic deletion of TAK1 has no effect on canonical NLRP3 activation and in fact, promoted spontaneous TLR-induced NLRP3 activation.29,270 In an alternative model, the Transient receptor potential melastatin 2 (TRPM2) channel was implicated in ROS-dependent Ca2+ influx to liposome/crystalline triggers.245 Discordant with the general idea that Ca2+ influx is required for NLRP3 activation, more recent studies have reported that: (i) cytosolic Ca2+ levels were neither essential nor sufficient for NLRP3 activation, compared to the critical effect of K+ efflux, (ii) direct engagement of calcium mobilizing G protein receptors failed to activate NLRP3, (iii) calcium chelator BAPTA-AM and channel blocker 2-APB blocked NLRP3 activation in a manner independent of perturbed Ca2+ levels,271 and (iv) high extracellular Ca2+ (in the presence of high phosphate ion levels) can crystalize and potentially act as a particulate to trigger lysosomal rupture45 (see Section 4.3.2). Therefore, the role of Ca2+ homeostasis in NLRP3 activation remains controversial despite significant genetic evidence for GPRC6A and TRPM2.

4.3.2 Lysosomal rupture

In contrast to soluble NLRP3 activators, particulate and crystalline stimuli, such as silica, alum, β-amyloid, and MSU, as well as bacteria, are phagocytosed and cause lysosomal rupture to promote cathepsin-dependent lysosomal cell death and NLRP3 activation.153,272,273 Recently, cytosolic DNA sensing by cGAS-STING was also shown to induce STING translocation to the lysosome, membrane rupture, and lysosomal cell death-triggered activation of the NLRP3 inflammasome.274 Despite the early belief that this pathway was highly distinct, it has now been established that all particulate/crystalline and lysosomotropic agents induce K+ efflux to activate NLRP3.45

Evidence for this mechanism has largely been gathered using chemical inhibitors that impact phagocytic and lysosomal activities. For example, the potent vacuolar-type H+-ATPase inhibitor bafilomycin A1, which prevents lysosomal acidification, blocked NLRP3 activation to particulate triggers, such as silica and alum.272 Conversely, “low-level” lysosome membrane permeabilization with the soluble lysosomotropic agent, LL-OMe promotes K+ efflux-dependent NLRP3 inflammasome assembly and activation in BMDMs and BMDCs.271 Curiously, doses of LL-OMe that cause extensive lysosomal membrane damage inhibited NLRP3 activation in dendritic cells, via an uncharacterized mechanism that may involve impaired deubiquitylation of NLRP3.271 Several early studies also showed that chemical inhibition of the lysosomal cysteine protease, cathepsin B, using CA-074-Me blocked NLRP3 activation; however, cathepsin B-deficient macrophages did not display major defects in NLRP3 activation to a range of particulates.153,273 This discrepancy now appears to most likely reflect functional redundancy, as CA-074-Me has been shown to target multiple cathepsins.275

5 CONCLUSIONS

There have been considerable attempts made in the last few years to gain a better understanding of the mechanisms behind NLRP3 activation and the downstream events that culminate in IL-1β secretion and cell death. Since the groundbreaking discovery of the noncanonical inflammasome, alternative and programmed cell death signaling pathways have been shown to induce NLRP3 inflammasome assembly and activation. It will therefore be of great interest to mechanistically tease apart these novel pathways, and ascertain their role, particularly specific BAX/BAK activation, in metabolic and common inflammatory diseases. It is worth mentioning that genetic loss or mutation of death receptor and TLR signaling machinery, including XIAP, A20, LUBAC components HOIL and HOIP and most recently RIPK1 and caspase-8, have all been linked to pathogen molecule-associated hyperinflammation linked to NLRP3 and IL-1β activation.110,276,277 A consensus on a universal event required to trigger NLRP3 activation to diverse activators also remains elusive. Based on our current knowledge, it seems clear that K+ efflux remains the most commonly described event precipitating in NLRP3 activation. Mitochondrial dysfunction has emerged as a highly attractive alternative mechanism for canonical NLRP3 activation; however, the large number of conflicting reports and caveats to the proposed models weakens its potential. Despite this controversy, there is considerable evidence in vivo during metabolic diseases that mitochondrial damage and oxidative stress may be involved in NLRP3 inflammasome activation and disease pathogenesis.

AUTHORSHIP

M.Y., D.J.C., D.S.S., and K.E.L. contributed to the writing of this review and generation of figures.

ACKNOWLEDGMENTS

This work was supported by a National Health and Medical Research Council Project grant 1145788 to K.E.L. and operational infrastructure grants through the Australian Government IRISS and the Victorian State Government OIS.

DISCLOSURES

The authors declare no conflicts of interest.

Abbreviations

     
  • AIM2

    absent in Melanoma

  •  
  • AMPK

    AMP-activated protein kinase

  •  
  • APAF-1

    apoptosis protease activating factor-1

  •  
  • ASC

    apoptosis-associated speck-like protein containing a caspase recruitment domain

  •  
  • BMDCs

    bone marrow dendritic cells

  •  
  • BMDMs

    bone marrow-derived macrophages

  •  
  • Ca2+

    calcium

  •  
  • CAPS

    Cryopyrin-Associated Periodic Syndromes

  •  
  • CARD

    caspase activation and recruitment domain

  •  
  • CaSR

    calcium sensing receptor

  •  
  • CCCP

    carbonyl cyanide m-chlorophenylhydrazone

  •  
  • c-FLIP

    cellular FADD-like IL-1β-converting enzyme-inhibitor protein

  •  
  • cGAS

    cyclic GMP-AMP Synthase

  •  
  • CINCA

    chronic infantile neurological, cutaneous, articular

  •  
  • CLICs

    chloride intracellular channels

  •  
  • DAMPS

    damage-associated molecular patterns

  •  
  • DPI

    diphyliodonium

  •  
  • DRP1

    dynamin-related protein 1

  •  
  • DUB

    deubiquitinase

  •  
  • ER-MAMs

    endoplasmic reticulum-mitochondria-associated membranes

  •  
  • FADD

    Fas-associated protein with death domain

  •  
  • FBXL2

    F-box L2

  •  
  • FCAS

    familial cold autoinflammatory syndrome

  •  
  • GBPs

    guanylate binding proteins

  •  
  • GPRC6A

    G protein coupled calcium sensing receptor 6A

  •  
  • GSDM

    gasdermin

  •  
  • HMGB1

    high mobility group box-1

  •  
  • IAP

    Inhibitor of Apoptosis

  •  
  • IAPP

    islet amyloid polypeptide

  •  
  • IP3R

    1,4,5-triphosphate receptor

  •  
  • IRAK

    IL-1 receptor associated kinase

  •  
  • IRE1

    inositol-required enzyme 1

  •  
  • LC3B

    light chain 3 beta

  •  
  • LL-OMe

    Leu-Leu methyl ester

  •  
  • LRR

    Leucine-rich repeat

  •  
  • LUBAC

    linear ubiquitination chain assembly complex

  •  
  • MARCH7

    membrane-associated RING finger protein 7

  •  
  • MDP

    muramyl dipeptide

  •  
  • MLKL

    Mixed lineage kinase domain-like

  •  
  • MPT

    mitochondrial permeability transition pore

  •  
  • MSU

    monosodium urate

  •  
  • mtDNA

    mitochondrial DNA

  •  
  • mtROS

    mitochondrial reactive oxygen species

  •  
  • MWS

    Muckle-Wells syndrome

  •  
  • NAC

    N-acetylcysteine

  •  
  • NAIP

    NLR family of apoptosis inhibitory protein

  •  
  • NASH

    nonalcoholic steatohepatitis

  •  
  • NEK7

    NIMA-related kinase 7

  •  
  • NLRP3

    NOD-like receptor protein 3

  •  
  • NLRP1

    Nucleotide-binding oligomerization domain (NOD)-like receptor 1

  •  
  • NOMID

    neonatal-onset multisystem inflammatory disease

  •  
  • OxPAPC

    oxidized phospholipids

  •  
  • P2X7R

    P2X7 receptor

  •  
  • PAMPS

    pathogen-associated molecular patterns

  •  
  • PGAM5

    PGAM family member 5

  •  
  • PLC

    phospholipase C

  •  
  • PRR

    pattern recognition receptors

  •  
  • PYD

    pyrin domain

  •  
  • RIPK

    receptor interacting protein kinase

  •  
  • ROS

    reactive oxygen species

  •  
  • SMAC

    second mitochondria-derived activator of caspases

  •  
  • SOCE

    store operated-Ca2+ entry

  •  
  • STING

    Stimulator of Interferon Genes

  •  
  • T3SS

    type III secretion system

  •  
  • TAK1

    TGF-β-activated kinase 1

  •  
  • TCA

    tricarboxylic acid

  •  
  • TFAM

    transcription factor A

  •  
  • TiO2

    titanium oxide

  •  
  • TNFR

    tumor necrosis factor receptor

  •  
  • TRIF

    Toll/IL-1R domain-containing adaptor-inducing IFN-β

  •  
  • TRIM

    tripartite motif

  •  
  • TRP

    transient receptor potential

  •  
  • TRPM2

    transient receptor potential melastatin 2

  •  
  • TRPV2

    TRP vanilloid 2

  •  
  • TXNIP

    thioredoxin-interacting protein

  •  
  • UMP-CMPK2

    uridine/cytidine monophosphate kinase-2

  •  
  • VDAC

    voltage-dependent anion channels

REFERENCES

1.

Broz
 
P
,
Dixit
 
VM
.
Inflammasomes: mechanism of assembly, regulation and signalling
.
Nat Rev Immunol
.
2016
;
16
:
407
420
.

2.

Shi
 
J
,
Zhao
 
Y
,
Wang
 
K
, et al.  
Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death
.
Nature
.
2015
;
526
:
660
665
.

3.

Kayagaki
 
N
,
Stowe
 
IB
,
Lee
 
BL
, et al.  
Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling
.
Nature
.
2015
;
526
:
666
671
.

4.

Martinon
 
F
,
Burns
 
K
,
Tschopp
 
J
.
The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta
.
Molecular Cell
.
2002
;
10
:
417
426
.

5.

