NLRP3 inflammasome activation, metabolic danger signals, and protein binding partners

in Journal of Endocrinology
Authors:
Sy-Ying Leu Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan, ROC

Search for other papers by Sy-Ying Leu in
Current site
Google Scholar
PubMed
Close
,
Yi-Ling Tsang Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
Institute of Physiological Chemistry and Pathobiochemistry and Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany

Search for other papers by Yi-Ling Tsang in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-9540-8847
,
Li-Chun Ho School of Medicine, I-Shou University, Kaohsiung, Taiwan, ROC
Division of General Medicine, Department of Internal Medicine, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan, ROC

Search for other papers by Li-Chun Ho in
Current site
Google Scholar
PubMed
Close
,
Ching-Chun Yang Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC

Search for other papers by Ching-Chun Yang in
Current site
Google Scholar
PubMed
Close
,
Ai-Ning Shao Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC

Search for other papers by Ai-Ning Shao in
Current site
Google Scholar
PubMed
Close
,
Chia-Yu Chang Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC

Search for other papers by Chia-Yu Chang in
Current site
Google Scholar
PubMed
Close
,
Hui-Kuan Lin Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA

Search for other papers by Hui-Kuan Lin in
Current site
Google Scholar
PubMed
Close
,
Pei-Jane Tsai Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC

Search for other papers by Pei-Jane Tsai in
Current site
Google Scholar
PubMed
Close
,
Junne-Ming Sung Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan, ROC

Search for other papers by Junne-Ming Sung in
Current site
Google Scholar
PubMed
Close
, and
Yau-Sheng Tsai Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan, Taiwan, ROC

Search for other papers by Yau-Sheng Tsai in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-5560-2661

Correspondence should be addressed to Y Tsai or P Tsai or J Sung: yaustsai@mail.ncku.edu.tw or peijtsai@mail.ncku.edu.tw or jmsung@mail.ncku.edu.tw
Free access

Sign up for journal news

The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is an oligomeric complex that assembles in response to exogenous signals of pathogen infection and endogenous danger signals of non-microbial origin. When NLRP3 inflammasome assembly activates caspase-1, it promotes the maturation and release of the inflammatory cytokines interleukin-1B and IL-18. Aberrant activation of the NLRP3 inflammasome has been implicated in various diseases, including chronic inflammatory, metabolic, and cardiovascular diseases. The NLRP3 inflammasome can be activated through several principal mechanisms, including K+ efflux, lysosomal damage, and the production of mitochondrial reactive oxygen species. Interestingly, metabolic danger signals activate the NLRP3 inflammasome to induce metabolic diseases. NLRP3 contains three crucial domains: an N-terminal pyrin domain, a central nucleotide-binding domain, and a C-terminal leucine-rich repeat domain. Protein–protein interactions act as a ‘pedal or brake’ to control the activation of the NLRP3 inflammasome. In this review, we present the mechanisms underlying NLRP3 inflammasome activation after induction by metabolic danger signals or via protein–protein interactions with NLRP3 that likely occur in metabolic diseases. Understanding these mechanisms will enable the development of specific inhibitors to treat NLRP3-related metabolic diseases.

Abstract

The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is an oligomeric complex that assembles in response to exogenous signals of pathogen infection and endogenous danger signals of non-microbial origin. When NLRP3 inflammasome assembly activates caspase-1, it promotes the maturation and release of the inflammatory cytokines interleukin-1B and IL-18. Aberrant activation of the NLRP3 inflammasome has been implicated in various diseases, including chronic inflammatory, metabolic, and cardiovascular diseases. The NLRP3 inflammasome can be activated through several principal mechanisms, including K+ efflux, lysosomal damage, and the production of mitochondrial reactive oxygen species. Interestingly, metabolic danger signals activate the NLRP3 inflammasome to induce metabolic diseases. NLRP3 contains three crucial domains: an N-terminal pyrin domain, a central nucleotide-binding domain, and a C-terminal leucine-rich repeat domain. Protein–protein interactions act as a ‘pedal or brake’ to control the activation of the NLRP3 inflammasome. In this review, we present the mechanisms underlying NLRP3 inflammasome activation after induction by metabolic danger signals or via protein–protein interactions with NLRP3 that likely occur in metabolic diseases. Understanding these mechanisms will enable the development of specific inhibitors to treat NLRP3-related metabolic diseases.

Introduction

The prevalence of metabolic diseases has increased significantly in recent years. The World Health Organization has identified obesity as a significant public health problem (Chooi et al. 2019). Moreover, type 2 diabetes remains one of the top ten global causes of death, and its prevalence has increased by approximately 70% since 2000 (Lin et al. 2020). Recent studies have suggested a critical role for non-microbial sterile inflammation in progressing metabolic diseases, including obesity, insulin resistance, type 2 diabetes, hepatic steatosis, cardiovascular and joint diseases, and certain types of cancer.

The release of pro-inflammatory cytokines after abnormal inflammasome activation has been well documented in the pathogenesis of metabolic diseases (Mangan et al. 2018). The inflammasome, especially the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, has been shown to recognise danger signals of non-microbial endogenous origin (Mangan et al. 2018). Thus, it is important to understand the activators and components of inflammasomes and their activation processes to prevent and treat metabolic diseases.

NLRP3 inflammasome

The NLRP3 (also known as NACHT, LRR, and PYD domain-containing protein 3; NALP3) inflammasome is an oligomeric complex that senses pathogen-associated molecular patterns (PAMPs) in infection conditions and damage-associated molecular patterns (DAMPs) in non-microbial inflammatory conditions. The NLRP3 inflammasome complex comprises three major components: NLRP3, apoptosis speck protein with caspase recruitment (ASC), and cysteine-dependent aspartate-directed protease-1 (caspase-1) (Broz & Dixit 2016) (Fig. 1). Structurally, NLRP3 contains three important domains: an N-terminal pyrin domain (PYD), a central nucleotide-binding domain (NBD), and a C-terminal leucine-rich repeat (LRR) domain (Broz & Dixit 2016). The adaptor protein ASC contains a PYD and a caspase recruitment domain (CARD), which facilitate interactions between NLRP3 and caspase-1 that carries a CARD (Broz & Dixit 2016). When NLRP3 responds to PAMPs or DAMPs, it interacts with ASC and caspase-1.

Figure 1
Figure 1

NLRP3 inflammasome activation. NLRP3 inflammasome activation comprises a two-step process. During the priming step, a Toll-like receptor mediates the downstream NF-kB translocation to the nucleus and produces NLRP3 and pro-IL-1B. During the following activation step, NLRP3 oligomerises and recruits ASC. Subsequently, pro-IL-1B, pro-IL-18, and gasdermin D (GSDMD) are cleaved by active caspase-1 and released. The released GSDMD triggers pyroptosis.

Citation: Journal of Endocrinology 257, 2; 10.1530/JOE-22-0184

NLRP3 inflammasome activation comprises a two-step pathway in rodent macrophages, but not in human macrophages or dendritic cells (Netea et al. 2009, Gaidt et al. 2016) or in rodent dendritic cells (He et al. 2013). In response to the first signal (step 1), inflammasome components (e.g. NLRP3 and pro-IL-1B) are primed through activation of the NF-kB transcription factor, a downstream signal of the toll-like receptor (TLR). In response to the second signal (step 2), NLRP3 oligomerises through the NBD and recruits ASC through the PYD to form an ASC speck. Oligomerised ASC then recruits caspase-1 through the CARD to activate caspase-1 via proximity-induced self-cleavage. Activated caspase-1 cleaves pro-IL-1B and pro-IL-18, leading to the maturation and release of IL-1B and IL-18 (Swanson et al. 2019). In addition, activated caspase-1 also cleaves gasdermin D (GSDMD), which inserts into the membrane to form pores and induce pyroptosis (Swanson et al. 2019). Both caspase-1-mediated pyroptosis and the secretion of IL-1B are critical for reducing microbial infection and improving tissue repair (Broz & Dixit 2016).

The role of the NLRP3 inflammasome in metabolic diseases

Elevated IL-1B and IL-18 levels are associated with obesity, insulin resistance, dyslipidaemia, and metabolic syndrome (Hung et al. 2005). When PAMPs or DAMPs trigger the innate immune system, the NLRP3 inflammasome is assembled to promote the maturation and secretion of IL-1B and IL-18 and initiate the inflammatory response. However, excessive inflammasome activation leads to tissue damage and promotes disease progression (Carlsson et al. 2010). Several metabolic diseases with dysregulated inflammatory responses are induced by aberrant inflammasome activation and excessive IL-1B production (Fig. 2). The mechanisms by which NRLP3 inflammasome is involved in metabolic diseases are addressed in each metabolic DAMP.

Figure 2
Figure 2

NLRP3 inflammasome-related metabolic diseases. Serial DAMPs activate the NLRP3 inflammasome and contribute to various diseases. Because obesity is a common precursor for NLRP3-mediated metabolic diseases, it seems to prime the NLRP3 inflammasome. In the progression of atherosclerosis, cholesterol crystals or serum amyloid A (SAA) can trigger NLRP3 inflammasome activation. Gout, an inflammatory disease that can be caused by uric acid crystal deposition, is also a potent NLRP3 activator. Amylin (IAPP) deposition and high glucose levels in diabetes are related to NLRP3 inflammasome activation. The obesity-associated factors, palmitate and ceramide, are activators of NLRP3. Pathogenesis of NASH also involves palmitate-induced release of danger signals and the activation of the NLRP3 inflammasome.

Citation: Journal of Endocrinology 257, 2; 10.1530/JOE-22-0184

Several mechanisms underlie the activation of the NLRP3 inflammasome in response to various stimuli (Fig. 3). For example, the change in cytoplasmic potassium (K+), chloride (Cl), or calcium (Ca2+) levels, the elevation of active cathepsin B in lysosomal disruption, and mitochondrial dysfunction and the release of reactive oxygen species (ROS) are all potent NLRP3 inflammasome activators (Swanson et al. 2019).

Figure 3
Figure 3

NLRP3 inflammasome activators. P2X7 is stimulated by ATP and serum amyloid A, subsequently regulating downstream K+ efflux and resulting in NLRP3 inflammasome activation. Uric acid and cholesterol crystals destabilise the lysosome, leading to the release of cathepsin B and the activation of the NLRP3 inflammasome. ROS stress derived from dysfunctional mitochondria and cytosol, which can be stimulated by glucose and amylin, is also a potent activator of the NLRP3 inflammasome. Palmitate activates the NLRP3 inflammasome by regulating the AMPK–autophagy–ROS pathway. Ceramide initiates mitochondrial dysfunction and activates the NLRP3 inflammasome.

Citation: Journal of Endocrinology 257, 2; 10.1530/JOE-22-0184

The contribution of ROS in NLRP3 inflammasome activation

Although ROS production represents an evolutionarily conserved pathway in response to infection, it is also a specific metabolic product of redox reactions. Most of NLRP3 activators, including the two well-known inducers, adenosine triphosphate (ATP) and particulate, trigger ROS production, while antioxidants inhibit NLRP3 inflammasome activation, suggesting that ROS is involved in NLRP3 activation (Dostert et al. 2008). A redox-sensitive protein can partly explain the link between ROS and inflammasome activation. For example, the generated ROS are sensed by thioredoxin and thioredoxin-interacting protein (TXNIP), which form a complex under the reducing condition. ROS induced by NLRP3 stimuli oxidises thioredoxin, leading to its dissociation from TXNIP. The released TXNIP then binds to NLRP3, triggering the activation of the NLRP3 inflammasome in THP-1 human macrophages. TXNIP deficiency impairs NLRP3-induced caspase-1 activation and IL-1B secretion in macrophages by several agonists, including R837, monosodium urate (MSU), alum or silica, and ATP (Zhou et al. 2010). While numerous NLRP3 activators induce inflammasome activation via the ROS-TXNIP pathway, this mechanism cannot count for all scenarios in ROS-mediated NLRP3-inflammasome activation. Moreover, the role of ROS in NLRP3 inflammasome activation has been challenged. While many cellular stresses produce ROS, not all activate the NLRP3 inflammasome. In addition, ROS inhibitors at high dosages have been shown to be involved in the signal-1, but not signal-2 pathway in mouse macrophages (Bauernfeind et al. 2011), suggesting that the conclusion of ROS resulting from chemical inhibitors should be made with caution. Thus, the detailed mechanism by which ROS is involved in NLRP3 inflammasome activation should be further verified.

