Next Article in Journal
Heterologous Expression of Jatropha curcas Fatty Acyl-ACP Thioesterase A (JcFATA) and B (JcFATB) Affects Fatty Acid Accumulation and Promotes Plant Growth and Development in Arabidopsis
Next Article in Special Issue
TRPA1 Role in Inflammatory Disorders: What Is Known So Far?
Previous Article in Journal
Molecular Imaging in Nanomedical Research
Previous Article in Special Issue
CD8+ T Cell Senescence: Lights and Shadows in Viral Infections, Autoimmune Disorders and Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Inflammasomes in Glomerulonephritis

by
Paula Anton-Pampols
1,2,†,
Clara Diaz-Requena
2,†,
Laura Martinez-Valenzuela
1,2,
Francisco Gomez-Preciado
1,
Xavier Fulladosa
1,2,3,
Anna Vidal-Alabro
2,†,
Joan Torras
1,2,3,*,
Núria Lloberas
2,4,‡ and
Juliana Draibe
1,2,‡
1
Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, 08907 Barcelona, Spain
2
IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, 08907 Barcelona, Spain
3
Clinical Sciences Department, Campus de Bellvitge, Barcelona University, Hospitalet de Llobregat, 08907 Barcelona, Spain
4
Department of Physiological Sciences, Campus de Bellvitge, Barcelona University, Hospitalet de Llobregat, 08907 Barcelona, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors share seniorship.
Int. J. Mol. Sci. 2022, 23(8), 4208; https://doi.org/10.3390/ijms23084208
Submission received: 28 February 2022 / Revised: 7 April 2022 / Accepted: 8 April 2022 / Published: 11 April 2022

Abstract

:
The inflammasome is an immune multiprotein complex that activates pro-caspase 1 in response to inflammation-inducing stimuli and it leads to IL-1β and IL-18 proinflammatory cytokine production. NLRP1 and NLRP3 inflammasomes are the best characterized and they have been related to several autoimmune diseases. It is well known that the kidney expresses inflammasome genes, which can influence the development of some glomerulonephritis, such as lupus nephritis, ANCA glomerulonephritis, IgA nephropathy and anti-GBM nephropathy. Polymorphisms of these genes have also been described to play a role in autoimmune and kidney diseases. In this review, we describe the main characteristics, activation mechanisms, regulation and functions of the different inflammasomes. Moreover, we discuss the latest findings about the role of the inflammasome in several glomerulonephritis from three different points of view: in vitro, animal and human studies.

1. The Inflammasome

The immune system is composed of two arms, the innate and adaptive immunity, that are responsible for both immediate and long-term immunity to pathogen- and non-pathogen-derived antigens. Innate immunity detects infections, changes in cellular homeostasis and tissue damage, subsequently generating inflammation, tissue repair and homeostatic balance restoration [1]. These effects are promoted by the recognition of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). PAMPs and DAMPs bind to pattern recognition receptors, which include Toll-like receptors (TLRs), cytoplasmic NOD-like receptors (NLRs) and absent in melanoma 2-like receptors (AIM2) [2]. Previous studies have demonstrated the role of several members of the NLR family in the formation of inflammasomes, multiprotein complexes capable of recognizing inflammation-inducing stimuli. These complexes activate pro-caspase-1, which is responsible for the cleavage of multiple substrates, mainly the proinflammatory cytokines IL-1β and IL-18 [3]. The release of these cytokines by the inflammasome can also be carried out through an inflammatory form of programmed cell death named pyroptosis [4]. Therefore, the activation of the inflammasome develops innate immunity activity in response to tissue infection. Noninfectious stimulus can also activate the inflammasome [5]. Although inflammasomes can be activated by many members of the NLR family, this review will focus mainly on the NLRP3 inflammasome and NLRP1, NLRC4 and proteins absent in melanoma 2 (AIM2) (Figure 1a), also important in many immune diseases [6]. Furthermore, the role of these complexes in different glomerulonephritis will be reviewed.

2. NLR Family Inflammasomes

The NLR family comprises 23 human genes. Members of this family show common structural elements: C-terminal series of leucine-rich repeats (LRRs) and central nucleotide binding domains (NBD), a component of the larger NACHT domain [7,8]. Furthermore, NLR family members can be divided into different subfamilies depending on their N-terminal effector domain: caspase-activation and recruitment domain (CARD), baculovirus inhibitor of apoptosis protein repeat (BIR) or pyrin domain (PYD). The NLRP and NLRC subfamilies are the most important, the former being the best-characterized subfamily of NLRs. The NLRP subfamily members have PYD domains at their N-terminal while the NLRC proteins have one or more CARD domains [8,9,10]. NLR family members NLRP1, NLRP3 and NLRC4 have been the best studied in inflammasome formation [11].