Man
 
SM
,
Kanneganti
 
T-D
.
Regulation of inflammasome activation
.
Immunol Rev
.
2015
;
265
:
6
21
.

6.

Agostini
 
L
,
Martinon
 
F
,
Burns
 
K
,
McDermott
 
MF
,
Hawkins
 
PN
,
Tschopp
 
J
.
NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder
.
Immunity
.
2004
;
20
:
319
325
.

7.

Mariathasan
 
S
,
Newton
 
K
,
Monack
 
DM
, et al.  
Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf
.
Nature
.
2004
;
430
:
213
218
.

8.

Elinav
 
E
,
Strowig
 
T
,
Kau
 
AL
, et al.  
NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis
.
Cell
.
2011
;
145
:
745
757
.

9.

Vladimer
 
GI
,
Weng
 
D
,
Paquette
 
SW
, et al.  
The NLRP12 inflammasome recognizes Yersinia pestis
.
Immunity
.
2012
;
37
:
96
107
.

10.

Khare
 
S
,
Dorfleutner
 
A
,
Bryan
 
NB
, et al.  
An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages
.
Immunity
.
2012
;
36
:
464
476
.

11.

Zhu
 
S
,
Ding
 
S
,
Wang
 
P
, et al.  
Nlrp9b inflammasome restricts rotavirus infection in intestinal epithelial cells
.
Nature
.
2017
;
546
:
667
670
.

12.

Hornung
 
V
,
Ablasser
 
A
,
Charrel-Dennis
 
M
, et al.  
AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC
.
Nature
.
2009
;
458
:
514
518
.

13.

Kerur
 
N
,
Veettil
 
MV
,
Sharma-Walia
 
N
, et al.  
IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi sarcoma-associated herpesvirus infection
.
Cell Host Microbe
.
2011
;
9
:
363
375
.

14.

Xu
 
H
,
Yang
 
J
,
Gao
 
W
, et al.  
Innate immune sensing of bacterial modifications of Rho GTPases by the Pyrin inflammasome
.
Nature
.
2014
;
513
:
237
241
.

15.

Levinsohn
 
JL
,
Newman
 
ZL
,
Hellmich
 
KA
, et al.  
Anthrax lethal factor cleavage of Nlrp1 is required for activation of the inflammasome
.
PLOS Pathog
.
2012
;
8
:
e1002638
.

16.

Hellmich
 
KA
,
Levinsohn
 
JL
,
Fattah
 
R
, et al.  
Anthrax lethal factor cleaves mouse nlrp1b in both toxin-sensitive and toxin-resistant macrophages
.
PLoS ONE
.
2012
;
7
:
e49741
.

17.

Fernandes-Alnemri
 
T
,
Yu
 
J-W
,
Datta
 
P
,
Wu
 
J
,
Alnemri
 
ES
.
AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA
.
Nature
.
2009
;
458
:
509
513
.

18.

Rathinam
 
VAK
,
Jiang
 
Z
,
Waggoner
 
SN
, et al.  
The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses
.
Nat Immunol
.
2010
;
11
:
395
402
.

19.

Zhao
 
Y
,
Yang
 
J
,
Shi
 
J
, et al.  
The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus
.
Nature
.
2011
;
477
:
596
600
.

20.

Kofoed
 
EM
,
Vance
 
RE
.
Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity
.
Nature
.
2011
;
477
:
592
595
.

21.

Yang
 
J
,
Zhao
 
Y
,
Shi
 
J
,
Shao
 
F
.
Human NAIP and mouse NAIP1 recognize bacterial type III secretion needle protein for inflammasome activation
.
Proc Natl Acad Sci
.
2013
;
110
:
14408
14413
.

22.

Reyes Ruiz
 
VM
,
Ramirez
 
J
,
Naseer
 
N
, et al.  
Broad detection of bacterial type III secretion system and flagellin proteins by the human NAIP/NLRC4 inflammasome
.
Proc Natl Acad Sci
.
2017
;
114
:
13242
13247
.

23.

Menu
 
P
,
Vince
 
JE
.
The NLRP3 inflammasome in health and disease: the good, the bad and the ugly
.
Clin Exp Immunol
.
2011
;
166
:
1
15
.

24.

Fernandes-Alnemri
 
T
,
Wu
 
J
,
Yu
 
J-W
, et al.  
The pyroptosome: a supramolecular assembly of ASC dimers mediating inflammatory cell death via caspase-1 activation
.
Cell Death Differ
.
2007
;
14
:
1590
1604
.

25.

Cai
 
X
,
Chen
 
J
,
Xu
 
H
, et al.  
Prion-like polymerization underlies signal transduction in antiviral immune defense and inflammasome activation
.
Cell
.
2014
;
156
:
1207
1222
.

26.

Lu
 
A
,
Magupalli
 
VG
,
Ruan
 
J
, et al.  
Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes
.
Cell
.
2014
;
156
:
1193
1206
.

27.

Dick
 
MS
,
Sborgi
 
L
,
Rühl
 
S
,
Hiller
 
S
,
Broz
 
P
.
ASC filament formation serves as a signal amplification mechanism for inflammasomes
.
Nat Commun
.
2016
;
7
:
11929
.

28.

Walsh
 
JG
,
Logue
 
SE
,
Lüthi
 
AU
,
Martin
 
SJ
.
Caspase-1 promiscuity is counterbalanced by rapid inactivation of processed enzyme
.
J Biol Chem
.
2011
;
286
:
32513
32524
.

29.

Vince
 
JE
,
Wong
 
WW-L
,
Gentle
 
I
, et al.  
Inhibitor of apoptosis proteins limit RIP3 kinase-dependent interleukin-1 activation
.
Immunity
.
2012
;
36
:
215
227
.

30.

Boucher
 
D
,
Monteleone
 
M
,
Coll
 
RC
, et al.  
Caspase-1 self-cleavage is an intrinsic mechanism to terminate inflammasome activity
.
J Exp Med
.
2018
;
215
:
827
840
.

31.

Broz
 
P
,
von Moltke
 
J
,
Jones
 
JW
,
Vance
 
RE
,
Monack
 
DM
.
Differential requirement for caspase-1 autoproteolysis in pathogen-induced cell death and cytokine processing
.
Cell Host Microbe
.
2010
;
8
:
471
483
.

32.

Guey
 
B
,
Bodnar
 
ML
,
Manié
 
SN
,
Tardivel
 
A
,
Petrilli
 
V
.
Caspase-1 autoproteolysis is differentially required for NLRP1b and NLRP3 inflammasome function
.
Proc Natl Acad Sci
.
2014
;
111
:
17254
17259
.

33.

Conos
 
SA
,
Lawlor
 
KE
,
Vaux
 
DL
,
Vince
 
JE
,
Lindqvist
 
LM
.
Cell death is not essential for caspase-1-mediated interleukin-1beta activation and secretion
.
Cell Death Differ
.
2016
;
23
:
1827
1838
.

34.

He
 
W-T
,
Wan
 
H
,
Hu
 
L
, et al.  
Gasdermin D is an executor of pyroptosis and required for interleukin-1beta secretion
.
Cell Res
.
2015
;
25
:
1285
1298
.

35.

Ding
 
J
,
Wang
 
K
,
Liu
 
W
, et al.  
Pore-forming activity and structural autoinhibition of the gasdermin family
.
Nature
.
2016
;
535
:
111
116
.

36.

Liu
 
X
,
Zhang
 
Z
,
Ruan
 
J
, et al.  
Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores
.
Nature
.
2016
;
535
:
153
158
.

37.

Chen
 
X
,
He
 
W-T
,
Hu
 
L
, et al.  
Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel-mediated necroptosis
.
Cell Res
.
2016
;
26
:
1007
1020
.

38.

Mulvihill
 
E
,
Sborgi
 
L
,
Mari
 
SA
,
Pfreundschuh
 
M
,
Hiller
 
S
,
Müller
 
DJ
.
Mechanism of membrane pore formation by human gasdermin-D
.
EMBO J
.
2018
;
37
:
e98321
.

39.

Russo
 
HM
,
Rathkey
 
J
,
Boyd-Tressler
 
A
,
Katsnelson
 
MA
,
Abbott
 
DW
,
Dubyak
 
GR
.
Active caspase-1 induces plasma membrane pores that precede pyroptotic lysis and are blocked by lanthanides
.
J Immunol
.
2016
;
197
:
1353
1367
.

40.

Evavold
 
CL
,
Ruan
 
J
,
Tan
 
Y
,
Xia
 
S
,
Wu
 
H
,
Kagan
 
JC
.
The pore-forming protein gasdermin D regulates interleukin-1 secretion from living macrophages
.
Immunity
.
2018
;
48
:
35
44.e6
.

41.

Schneider
 
KS
,
Groß
 
CJ
,
Dreier
 
RF
, et al.  
The Inflammasome drives GSDMD-independent secondary pyroptosis and IL-1 release in the absence of caspase-1 protease activity
.
Cell Rep
.
2017
;
21
:
3846
3859
.

42.

Shu
 
F
,
Chen
 
J
,
Ma
 
X
, et al.  
Cholesterol crystal-mediated inflammation is driven by plasma membrane destabilization
.
Front Immunol
.
2018
;
9
:
1163
.

43.

Sagulenko
 
V
,
Thygesen
 
SJ
,
Sester
 
DP
, et al.  
AIM2 and NLRP3 inflammasomes activate both apoptotic and pyroptotic death pathways via ASC
.
Cell Death Differ
.
2013
;
20
:
1149
1160
.

44.

Lima
 
H
, Jr,
Jacobson
 
L
,
Goldberg
 
M
, et al.  
Role of lysosome rupture in controlling Nlrp3 signaling and necrotic cell death
.
Cell Cycle
.
2013
;
12
:
1868
1878
.

45.

Muñoz-Planillo
 
RL
,
Kuffa
 
P
,
Martínez-Colón
 
G
,
Smith
 
BL
,
Rajendiran
 
TM
,
Núñez
 
G
.
K(+) efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter
.
Immunity
.
2013
;
38
:
1142
1153
.

46.

Orlowski
 
GM
,
Sharma
 
S
,
Colbert
 
JD
, et al.  
Frontline Science: multiple cathepsins promote inflammasome-independent, particle-induced cell death during NLRP3-dependent IL-1beta activation
.
J Leukoc Biol
.
2017
;
102
:
7
17
.