ROS are composed of various species, including hydroxyl radical (HO), hydroxide ion (HO), triplet oxygen (O22•), superoxide anion (O2•−), peroxide ion (O22−), hydrogen peroxide (H2O2), peroxynitrite (ONOO), and nitric oxide (NO). There exists evidence of the involvement of specific species of ROS on NLRP3 inflammasome activation. For example, direct H2O2 treatment is sufficient to trigger NLRP3 inflammasome activation and IL-1B secretion in THP-1 cells (Zhou et al. 2010). Treatment of hydrogen, an antioxidant that can selectively scavenge HO, suppresses IL-1B production and caspase-1 activity in LPS-stimulated RAW264.7 cells (Ren et al. 2016). A peroxynitrite ONOO scavenger 5,10,15,20-Tetrakis(4-sulfonatophenyl) porphyrinato iron (III) chloride (FeTPPS) attenuates nigericin-induced caspase-1 activation and IL-1B secretion in human monocytes (Hewinson et al. 2008). Scavenging of O2•− by TEMPOL (4-hydroxy-2,2,6,6-tetramethylpo-peridine 1-oxyl) inhibits homocysteine-induced NLRP3 inflammasome activation and IL-1B production in mouse podocytes (Abais et al. 2014). Therefore, these results suggest that various oxidant species can activate the NLRP3 inflammasome. More studies using species-specific scavenger or gene knockout mice would be required to dissect various ROS species' contributions to NLRP3 inflammasome activation.

The source of ROS generated during NLRP3 inflammasome activation remains debated. ROS can be derived from mitochondria, NADPH oxidases (NOX), or xanthine oxidase. Cytosolic ROS are previously thought to be one of the common triggers in NLRP3 inflammasome activation (Dostert et al. 2008). However, attenuation of cytosolic ROS generation by loss of NADPH oxidase activity retains normal activation of NLRP3 inflammasome in mouse peritoneal macrophages and bone marrow-derived macrophages (BMDMs) (Hornung et al. 2008, Bulua et al. 2011). These studies argue the role of cytosolic ROS in NLRP3 inflammasome activation. Another source of ROS is derived from xanthine oxidase and has been linked to the activation of NLRP3 inflammasome. For example, a xanthine oxidase inhibitor allopurinol abrogates TLR7/8 activation-induced ROS production, caspase-1 and IL-1B processing in THP-1 cells and in mice (Nicholas et al. 2011). Treatment of vascular smooth muscle cells and THP-1 cells with xanthine oxidase activates NLRP3 and results in form cell formation in concert with ROS generation (Dai et al. 2017).

Mitochondria are another significant source of cellular ROS via the mitochondrial respiratory chain. Lately, mitochondrial ROS (mtROS), generated through dysfunctional mitochondria during cellular stress, are thought to be the predominant ROS in the activation of the NLRP3 inflammasome (Xue et al. 2017). The study in THP-1 cells using mitochondrial complex inhibitors for mtROS production was initially proposed to be the link between mitochondrial dysfunction and NLRP3 inflammasome activation (Zhou et al. 2011). Interestingly, mtROS are critical for NLRP3 inflammasome activation in one study using BMDMs (Gross et al. 2016) but appear dispensable in another study using the same cells (Munoz-Planillo et al. 2013). The involvement of mitochondrial perturbation and associated mtROS production remains debated for NLRP3 inflammasome activation.

In addition to mtROS, other materials released from dysfunctional mitochondria are potent triggers for NLRP3 inflammasome activation. For example, mitochondrial DNA released into the cytosol has been shown to interact and activate NLRP3 inflammasome in BMDMs (Shimada et al. 2012). Cardiolipin, a specific lipid within the mitochondrial inner membrane, is flipped to the outer membrane and interacts with the LRR of NLRP3 in mouse macrophage J774A.1 cells (Iyer et al. 2013). The mitochondrial antiviral signalling protein (MAVS), an outer mitochondrial membrane-associated protein, mediates NLRP3 mitochondrial recruitment through the direct NLRP3–MAVS interaction. MAVS is required not only for NLRP3 localisation to the mitochondria but also for NLRP3 oligomerisation in response to mtROS in BMDMs (Subramanian et al. 2013, Buskiewicz et al. 2016). Thus, perturbation of mitochondria is pivotal for NLRP3 inflammasome activation for more than the generation of mtROS.

Metabolic DAMPs in NLRP3 inflammasome activation

Several metabolic DAMPs directly activate the NLRP3 inflammasome. First, ATP and serum amyloid A (SAA) can stimulate P2X7 channels and promote K+ efflux to induce activation of NLRP3 inflammasome (Niemi et al. 2011, Gustin et al. 2015). Second, crystals and particulate structures (e.g. uric acid and cholesterol) lead to lysosomal dysfunction, destabilising lysosomes and releasing the protease cathepsin B. NLRP3 then senses the leakage of cathepsin B to induce inflammasome activation (Hornung et al. 2008). Third, mitochondrial dysfunction and mtROS release into the cytosol are critical events in NLRP3 activation. Obesity-associated metabolic DAMPs (e.g. palmitate and ceramide) damage mitochondria and increase mtROS production, which activates the NLRP3 inflammasome (Sharma & Kanneganti 2021). Additionally, other metabolic DAMPs (e.g. amylin and glucose) stimulate NLRP3 to assemble the inflammasome complex in multiple pathways involving the production of ROS (Masters et al. 2010, Zhou et al. 2010). In the following section, we discuss metabolic DAMPs that can potentially serve as great sources and potent inducers of activation of the NLRP3 inflammasome.

Adenosine triphosphate

ATP serves as both an energy and a signalling molecule. Extracellular ATP (at concentrations of ~0.05–1 mM) activates the purinergic receptor P2X7, resulting in plasma membrane depolarisation, rapid influx of Na+ and Ca2+, and efflux of K+ and other cations (Surprenant et al. 1996). K+ efflux is a key step in NLRP3 activation. Thus, blockade with high extracellular K+ or P2X7 inhibition suppresses ATP-induced IL-1B and caspase-1 release in mouse macrophage J774A.1 cells. These results suggest a mechanism whereby P2X7 activation triggered by ATP regulates K+ efflux and subsequent activation of NLRP3 inflammasome (Yaron et al. 2015). A decrease in intracellular K+ concentration facilitates a structural change in NLRP3 during its activation in BMDMs. A unique linker sequence and a short FISNA (fish-specific NACHT associated) domain located between PYD and NBD domains of NLRP3 are important for its sensing to decrease intracellular K+ (Tapia-Abellan et al. 2021), and this regulation is mediated through the essential mediator NIMA-related kinase-7 (NEK7) (He et al. 2016).

Serum amyloid A

SAA is an acute-phase serum protein produced by hepatocytes and adipose tissues. SAA induces the secretion of IL-1B from macrophages, dendritic cells, and neutrophils by activating the NLRP3 inflammasome and caspase-1 (Niemi et al. 2011, Shridas et al. 2018). However, different mechanisms are involved in the SAA-induced activation of the NLRP3 inflammasome. For example, oxidised ATP and KN-62, both P2X7 inhibitors, significantly reduced SAA-induced IL-1B release from human macrophages, suggesting that P2X7 signalling is involved in SAA-mediated inflammasome activation (Niemi et al. 2011). Moreover, SAA induces the secretion of caspase-1, ASC, and the active form of cathepsin B, and these processes have been detected after stimulation with P2X7 (Niemi et al. 2011).

Although SAA-induced activation of NLRP3 inflammasome is mediated through cathepsin B, it does not necessarily involve phagocytosis of SAA and lysosomal destabilisation (Niemi et al. 2011). Thus, SAA-induced NLRP3 inflammasome activation occurs via a unique P2X7–cathepsin B-dependent mechanism. However, SAA-induced inflammasome activation has also been reported to occur through a P2X7-independent mechanism. In mouse J774.2 macrophage-like cells, specific P2X7 receptor antagonists (AZ10606120 and A438079) failed to reduce SAA-induced IL-1B production. Despite different P2X7 dependencies, SAA mediates the activation of NLRP3 inflammasome via a mechanism involving cathepsin B activity in macrophage-like cells (Shridas et al. 2018).

Serum SAA level is elevated in many chronic inflammatory diseases, including obesity, type 2 diabetes, rheumatic diseases, and atherosclerosis, where IL-1B is predominant (Shridas et al. 2018). Overexpression of SAA increases atherosclerosis through increased inflammatory cell infiltration in ApoE−/− mice (Dong et al. 2011). In addition to activating the NLRP3 inflammasome, SAA alters vascular function, affects HDL function, and increases thrombosis in mice (Shridas et al. 2018). Although there is evidence that increased SAA levels are associated with cardiovascular diseases, it is not yet clear how SAA initiates and develops atherosclerosis through NLRP3 inflammasome activation.

Uric acid

Uric acid is a by-product of purine metabolism. MSU, the crystal form of uric acid, requires phagocytosis for uptake. Crystalline and particulate activators of the inflammasome cause destabilisation of the lysosomal compartment and release of cathepsin B. NLRP3 then senses the leakage of cathepsin B to trigger activation of the inflammasome (Jin & Flavell 2010). This causes the activation of caspase-1 and the release of mature IL-1B in human peripheral blood mononuclear cells (Hornung et al. 2008). Cathepsin B deficiency inhibits ASC oligomerisation and the formation of the NLRP3 inflammasome complex induced by MSU in BMDMs. These findings illustrate the importance of the lysosome-cathepsin B pathway in NLRP3-mediated ASC oligomerisation, caspase-1 cleavage, and IL-1B maturation (Chevriaux et al. 2020). Moreover, the co-localisation of NLRP3 and cathepsin B at the endoplasmic reticulum has been demonstrated in BMDMs treated with MSU (Chevriaux et al. 2020), suggesting that cathepsin B needs to be localised outside of the lysosome to contact NLRP3 at its inactive location in the endoplasmic reticulum. Moreover, the lysosome–cathepsin B pathway has been shown to activate the ROS pathway (Chevriaux et al. 2020). Hence, MSU-induced NLRP3 inflammasome complex formation involves the lysosome/cathepsin B pathway, with some contribution by ROS.

IL-1B, a key regulatory pro-inflammatory cytokine in gout, promotes neutrophil influx into the synovium and joint fluid, the pathological hallmark of gout attack. In the pathogenesis of gout, MSU uptake by macrophages activates the NLRP3 inflammasome, increasing IL-1B levels, recruiting neutrophils, and subsequently causing tissue damage in the joint (Martinon et al. 2006). NLRP3 inflammasome inhibitors and IL-1B inhibitors, such as anakinra (an IL-1R antagonist), have been tested in clinical trials to treat acute and chronic gout patients (Yang et al. 2022).

Cholesterol

Cholesterol, a type of lipid, is an essential structural component of cell membranes. Cholesterol crystals are solid, crystalline forms of cholesterol found in gallstones and atherosclerotic plaques. It has been reported that cholesterol crystals can induce activation of the NLRP3 inflammasome via signal-1 and signal-2 pathways. In the signal-1 pathway, treatment with cholesterol crystals exacerbates lipopolysaccharide (LPS)-induced expression of inflammasome-related genes in primary human macrophages (Rajamaki et al. 2010, Baldrighi et al. 2017). In the signal-2 pathway, cholesterol crystals destabilise lysosomes and cause leakage of cathepsin B into the cytoplasm, resulting in activation of the NLRP3 inflammasome in THP-1 cells (Rajamaki et al. 2010). The enrichment of cholesterol crystals in lysosomes can directly alter the content of the cholesterol membrane, leading to lysosomal instability in THP-1 cells (Thacker et al. 2016). Therefore, the formation of cholesterol crystals in LPS-primed murine macrophages induces lysosomal damage and subsequent NLRP3 inflammasome activation, resulting in the production of IL-1B and IL-18 (Duewell et al. 2010). Moreover, cholesterol crystals can enhance NLRP3 inflammasome activation by increasing the expression of CD36 in BMDMs. CD36 is responsible for the uptake of oxidised (ox)LDL into macrophages, leading to further augmentation of intracellular cholesterol crystallisation and activation of the NLRP3 inflammasome (Sheedy et al. 2013). These results highlight the dual role of cholesterol crystals in the priming of NLRP3 and activation of the NLRP3 inflammasome.