47.

Mascarenhas
 
DPA
,
Cerqueira
 
DM
,
Pereira
 
MSF
, et al.  
Inhibition of caspase-1 or gasdermin-D enable caspase-8 activation in the Naip5/NLRC4/ASC inflammasome
.
PLOS Pathog
.
2017
;
13
:
e1006502
.

48.

Monteleone
 
M
,
Stanley
 
AC
,
Chen
 
KW
, et al.  
Interleukin-1beta maturation triggers its relocation to the plasma membrane for gasdermin-D-dependent and -independent secretion
.
Cell Rep
.
2018
;
24
:
1425
1433
.

49.

Man
 
SM
,
Tourlomousis
 
P
,
Hopkins
 
L
,
Monie
 
TP
,
Fitzgerald
 
KA
,
Bryant
 
CE
.
Salmonella infection induces recruitment of caspase-8 to the inflammasome to modulate IL-1beta production
.
J Immunol
.
2013
;
191
:
5239
5246
.

50.

Antonopoulos
 
C
,
Russo
 
HM
,
El Sanadi
 
C
, et al.  
Caspase-8 as an effector and regulator of NLRP3 inflammasome signaling
.
J Biol Chem
.
2015
;
290
:
20167
20184
.

51.

Sagulenko
 
V
,
Vitak
 
N
,
Vajjhala
 
PR
,
Vince
 
JE
,
Stacey
 
KJ
.
Caspase-1 Is an apical caspase leading to caspase-3 cleavage in the AIM2 inflammasome response, independent of caspase-8
.
J Mol Biol
.
2018
;
430
:
238
247
.

52.

Taabazuing
 
CY
,
Okondo
 
MC
,
Bachovchin
 
DA
.
Pyroptosis and apoptosis pathways engage in bidirectional crosstalk in monocytes and macrophages
.
Cell Chem Biol
.
2017
;
24
:
507
514.e4
. e504.

53.

Gaidt
 
MM
,
Ebert
 
TS
,
Chauhan
 
D
, et al.  
Human Monocytes Engage an Alternative Inflammasome Pathway
.
Immunity
.
2016
;
44
:
833
846
.

54.

Moriwaki
 
K
,
Bertin
 
J
,
Gough
 
PJ
,
Chan
 
FK-M
.
A RIPK3-caspase 8 complex mediates atypical pro-IL-1beta processing
.
J Immunol
.
2015
;
194
:
1938
1944
.

55.

He
 
Y
,
Franchi
 
L
,
Nunez
 
G
.
TLR Agonists stimulate Nlrp3-dependent IL-1beta production independently of the purinergic P2X7 receptor in dendritic cells and in vivo
.
J Immunol
.
2013
;
190
:
334
339
.

56.

Shi
 
H
,
Wang
 
Y
,
Li
 
X
, et al.  
NLRP3 activation and mitosis are mutually exclusive events coordinated by NEK7, a new inflammasome component
.
Nat Immunol
.
2016
;
17
:
250
258
.

57.

He
 
Y
,
Zeng
 
MY
,
Yang
 
D
,
Motro
 
B
,
Núñez
 
G
.
NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux
.
Nature
.
2016
;
530
:
354
357
.

58.

Schmid-Burgk
 
JL
,
Chauhan
 
D
,
Schmidt
 
T
, et al.  
A genome-wide CRISPR (clustered regularly interspaced short palindromic repeats) screen identifies NEK7 as an essential component of NLRP3 inflammasome activation
.
J Biol Chem
.
2016
;
291
:
103
109
.

59.

Shi
 
C-S
,
Kehrl
 
JH
.
Cytochrome c negatively regulates NLRP3 inflammasomes
.
PLoS ONE
.
2016
;
11
:
e0167636
.

60.

He
 
H
,
Jiang
 
H
,
Chen
 
Y
, et al.  
Oridonin is a covalent NLRP3 inhibitor with strong anti-inflammasome activity
.
Nat Commun
.
2018
;
9
:
2550
.

61.

Bauernfeind
 
FG
,
Horvath
 
G
,
Stutz
 
A
, et al.  
Cutting edge: nF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression
.
J Immunol
.
2009
;
183
:
787
791
.

62.

Schroder
 
K
,
Sagulenko
 
V
,
Zamoshnikova
 
A
, et al.  
Acute lipopolysaccharide priming boosts inflammasome activation independently of inflammasome sensor induction
.
Immunobiology
.
2012
;
217
:
1325
1329
.

63.

Juliana
 
C
,
Fernandes-Alnemri
 
T
,
Kang
 
S
,
Farias
 
A
,
Qin
 
F
,
Alnemri
 
ES
.
Non-transcriptional priming and deubiquitination regulate NLRP3 inflammasome activation
.
J Biol Chem
.
2012
;
287
:
36617
36622
.

64.

Fernandes-Alnemri
 
T
,
Kang
 
S
,
Anderson
 
C
,
Sagara
 
J
,
Fitzgerald
 
KA
,
Alnemri
 
ES
.
Cutting edge: tLR signaling licenses IRAK1 for rapid activation of the NLRP3 inflammasome
.
J Immunol
.
2013
;
191
:
3995
3999
.

65.

Lin
 
K-M
,
Hu
 
W
,
Troutman
 
TD
, et al.  
IRAK-1 bypasses priming and directly links TLRs to rapid NLRP3 inflammasome activation
.
Proc Natl Acad Sci
.
2014
;
111
:
775
780
.

66.

Py
 
BNDF
,
Kim
 
M-S
,
Vakifahmetoglu-Norberg
 
H
,
Yuan
 
J
.
Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity
.
Molecular Cell
.
2013
;
49
:
331
338
.

67.

Chi
 
W
,
Hua
 
X
,
Chen
 
X
, et al.  
Mitochondrial DNA oxidation induces imbalanced activity of NLRP3/NLRP6 inflammasomes by activation of caspase-8 and BRCC36 in dry eye
.
J Autoimmun
.
2017
;
80
:
65
76
.

68.

Rodgers
 
MA
,
Bowman
 
JW
,
Fujita
 
H
, et al.  
The linear ubiquitin assembly complex (LUBAC) is essential for NLRP3 inflammasome activation
.
J Exp Med
.
2014
;
211
:
1333
1347
.

69.

Gurung
 
P
,
Lamkanfi
 
M
,
Kanneganti
 
T-D
.
Cutting edge: sHARPIN is required for optimal NLRP3 inflammasome activation
.
J Immunol
.
2015
;
194
:
2064
2067
.

70.

Duong
 
BH
,
Onizawa
 
M
,
Oses-Prieto
 
JA
, et al.  
A20 restricts ubiquitination of pro-interleukin-1beta protein complexes and suppresses NLRP3 inflammasome activity
.
Immunity
.
2015
;
42
:
55
67
.

71.

Yan
 
Y
,
Jiang
 
W
,
Liu
 
L
, et al.  
Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome
.
Cell
.
2015
;
160
:
62
73
.

72.

Han
 
S
,
Lear
 
TB
,
Jerome
 
JA
, et al.  
Lipopolysaccharide primes the NALP3 inflammasome by inhibiting its ubiquitination and degradation mediated by the SCFFBXL2 E3 ligase
.
J Biol Chem
.
2015
;
290
:
18124
18133
.

73.

Song
 
H
,
Liu
 
B
,
Huai
 
W
, et al.  
The E3 ubiquitin ligase TRIM31 attenuates NLRP3 inflammasome activation by promoting proteasomal degradation of NLRP3
.
Nat Commun
.
2016
;
7
:
13727
.

74.

Poudel
 
B
,
Gurung
 
P
.
An update on cell intrinsic negative regulators of the NLRP3 inflammasome
.
J Leukoc Biol
.
2018
;
103
:
1165
1177
.

75.

Kayagaki
 
N
,
Warming
 
SR
,
Lamkanfi
 
M
, et al.  
Non-canonical inflammasome activation targets caspase-11
.
Nature
.
2011
;
479
:
117
121
.

76.

Kang
 
S-J
,
Wang
 
S
,
Hara
 
H
, et al.  
Dual role of caspase-11 in mediating activation of caspase-1 and caspase-3 under pathological conditions
.
J Cell Biol
.
2000
;
149
:
613
622
.

77.

Kayagaki
 
N
,
Wong
 
MT
,
Stowe
 
IB
, et al.  
Noncanonical inflammasome activation by intracellular LPS independent of TLR4
.
Science
.
2013
;
341
:
1246
1249
.

78.

Gurung
 
P
,
Malireddi
 
RKS
,
Anand
 
PK
, et al.  
Toll or interleukin-1 receptor (TIR) domain-containing adaptor inducing interferon-beta (TRIF)-mediated caspase-11 protease production integrates Toll-like receptor 4 (TLR4) protein- and Nlrp3 inflammasome-mediated host defense against enteropathogens
.
J Biol Chem
.
2012
;
287
:
34474
34483
.

79.

Case
 
CL
,
Kohler
 
LJ
,
Lima
 
JB
, et al.  
Caspase-11 stimulates rapid flagellin-independent pyroptosis in response to Legionella pneumophila
.
Proc Natl Acad Sci
.
2013
;
110
:
1851
1856
.

80.

Aachoui
 
Y
,
Leaf
 
IA
,
Hagar
 
JA
, et al.  
Caspase-11 protects against bacteria that escape the vacuole
.
Science
.
2013
;
339
:
975
978
.

81.

Knodler
 
LA
,
Crowley
 
SM
,
Sham
 
HP
, et al.  
Noncanonical inflammasome activation of caspase-4/caspase-11 mediates epithelial defenses against enteric bacterial pathogens
.
Cell Host Microbe
.
2014
;
16
:
249
256
.

82.

Demon
 
D
,
Kuchmiy
 
A
,
Fossoul
 
A
,
Zhu
 
Q
,
Kanneganti
 
T-D
,
Lamkanfi
 
M
.
Caspase-11 is expressed in the colonic mucosa and protects against dextran sodium sulfate-induced colitis
.
Mucosal Immunol
.
2014
;
7
:
1480
1491
.

83.

Oficjalska
 
K
,
Raverdeau
 
M
,
Aviello
 
G
, et al.  
Protective role for caspase-11 during acute experimental murine colitis
.
J Immunol
.
2015
;
194
:
1252
1260
.

84.

Shi
 
J
,
Zhao
 
Y
,
Wang
 
Y
, et al.  
Inflammatory caspases are innate immune receptors for intracellular LPS
.
Nature
.
2014
;
514
:
187
192
.

85.

Baker
 
PJ
,
Boucher
 
D
,
Bierschenk
 
D
, et al.  
NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase-5
.
Eur J Immunol
.
2015
;
45
:
2918
2926
.

86.

Casson
 
CN
,
Yu
 
J
,
Reyes
 
VM
, et al.  
Human caspase-4 mediates noncanonical inflammasome activation against gram-negative bacterial pathogens
.
Proc Natl Acad Sci
.
2015
;
112
:
6688
6693
.

87.

Hagar
 
JA
,
Powell
 
DA
,
Aachoui
 
Y
,
Ernst
 
RK
,
Miao
 
EA
.
Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock
.
Science
.
2013
;
341
:
1250
1253
.

88.

Zanoni
 
I
,
Tan
 
Y
,
Di Gioia
 
M
, et al.  
An endogenous caspase-11 ligand elicits interleukin-1 release from living dendritic cells
.
Science
.
2016
;
352
:
1232
1236
.

89.

Kerur
 
N
,
Fukuda
 
S
,
Banerjee
 
D
, et al.  
cGAS drives noncanonical-inflammasome activation in age-related macular degeneration
.
Nat Med
.
2018
;
24
:
50
61
.

90.

Zanoni
 
I
,
Tan
 
Y
,
Di Gioia
 
M
,
Springstead
 
JR
,
Kagan
 
JC
.
By capturing inflammatory lipids released from dying cells, the receptor CD14 induces inflammasome-dependent phagocyte hyperactivation
.
Immunity
.
2017
;
47
:
697
709.e3
. e693.

91.

Yeon
 
SH
,
Yang
 
G
,
Lee
 
HE
,
Lee
 
JY
.
Oxidized phosphatidylcholine induces the activation of NLRP3 inflammasome in macrophages
.
J Leukoc Biol
.
2017
;
101
:
205
215
.

92.

Chu
 
LH
,
Indramohan
 
M
,
Ratsimandresy
 
RA
, et al.  
The oxidized phospholipid oxPAPC protects from septic shock by targeting the non-canonical inflammasome in macrophages
.
Nat Commun
.
2018
;
9
:
996
.

93.

Bitto
 
NJ
,
Baker
 
PJ
,
Dowling
 
JK
, et al.  
Membrane vesicles from Pseudomonas aeruginosa activate the non-canonical inflammasome through caspase-5 in human monocytes
.
Immunol Cell Biol
.
2018
;
96
:
1120
1130
.

94.

Wacker
 
MA
,
Teghanemt
 
A
,
Weiss
 
JP
,
Barker
 
JH
.
High-affinity caspase-4 binding to LPS presented as high molecular mass aggregates or in outer membrane vesicles
.
Innate Immun
.
2017
;
23
:
336
344
.

95.

Yang
 
D
,
He
 
Y
,
Muñoz-Planillo
 
R
,
Liu
 
Q
,
Núñez
 
G
.
Caspase-11 requires the pannexin-1 channel and the purinergic P2X7 pore to mediate pyroptosis and endotoxic shock
.
Immunity
.
2015
;
43
:
923
932
.

96.

Rühl
 
S
,
Broz
 
P
.
Caspase-11 activates a canonical NLRP3 inflammasome by promoting K(+) efflux
.
Eur J Immunol
.
2015
;
45
:
2927
2936
.

97.

Cheng
 
KT
,
Xiong
 
S
,
Ye
 
Z
, et al.  
Caspase-11-mediated endothelial pyroptosis underlies endotoxemia-induced lung injury
.
J Clin Invest
.
2017
;
127
:
4124
4135
.

98.

Mandal
 
P
,
Feng
 
Y
,
Lyons
 
JD
, et al.  
Caspase-8 collaborates with caspase-11 to drive tissue damage and execution of endotoxic shock
.
Immunity
.
2018
;
49
:
42
55.e6
. e46.

100.

Rathinam
 
VAK
,
Vanaja
 
SK
,
Waggoner
 
L
, et al.  
TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by gram-negative bacteria
.
Cell
.
2012
;
150
:
606
619
.

101.

Napier
 
BA
,
Brubaker
 
SW
,
Sweeney
 
TE
, et al.  
Complement pathway amplifies caspase-11-dependent cell death and endotoxin-induced sepsis severity
.
J Exp Med
.
2016
;
213
:
2365
2382
.

102.

Aachoui
 
Y
,
Kajiwara
 
Y
,
Leaf
 
IA
, et al.  
Canonical inflammasomes drive IFN-gamma to prime caspase-11 in defense against a cytosol-invasive bacterium
.
Cell Host Microbe
.
2015
;
18
:
320
332
.

103.

Pilla
 
DM
,
Hagar
 
JA
,
Haldar
 
AK
, et al.  
Guanylate binding proteins promote caspase-11-dependent pyroptosis in response to cytoplasmic LPS
.
Proc Natl Acad Sci
.
2014
;
111
:
6046
6051
.

104.

Santos
 
JC
,
Dick
 
MS
,
Lagrange
 
B
, et al.  
LPS targets host guanylate-binding proteins to the bacterial outer membrane for non-canonical inflammasome activation
.
EMBO J
.
2018
;
37
:
e98089
.

105.

Finethy
 
R
,
Jorgensen
 
I
,
Haldar
 
AK
, et al.  
Guanylate binding proteins enable rapid activation of canonical and noncanonical inflammasomes in Chlamydia-infected macrophages
.
Infect Immun
.
2015
;
83
:
4740
4749
.

106.

Man
 
SM
,
Karki
 
R
,
Sasai
 
M
, et al.  
IRGB10 liberates bacterial ligands for sensing by the AIM2 and caspase-11-NLRP3 inflammasomes
.
Cell
.
2016
;
167
:
382
396.e17
.

107.

Li
 
P
,
Jiang
 
W
, et al.  
Ubiquitination and degradation of GBPs by a Shigella effector to suppress host defence
.
Nature
.
2017
;
551
:
378
383
.

108.

Delbridge
 
ARD
,
Grabow
 
S
,
Strasser
 
A
,
Vaux
 
DL
.
Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies
.
Nat Rev Cancer
.
2016
;
16
:
99
109
.

109.

Murphy
 
JM
,
Vince
 
JE
.
Post-translational control of RIPK3 and MLKL mediated necroptotic cell death
.
F1000Research
.
2015
;
4
:
F1000
. Faculty Rev-1297.

110.

Feltham
 
R
,
Vince
 
JE
,
Lawlor
 
KE
.
Caspase-8: not so silently deadly
.
Clin Transl Immunol
.
2017
;
6
:
e124
.

111.

Maelfait
 
J
,
Vercammen
 
E
,
Janssens
 
S
, et al.  
Stimulation of Toll-like receptor 3 and 4 induces interleukin-1beta maturation by caspase-8
.
J Exp Med
.
2008
;
205
:
1967
1973
.

112.

Bossaller
 
L
,
Chiang
 
P-I
,
Schmidt-Lauber
 
C
, et al.  
Cutting Edge: fAS (CD95) mediates noncanonical IL-1beta and IL-18 maturation via caspase-8 in an RIP3-independent manner
.
J Immunol
.
2012
;
189
:
5508
5512
.

113.

Antonopoulos
 
C
,
El Sanadi
 
C
,
Kaiser
 
WJ
,
Mocarski
 
ES
,
Dubyak
 
GR
.
Proapoptotic chemotherapeutic drugs induce noncanonical processing and release of IL-1beta via caspase-8 in dendritic cells
.
J Immunol
.
2013
;
191
:
4789
4803
.

114.

Wang
 
L
,
Du
 
F
,
Wang
 
X
.
TNF-alpha induces two distinct caspase-8 activation pathways
.
Cell
.
2008
;
133
:
693
703
.

115.

Yabal
 
M
,
Müller
 
N
,
Adler
 
H
, et al.  
XIAP restricts TNF- and RIP3-dependent cell death and inflammasome activation
.
Cell Rep
.
2014
;
7
:
1796
1808
.

116.

Lawlor
 
KE
,
Khan
 
N
,
Mildenhall
 
A
, et al.  
RIPK3 promotes cell death and NLRP3 inflammasome activation in the absence of MLKL
.
Nat Commun
.
2015
;
6
:
6282
.

117.

Wicki
 
S
,
Gurzeler
 
U
,
Wei-Lynn Wong
 
W
,
Jost
 
PJ
,
Bachmann
 
D
,
Kaufmann
 
T
.
Loss of XIAP facilitates switch to TNFalpha-induced necroptosis in mouse neutrophils
.
Cell Death Dis
.
2016
;
7
:
e2422
.

118.

Chen
 
KW
,
Lawlor
 
KE
,
von Pein
 
JB
, et al.  
Cutting edge: blockade of inhibitor of apoptosis proteins sensitizes neutrophils to TNF- but not lipopolysaccharide-mediated cell death and IL-1beta secretion
.
J Immunol
.
2018
;
200
:
3341
3346
.

119.

Lawlor
 
KE
,
Feltham
 
R
,
Yabal
 
M
, et al.  
XIAP Loss triggers RIPK3- and caspase-8-driven IL-1beta activation and cell death as a consequence of TLR-MyD88-induced cIAP1-TRAF2 degradation
.
Cell Rep
.
2017
;
20
:
668
682
.

120.

Wu
 
Y-H
,
Kuo
 
W-C
,
Wu
 
Y-J
, et al.  
Participation of c-FLIP in NLRP3 and AIM2 inflammasome activation
.
Cell Death Differ
.
2014
;
21
:
451
461
.

121.

Gurung
 
P
,
Anand
 
PK
,
Malireddi
 
RKS
, et al.  
FADD and caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes
.
J Immunol
.
2014
;
192
:
1835
1846
.

122.