Cholesterol crystal deposition in the artery has been a hallmark of the early atherosclerotic lesion. Cholesterol crystals activate the NLRP3 inflammasome and consequent IL-1B production in minimally modified low-density lipoprotein (mmLDL)-primed murine macrophages (Duewell et al. 2010). Increased IL-1B levels promote neutrophil infiltration in atherosclerotic lesions and atherosclerosis progression (Duewell et al. 2010).

Palmitate

Obesity-associated danger signals, such as palmitate and ceramide, induce activation of the NLRP3 inflammasome in a ROS-dependent manner. Saturated fatty acid palmitate results in NLRP3 inflammasome activation and IL-1B release in a mtROS-dependent manner in BMDMs. The AMPK activator reduces palmitate-induced mtROS generation, suggesting that AMPK negatively controls mtROS levels induced by palmitate treatment. However, palmitate inactivates AMPK, leading to defective autophagy and increased mtROS, presumably due to a deficiency in the clearance of dysfunctional mitochondria. Thus, palmitate causes activation of the NLRP3 inflammasome and release of active IL-1B via an AMPK–autophagy–mtROS pathway (Wen et al. 2011).

In the development of insulin resistance induced by a high-fat diet, accumulation of saturated fatty acids, such as palmitate, induces NLRP3 inflammasome activation, causing maturation of caspase-1 and production of IL-1B and IL-18 in BMDMs (Wen et al. 2011). NLRP3 inflammasome activation in macrophages further impairs insulin signalling in target tissues, reducing insulin sensitivity and glucose tolerance. In non-alcoholic steatohepatitis (NASH), palmitate-induced apoptosis causes the release of danger signals from mouse hepatocytes, which in turn activate the NLRP3 inflammasome and increase IL-1B levels in Kupffer cells (Csak et al. 2011).

Ceramide

In lipid metabolism, palmitate enters the non-oxidative pathway and is converted into ceramide via irreversible condensation with l-serine. Ceramides and other sphingolipids are structural components of membranes and signalling molecules that regulate cell homeostasis (Gault et al. 2010). In BMDMs, ceramide induces mitochondrial oxidative stress to drive NLRP3 and caspase-1 activation (Camell et al. 2015). Aberrant ceramide accumulation is found in obese rodents and in insulin-resistance human subjects (Adams et al. 2004, Bijl et al. 2008). Thus, in the progression of obesity, lipotoxicity-associated ceramide activates the NLRP3 inflammasome to cleave caspase-1 in mouse macrophages and adipose tissues (Vandanmagsar et al. 2011). NLRP3 deletion in mice attenuates obesity-induced inflammation and enhances insulin signalling in adipose tissues and liver (Vandanmagsar et al. 2011).

Amylin/islet amyloid polypeptide

Amylin (also known as islet amyloid polypeptide (IAPP)), a hormone secreted by β-cells, forms amyloid deposits in the pancreas during the development of type 2 diabetes. Phagocytosis of amylin by BMDMs results in ROS generation, triggering NLRP3 inflammasome activation and mature IL-1B release, which requires priming with mmLDL (Masters et al. 2010). The IL-1B released by the infiltrated macrophages causes pancreatic β-cell death in vitro and in vivo (Dinarello et al. 2010). Along with the ROS-mediated mechanism, amylin has also been shown in BMDMs to prime the NLRP3 inflammasome for IL-1B production through MyD88 (Westwell-Roper et al. 2011). Additionally, amylin directly binds to the LRR domain of NLRP3 in β-cells to contribute to inflammation and β-cell death in pancreatic islets of patients with type 2 diabetes (Morikawa et al. 2018). Thus, amylin-mediated NLRP3 inflammasome activation and pancreatic β-cell death in type 2 diabetes involve ROS-sensitive and ROS-independent mechanisms. In the pathogenesis of type 2 diabetes, amylin activates the NLRP3 inflammasome and produces IL-1B in pancreatic dendritic cells and macrophages; increased IL-1B levels further promote β-cell death (Masters et al. 2010).

Glucose

Glucose is the most critical energy source in living cells. High glucose levels have been shown to induce ROS generation (Zhou et al. 2010), suggesting that hyperglycaemia causes activation of the NLRP3 inflammasome through oxidative stress. Moreover, the ROS inhibitor (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate (APDC) attenuates the activation of the NLRP3 inflammasome and secretion of IL-1B induced by glucose (Zhou et al. 2010). After a glucose-induced increase in ROS levels, TXNIP is dissociated from oxidised thioredoxin and subsequently binds to NLRP3 (Zhou et al. 2010). Additionally, glucose-induced ROS production is sufficient to act as signal-2 to promote NLRP3 inflammasome activation for processing caspase-1 and IL-1B in mouse pancreatic islets (Feng et al. 2016).

In pancreatic islet failure during type 2 diabetes, glucose induces ROS production, which triggers TXNIP-thioredoxin dissociation and NLRP3 inflammasome activation in β-cells. Together with the previously addressed effect of amylin on NLRP3 inflammasome activation in BMDMs, which is independent of TXNIP (Masters et al. 2010), IL-1B secreted from both sources accelerates β-cell mass loss and promotes further immune cell infiltration, leading to pancreatic islet failure and progression to diabetes.

In conclusion, activation of the NLRP3 inflammasome and subsequent maturation and secretion of its downstream product (IL-1B) are involved in the interplay between macrophages and their local target cells, thereby participating in the pathogenesis of several metabolic diseases. Thus, identification of the unknown metabolic DAMPs and dissection of the novel mechanism should ultimately help design therapeutic targets for metabolic diseases associated with activating the NLRP3 inflammasome.

Endogenous ‘pedal and brake’ for NLRP3 inflammasome activation

Each of the three domains of NLRP3 (PYD, NBD, and LRR) has a unique feature in regulating inflammasome activation. For example, PYD forms self-association or homotypic interactions (PYD–PYD) with other members of this family (Abderrazak et al. 2015). NBD, associated with NLRP3 self-oligomerisation, is a domain involved with nucleotides, ATP binding, and hydrolysis (Lechtenberg et al. 2014). The LRR domain acts as a self-inhibitory domain that curves back to block NBD (Lechtenberg et al. 2014) and a ligand-detecting domain that breaks down the self-inhibitory interaction between NBD and LRR to promote inflammasome activation (Latz 2010).

Because NLRP3 serves as a converging point in recognising many danger signals, numerous NLRP3 regulating proteins have been discovered over the last few years. For example, pyrin-only proteins interact with ASC to block inflammasome formation (Bedoya et al. 2007), whereas CARD-only proteins interact with caspase-1 to reduce caspase-1 activity during inflammasome activation (Latz et al. 2013). Therefore, protein–protein interactions are crucial and potential regulators of NLRP3 inflammasome activation. Here, we demonstrated that several proteins regulate NLRP3 inflammasome activation via domain–domain interactions, including some regarded as ‘a pedal to accelerate’ and some as ‘a brake to slow down’ inflammasome activation (Fig. 4).

Figure 4
Figure 4

NLRP3 binding partners and domain interaction. NLRP3 is composed of three domains, LRR, NBD, and PYD. NLRP3 inflammasome activation is mediated by various proteins with different domains. GBP5 activates the NLRP3 inflammasome by binding to PYD, MARK4 by binding to NBD and PYD, HSP90 to LRR, and TXNIP and NEK7 to LRR and NBD. GNB1 and SHP inhibit the NLRP3 inflammasome by binding to PYD, and PPARG by binding to LRR and NBD. The inhibitory domain of NLRP3 mediated by HSP70 remains unclear. Metabolic DAMPs and related metabolic diseases associated with NLRP3 binding partners are shown.

Citation: Journal of Endocrinology 257, 2; 10.1530/JOE-22-0184

Guanylate-binding protein 5

Guanylate-binding protein 5 (GBP5) belongs to the GTPase family induced by LPS and interferon-gamma and is a key contributor to protective immunity against microbial and viral pathogens. GBP5 is critical in NLRP3 inflammasome activation induced by ATP, MSU, and pathogenic bacteria, but not particulate (Shenoy et al. 2012). GBP5 promotes bacteria-induced activation of the NLRP3 inflammasome through binding to the PYD of NLRP3. GBP5 deficiency attenuated LPS-induced caspase-1 cleavage and release of IL-1B and IL-18 in BMDMs and impaired host defence (Shenoy et al. 2012). However, another study shows contradictory results in unprimed BMDMs (Man et al. 2015). Although GBP5 is an activator for the NLRP3-ASC oligomerisation complex, further clarification is required. One recent study showed a significantly increased expression of NEK7 and GBP5 in a diabetic mouse heart, suggesting the involvement of the NLRP3-NEK7-GBP5 pathway in the diabetic heart. Their study suggests that inflammation-induced cardiac cell pyroptosis mediated by GBP5 is the key player in the development and progression of diabetic cardiomyopathy (Elmadbouh & Singla 2021).

Microtubule affinity-regulating kinase 4

Microtubule affinity-regulating kinase 4 (MARK4) belongs to the serine/threonine kinase family that phosphorylates microtubule-associated proteins to bring about their microtubule detachment. Interestingly, MARK4 interacts with the PYD and NBD domains of NLRP3 to induce activation of NLRP3 inflammasome and release of IL-1B. Mark4−/− BMDMs reduced IL-1B production, specifically after activation by NLRP3 stimuli. Moreover, MARK4-deficient cells exhibited significantly reduced NLRP3–ASC association, suggesting that MARK4 is involved in NLRP3 complex formation. Furthermore, the MARK4 catalytic kinase and NLRP3 PYD–NBD domains are essential for this interaction. IL-1B production and neutrophil accumulation in the peritoneal lavage were markedly reduced in Mark4−/− mice challenged with MSU. Overall, MARK4 is required for NLRP3-mediated IL-1B production and inflammasome activation. A recent study showed that MARK4-dependent NLRP3 inflammasome activation in haematopoietic cells regulates atherosclerosis development (Clement et al. 2019). Moreover, circulating IL-1B, IL-18, NLRP3 inflammasome, and MARK4 expression are increased in diabetic patients and rats. High glucose mediated NLRP3 inflammasome activation via upregulation of MARK4 in human umbilical vein endothelial cells (Wang et al. 2020).

Heat shock protein 90

Heat shock protein 90 (HSP90), a molecular chaperone that facilitates protein folding, is required to protect NLRP3 from proteasome and autophagic degradations (Mayor et al. 2007, Piippo et al. 2018). HSP90 forms a complex with NLRP3 and retains it in a competent form to receive the activation signal. The interaction between the LRR domain of NLRP3 and HSP90 requires another protein suppressor of the G2 allele of SKP1 (SGT1) in THP-1 cells (Mayor et al. 2007). Treatment with the HSP90 inhibitor geldanamycin attenuated caspase-1 activity and IL-1B release in a human retinal pigment epithelial cell line exposed to MG-132 + BafA, suggesting that HSP90 inhibition attenuates inflammasome activation (Piippo et al. 2018). Hence, a mechanism involving SGT1, HSP90, and NLRP3 regulates innate immune responses.

Thioredoxin-interacting protein

TXNIP is a metabolic protein involved in redox regulation. TXNIP acts as a redox sensor by binding to thioredoxin under reducing conditions. ROS dissociates the complex and releases TXNIP to bind to the NBD and LRR of NLRP3 to promote activation of the NLRP3 inflammasome. TXNIP ablation reduces the secretion of active caspase-1 and IL-1B from the LPS-primed BMDM cells stimulated with various NLRP3 inflammasome activators (e.g. MSU, alum, silica, and ATP) (Zhou et al. 2010). However, another study shows a dispensable role of TXNIP in NLRP3 inflammasome activation induced by ATP, MSU, and amylin in BMDMs (Masters et al. 2010). Txnip−/− mice showed attenuated responses to MSU-induced neutrophil influx and IL-1B production (Zhou et al. 2010). Moreover, TXNIP deficiency protects the mice from diet-induced insulin resistance and type 2 diabetes (Alhawiti et al. 2017). Thus, TXNIP is a critical promoter of ROS-mediated activation of NLRP3 inflammasome and plays an essential role in regulating insulin resistance and pancreatic islet failure.