Philip
 
NH
,
Dillon
 
CP
,
Snyder
 
AG
, et al.  
Caspase-8 mediates caspase-1 processing and innate immune defense in response to bacterial blockade of NF-kappaB and MAPK signaling
.
Proc Natl Acad Sci
.
2014
;
111
:
7385
7390
. https://doi.org/10.1073/pnas.1403252111.

123.

Allam
 
R
,
Lawlor
 
KE
,
Yu
 
EC-W
, et al.  
Mitochondrial apoptosis is dispensable for NLRP3 inflammasome activation but non-apoptotic caspase-8 is required for inflammasome priming
.
EMBO Reports
.
2014
;
15
:
982
990
.

124.

Weng
 
D
,
Marty-Roix
 
R
,
Ganesan
 
S
, et al.  
Caspase-8 and RIP kinases regulate bacteria-induced innate immune responses and cell death
.
Proc Natl Acad Sci
.
2014
;
111
:
7391
7396
.

125.

Zhang
 
X
,
Fan
 
C
,
Zhang
 
H
, et al.  
MLKL and FADD are critical for suppressing progressive lymphoproliferative disease and activating the NLRP3 inflammasome
.
Cell Rep
.
2016
;
16
:
3247
3259
.

126.

Conos
 
SA
,
Chen
 
KW
,
De Nardo
 
D
, et al.  
Active MLKL triggers the NLRP3 inflammasome in a cell-intrinsic manner
.
Proc Natl Acad Sci
.
2017
;
114
:
E961
E969
.

127.

Kang
 
T-B
,
Yang
 
S-H
,
Toth
 
B
,
Kovalenko
 
A
,
Wallach
 
D
.
Caspase-8 blocks kinase RIPK3-mediated activation of the NLRP3 inflammasome
.
Immunity
.
2013
;
38
:
27
40
.

128.

Moriwaki
 
K
,
Farias Luz
 
N
,
Balaji
 
S
, et al.  
The mitochondrial phosphatase PGAM5 is dispensable for necroptosis but promotes inflammasome activation in macrophages
.
J Immunol
.
2016
;
196
:
407
415
.

129.

Rickard
 
JA
,
O'Donnell
 
JA
,
Evans
 
JM
, et al.  
RIPK1 regulates RIPK3-MLKL driven systemic inflammation and emergency hematopoiesis
.
Cell
.
2014
;
157
:
1175
1188
.

130.

Speir
 
M
,
Lawlor
 
KE
,
Glaser
 
SP
, et al.  
Eliminating Legionella by inhibiting BCL-XL to induce macrophage apoptosis
.
Nat Microbiol
.
2016
;
1
:
15034
.

131.

Vince
 
JE
,
De Nardo
 
D
,
Gao
 
W
, et al.  
The mitochondrial apoptotic effectors BAX/BAK activate caspase-3 and -7 to trigger NLRP3 inflammasome and caspase-8 driven IL-1β activation
.
Cell Rep
.
2018
;
25
:
2339
2353.e4
.

132.

Chauhan
 
D
,
Bartok
 
E
,
Gaidt
 
MM
, et al.  
BAX/BAK Induced apoptosis results in caspase-8 dependent IL-1β maturation in macrophages
.
Cell Rep
.
2018
;
25
:
2354
2368.e5
.

133.

Wang
 
Y
,
Gao
 
W
,
Shi
 
X
, et al.  
Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin
.
Nature
.
2017
;
547
:
99
103
.

134.

Rogers
 
C
,
Fernandes-Alnemri
 
T
,
Mayes
 
L
,
Alnemri
 
D
,
Cingolani
 
G
,
Alnemri
 
ES
.
Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death
.
Nat Commun
.
2017
;
8
:
14128
.

135.

Hoffman
 
HM
,
Mueller
 
JL
,
Broide
 
DH
,
Wanderer
 
AA
,
Kolodner
 
RD
.
Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome
.
Nat Genet
.
2001
;
29
:
301
305
.

136.

Aksentijevich
 
I
,
Nowak
 
M
,
Mallah
 
M
, et al.  
De novo CIAS1 mutations, cytokine activation, and evidence for genetic heterogeneity in patients with neonatal-onset multisystem inflammatory disease (NOMID): a new member of the expanding family of pyrin-associated autoinflammatory diseases
.
Arthritis Rheum
.
2002
;
46
:
3340
3348
.

137.

Martorana
 
D
,
Bonatti
 
F
,
Mozzoni
 
P
,
Vaglio
 
A
,
Percesepe
 
A
.
Monogenic autoinflammatory diseases with mendelian inheritance: genes, mutations, and genotype/phenotype correlations
.
Front Immunol
.
2017
;
8
:
344
.

138.

Rowczenio
 
DM
,
Gomes
 
SNM
,
Ar?stegui
 
JI
, et al.  
Late-onset cryopyrin-associated periodic syndromes caused by somatic NLRP3 mosaicism—UK Single Center Experience
.
Front Immunol
.
2017
;
8
:
1410
.

139.

Brydges
 
SD
,
Mueller
 
JL
,
McGeough
 
MD
, et al.  
Inflammasome-mediated disease animal models reveal roles for innate but not adaptive immunity
.
Immunity
.
2009
;
30
:
875
887
.

140.

Lachmann
 
HJ
,
Kone-Paut
 
I
,
Kuemmerle-Deschner
 
JB
, et al.  
Use of canakinumab in the cryopyrin-associated periodic syndrome
.
N Engl J Med
.
2009
;
360
:
2416
2425
.

141.

Paramel
 
GV
,
Sirsjö
 
A
,
Frans?n
 
K
.
Role of genetic alterations in the NLRP3 and CARD8 genes in health and disease
.
Mediators Inflamm
.
2015
. 846782.

142.

Heneka
 
MT
,
Kummer
 
MP
,
Stutz
 
A
, et al.  
NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice
.
Nature
.
2013
;
493
:
674
678
.

143.

Jankovic
 
D
,
Ganesan
 
J
,
Bscheider
 
M
, et al.  
The Nlrp3 inflammasome regulates acute graft-versus-host disease
.
J Exp Med
.
2013
;
210
:
1899
1910
.

144.

Coll
 
RC
,
Robertson
 
AAB
,
Chae
 
JJ
, et al.  
A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases
.
Nat Med
.
2015
;
21
:
248
255
.

145.

Wree
 
A
,
Eguchi
 
A
,
McGeough
 
MD
, et al.  
NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice
.
Hepatology
.
2014
;
59
:
898
910
.

146.

Martinon
 
F
,
P?trilli
 
V
,
Mayor
 
A
,
Tardivel
 
A
,
Tschopp
 
JR
.
Gout-associated uric acid crystals activate the NALP3 inflammasome
.
Nature
.
2006
;
440
:
237
241
.

147.

Vandanmagsar
 
B
,
Youm
 
Y-H
,
Ravussin
 
A
, et al.  
The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance
.
Nat Med
.
2011
;
17
:
179
188
.

148.

Duewell
 
P
,
Kono
 
H
,
Rayner
 
KJ
, et al.  
NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals
.
Nature
.
2010
;
464
:
1357
1361
.

149.

Ridker
 
PM
,
MacFadyen
 
JG
,
Everett
 
BM
, et al.  
Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial
.
Lancet
.
2018
;
391
:
319
328
.

150.

Ridker
 
PM
,
Everett
 
BM
,
Thuren
 
T
, et al.  
Antiinflammatory therapy with canakinumab for atherosclerotic disease
.
N Engl J Med
.
2017
;
377
:
1119
1131
.

151.

Mangan
 
MSJ
,
Olhava
 
EJ
,
Roush
 
WR
,
Seidel
 
HM
,
Glick
 
GD
,
Latz
 
E
.
Targeting the NLRP3 inflammasome in inflammatory diseases
.
Nat Rev Drug Discov
.
2018
;
17
:
588
606
.

152.

Masters
 
SL
,
Dunne
 
A
,
Subramanian
 
SL
, et al.  
Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1beta in type 2 diabetes
.
Nat Immunol
.
2010
;
11
:
897
904
.

153.

Halle
 
A
,
Hornung
 
V
,
Petzold
 
GC
, et al.  
The NALP3 inflammasome is involved in the innate immune response to amyloid-beta
.
Nat Immunol
.
2008
;
9
:
857
865
.

154.

Nsiah-Sefaa
 
A
,
McKenzie
 
M
.
Combined defects in oxidative phosphorylation and fatty acid beta-oxidation in mitochondrial disease
.
Biosci Rep
.
2016
;
36
:
e00313
e00313
.

155.

Bratic
 
A
,
Larsson
 
N-GR
.
The role of mitochondria in aging
.
J Clin Invest
.
2013
;
123
:
951
957
.

156.

Montgomery
 
MK
,
Turner
 
N
.
Mitochondrial dysfunction and insulin resistance: an update
.
Endocr Connect
.
2015
;
4
:
R1
R15
.

157.

Kelley
 
DE
,
He
 
J
,
Menshikova
 
EV
,
Ritov
 
VB
.
Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes
.
Diabetes
.
2002
;
51
:
2944
2950
.

158.

Pendergrass
 
M
,
Koval
 
J
,
Vogt
 
C
, et al.  
Insulin-induced hexokinase II expression is reduced in obesity and NIDDM
.
Diabetes
.
1998
;
47
:
387
394
.

159.

Wolf
 
AJ
,
Reyes
 
CN
,
Liang
 
W
, et al.  
Hexokinase is an innate immune receptor for the detection of bacterial peptidoglycan
.
Cell
.
2016
;
166
:
624
636
.

160.

Zhou
 
R
,
Tardivel
 
A
,
Thorens
 
B
,
Choi
 
I
,
Tschopp
 
JR
.
Thioredoxin-interacting protein links oxidative stress to inflammasome activation
.
Nat Immunol
.
2010
;
11
:
136
140
.

161.

Zhou
 
R
,
Yazdi
 
AS
,
Menu
 
P
,
Tschopp
 
JR
.
A role for mitochondria in NLRP3 inflammasome activation
.
Nature
.
2011
;
469
:
221
225
.

162.

Listenberger
 
LL
,
Han
 
X
,
Lewis
 
SE
, et al.  
Triglyceride accumulation protects against fatty acid-induced lipotoxicity
.
Proc Natl Acad Sci
.
2003
;
100
:
3077
3082
.

163.

Yuzefovych
 
L
,
Wilson
 
G
,
Rachek
 
L
.
Different effects of oleate vs. palmitate on mitochondrial function, apoptosis, and insulin signaling in L6 skeletal muscle cells: role of oxidative stress
.
Am J Physiol-Endocrinol Metabol
.
2010
;
299
:
E1096
E1105
.

164.

Zorzano
 
A
,
Liesa
 
M
,
Palacín
 
M
.
Role of mitochondrial dynamics proteins in the pathophysiology of obesity and type 2 diabetes
.
Int J Biochem Cell Biol
.
2009
;
41
:
1846
1854
.

165.

Li
 
A
,
Zhang
 
S
,
Li
 
J
,
Liu
 
K
,
Huang
 
F
,
Liu
 
B
.
Metformin and resveratrol inhibit Drp1-mediated mitochondrial fission and prevent ER stress-associated NLRP3 inflammasome activation in the adipose tissue of diabetic mice
.
Mol Cell Endocrinol
.
2016
;
434
:
36
47
.

166.

Youm
 
Y-H
,
Grant
 
RW
,
McCabe
 
LR
, et al.  
Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging
.
Cell Metabol
.
2013
;
18
:
519
532
.

167.

Youm
 
Y-H
,
Kanneganti
 
T-D
,
Vandanmagsar
 
B
, et al.  
The Nlrp3 inflammasome promotes age-related thymic demise and immunosenescence
.
Cell Rep
.
2012
;
1
:
56
68
.

168.

Bauernfeind
 
F
,
Niepmann
 
S
,
Knolle
 
PA
,
Hornung
 
V
.
Aging-associated TNF production primes inflammasome activation and NLRP3-related metabolic disturbances
.
J Immunol
.
2016
;
197
:
2900
2908
.

169.

Spadaro
 
O
,
Goldberg
 
EL
,
Camell
 
CD
, et al.  
Growth hormone receptor deficiency protects against age-related NLRP3 inflammasome activation and immune senescence
.
Cell Rep
.
2016
;
14
:
1571
1580
.

170.

Camell
 
CD
,
Sander
 
J
, et al.  
Inflammasome-driven catecholamine catabolism in macrophages blunts lipolysis during ageing
.
Nature
.
2017
;
550
:
119
123
.

171.

Acosta
 
JC
,
Banito
 
A
,
Wuestefeld
 
T
, et al.  
A complex secretory program orchestrated by the inflammasome controls paracrine senescence
.
Nat Cell Biol
.
2013
;
15
:
978
990
.

172.

Rheinheimer
 
J
,
de Souza
 
BM
,
Cardoso
 
NS
,
Bauer
 
AC
,
Crispim
 
D
.
Current role of the NLRP3 inflammasome on obesity and insulin resistance: a systematic review
.
Metabolism
.
2017
;
74
:
1
9
.

173.

Youm
 
Y-H
,
Adijiang
 
A
,
Vandanmagsar
 
B
,
Burk
 
D
,
Ravussin
 
A
,
Dixit
 
VD
.
Elimination of the NLRP3-ASC inflammasome protects against chronic obesity-induced pancreatic damage
.
Endocrinology
.
2011
;
152
:
4039
4045
.

174.

Stienstra
 
R
,
van Diepen
 
JA
,
Tack
 
CJ
, et al.  
Inflammasome is a central player in the induction of obesity and insulin resistance
.
Proc Natl Acad Sci
.
2011
;
108
:
15324
15329
.

175.

Pavillard
 
LSE
,
Cañadas-Lozano
 
D
,
Alcocer-Gómez
 
ES
, et al.  
NLRP3-inflammasome inhibition prevents high fat and high sugar diets-induced heart damage through autophagy induction
.
Oncotarget
.
2017
;
8
:
99740
99756
.

176.

van der Heijden
 
T
,
Kritikou
 
E
,
Venema
 
W
, et al.  
NLRP3 inflammasome inhibition by MCC950 reduces atherosclerotic lesion development in apolipoprotein E-deficient mice-brief report
.
Arterioscler Thromb Vasc Biol
.
2017
;
37
:
1457
1461
.

177.

Sanz
 
Y
,
Moya-Perez
 
A
.
Microbiota, inflammation and obesity
.
Adv Exp Med Biol
.
2014
;
817
:
291
317
.

178.

Rainone
 
V
,
Schneider
 
L
,
Saulle
 
I
, et al.  
Upregulation of inflammasome activity and increased gut permeability are associated with obesity in children and adolescents
.
Int J Obes
.
2016
;
40
:
1026
1033
.

179.

Kim
 
K-A
,
Gu
 
W
,
Lee
 
I-A
,
Joh
 
E-H
,
Kim
 
D-H
.
High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway
.
PLoS ONE
.
2012
;
7
:
e47713
.

180.

Griffin
 
C
,
Eter
 
L
,
Lanzetta
 
N
, et al.  
TLR4, TRIF, and MyD88 are essential for myelopoiesis and CD11c(+) adipose tissue macrophage production in obese mice
.
J Biol Chem
.
2018
;
293
:
8775
8786
.

181.

Lu
 
Z
,
Zhang
 
X
,
Li
 
Y
,
Lopes-Virella
 
MF
,
Huang
 
Y
.
TLR4 antagonist attenuates atherogenesis in LDL receptor-deficient mice with diet-induced type 2 diabetes
.
Immunobiology
.
2015
;
220
:
1246
1254
.

182.

He
 
W
,
Yuan
 
T
,
Choezom
 
D
, et al.  
Ageing potentiates diet-induced glucose intolerance, beta-cell failure and tissue inflammation through TLR4
.
Sci Rep
.
2018
;
8
:
2767
.

183.

Reynolds
 
CM
,
McGillicuddy
 
FC
,
Harford
 
KA
,
Finucane
 
OM
,
Mills
 
KHG
,
Roche
 
HM
.
Dietary saturated fatty acids prime the NLRP3 inflammasome via TLR4 in dendritic cells-implications for diet-induced insulin resistance
.
Mol Nutr Food Res
.
2012
;
56
:
1212
1222
.

184.

Sheedy
 
FJ
,
Grebe
 
A
,
Rayner
 
KJ
, et al.  
CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation
.
Nat Immunol
.
2013
;
14
:
812
820
.

185.

Schilling
 
JD
,
Machkovech
 
HM
,
He
 
L
, et al.  
Palmitate and lipopolysaccharide trigger synergistic ceramide production in primary macrophages
.
J Biol Chem
.
2013
;
288
:
2923
2932
.

186.

Wen
 
H
,
Gris
 
D
,
Lei
 
Y
, et al.  
Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling
.
Nat Immunol
.
2011
;
12
:
408
415
.

187.

Robblee
 
MM
,
Kim
 
CC
,
Abate
 
JP
, et al.  
Saturated fatty acids engage an IRE1alpha-dependent pathway to activate the NLRP3 inflammasome in myeloid cells
.
Cell Rep
.
2016
;
14
:
2611
2623
.

188.

Tufanli
 
O
,
Telkoparan Akillilar
 
P
,
Acosta-Alvear
 
D
, et al.  
Targeting IRE1 with small molecules counteracts progression of atherosclerosis
.
Proc Natl Acad Sci
.
2017
;
114
:
E1395
E1404
.

189.

Karasawa
 
T
,
Kawashima
 
A
,
Usui-Kawanishi
 
F
, et al.  
Saturated fatty acids undergo intracellular crystallization and activate the NLRP3 inflammasome in macrophages
.
Arter Thromb Vasc Biol
.
2018
;
38
:
744
756
.

190.

Yan
 
Y
,
Jiang
 
W
,
Spinetti
 
T
, et al.  
Omega-3 fatty acids prevent inflammation and metabolic disorder through inhibition of NLRP3 inflammasome activation
.
Immunity
.
2013
;
38
:
1154
1163
.

191.

Williams-Bey
 
Y
,
Boularan
 
C
,
Vural
 
A
, et al.  
Omega-3 free fatty acids suppress macrophage inflammasome activation by inhibiting NF-kappaB activation and enhancing autophagy
.
PLoS ONE
.
2014
;
9
:
e97957
.

192.

Youm
 
Y-H
,
Nguyen
 
KY
,
Grant
 
RW
, et al.  
The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease
.
Nat Med
.
2015
;
21
:
263
269
.

193.

Goldberg
 
EL
,
Asher
 
JL
,
Molony
 
RD
, et al.  
Beta-hydroxybutyrate deactivates neutrophil NLRP3 inflammasome to relieve gout flares
.
Cell Rep
.
2017
;
18
:
2077
2087
.

194.

Parikh
 
H
,
Carlsson
 
E
,
Chutkow
 
WA
, et al.  
TXNIP regulates peripheral glucose metabolism in humans
.
PLoS Med
.
2007
;
4
:
e158
.

195.

van Greevenbroek
 
MMJ
,
Vermeulen
 
VMM-J
,
Feskens
 
EJM
, et al.  
Genetic variation in thioredoxin interacting protein (TXNIP) is associated with hypertriglyceridaemia and blood pressure in diabetes mellitus
.
Diabetic Med
.
2007
;
24
:
498
504
.

196.

Koenen
 
TB
,
Stienstra
 
R
,
van Tits
 
LJ
, et al.  
Hyperglycemia activates caspase-1 and TXNIP-mediated IL-1beta transcription in human adipose tissue
.
Diabetes
.
2011
;
60
:
517
524
.

197.

Minn
 
AlexandraH
,
Hafele
 
Christia
,
Shalev
 
Anat
.
Thioredoxin-interacting protein is stimulated by glucose through a carbohydrate response element and induces beta-cell apoptosis
.
Endocrinology
.
2005
;
146
:
2397
2405
.

198.

Chen
 
J
,
Fontes
 
G
,
Saxena
 
G
,
Poitout
 
V
,
Shalev
 
A
.
Lack of TXNIP protects against mitochondria-mediated apoptosis but not against fatty acid-induced ER stress-mediated beta-cell death
.
Diabetes
.
2010
;
59
:
440
447
.

199.

Chutkow
 
WA
,
Birkenfeld
 
AL
,
Brown
 
JD
, et al.  
Deletion of the alpha-arrestin protein Txnip in mice promotes adiposity and adipogenesis while preserving insulin sensitivity
.
Diabetes
.
2010
;
59
:
1424
1434
.