NIMA-related kinase-7

NIMA-related kinase-7 (NEK7) is a serine/threonine kinase that controls the cell cycle. Three groups independently identified NEK7 as a critical regulator for NLRP3 inflammasome activation (He et al. 2016, Schmid-Burgk et al. 2016, Shi et al. 2016). NEK7 is a core component specific to NLRP3 but not NLRC4 and AIM2 inflammasomes. Activation of the NLRP3 inflammasome in BMDMs requires binding the catalytic domain of NEK7 to the NBD and LRR domain of NLRP3, which is independent of NEK7 kinase activity (He et al. 2016). This binding is necessary for NLRP3–ASC complex formation, ASC oligomerisation, and caspase-1 cleavage in BMDMs (Shi et al. 2016). NEK7 is required for NLRP3 inflammasome activation that is induced by ATP, nigericin, MSU, and particulate in BMDMs and mouse peritoneal macrophages (He et al. 2016, Shi et al. 2016). Moreover, NEK7 acts as a downstream target of K+ efflux in regulating NLRP3 activation in BMDMs (He et al. 2016). Thus, K+ efflux promotes the interaction between NLRP3 and NEK7, essential for forming ASC speck and cleavage of caspase-1. The process examined in HEK293T cells is mediated via NEK7 interaction with the NBD and LRR domains of NLRP3 (He et al. 2016). Thus, NEK7 is essential for assembling and activating the NLRP3 inflammasome, particularly under K+ efflux conditions. In addition to the involvement of the NLRP3-NEK7 pathway in the diabetic mouse heart (Elmadbouh & Singla 2021), one recent study showed that protein levels of NEK7 and the NLRP3 inflammasome components, including NLRP3, ASC, and caspase-1, are significantly increased in the vasculature of patients with diabetic foot. Increased serum levels of IL-1B and IL-18, an indication of chronic inflammatory status, accompany this. These results suggest that NEK7 might mediate NLRP3 inflammasome activation in the pathogenesis of diabetic lower extremity arterial disease (Cai et al. 2020).

G protein subunit B1

G protein subunit B1 (GNB1) is a component of heterotrimeric G proteins, with mutations in GNB1 causing severe neurodevelopmental disabilities, hypotonia, and seizures (Petrovski et al. 2016). Although G protein-coupled receptors are associated with NLRP3 inflammasome activation (Rossol et al. 2012), whether they act through GNB1 to regulate the NLRP3 inflammasome remains to be clarified. GNB1 negatively regulates NLRP3 inflammasome activation by interacting with the PYD of NLRP3 and suppressing ASC oligomerisation. GNB1 knockdown in BMDMs did not affect the expression of NLRP3 and pro-IL-1B induced following LPS treatment but significantly enhanced caspase-1 activation and IL-1B release following ATP treatment. These results suggest that GNB1 is a negative regulator of NLRP3 inflammasome activation (Murakami et al. 2019).

Small heterodimer partner

Small heterodimer partner (SHP), an orphan nuclear receptor, regulates glucose and cholesterol metabolism. SHP has been shown to influence its target genes directly or indirectly (Zhang et al. 2011). Interestingly, SHP interacted with the PYD of NLRP3 to inhibit the NLRP3–ASC interaction, leading to decreased IL-1B and IL-18 secretion in BMDMs. SHP is involved in protection mechanisms against inflammation-related disease progression in several mouse models, including MSU-mediated peritonitis and acute renal tubular injury. SHP acts as a negative regulator for NLRP3 inflammasome activation (Yang et al. 2015).

Peroxisome proliferator-activated receptor gamma

Peroxisome proliferator-activated receptor gamma (PPARG), a ligand-activated nuclear receptor, plays a major role in the transcriptional regulation of energy metabolism by promoting adipocyte differentiation and insulin-sensitising potential. PPARG is known for its anti-inflammatory potential, mediated through NF-kB transrepression and the subsequent inhibitory effect on the expression of inflammatory cytokines. We have recently shown an additional anti-inflammatory role of PPARG in suppressing NLRP3 inflammasome activation, which is mediated through binding to NLRP3. PPARG inhibits NLRP3 inflammasome assembly by damping interactions between NLRP3–ASC and NLRP3–NLRP3 and NLRP3-dependent ASC oligomerisation. The interaction between the NBD and LRR domains of NLRP3 and the DNA-binding domain of PPARG mediates this inhibitory effect. Our study further found that PPARγ is required to limit NLRP3 inflammasome activation induced by metabolic DAMP, such as palmitate, in mouse macrophages. There exists a negative correlation between PPARγ and mature caspase-1 in circulating mononuclear cells of obese patients. Thus, PPARγ agonism may be a therapeutic option for targeting-NLRP3-related metabolic diseases (Yang et al. 2021).

Heat shock protein 70

Heat shock protein 70 (HSP70), another molecular chaperone, protects cells from the adverse effects of physiological stresses. Extracellular HSP70 functions as a DAMP to stimulate immune and inflammatory responses through TLRs, TLR2 and TLR4, resulting in sterile inflammation and amplification of current inflammation (Hulina et al. 2018). In contrast, intracellular HSP70 has been reported to bind to NLRP3 and downregulate NLRP3 inflammasome activation. However, the domains mediating the interaction between NLRP3 and HSP70 remain to be identified. Moreover, LPS is sufficient to induce ASC/NLRP3 speck formation in Hsp70−/− BMDMs but not in wild-type control cells (Martine et al. 2019).

Therapeutic targeting NLRP3 for metabolic diseases

Metabolic diseases are increasingly recognised as having an inflammatory component that contributes significantly to the disease progression. This is particularly important when aberrant NLRP3 inflammasome activation is predominantly involved in the pathogenesis of metabolic diseases. Several available strategies targeting NLRP3 inflammasome product, IL-1B, have been applied in treating metabolic diseases. For example, the IL-1 receptor antagonist anakinra, IL-1B neutralising antibody canakinumab, and the decoy IL-1 receptor rilonacept have beneficial effects on gout and type 2 diabetes (Larsen et al. 2007, Cavelti-Weder et al. 2011). It is promising to see the effects of such treatments on other inflammasome-mediated metabolic diseases, such as atherosclerosis, insulin resistance, and NASH. Thus, the clinical indications of IL-1 blockade on metabolic diseases are continuously expanding. It is worth mentioning that the involvement of inflammation likely varies between metabolic diseases. Certain metabolic diseases with a more prominent role of NLRP3-mediated IL-1B may benefit more from NLRP3 targeting therapy. Thus, targeting NLRP3-mediated IL-1B may only be effective in some metabolic diseases involving NLRP3 predominantly.

In addition to blocking the outcomes of inflammasome activation, therapies directly targeting NLRP3 or inflammasome components are of interest. Several lines of small molecules directly targeting NLRP3 have been discovered. For example, the diarylsulfonylurea compound MCC950 has therapeutic efficacy against several preclinical immunopathological models, including atherosclerosis (van der Heijden et al. 2017), type 2 diabetes (Zhai et al. 2018), and NASH (Mridha et al. 2017). In addition, CY-09, an analogue of C172 used for cystic fibrosis (Jiang et al. 2017), tranilast, an analogue of a tryptophan metabolite, used for allergy, asthma, and hypertrophic scars (Darakhshan & Pour 2015), and oridonin, the main ingredient of the herbal medicine used for inflammatory diseases (Ma et al. 2011), exhibit therapeutic potential in mouse models of type 2 diabetes. Since these inhibitors are initially recognised for treating various inflammatory diseases, their applications in metabolic diseases require more exploration.

Glyburide, the first compound identified to inhibit NLRP3 inflammasome, is a second-line antidiabetic medication that stimulates insulin secretion in pancreatic β-cells (Lamkanfi et al. 2009). Although the effect of glyburide on the inhibition of NLRP3 inflammasome activation in vitro is potent, its effect in animals requires a high dosage (Mangan et al. 2018). While its mechanism of action remains unknown, it is likely to function downstream of the P2X7 receptor and upstream of NLRP3. Moreover, several P2X7 inhibitors and caspase-1 inhibiting pro-drugs have been discovered (Swanson et al. 2019). However, their effects on metabolic diseases await future studies.

Anti-inflammatory effect of metabolic drugs

In addition to targeting inflammation, NLRP3, and inflammasome components, several metabolic drugs show promising effects on attenuating NLRP3 inflammasome activation. Interestingly, the anti-inflammatory effect is frequently seen when treating these metabolic drugs, suggesting a close link between metabolic danger signals and inflammatory conditions. Thus, the anti-inflammatory effect of these metabolic drugs could result from resolving the accumulated metabolic DAMPs, such as glucose, palmitate, and cholesterol, by treatment.

Metformin, a first-line antidiabetic medication, exerts anti-inflammatory effects (Bulcao et al. 2007). For example, metformin treatment attenuated the upregulation of NLRP3 inflammasome activation in circulating macrophages of type 2 diabetic patients (Lee et al. 2013). Metformin also reverses the decreased expression of thioredoxin in the aorta of ApoE−/− mice (Tang et al. 2019). Thus, the beneficial effects of metformin on the amelioration of both inflammation and metabolic disorder could involve an NLRP3-based mechanism.

Sodium-glucose cotransporter-2 inhibitors (SGLT2i) reduce circulating glucose levels in type 2 diabetic patients by increasing glucose excretion via inhibiting renal SGLT2 reabsorption. Treatment with SGLT2i dapagliflozin reduced IL-1B secretion in the macrophages of type 2 diabetic patients and NLRP3 inflammasome activation in activated B cells (Kim et al. 2020). Another SGLT2i empagliflozin decreased NLRP3 inflammasome activation and downstream inflammatory responses in the diabetic mouse kidney (Benetti et al. 2016). Thus, SGLT2i appears to exert anti-inflammatory effects by inhibiting NLRP3 inflammasome activation.

Dipeptidyl peptidase-4 inhibitors (DPP4i) exhibit glucose-lowering effects by inhibiting glucagon release and increasing insulin secretion. Treatment with DPP4i saxagliptin attenuated renal NLRP3 activity and injury in diabetic mice (Birnbaum et al. 2016). Resveratrol, a plant polyphenol found in red grapes, is known to have anti-inflammatory effects and exhibits glucose-lowering effects in clinal trials (Berman et al. 2017, Ozturk et al. 2017). Treatment with resveratrol attenuated TXNIP-mediated NLRP3 inflammasome activation in diabetic rats subjected to acute kidney injury and in human kidney proximal tubular HK-2 cells in high glucose condition (30 mM) subjected to hypoxia/reoxygenation (Xiao et al. 2016). Therefore, DPP4i and resveratrol both benefit by involving an NLRP3-related mechanism.

PPARγ agonists, an insulin sensitiser mentioned in the previous section, are well known for its anti-inflammatory effect. The anti-inflammatory role of PPARγ is initially thought to mediate through the transrepression of NF-κB and subsequent inhibition of cytokine transcription. Our group discovered an additional anti-inflammatory role for PPARγ that specifically targets NLRP3 inflammasome activation (Yang et al. 2021). In addition to the reduction of metabolic DAMPs (glucose and lipid), we also revealed a protein (NLRP3)-protein (PPARγ) interaction via the target protein of these metabolic drugs (Yang et al. 2021). Thus, the effects of metabolic drugs on the cure of metabolic diseases associated with the reduction in inflammatory status likely involve more than one mechanism. This highlights the crosstalk between metabolism and inflammation. Thus, inhibition of NLRP3 inflammasome activation can be achieved by improving metabolic pathways via various metabolic drugs.