200.

Panse
 
Madhur
,
Kluth
 
Olive
,
Lorza-Gil
 
Estel
, et al.  
Palmitate and insulin counteract glucose-induced thioredoxin interacting protein (TXNIP) expression in insulin secreting cells via distinct mechanisms
.
PLoS ONE
.
2018
;
13
:
e0198016
.

201.

Litwak
 
SaraA
,
Wali
 
JibranA
,
Pappas
 
EvanG
, et al.  
Lipotoxic stress induces pancreatic beta-cell apoptosis through modulation of Bcl-2 proteins by the ubiquitin-proteasome system
.
J Diabetes Res
.
2015
;
2015
:
1
.

202.

Tseng
 
HisaHuiLin
,
Vong
 
ChiTen
,
Kwan
 
YiuW
,
Lee
 
SimonMing-Yue
,
Hoi
 
MaggiePuiMa
.
TRPM2 regulates TXNIP-mediated NLRP3 inflammasome activation via interaction with p47 phox under high glucose in human monocytic cells
.
Sci Rep
.
2016
;
6
:
35016
.

203.

Wali
 
JibranA
,
Gurzov
 
EstebanN
,
Fynch
 
Stace
, et al.  
Activation of the NLRP3 inflammasome complex is not required for stress-induced death of pancreatic islets
.
PLoS ONE
.
2014
;
9
:
e113128
.

204.

Lebreton
 
Fann
,
Berishvili
 
Ekaterin
,
Parnaud
 
Géraldin
, et al.  
NLRP3 inflammasome is expressed and regulated in human islets
.
Cell Death Dis
.
2018
;
9
:
726
.

205.

Chen
 
Junqi
,
Hui
 
SimonT
,
Couto
 
FrancescaM
, et al.  
Thioredoxin-interacting protein deficiency induces Akt/Bcl-xL signaling and pancreatic beta-cell mass and protects against diabetes
.
FASEB J
.
2008
;
22
:
3581
3594
.

206.

Bedarida
 
Tatian
,
Domingues
 
Aliso
,
Baron
 
Stephani
, et al.  
Reduced endothelial thioredoxin-interacting protein protects arteries from damage induced by metabolic stress in vivo
.
FASEB J
.
2018
;
32
:
3108
3118
.

207.

Ding
 
Chunchu
,
Zhao
 
Ya
,
Shi
 
Xu
, et al.  
New insights into salvianolic acid A action: regulation of the TXNIP/NLRP3 and TXNIP/ChREBP pathways ameliorates HFD-induced NAFLD in rats
.
Sci Rep
.
2016
;
6
:
28734
.

208.

Groß
 
CJ
,
Mishra
 
R
,
Schneider
 
KS
, et al.  
K(+) efflux-independent NLRP3 inflammasome activation by small molecules targeting mitochondria
.
Immunity
.
2016
;
45
:
761
773
.

209.

Perregaux
 
D
,
Gabel
 
CA
.
Interleukin-1 beta maturation and release in response to ATP and nigericin. Evidence that potassium depletion mediated by these agents is a necessary and common feature of their activity
.
J Biol Chem
.
1994
;
269
:
15195
15203
.

210.

Mariathasan
 
Sanjee
,
Weiss
 
DavidS
,
Newton
 
Ki
, et al.  
Cryopyrin activates the inflammasome in response to toxins and ATP
.
Nature
.
2006
;
440
:
228
232
.

211.

Pétrilli
 
V
,
Papin
 
S
,
Dostert
 
C
,
Mayor
 
A
,
Martinon
 
F
,
Tschopp
 
J
.
Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration
.
Cell Death Differ
.
2007
;
14
:
1583
1589
.

212.

Lamkanfi
 
Mohame
,
Mueller
 
JamesL
,
Vitari
 
Alberto C
, et al.  
Glyburide inhibits the cryopyrin/Nalp3 inflammasome
.
J Cell Biol
.
2009
;
187
:
61
70
.

213.

Pelegrin
 
Pabl
,
Surprenant
 
Annmari
.
Pannexin-1 mediates large pore formation and interleukin-1beta release by the ATP-gated P2X7 receptor
.
EMBO J
.
2006
;
25
:
5071
5082
.

214.

Pelegrin
 
Pabl
,
Surprenant
 
Annmari
.
Pannexin-1 couples to maitotoxin- and nigericin-induced interleukin-1beta release through a dye uptake-independent pathway
.
J Biol Chem
.
2007
;
282
:
2386
2394
.

215.

Compan
 
V
,
Baroja-Mazo
 
A
,
López-Castejón
 
Glor
.
Cell volume regulation modulates NLRP3 inflammasome activation
.
Immunity
.
2012
;
37
:
487
500
.

216.

Qu
 
Y
,
Misaghi
 
S
,
Newton
 
K
.
Pannexin-1 is required for ATP release during apoptosis but not for inflammasome activation
.
J Immunol
.
2011
;
186
:
6553
6561
.

217.

Di
 
A
,
Xiong
 
S
,
Ye
 
Z
.
The TWIK2 potassium efflux channel in macrophages mediates NLRP3 inflammasome-induced inflammation
.
Immunity
.
2018
;
49
:
56
65.e4
.

218.

Tang
 
T
,
Lang
 
X
,
Xu
 
C
.
CLICs-dependent chloride efflux is an essential and proximal upstream event for NLRP3 inflammasome activation
.
Nat Commun
.
2017
;
8
:
202
.

219.

Domingo-Fernández
 
R
,
Coll
 
RC
,
Kearney
 
J
,
Breit
 
S
,
O'Neill
 
LAJ
.
The intracellular chloride channel proteins CLIC1 and CLIC4 induce IL-1beta transcription and activate the NLRP3 inflammasome
.
J Biol Chem
.
2017
;
292
:
12077
12087
.

220.

Sanman
 
LE
,
Qian
 
Y
,
Eisele
 
NA
.
Disruption of glycolytic flux is a signal for inflammasome signaling and pyroptotic cell death
.
eLife
.
2016
;
5
:
e13663
.

221.

Seth
 
RB
,
Sun
 
L
,
Ea
 
C-K
,
Chen
 
ZJ
.
Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3
.
Cell
.
2005
;
122
:
669
682
.

222.

White
 
MJ
,
McArthur
 
K
,
Metcalf
 
D
.
Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production
.
Cell
.
2014
;
159
:
1549
1562
.

223.

Rongvaux
 
A
,
Jackson
 
R
,
Harman
 
CCD
.
Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA
.
Cell
.
2014
;
159
:
1563
1577
.

224.

Bruey
 
J-M
,
Bruey-Sedano
 
N
,
Luciano
 
F
.
Bcl-2 and Bcl-XL regulate proinflammatory caspase-1 activation by interaction with NALP1
.
Cell
.
2007
;
129
:
45
56
.

225.

Gerlic
 
M
,
Faustin
 
B
,
Postigo
 
A
.
Vaccinia virus F1L protein promotes virulence by inhibiting inflammasome activation
.
Proc Natl Acad Sci
.
2013
;
110
:
7808
7813
.

226.

Yu
 
C-H
,
Moecking
 
J
,
Geyer
 
M
,
Masters
 
SL
.
Mechanisms of NLRP1-mediated autoinflammatory disease in humans and mice
.
J Mol Biol
.
2018
;
430
:
142
152
.

227.

Kovarova
 
M
,
Hesker
 
PR
,
Jania
 
L
.
NLRP1-dependent pyroptosis leads to acute lung injury and morbidity in mice
.
J Immunol
.
2012
;
189
:
2006
2016
.

228.

Yazdi
 
AS
,
Guarda
 
G
,
Riteau
 
N
, et al.  
Nanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1alpha and IL-1beta
.
Proc Natl Acad Sci
.
2010
;
107
:
19449
19454
.

229.

Shimada
 
K
,
Crother
 
TR
,
Karlin
 
J
.
Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis
.
Immunity
.
2012
;
36
:
401
414
.

230.

Lawlor
 
KE
,
van Nieuwenhuijze
 
A
,
Parker
 
KL
.
Bcl-2 overexpression ameliorates immune complex-mediated arthritis by altering FcgammaRIIb expression and monocyte homeostasis
.
J Leukoc Biol
.
2013
;
93
:
585
597
.

231.

Saitoh
 
T
,
Fujita
 
N
,
Jang
 
MH
.
Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production
.
Nature
.
2008
;
456
:
264
268
.

232.

Nakahira
 
K
,
Haspel
 
JA
,
Rathinam
 
VAK
.
Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome
.
Nat Immunol
.
2011
;
12
:
222
230
.

233.

Jessop
 
F
,
Hamilton
 
RF
,
Rhoderick
 
JF
,
Shaw
 
PK
,
Holian
 
A
.
Autophagy deficiency in macrophages enhances NLRP3 inflammasome activity and chronic lung disease following silica exposure
.
Toxicol Appl Pharmacol
.
2016
;
309
:
101
110
.

234.

Harris
 
J
,
Hartman
 
M
,
Roche
 
C
.
Autophagy controls IL-1beta secretion by targeting pro-IL-1beta for degradation
.
J Biol Chem
.
2011
;
286
:
9587
9597
.

235.

Kimura
 
T
,
Jain
 
A
,
Choi
 
SW
.
TRIM-mediated precision autophagy targets cytoplasmic regulators of innate immunity
.
J Cell Biol
.
2015
;
210
:
973
989
.

236.

Jabir
 
MS
,
Ritchie
 
ND
,
Li
 
D
.
Caspase-1 cleavage of the TLR adaptor TRIF inhibits autophagy and beta-interferon production during Pseudomonas aeruginosa infection
.
Cell Host Microbe
.
2014
;
15
:
214
227
.

237.

Yu
 
J
,
Nagasu
 
H
,
Murakami
 
T
, et al.  
Inflammasome activation leads to caspase-1-dependent mitochondrial damage and block of mitophagy
.
Proc Natl Acad Sci
.
2014
;
111
:
15514
15519
.

238.

Jamilloux
 
Y
,
Lagrange
 
B
,
Di Micco
 
A
.
A proximity-dependent biotinylation (BioID) approach flags the p62/sequestosome-1 protein as a caspase-1 substrate
.
J Biol Chem
.
2018
;
293
:
12563
12575
.

239.

Dostert
 
C
,
Petrilli
 
V
,
Van Bruggen
 
R
,
Steele
 
C
,
Mossman
 
BT
,
Tschopp
 
J
.
Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica
.
Science
.
2008
;
320
:
674
677
.

240.

Bauernfeind
 
F
,
Bartok
 
E
,
Rieger
 
A
,
Franchi
 
L
,
Nunez
 
G
,
Hornung
 
V
.
Cutting edge: reactive oxygen species inhibitors block priming, but not activation, of the NLRP3 inflammasome
.
J Immunol
.
2011
;
187
:
613
617
.

241.

van Bruggen
 
R
,
Koker
 
MY
,
Jansen
 
M
.
Human NLRP3 inflammasome activation is Nox1-4 independent
.
Blood
.
2010
;
115
:
5398
5400
.

242.

Murakami
 
T
,
Ockinger
 
J
,
Yu
 
J
.
Critical role for calcium mobilization in activation of the NLRP3 inflammasome
.
Proc Natl Acad Sci
.
2012
;
109
:
11282
11287
.

243.

Won
 
J-H
,
Park
 
S
,
Hong
 
S
,
Son
 
S
,
Yu
 
J-Wo
.
Rotenone-induced impairment of mitochondrial electron transport chain confers a selective priming signal for NLRP3 inflammasome activation
.
J Biol Chem
.
2015
;
290
:
27425
27437
.

244.

Park
 
Y-J
,
Yoon
 
S-J
,
Suh
 
H-W
.
TXNIP deficiency exacerbates endotoxic shock via the induction of excessive nitric oxide synthesis
.
PLOS Pathog
.
2013
;
9
:
e1003646
.

245.

Zhong
 
Z
,
Zhai
 
Y
,
Liang
 
S
, et al.  
TRPM2 links oxidative stress to NLRP3 inflammasome activation
.
Nat Commun
.
2013
;
4
:
1611
.

246.

Ito
 
M
,
Yanagi
 
Y
,
Ichinohe
 
T
.
Encephalomyocarditis virus viroporin 2B activates NLRP3 inflammasome
.
PLOS Pathog
.
2012
;
8
:
e1002857
.

247.

Ichinohe
 
T
,
Yamazaki
 
T
,
Koshiba
 
T
,
Yanagi
 
Y
.
Mitochondrial protein mitofusin 2 is required for NLRP3 inflammasome activation after RNA virus infection
.
Proc Natl Acad Sci
.
2013
;
110
:
17963
17968
.

248.

Bronner
 
DN
,
Abuaita
 
BH
,
Chen
 
X
.
Endoplasmic reticulum stress activates the inflammasome via NLRP3- and caspase-2-driven mitochondrial damage
.
Immunity
.
2015
;
43
:
451
462
.

249.

Groß
 
O
,
Yazdi
 
AS
,
Thomas
 
CJ
.
Inflammasome activators induce interleukin-1alpha secretion via distinct pathways with differential requirement for the protease function of caspase-1
.
Immunity
.
2012
;
36
:
388
400
.

250.

Narendra
 
D
,
Tanaka
 
A
,
Suen
 
D-F
,
Youle
 
RJ
.
Parkin is recruited selectively to impaired mitochondria and promotes their autophagy
.
J Cell Biol
.
2008
;
183
:
795
803
.

251.

Zhong
 
Z
,
Umemura
 
A
,
Sanchez-Lopez
 
E
.
NF-kappaB restricts inflammasome activation via elimination of damaged mitochondria
.
Cell
.
2016
;
164
:
896
910
.

252.

Wang
 
Y
,
Shan
 
B
,
Liang
 
Y
,
Wei
 
H
,
Yuan
 
J
.
Parkin regulates NF-kappaB by mediating site-specific ubiquitination of RIPK1
.
Cell Death Dis
.
2018
;
9
:
732
.

253.

Zhong
 
Z
,
Liang
 
S
,
Sanchez-Lopez
 
E
.
New mitochondrial DNA synthesis enables NLRP3 inflammasome activation
.
Nature
.
2018
;
560
:
198
203
.

254.

West
 
AP
,
Khoury-Hanold
 
W
,
Staron
 
M
.
Mitochondrial DNA stress primes the antiviral innate immune response
.
Nature
.
2015
;
520
:
553
557
.

255.

Man
 
SM
,
Karki
 
R
,
Malireddi
 
RKS
.
The transcription factor IRF1 and guanylate-binding proteins target activation of the AIM2 inflammasome by Francisella infection
.
Nat Immunol
.
2015
;
16
:
467
475
.

256.

Dang
 
EV
,
McDonald
 
JG
,
Russell
 
DW
,
Cyster
 
JG
.
Oxysterol restraint of cholesterol synthesis prevents AIM2 inflammasome activation
.
Cell
.
2017
;
171
:
1057
1071.e11
.

257.

Misawa
 
T
,
Takahama
 
M
,
Kozaki
 
T
.
Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome
.
Nat Immunol
.
2013
;
14
:
454
460
.

258.

Bryan
 
NB
,
Dorfleutner
 
A
,
Rojanasakul
 
Y
,
Stehlik
 
C
.
Activation of inflammasomes requires intracellular redistribution of the apoptotic speck-like protein containing a caspase recruitment domain
.
J Immunol
.
2009
;
182
:
3173
3182
.

259.

Subramanian
 
N
,
Natarajan
 
K
,
Clatworthy
 
MR
,
Wang
 
Z
,
Germain
 
RN
.
The adaptor MAVS promotes NLRP3 mitochondrial localization and inflammasome activation
.
Cell
.
2013
;
153
:
348
361
.

260.

Park
 
S
,
Juliana
 
C
,
Hong
 
S
.
The mitochondrial antiviral protein MAVS associates with NLRP3 and regulates its inflammasome activity
.
J Immunol
.
2013
;
191
:
4358
4366
.

261.

Franchi
 
L
,
Eigenbrod
 
T
,
Muñoz-Planillo
 
RL
.
Cytosolic double-stranded RNA activates the NLRP3 inflammasome via MAVS-induced membrane permeabilization and K+ efflux
.
J Immunol
.
2014
;
193
:
4214
4222
.

262.

Iyer
 
SS
,
He
 
Q
,
Janczy
 
JR
.
Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation
.
Immunity
.
2013
;
39
:
311
323
.

263.

Elliott
 
EI
,
Miller
 
AN
,
Banoth
 
B
.
Cutting edge: mitochondrial assembly of the NLRP3 inflammasome complex is initiated at priming
.
J Immunol
.
2018
;
200
:
3047
3052
.

264.

Wang
 
Y
,
Yang
 
C
,
Mao
 
K
,
Chen
 
S
,
Meng
 
G
,
Sun
 
B
.
Cellular localization of NLRP3 inflammasome
.
Protein Cell
.
2013
;
4
:
425
431
.

265.

Wang
 
X
,
Jiang
 
W
,
Yan
 
Y
, et al.  
RNA viruses promote activation of the NLRP3 inflammasome through a RIP1-RIP3-DRP1 signaling pathway
.
Nat Immunol
.
2014
;
15
:
1126
1133
.

266.

Kang
 
S
,
Fernandes-Alnemri
 
T
,
Rogers
 
C
.
Caspase-8 scaffolding function and MLKL regulate NLRP3 inflammasome activation downstream of TLR3
.
Nat Commun
.
2015
;
6
:
7515
.

267.

Brough
 
D
,
Le Feuvre
 
RA
,
Wheeler
 
RD
, et al.  
Ca2+ stores and Ca2+ entry differentially contribute to the release of IL-1 beta and IL-1 alpha from murine macrophages
.
J Immunol
.
2003
;
170
:
3029
3036
.

268.

Lee
 
G-S
,
Subramanian
 
N
,
Kim
 
AI
.
The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP
.
Nature
.
2012
;
492
:
123
127
.

269.

Rossol
 
M
,
Pierer
 
M
,
Raulien
 
N
.
Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G protein-coupled calcium sensing receptors
.
Nat Commun
.
2012
;
3
:
1329
.

270.

Malireddi
 
RKS
,
Gurung
 
P
,
Mavuluri
 
J
.
TAK1 restricts spontaneous NLRP3 activation and cell death to control myeloid proliferation
.
J Exp Med
.
2018
;
215
:
1023
1034
.

271.

Katsnelson
 
MA
,
Rucker
 
LG
,
Russo
 
HM
,
Dubyak
 
GR
.
K+ efflux agonists induce NLRP3 inflammasome activation independently of Ca2+ signaling
.
J Immunol
.
2015
;
194
:
3937
3952
.

272.

Hornung
 
V
,
Bauernfeind
 
F
,
Halle
 
A
.
Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization
.
Nat Immunol
.
2008
;
9
:
847
856
.

273.

Dostert
 
C
,
Guarda
 
G
,
Romero
 
JF
.
Malarial hemozoin is a Nalp3 inflammasome activating danger signal
.
PLoS ONE
.
2009
;
4
:
e6510
.

274.

Gaidt
 
MM
,
Ebert
 
TS
,
Chauhan
 
D
.
The DNA inflammasome in human myeloid cells is initiated by a STING-cell death program upstream of NLRP3
.
Cell
.
2017
;
171
:
1110
1124.e18
.

275.

Orlowski
 
GM
,
Colbert
 
JD
,
Sharma
 
S
,
Bogyo
 
M
,
Robertson
 
SA
,
Rock
 
KL
.
Multiple cathepsins promote pro-IL-1beta synthesis and NLRP3-mediated IL-1beta activation
.
J Immunol
.
2015
;
195
:
1685
1697
.

276.

Cuchet-Lourenço
 
D
,
Eletto
 
D
,
Wu
 
C
.
Biallelic RIPK1 mutations in humans cause severe immunodeficiency, arthritis, and intestinal inflammation
.
Science
.
2018
;
361
:
810
813
.

277.

Lehle
 
AS
,
Farin
 
HF
,
Marquardt
 
B
.
Intestinal inflammation and dysregulated immunity in patients with inherited caspase-8 deficiency
.
Gastroenterology
.
2018
. https://doi.org/10.1053/j.gastro.2018.09.041.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)