Main gaps and recent novel development

In the past few years, inhibitors targeting IL-1B are increasing rapidly. Several of them are currently used in the clinical treatment of multiple inflammatory diseases. Several similar agents, such as neutralizing and blocking antibodies, are currently under development. However, several points should be noted. As IL-1B is not the only product of inflammasome activation, the involvement of other secretory mediators, such as IL-18 and HMGB1, in metabolic diseases cannot be neglected. Further, IL-1B can be produced from NLRP3 inflammasome-independent pathways or through the activation of other inflammasomes. Lastly, inhibitors targeting IL-1B can have detrimental effects on immune defence, and should be applied cautiously in the context of metabolic diseases.

Lately, a number of small molecules directly targeting NLRP3 or inflammasome components or its related signaling have been developed. As the inhibitory mechanisms or precise targets remain to be defined, most of these inhibitors raise concerns regarding off-target effects. Therefore, their potency for in vivo usage requires further evaluation. Although some of these inhibitors are being used in clinical practice, some have been suspended at phase II clinical trials because of hepatic toxicity (MacKenzie et al. 2010, Mangan et al. 2018). Therefore, the strategies for targeting NLRP3 inflammasomes as a therapeutic target for metabolic diseases need to be refined.

The application of metabolic drugs for inhibiting NLRP3 inflammasome activation is promising. Most of these metabolic drugs are being used in clinical practice or are being investigated in clinical trials with relatively high safety. In the past few years, the development of inhibitors targeting IL-1B and NLRP3 as therapeutics for multiple diseases has undergone major advances. The main limitations in developing NLRP3-related metabolic drugs are time-consumption and cost-effectiveness. Therefore, instead of repurposing of NLRP3 inhibitors for metabolic diseases, it may be more practical and feasible to repurpose metabolic drugs for targeting the NLRP3 inflammasome.

Conclusions and perspectives

Improving metabolic pathways has been associated with alleviating NLRP3 inflammasome activation, which might be related to a reduction in particular nutrients or metabolites acting as metabolic DAMPs to activate the inflammasome. This has received much attention in studying TLRs that sense various nutrients or metabolites in addition to the canonical activator PAMPs. Aberrant accumulation of these nutrients or metabolites in metabolic diseases indeed are endogenous DAMPs that are shown to be direct activators of the NLRP3 inflammasome, mainly involved in the initiation and progression of these diseases. Therefore, more nutrients or metabolites would be candidates for the accelerators of NLRP3 inflammasome activation.

Although not all of the abovementioned protein binding partners for NLRP3 have been directly linked to metabolic diseases, most are identified with inflammation. Because of their characteristics in physical contact with NLRP3, the involvement of these binding partners in metabolic diseases warrants future investigations. Based on the concept of the domain-domain interaction and the unique feature of the binding domain, other proteins with similar domain features may be candidates for searching for more NLRP3 binding partners. Moreover, small molecules targeting NLRP3 with known inhibitory domains on NLRP3 can be applied to stop the pedal action of binding partners listed in Fig. 4. Finally, as illustrated in Fig. 4, metabolic DAMPs may affect NLRP3 inflammasome activation via these binding partners. Attention can be focused on the crosstalk between metabolic DAMPs and protein binding partners.

In recent years, immuno-metabolism research has gained considerable attention due to the interplay between inflammatory responses and metabolism. Several metabolites, danger signals, enzymes, and nuclear receptors essential for energy sensing or metabolism have been implicated in the modulation of inflammatory responses. Therefore, it is necessary to dissect the link between metabolic factors and inflammation and to identify novel metabolic danger signals. This review highlights the induction by metabolic danger signals and regulation by protein binding partners of the NLRP3 inflammasome as potential therapeutic targets for metabolic diseases. Understanding how, what, and when metabolic factors modulate NLRP3 inflammasome activation could provide novel therapeutic approaches through the cautious manipulation of these metabolic factors.

Declaration of interest

The authors have declared that no competing interest exists.

Funding

This work was supported by grants from the Ministry of Science and Technology, Taiwan (MOST-107-2320-B-006-063-MY3, MOST-110-2320-B-006-017-MY3, and MOST-111-2320-B-006-022-MY3), National Health Research Institutes (NHRI-EX107-10511SI), National Cheng Kung University Hospital (NCKUH-10909039), and National Cheng Kung University and E-DA Hospital Research Project.

Author contribution statement

Sy-Ying Leu, Ching-Chun Yang, Li-Chun Ho, Ai-Ning Shao, and Chia-Yu Chang wrote the first draft; Yi-Ling Tsang drew the graphical illustration; Hui-Kuan Lin, Pei-Jane Tsai, and Junne-Ming Sung provided suggestions and discussion; Yau-Sheng Tsai made critical review of the manuscript; All authors read and approved the final manuscript.

Acknowledgements

The authors thank Editage for language editing.

References

  • Abais JM, Xia M, Li G, Gehr TW, Boini KM & & Li PL 2014 Contribution of endogenously produced reactive oxygen species to the activation of podocyte NLRP3 inflammasomes in hyperhomocysteinemia. Free Radical Biology and Medicine 67 211220. (https://doi.org/10.1016/j.freeradbiomed.2013.10.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Abderrazak A, Syrovets T, Couchie D, El Hadri K, Friguet B, Simmet T & & Rouis M 2015 NLRP3 inflammasome: from a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases. Redox Biology 4 296307. (https://doi.org/10.1016/j.redox.2015.01.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Adams JM 2nd, Pratipanawatr T, Berria R, Wang E, DeFronzo RA, Sullards MC & & Mandarino LJ 2004 Ceramide content is increased in skeletal muscle from obese insulin-resistant humans. Diabetes 53 2531. (https://doi.org/10.2337/diabetes.53.1.25)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Alhawiti NM, Al Mahri S, Aziz MA, Malik SS & & Mohammad S 2017 TXNIP in metabolic regulation: physiological role and therapeutic outlook. Current Drug Targets 18 10951103. (https://doi.org/10.2174/1389450118666170130145514)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baldrighi M, Mallat Z & & Li X 2017 NLRP3 inflammasome pathways in atherosclerosis. Atherosclerosis 267 127138. (https://doi.org/10.1016/j.atherosclerosis.2017.10.027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bauernfeind F, Bartok E, Rieger A, Franchi L, Nunez G & & Hornung V 2011, Cutting edge: reactive oxygen species inhibitors block priming, but not activation, of the NLRP3 inflammasome. Journal of Immunology 187 613617. (https://doi.org/10.4049/jimmunol.1100613)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bedoya F, Sandler LL & & Harton JA 2007 Pyrin-only protein 2 modulates NF-kappaB and disrupts ASC:CLR interactions. Journal of Immunology 178 38373845. (https://doi.org/10.4049/jimmunol.178.6.3837)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Benetti E, Mastrocola R, Vitarelli G, Cutrin JC, Nigro D, Chiazza F, Mayoux E, Collino M & & Fantozzi R 2016 Empagliflozin protects against diet-induced NLRP-3 inflammasome activation and lipid accumulation. Journal of Pharmacology and Experimental Therapeutics 359 4553. (https://doi.org/10.1124/jpet.116.235069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Berman AY, Motechin RA, Wiesenfeld MY & & Holz MK 2017 The therapeutic potential of resveratrol: a review of clinical trials. npj Precision Oncology 1 35. (https://doi.org/10.1038/s41698-017-0038-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bijl N, Scheij S, Houten S, Boot RG, Groen AK & & Aerts JM 2008, The glucosylceramide synthase inhibitor N-(5-adamantane-1-yl-methoxy-pentyl)-deoxynojirimycin induces sterol regulatory element-binding protein-regulated gene expression and cholesterol synthesis in HepG2 cells. Journal of Pharmacology and Experimental Therapeutics 326 849855. (https://doi.org/10.1124/jpet.108.139394)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Birnbaum Y, Bajaj M, Qian J & & Ye Y 2016 Dipeptidyl peptidase-4 inhibition by saxagliptin prevents inflammation and renal injury by targeting the Nlrp3/ASC inflammasome. BMJ Open Diabetes Research and Care 4 e000227. (https://doi.org/10.1136/bmjdrc-2016-000227)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Broz P & & Dixit VM 2016 Inflammasomes: mechanism of assembly, regulation, and signalling. Nature Reviews. Immunology 16 407420. (https://doi.org/10.1038/nri.2016.58)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulcao C, Ribeiro-Filho, FF, Sanudo A & & Roberta Ferreira SG, 2007 Effects of simvastatin and metformin on inflammation and insulin resistance in individuals with mild metabolic syndrome. American Journal of Cardiovascular Drugs 7 219224. (https://doi.org/10.2165/00129784-200707030-00007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulua AC, Simon A, Maddipati R, Pelletier M, Park H, Kim KY, Sack MN, Kastner DL & & Siegel RM 2011 Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). Journal of Experimental Medicine 208 519533. (https://doi.org/10.1084/jem.20102049)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buskiewicz IA, Montgomery T, Yasewicz EC, Huber SA, Murphy MP, Hartley RC, Kelly R, Crow MK, Perl A, Budd RC, et al.2016 Reactive oxygen species induced virus-independent MAVS oligomerization in systemic lupus erythematosus. Science Signaling 9 ra115. (https://doi.org/10.1126/scisignal.aaf1933)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cai H, Wang P, Zhang B & & Dong X 2020 Expression of the NEK7/NLRP3 inflammasome pathway in patients with diabetic lower extremity arterial disease. BMJ Open Diabetes Research and Care 8 e001808. (https://doi.org/10.1136/bmjdrc-2020-001808)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Camell CD, Nguyen KY, Jurczak MJ, Christian BE, Shulman GI, Shadel GS & & Dixit VD 2015 Macrophage-specific de novo synthesis of ceramide is dispensable for inflammasome-driven inflammation and insulin resistance in obesity. Journal of Biological Chemistry 290 2940229413. (https://doi.org/10.1074/jbc.M115.680199)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carlsson F, Kim J, Dumitru C, Barck KH, Carano RA, Sun M, Diehl L & & Brown EJ 2010 Host-detrimental role of Esx-1-mediated inflammasome activation in mycobacterial infection. PLOS Pathogen 6 e1000895. (https://doi.org/10.1371/journal.ppat.1000895)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cavelti-Weder C, Furrer R, Keller C, Babians-Brunner A, Solinger AM, Gast H, Fontana A, Donath MY & & Penner IK 2011 Inhibition of IL-1beta improves fatigue in type 2 diabetes. Diabetes Care 34 e158. (https://doi.org/10.2337/dc11-1196)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chevriaux A, Pilot T, Derangere V, Simonin H, Martine P, Chalmin F, Ghiringhelli F & & Rebe C 2020 Cathepsin B is required for NLRP3 inflammasome activation in macrophages, through NLRP3 interaction. Frontiers in Cell and Developmental Biology 8 167. (https://doi.org/10.3389/fcell.2020.00167)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chooi YC, Ding C & & Magkos F 2019, The epidemiology of obesity. Metabolism: Clinical and Experimental 92 610. (https://doi.org/10.1016/j.metabol.2018.09.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clement M, Chen X, Chenoweth HL, Teng Z, Thome S, Newland SA, Harrison J, Yu X, Finigan AJ, Mallat Z, et al.2019 MARK4 (microtubule affinity-regulating kinase 4)-dependent inflammasome activation promotes atherosclerosis brief report. Arteriosclerosis, Thrombosis, and Vascular Biology 39 16451651.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Csak T, Ganz M, Pespisa J, Kodys K, Dolganiuc A & & Szabo G 2011 Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells. Hepatology 54 133144. (https://doi.org/10.1002/hep.24341)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dai Y, Cao Y, Zhang Z, Vallurupalli S & & Mehta JL 2017 Xanthine oxidase induces foam cell formation through LOX-1 and NLRP3 activation. Cardiovascular Drugs and Therapy 31 1927. (https://doi.org/10.1007/s10557-016-6706-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Darakhshan S & & Pour AB 2015 Tranilast: a review of its therapeutic applications. Pharmacological Research 91 1528. (https://doi.org/10.1016/j.phrs.2014.10.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dinarello CA, Donath MY & & Mandrup-Poulsen T, 2010 Role of IL-1beta in type 2 diabetes. Current Opinion in Endocrinology, Diabetes, and Obesity 17 314321. (https://doi.org/10.1097/MED.0b013e32833bf6dc)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dong Z, Wu T, Qin W, An C, Wang Z, Zhang M, Zhang Y, Zhang C & & An F 2011 Serum amyloid A directly accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice. Molecular Medicine 17 13571364. (https://doi.org/10.2119/molmed.2011.00186)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT & & Tschopp J 2008 Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 320 674677. (https://doi.org/10.1126/science.1156995)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nunez G, Schnurr M, et al.2010 NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464 13571361. (https://doi.org/10.1038/nature08938)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Elmadbouh I & & Singla DK 2021 BMP-7 attenuates inflammation-induced pyroptosis and improves cardiac repair in diabetic cardiomyopathy. Cells 10 2640. (https://doi.org/10.3390/cells10102640)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Feng H, Gu J, Gou F, Huang W, Gao C, Chen G, Long Y, Zhou X, Yang M, Liu S, et al.2016 High glucose and lipopolysaccharide prime NLRP3 inflammasome via ROS/TXNIP pathway in mesangial cells. Journal of Diabetes Research 2016 6973175. (https://doi.org/10.1155/2016/6973175)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaidt MM, Ebert TS, Chauhan D, Schmidt T, Schmid-Burgk JL, Rapino F, Robertson AA, Cooper MA, Graf T & & Hornung V 2016 Human monocytes engage an alternative inflammasome pathway. Immunity 44 833846. (https://doi.org/10.1016/j.immuni.2016.01.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gault CR, Obeid LM & & Hannun YA 2010, An overview of sphingolipid metabolism: from synthesis to breakdown. Advances in Experimental Medicine and Biology 688 123. (https://doi.org/10.1007/978-1-4419-6741-1_1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gross CJ, Mishra R, Schneider KS, Medard G, Wettmarshausen J, Dittlein DC, Shi H, Gorka O, Koenig PA, Fromm S, et al.2016 k K+ Efflux-Independent NLRP3 Inflammasome Activation by Small Molecules Targeting Mitochondria. Immunity 45 761773. (https://doi.org/10.1016/j.immuni.2016.08.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gustin A, Kirchmeyer M, Koncina E, Felten P, Losciuto S, Heurtaux T, Tardivel A, Heuschling P & & Dostert C 2015 NLRP3 inflammasome is expressed and functional in mouse brain microglia but not in astrocytes. PLoS One 10 e0130624. (https://doi.org/10.1371/journal.pone.0130624)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • He Y, Franchi L & & Nunez G 2013 TLR agonists stimulate Nlrp3-dependent IL-1beta production independently of the purinergic P2X7 receptor in dendritic cells and in vivo. Journal of Immunology 190 334339. (https://doi.org/10.4049/jimmunol.1202737)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • He Y, Zeng MY, Yang D, Motro B & & Nunez G 2016 NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature 530 354357. (https://doi.org/10.1038/nature16959)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hewinson J, Moore SF, Glover C, Watts AG & & MacKenzie AB, 2008 A key role for redox signaling in rapid P2X7 receptor-induced IL-1 beta processing in human monocytes. Journal of Immunology 180 84108420. (https://doi.org/10.4049/jimmunol.180.12.8410)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA & & Latz E 2008 Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nature Immunology 9 847856. (https://doi.org/10.1038/ni.1631)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hulina A, Grdic Rajkovic M, Jakšić Despot D, Jelić D, Dojder A, Čepelak I & & Rumora L 2018 Extracellular Hsp70 induces inflammation and modulates LPS/LTA-stimulated inflammatory response in THP-1 cells. Cell Stress and Chaperones 23 373384. (https://doi.org/10.1007/s12192-017-0847-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hung J, McQuillan BM, Chapman CM, Thompson PL & & Beilby JP 2005, Elevated interleukin-18 levels are associated with the metabolic syndrome independent of obesity and insulin resistance. Arteriosclerosis, Thrombosis, and Vascular Biology 25 12681273. (https://doi.org/10.1161/01.ATV.0000163843.70369.12)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, Sadler JJ, Knepper-Adrian V, Han R, Qiao L, et al.2013 Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity 39 311323. (https://doi.org/10.1016/j.immuni.2013.08.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang H, He H, Chen Y, Huang W, Cheng J, Ye J, Wang A, Tao J, Wang C, Liu Q, et al.2017 Identification of a selective and direct NLRP3 inhibitor to treat inflammatory disorders. Journal of Experimental Medicine 214 32193238. (https://doi.org/10.1084/jem.20171419)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jin C & & Flavell RA 2010 Molecular mechanism of NLRP3 inflammasome activation. Journal of Clinical Immunology 30 628631. (https://doi.org/10.1007/s10875-010-9440-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim SR, Lee SG, Kim SH, Kim JH, Choi E, Cho W, Rim JH, Hwang I, Lee CJ, Lee M, et al.2020 SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nature Communications 11 2127. (https://doi.org/10.1038/s41467-020-15983-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lamkanfi M, Mueller JL, Vitari AC, Misaghi S, Fedorova A, Deshayes K, Lee WP, Hoffman HM & & Dixit VM 2009 Glyburide inhibits the cryopyrin/Nalp3 inflammasome. Journal of Cell Biology 187 6170. (https://doi.org/10.1083/jcb.200903124)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, Mandrup-Poulsen T & & Donath MY 2007 Interleukin-1-receptor antagonist in type 2 diabetes mellitus. New England Journal of Medicine 356 15171526. (https://doi.org/10.1056/NEJMoa065213)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Latz E 2010 inflammasomes: mechanisms of activation and function. Current Opinion in Immunology 22 2833. (https://doi.org/10.1016/j.coi.2009.12.004)

  • Latz E, Xiao TS & & Stutz A 2013 Activation and regulation of the inflammasomes. Nature Reviews. Immunology 13 397411. (https://doi.org/10.1038/nri3452)

  • Lechtenberg BC, Mace PD & & Riedl SJ 2014 Structural mechanisms in NLR inflammasome signaling. Current Opinion in Structural Biology 29 1725. (https://doi.org/10.1016/j.sbi.2014.08.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee HM, Kim JJ, Kim HJ, Shong M, Ku BJ & & Jo EK 2013 Upregulated NLRP3 inflammasome activation in patients with type 2 diabetes. Diabetes 62 194204. (https://doi.org/10.2337/db12-0420)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin X, Xu Y, Pan X, Xu J, Ding Y, Sun X, Song X, Ren Y & & Shan PF 2020 Global, regional, and national burden and trend of diabetes in 195 countries and territories: an analysis from 1990 to 2025. Scientific Reports 10 14790. (https://doi.org/10.1038/s41598-020-71908-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ma Z, Hu C & & Zhang Y 2011 Therapeutic effect of Rabdosia rubescens aqueous extract on chronic pharyngitis and its safety. Zhong Nan Da Xue Xue Bao Yi Xue Ban 36 170173. (https://doi.org/10.3969/j.issn.1672-7347.2011.02.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • MacKenzie SH, Schipper JL & & Clark AC 2010 The potential for caspases in drug discovery. Current Opinion in Drug Discovery and Development 13 568576.

  • Man SM, Karki R, Malireddi RK, Neale G, Vogel P, Yamamoto M, Lamkanfi M & & Kanneganti TD 2015 The transcription factor IRF1 and guanylate-binding proteins target activation of the AIM2 inflammasome by Francisella infection. Nature Immunology 16 467475. (https://doi.org/10.1038/ni.3118)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mangan MSJ, Olhava EJ, Roush WR, Seidel HM, Glick GD & & Latz E 2018 Targeting the NLRP3 inflammasome in inflammatory diseases. Nature Reviews. Drug Discovery 17 688. (https://doi.org/10.1038/nrd.2018.149)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martine P, Chevriaux A, Derangere V, Apetoh L, Garrido C, Ghiringhelli F & & Rebe C 2019 HSP70 is a negative regulator of NLRP3 inflammasome activation. Cell Death and Disease 10 256. (https://doi.org/10.1038/s41419-019-1491-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martinon F, Petrilli V, Mayor A, Tardivel A & & Tschopp J 2006 Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440 237241. (https://doi.org/10.1038/nature04516)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Masters SL, Dunne A, Subramanian SL, Hull RL, Tannahill GM, Sharp FA, Becker C, Franchi L, Yoshihara E, Chen Z, et al.2010 Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1beta in type 2 diabetes. Nature Immunology 11 897904. (https://doi.org/10.1038/ni.1935)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mayor A, Martinon F, De Smedt T, Petrilli V & & Tschopp J 2007 A crucial function of SGT1 and HSP90 in inflammasome activity linking mammalian and plant innate immune responses. Nature Immunology 8 497503. (https://doi.org/10.1038/ni1459)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Morikawa S, Kaneko N, Okumura C, Taguchi H, Kurata M, Yamamoto T, Osawa H, Nakanishi A, Zako T & & Masumoto J 2018 IAPP/amylin deposition, which is correlated with expressions of ASC and IL-1beta in beta-cells of Langerhans' islets, directly initiates NLRP3 inflammasome activation. International Journal of Immunopathology and Pharmacology 32 2058738418788749. (https://doi.org/10.1177/2058738418788749)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mridha AR, Wree A, Robertson AAB, Yeh MM, Johnson CD, Van Rooyen DM, Haczeyni F, Teoh NC, Savard C, Ioannou GN, et al.2017 NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. Journal of Hepatology 66 10371046. (https://doi.org/10.1016/j.jhep.2017.01.022)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Munoz-Planillo R, Kuffa P, Martinez-Colon G, Smith BL, Rajendiran TM & & Nunez G 2013 K⁺ efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity 38 11421153. (https://doi.org/10.1016/j.immuni.2013.05.016)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Murakami T, Ruengsinpinya L, Nakamura E, Takahata Y, Hata K, Okae H, Taniguchi S, Takahashi M & & Nishimura R 2019 Cutting Edge: G protein subunit beta 1 negatively regulates NLRP3 inflammasome activation. Journal of Immunology 202 19421947. (https://doi.org/10.4049/jimmunol.1801388)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Netea MG, Nold-Petry CA, Nold MF, Joosten LA, Opitz B, van der Meer JH, van de Veerdonk FL, Ferwerda G, Heinhuis B, Devesa I, et al.2009 Differential requirement for the activation of the inflammasome for processing and release of IL-1beta in monocytes and macrophages. Blood 113 23242335. (https://doi.org/10.1182/blood-2008-03-146720)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nicholas SA, Bubnov VV, Yasinska IM & & Sumbayev VV 2011 Involvement of xanthine oxidase and hypoxia-inducible factor 1 in toll-like receptor 7/8-mediated activation of caspase 1 and interleukin-1beta. Cellular and Molecular Life Sciences 68 151158. (https://doi.org/10.1007/s00018-010-0450-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Niemi K, Teirila L, Lappalainen J, Rajamaki K, Baumann MH, Oorni K, Wolff H, Kovanen PT, Matikainen S & & Eklund KK 2011 Serum amyloid A activates the NLRP3 inflammasome via P2X7 receptor and a cathepsin B-sensitive pathway. Journal of Immunology 186 61196128. (https://doi.org/10.4049/jimmunol.1002843)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ozturk E, Arslan AKK, Yerer MB & & Bishayee A 2017 Resveratrol and diabetes: a critical review of clinical studies. Biomedicine and Pharmacotherapy 95 230234. (https://doi.org/10.1016/j.biopha.2017.08.070)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Petrovski S, Kury S, Myers CT, Anyane-Yeboa K, Cogne B, Bialer M, Xia F, Hemati P, Riviello J, Mehaffey M, et al.2016 Germline de novo mutations in GNB1 cause severe neurodevelopmental disability, hypotonia, and seizures. American Journal of Human Genetics 98 10011010. (https://doi.org/10.1016/j.ajhg.2016.03.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Piippo N, Korhonen E, Hytti M, Skottman H, Kinnunen K, Josifovska N, Petrovski G, Kaarniranta K & & Kauppinen A 2018 Hsp90 inhibition as a means to inhibit activation of the NLRP3 inflammasome. Scientific Reports 8 6720. (https://doi.org/10.1038/s41598-018-25123-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rajamaki K, Lappalainen J, Oorni K, Valimaki E, Matikainen S, Kovanen PT & & Eklund KK 2010 Cholesterol crystals activate the NLRP3 inflammasome in human macrophages: a novel link between cholesterol metabolism and inflammation. PLoS One 5 e11765. (https://doi.org/10.1371/journal.pone.0011765)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ren JD, Wu XB, Jiang R, Hao DP & & Liu Y 2016 Molecular hydrogen inhibits lipopolysaccharide-triggered NLRP3 inflammasome activation in macrophages by targeting the mitochondrial reactive oxygen species. Biochimica et Biophysica Acta, 1863 5055. (https://doi.org/10.1016/j.bbamcr.2015.10.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rossol M, Pierer M, Raulien N, Quandt D, Meusch U, Rothe K, Schubert K, Schoneberg T, Schaefer M, Krugel U, et al.2012 Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G protein-coupled calcium sensing receptors. Nature Communications 3 1329. (https://doi.org/10.1038/ncomms2339)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schmid-Burgk JL, Chauhan D, Schmidt T, Ebert TS, Reinhardt J, Endl E & & Hornung V 2016 A genome-wide CRISPR (clustered regularly interspaced short palindromic repeats) screen identifies NEK7 as an essential component of NLRP3 inflammasome activation. Journal of Biological Chemistry 291 103109. (https://doi.org/10.1074/jbc.C115.700492)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sharma BR & & Kanneganti TD 2021 NLRP3 inflammasome in cancer and metabolic diseases. Nature Immunology 22 550559. (https://doi.org/10.1038/s41590-021-00886-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sheedy FJ, Grebe A, Rayner KJ, Kalantari P, Ramkhelawon B, Carpenter SB, Becker CE, Ediriweera HN, Mullick AE, Golenbock DT, et al.2013 CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nature Immunology 14 812820. (https://doi.org/10.1038/ni.2639)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shenoy AR, Wellington DA, Kumar P, Kassa H, Booth CJ, Cresswell P & & MacMicking JD 2012 GBP5 promotes NLRP3 inflammasome assembly and immunity in mammals. Science 336 481485. (https://doi.org/10.1126/science.1217141)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shi H, Wang Y, Li X, Zhan X, Tang M, Fina M, Su L, Pratt D, Bu CH, Hildebrand S, et al.2016 NLRP3 activation and mitosis are mutually exclusive events coordinated by NEK7, a new inflammasome component. Nature Immunology 17 250258. (https://doi.org/10.1038/ni.3333)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L, Ojcius DM, et al.2012 Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 36 401414. (https://doi.org/10.1016/j.immuni.2012.01.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shridas P, De Beer MC & & Webb NR 2018 High-density lipoprotein inhibits serum amyloid A-mediated reactive oxygen species generation and NLRP3 inflammasome activation. Journal of Biological Chemistry 293 1325713269. (https://doi.org/10.1074/jbc.RA118.002428)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Subramanian N, Natarajan K, Clatworthy MR, Wang Z & & Germain RN 2013, The adaptor MAVS promotes NLRP3 mitochondrial localization and inflammasome activation. Cell 153 348361. (https://doi.org/10.1016/j.cell.2013.02.054)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Surprenant A, Rassendren F, Kawashima E, North RA & & Buell G 1996 The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7) Science 272 735738. (https://doi.org/10.1126/science.272.5262.735)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Swanson KV, Deng M & & Ting JP 2019 The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nature Reviews. Immunology 19 477489. (https://doi.org/10.1038/s41577-019-0165-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tang G, Duan F, Li W, Wang Y, Zeng C, Hu J, Li H, Zhang X, Chen Y & & Tan H 2019 Metformin inhibited Nod-like receptor protein 3 inflammasome activation and suppressed diabetes-accelerated atherosclerosis in apoE(-/-) mice. Biomedicine and Pharmacotherapy 119 109410. (https://doi.org/10.1016/j.biopha.2019.109410)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tapia-Abellan A, Angosto-Bazarra D, Alarcon-Vila C, Banos MC, Hafner-Bratkovic I, Oliva B & & Pelegrin P 2021 Sensing low intracellular potassium by NLRP3 results in a stable open structure that promotes inflammasome activation. Science Advances 7 eabf4468. (https://doi.org/10.1126/sciadv.abf4468)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Thacker SG, Zarzour A, Chen Y, Alcicek MS, Freeman LA, Sviridov DO, Demosky SJ & & Remaley AT 2016 High-density lipoprotein reduces inflammation from cholesterol crystals by inhibiting inflammasome activation. Immunology 149 306319. (https://doi.org/10.1111/imm.12638)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • van der Heijden T, Kritikou E, Venema W, van Duijn J, van Santbrink PJ, Slutter B, Foks AC, Bot I & & Kuiper J 2017 NLRP3 inflammasome inhibition by MCC950 reduces atherosclerotic lesion development in apolipoprotein E-deficient mice-brief report. Arteriosclerosis, Thrombosis, and Vascular Biology 37 14571461. (https://doi.org/10.1161/ATVBAHA.117.309575)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, Ravussin E, Stephens JM & & Dixit VD 2011 The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nature Medicine 17 179188. (https://doi.org/10.1038/nm.2279)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang J, Shen X, Liu J, Chen W, Wu F, Wu W, Meng Z, Zhu M & & Miao C 2020 High glucose mediated NLRP3 inflammasome activation via upregulation of ELF3 expression. Cell Death and Disease 11 383. (https://doi.org/10.1038/s41419-020-2598-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wen H, Gris D, Lei Y, Jha S, Zhang L, Huang MT, Brickey WJ & & Ting JP 2011 Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nature Immunology 12 408415. (https://doi.org/10.1038/ni.2022)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Westwell-Roper C, Dai DL, Soukhatcheva G, Potter KJ, van Rooijen N, Ehses JA & & Verchere CB 2011 IL-1 blockade attenuates islet amyloid polypeptide-induced proinflammatory cytokine release and pancreatic islet graft dysfunction. Journal of Immunology 187 27552765. (https://doi.org/10.4049/jimmunol.1002854)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xiao YD, Huang YY, Wang HX, Wu Y, Leng Y, Liu M, Sun Q & & Xia ZY 2016 Thioredoxin-interacting protein mediates NLRP3 inflammasome activation involved in the susceptibility to ischemic acute kidney injury in diabetes. Oxidative Medicine and Cellular Longevity 2016 2386068. (https://doi.org/10.1155/2016/2386068)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xue Y, Du M & & Zhu MJ 2017 Quercetin suppresses NLRP3 inflammasome activation in epithelial cells triggered by Escherichia coli O157:H7. Free Radical Biology and Medicine 108 760769. (https://doi.org/10.1016/j.freeradbiomed.2017.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang CS, Kim JJ, Kim TS, Lee PY, Kim SY, Lee HM, Shin DM, Nguyen LT, Lee MS, Jin HS, et al.2015 Small heterodimer partner interacts with NLRP3 and negatively regulates activation of the NLRP3 inflammasome. Nature Communications 6 6115. (https://doi.org/10.1038/ncomms7115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang CC, Wu CH, Lin TC, Cheng YN, Chang CS, Lee KT, Tsai PJ & & Tsai YS 2021 Inhibitory effect of PPARgamma on NLRP3 inflammasome activation. Theranostics 11 24242441. (https://doi.org/10.7150/thno.46873)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang G, Kang HC, Cho YY, Lee HS & & Lee JY 2022 Inflammasomes and their roles in arthritic disease pathogenesis. Frontiers in Molecular Biosciences 9 1027917. (https://doi.org/10.3389/fmolb.2022.1027917)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yaron JR, Gangaraju S, Rao MY, Kong X, Zhang L, Su F, Tian Y, Glenn HL & & Meldrum DR 2015 K(+) regulates Ca(2+) to drive inflammasome signaling: dynamic visualization of ion flux in live cells. Cell Death and Disease 6 e1954. (https://doi.org/10.1038/cddis.2015.277)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhai Y, Meng X, Ye T, Xie W, Sun G & & Sun X 2018 Inhibiting the NLRP3 inflammasome Activation with MCC950 Ameliorates Diabetic encephalopathy in db/db Mice. Molecules 23 522. (https://doi.org/10.3390/molecules23030522)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang Y, Hagedorn CH & & Wang L 2011 Role of nuclear receptor SHP in metabolism and cancer. Biochimica et Biophysica Acta 1812 893908. (https://doi.org/10.1016/j.bbadis.2010.10.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou R, Tardivel A, Thorens B, Choi I & & Tschopp J 2010 Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nature Immunology 11 136140. (https://doi.org/10.1038/ni.1831)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou R, Yazdi AS, Menu P & & Tschopp J 2011 A role for mitochondria in NLRP3 inflammasome activation. Nature 469 221225. (https://doi.org/10.1038/nature09663)

 

  • Collapse
  • Expand
  • Figure 1

    NLRP3 inflammasome activation. NLRP3 inflammasome activation comprises a two-step process. During the priming step, a Toll-like receptor mediates the downstream NF-kB translocation to the nucleus and produces NLRP3 and pro-IL-1B. During the following activation step, NLRP3 oligomerises and recruits ASC. Subsequently, pro-IL-1B, pro-IL-18, and gasdermin D (GSDMD) are cleaved by active caspase-1 and released. The released GSDMD triggers pyroptosis.

  • Figure 2

    NLRP3 inflammasome-related metabolic diseases. Serial DAMPs activate the NLRP3 inflammasome and contribute to various diseases. Because obesity is a common precursor for NLRP3-mediated metabolic diseases, it seems to prime the NLRP3 inflammasome. In the progression of atherosclerosis, cholesterol crystals or serum amyloid A (SAA) can trigger NLRP3 inflammasome activation. Gout, an inflammatory disease that can be caused by uric acid crystal deposition, is also a potent NLRP3 activator. Amylin (IAPP) deposition and high glucose levels in diabetes are related to NLRP3 inflammasome activation. The obesity-associated factors, palmitate and ceramide, are activators of NLRP3. Pathogenesis of NASH also involves palmitate-induced release of danger signals and the activation of the NLRP3 inflammasome.

  • Figure 3

    NLRP3 inflammasome activators. P2X7 is stimulated by ATP and serum amyloid A, subsequently regulating downstream K+ efflux and resulting in NLRP3 inflammasome activation. Uric acid and cholesterol crystals destabilise the lysosome, leading to the release of cathepsin B and the activation of the NLRP3 inflammasome. ROS stress derived from dysfunctional mitochondria and cytosol, which can be stimulated by glucose and amylin, is also a potent activator of the NLRP3 inflammasome. Palmitate activates the NLRP3 inflammasome by regulating the AMPK–autophagy–ROS pathway. Ceramide initiates mitochondrial dysfunction and activates the NLRP3 inflammasome.

  • Figure 4

    NLRP3 binding partners and domain interaction. NLRP3 is composed of three domains, LRR, NBD, and PYD. NLRP3 inflammasome activation is mediated by various proteins with different domains. GBP5 activates the NLRP3 inflammasome by binding to PYD, MARK4 by binding to NBD and PYD, HSP90 to LRR, and TXNIP and NEK7 to LRR and NBD. GNB1 and SHP inhibit the NLRP3 inflammasome by binding to PYD, and PPARG by binding to LRR and NBD. The inhibitory domain of NLRP3 mediated by HSP70 remains unclear. Metabolic DAMPs and related metabolic diseases associated with NLRP3 binding partners are shown.

  • Abais JM, Xia M, Li G, Gehr TW, Boini KM & & Li PL 2014 Contribution of endogenously produced reactive oxygen species to the activation of podocyte NLRP3 inflammasomes in hyperhomocysteinemia. Free Radical Biology and Medicine 67 211220. (https://doi.org/10.1016/j.freeradbiomed.2013.10.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Abderrazak A, Syrovets T, Couchie D, El Hadri K, Friguet B, Simmet T & & Rouis M 2015 NLRP3 inflammasome: from a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases. Redox Biology 4 296307. (https://doi.org/10.1016/j.redox.2015.01.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Adams JM 2nd, Pratipanawatr T, Berria R, Wang E, DeFronzo RA, Sullards MC & & Mandarino LJ 2004 Ceramide content is increased in skeletal muscle from obese insulin-resistant humans. Diabetes 53 2531. (https://doi.org/10.2337/diabetes.53.1.25)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Alhawiti NM, Al Mahri S, Aziz MA, Malik SS & & Mohammad S 2017 TXNIP in metabolic regulation: physiological role and therapeutic outlook. Current Drug Targets 18 10951103. (https://doi.org/10.2174/1389450118666170130145514)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baldrighi M, Mallat Z & & Li X 2017 NLRP3 inflammasome pathways in atherosclerosis. Atherosclerosis 267 127138. (https://doi.org/10.1016/j.atherosclerosis.2017.10.027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bauernfeind F, Bartok E, Rieger A, Franchi L, Nunez G & & Hornung V 2011, Cutting edge: reactive oxygen species inhibitors block priming, but not activation, of the NLRP3 inflammasome. Journal of Immunology 187 613617. (https://doi.org/10.4049/jimmunol.1100613)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bedoya F, Sandler LL & & Harton JA 2007 Pyrin-only protein 2 modulates NF-kappaB and disrupts ASC:CLR interactions. Journal of Immunology 178 38373845. (https://doi.org/10.4049/jimmunol.178.6.3837)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Benetti E, Mastrocola R, Vitarelli G, Cutrin JC, Nigro D, Chiazza F, Mayoux E, Collino M & & Fantozzi R 2016 Empagliflozin protects against diet-induced NLRP-3 inflammasome activation and lipid accumulation. Journal of Pharmacology and Experimental Therapeutics 359 4553. (https://doi.org/10.1124/jpet.116.235069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Berman AY, Motechin RA, Wiesenfeld MY & & Holz MK 2017 The therapeutic potential of resveratrol: a review of clinical trials. npj Precision Oncology 1 35. (https://doi.org/10.1038/s41698-017-0038-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bijl N, Scheij S, Houten S, Boot RG, Groen AK & & Aerts JM 2008, The glucosylceramide synthase inhibitor N-(5-adamantane-1-yl-methoxy-pentyl)-deoxynojirimycin induces sterol regulatory element-binding protein-regulated gene expression and cholesterol synthesis in HepG2 cells. Journal of Pharmacology and Experimental Therapeutics 326 849855. (https://doi.org/10.1124/jpet.108.139394)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Birnbaum Y, Bajaj M, Qian J & & Ye Y 2016 Dipeptidyl peptidase-4 inhibition by saxagliptin prevents inflammation and renal injury by targeting the Nlrp3/ASC inflammasome. BMJ Open Diabetes Research and Care 4 e000227. (https://doi.org/10.1136/bmjdrc-2016-000227)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Broz P & & Dixit VM 2016 Inflammasomes: mechanism of assembly, regulation, and signalling. Nature Reviews. Immunology 16 407420. (https://doi.org/10.1038/nri.2016.58)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulcao C, Ribeiro-Filho, FF, Sanudo A & & Roberta Ferreira SG, 2007 Effects of simvastatin and metformin on inflammation and insulin resistance in individuals with mild metabolic syndrome. American Journal of Cardiovascular Drugs 7 219224. (https://doi.org/10.2165/00129784-200707030-00007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bulua AC, Simon A, Maddipati R, Pelletier M, Park H, Kim KY, Sack MN, Kastner DL & & Siegel RM 2011 Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). Journal of Experimental Medicine 208 519533. (https://doi.org/10.1084/jem.20102049)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buskiewicz IA, Montgomery T, Yasewicz EC, Huber SA, Murphy MP, Hartley RC, Kelly R, Crow MK, Perl A, Budd RC, et al.2016 Reactive oxygen species induced virus-independent MAVS oligomerization in systemic lupus erythematosus. Science Signaling 9 ra115. (https://doi.org/10.1126/scisignal.aaf1933)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cai H, Wang P, Zhang B & & Dong X 2020 Expression of the NEK7/NLRP3 inflammasome pathway in patients with diabetic lower extremity arterial disease. BMJ Open Diabetes Research and Care 8 e001808. (https://doi.org/10.1136/bmjdrc-2020-001808)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Camell CD, Nguyen KY, Jurczak MJ, Christian BE, Shulman GI, Shadel GS & & Dixit VD 2015 Macrophage-specific de novo synthesis of ceramide is dispensable for inflammasome-driven inflammation and insulin resistance in obesity. Journal of Biological Chemistry 290 2940229413. (https://doi.org/10.1074/jbc.M115.680199)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carlsson F, Kim J, Dumitru C, Barck KH, Carano RA, Sun M, Diehl L & & Brown EJ 2010 Host-detrimental role of Esx-1-mediated inflammasome activation in mycobacterial infection. PLOS Pathogen 6 e1000895. (https://doi.org/10.1371/journal.ppat.1000895)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cavelti-Weder C, Furrer R, Keller C, Babians-Brunner A, Solinger AM, Gast H, Fontana A, Donath MY & & Penner IK 2011 Inhibition of IL-1beta improves fatigue in type 2 diabetes. Diabetes Care 34 e158. (https://doi.org/10.2337/dc11-1196)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chevriaux A, Pilot T, Derangere V, Simonin H, Martine P, Chalmin F, Ghiringhelli F & & Rebe C 2020 Cathepsin B is required for NLRP3 inflammasome activation in macrophages, through NLRP3 interaction. Frontiers in Cell and Developmental Biology 8 167. (https://doi.org/10.3389/fcell.2020.00167)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chooi YC, Ding C & & Magkos F 2019, The epidemiology of obesity. Metabolism: Clinical and Experimental 92 610. (https://doi.org/10.1016/j.metabol.2018.09.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clement M, Chen X, Chenoweth HL, Teng Z, Thome S, Newland SA, Harrison J, Yu X, Finigan AJ, Mallat Z, et al.2019 MARK4 (microtubule affinity-regulating kinase 4)-dependent inflammasome activation promotes atherosclerosis brief report. Arteriosclerosis, Thrombosis, and Vascular Biology 39 16451651.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Csak T, Ganz M, Pespisa J, Kodys K, Dolganiuc A & & Szabo G 2011 Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells. Hepatology 54 133144. (https://doi.org/10.1002/hep.24341)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dai Y, Cao Y, Zhang Z, Vallurupalli S & & Mehta JL 2017 Xanthine oxidase induces foam cell formation through LOX-1 and NLRP3 activation. Cardiovascular Drugs and Therapy 31 1927. (https://doi.org/10.1007/s10557-016-6706-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Darakhshan S & & Pour AB 2015 Tranilast: a review of its therapeutic applications. Pharmacological Research 91 1528. (https://doi.org/10.1016/j.phrs.2014.10.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dinarello CA, Donath MY & & Mandrup-Poulsen T, 2010 Role of IL-1beta in type 2 diabetes. Current Opinion in Endocrinology, Diabetes, and Obesity 17 314321. (https://doi.org/10.1097/MED.0b013e32833bf6dc)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dong Z, Wu T, Qin W, An C, Wang Z, Zhang M, Zhang Y, Zhang C & & An F 2011 Serum amyloid A directly accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice. Molecular Medicine 17 13571364. (https://doi.org/10.2119/molmed.2011.00186)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT & & Tschopp J 2008 Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 320 674677. (https://doi.org/10.1126/science.1156995)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nunez G, Schnurr M, et al.2010 NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464 13571361. (https://doi.org/10.1038/nature08938)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Elmadbouh I & & Singla DK 2021 BMP-7 attenuates inflammation-induced pyroptosis and improves cardiac repair in diabetic cardiomyopathy. Cells 10 2640. (https://doi.org/10.3390/cells10102640)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Feng H, Gu J, Gou F, Huang W, Gao C, Chen G, Long Y, Zhou X, Yang M, Liu S, et al.2016 High glucose and lipopolysaccharide prime NLRP3 inflammasome via ROS/TXNIP pathway in mesangial cells. Journal of Diabetes Research 2016 6973175. (https://doi.org/10.1155/2016/6973175)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaidt MM, Ebert TS, Chauhan D, Schmidt T, Schmid-Burgk JL, Rapino F, Robertson AA, Cooper MA, Graf T & & Hornung V 2016 Human monocytes engage an alternative inflammasome pathway. Immunity 44 833846. (https://doi.org/10.1016/j.immuni.2016.01.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gault CR, Obeid LM & & Hannun YA 2010, An overview of sphingolipid metabolism: from synthesis to breakdown. Advances in Experimental Medicine and Biology 688 123. (https://doi.org/10.1007/978-1-4419-6741-1_1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gross CJ, Mishra R, Schneider KS, Medard G, Wettmarshausen J, Dittlein DC, Shi H, Gorka O, Koenig PA, Fromm S, et al.2016 k K+ Efflux-Independent NLRP3 Inflammasome Activation by Small Molecules Targeting Mitochondria. Immunity 45 761773. (https://doi.org/10.1016/j.immuni.2016.08.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gustin A, Kirchmeyer M, Koncina E, Felten P, Losciuto S, Heurtaux T, Tardivel A, Heuschling P & & Dostert C 2015 NLRP3 inflammasome is expressed and functional in mouse brain microglia but not in astrocytes. PLoS One 10 e0130624. (https://doi.org/10.1371/journal.pone.0130624)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • He Y, Franchi L & & Nunez G 2013 TLR agonists stimulate Nlrp3-dependent IL-1beta production independently of the purinergic P2X7 receptor in dendritic cells and in vivo. Journal of Immunology 190 334339. (https://doi.org/10.4049/jimmunol.1202737)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • He Y, Zeng MY, Yang D, Motro B & & Nunez G 2016 NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature 530 354357. (https://doi.org/10.1038/nature16959)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hewinson J, Moore SF, Glover C, Watts AG & & MacKenzie AB, 2008 A key role for redox signaling in rapid P2X7 receptor-induced IL-1 beta processing in human monocytes. Journal of Immunology 180 84108420. (https://doi.org/10.4049/jimmunol.180.12.8410)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA & & Latz E 2008 Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nature Immunology 9 847856. (https://doi.org/10.1038/ni.1631)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hulina A, Grdic Rajkovic M, Jakšić Despot D, Jelić D, Dojder A, Čepelak I & & Rumora L 2018 Extracellular Hsp70 induces inflammation and modulates LPS/LTA-stimulated inflammatory response in THP-1 cells. Cell Stress and Chaperones 23 373384. (https://doi.org/10.1007/s12192-017-0847-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hung J, McQuillan BM, Chapman CM, Thompson PL & & Beilby JP 2005, Elevated interleukin-18 levels are associated with the metabolic syndrome independent of obesity and insulin resistance. Arteriosclerosis, Thrombosis, and Vascular Biology 25 12681273. (https://doi.org/10.1161/01.ATV.0000163843.70369.12)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, Sadler JJ, Knepper-Adrian V, Han R, Qiao L, et al.2013 Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity 39 311323. (https://doi.org/10.1016/j.immuni.2013.08.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jiang H, He H, Chen Y, Huang W, Cheng J, Ye J, Wang A, Tao J, Wang C, Liu Q, et al.2017 Identification of a selective and direct NLRP3 inhibitor to treat inflammatory disorders. Journal of Experimental Medicine 214 32193238. (https://doi.org/10.1084/jem.20171419)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jin C & & Flavell RA 2010 Molecular mechanism of NLRP3 inflammasome activation. Journal of Clinical Immunology 30 628631. (https://doi.org/10.1007/s10875-010-9440-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim SR, Lee SG, Kim SH, Kim JH, Choi E, Cho W, Rim JH, Hwang I, Lee CJ, Lee M, et al.2020 SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nature Communications 11 2127. (https://doi.org/10.1038/s41467-020-15983-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lamkanfi M, Mueller JL, Vitari AC, Misaghi S, Fedorova A, Deshayes K, Lee WP, Hoffman HM & & Dixit VM 2009 Glyburide inhibits the cryopyrin/Nalp3 inflammasome. Journal of Cell Biology 187 6170. (https://doi.org/10.1083/jcb.200903124)

    • PubMed