2.1. NLRP Subfamily

The NLRP subfamily is composed of 14 members in human genome, plus 3 paralogs in mouse being NLRP1 (NALP1/CARD7) the first to be described in forming inflammasomes [12]. Its structure consists of a N-terminal PYD followed by a NACHT domain and LRRs. This is also contributed by a C-terminal extension containing a function-to-find domain (FIIND), which auto processes NLRP1 into two polypeptide chains, and a CARD domain, that leads to caspase-1 activation and the consequent proinflammatory cytokine release [13,14]. It has been reported that NLRP1 mutations can play a role in inflammatory diseases such as psoriasis [15], rheumatoid arthritis (RA) [16] or in systemic lupus erythematosus (SLE) [17].
NLRP3 inflammasome (Cryopyrin/Nalp3/Cias1/Pypaf1) is the most widely studied and is the only known member to be activated by numerous pathogenic and sterile inflammatory signals. Furthermore, NLRP3 plays a role in the regulation of IL-1β production in macrophages [18,19]. NLRP3 is composed of the NLRP3 scaffold, an adaptor apoptosis speck-like protein (ASC) and the effector procaspase-1. It interacts with ASC via PYD-PYD homotypic interactions to promote the formation of the inflammasome by recruiting and activating procaspase-1 to generate active caspase-1 (Figure 1b). This effector protein leads the conversion of the cytokine precursors pro-IL-1β and pro-IL-18 into mature and biologically active IL-1β and IL-18 [9,20]. The main attention given to the NLRP3 inflammasome has been due especially to its implication in the pathogenesis of several human inflammatory diseases, particularly of the cryopyrin-associated periodic syndromes (CAPS) [21]. Focusing on its critical role in regulating inflammation, the NLRP3 inflammasome could be of great importance to therapies targeting inflammation [22].

2.2. IPAF-NAIP Subfamily

Its most well-studied element, NLRC4 (IPAF/CARD12), was previously characterized as an ICE-protease activating factor (IPAF) regarding its capacity for activating caspase-1. Nevertheless, posterior studies clearly placed its domain structure in the NLR family, and as it possessed a CARD domain, it was renamed NLRC4 [23]. The CARD domain allows it to directly bind to the CARD of caspase-1 without the participation of ASC [24]. However, NLRC4 is able to bind to ASC and efficiently activate caspase-1, as well as caspase-8, an apoptotic caspase [25].
NLRC5 is a less well-known inflammasome that links both innate and adaptive immune responses by regulating major histocompatibility complex (MHC) I class expression [26]. It is expressed in macrophages, dendritic cells, T cells, B cells and fibroblasts [27]. Moreover, an observed interaction with the NLRP3 inflammasome seemed to have a synergistic effect on IL-1β cleavage, thus it may positively modulate NLRP3 inflammasome activation [28]. Therefore, NLRC5 could form a functional inflammasome, but more studies are needed to know its physiological function more accurately.
Additionally, NOD1 is the founding member of the NLR family, and together with NOD2, they were the first NLRs identified as sensors for PAMPs [29]. NOD1 (NLRC1) and NOD2 (NLRC2) receptors can activate NF-κB and lead the production of inflammatory cytokines. Nevertheless, they have not been described to form an inflammasome complex [30].

3. Non-NLR Family Inflammasomes

Recently, other inflammasomes not belonging to the NLR family have been widely described, such as the proteins absent in melanoma 2 (AIM2) and pyrin inflammasomes. AIM2 was described as a sensor able to trigger inflammasome activation, pyroptosis and release of IL-1β and IL-18 in response to intracellularly delivered double-stranded DNA (dsDNA) detection [31]. AIM2 is a member of the ALR family of proteins, composed of an N-terminal PYD domain and a C-terminal HIN (hematopoietic, interferon-inducible and nuclear localization) domain [32]. Moreover, it negatively regulates inflammation and type I interferon (IFN) responses independent of its inflammasome function [33]. Different studies have elucidated a link between increased AIM2 expression and several human diseases, such as atherosclerosis, skin disease or chronic kidney disease [34].

4. Mechanisms of NLRP3 Inflammasome Activation

The inflammasome can be understood as a two-sides element and it regulates pathogen infection, but when the immune response triggered is not tightly regulated, it can be involved in pathologies such as CAPS and autoinflammatory disorders [21]. Inflammasomes can recognize a wide variety of endogenous or exogenous, sterile or infectious stimuli within the cell (PAMPs and DAMPs), which trigger its assembly and activation. This process can be explained by considering the upstream sensors recognizing activating signals, the adapters and the downstream effectors [35]. The unfeasibility of a direct interaction between NLRP3 and this diversity of stimuli led to a cellular event producing a conformational change in NLRP3, converting it into an active form. Nevertheless, there is no unique mechanism for the activation of the NLRP3 inflammasome [36]. NLRP3 activation can be triggered by PAMPs and DAMPs detection via PRRs, such as TLRs and NLRs, by cytokine stimulation via IL-1 receptor (IL-1R) or through TNF link to tumor necrosis factor (TNF) receptors TNFR1 and TNFR2 [37]. Moreover, there are mediators that facilitate signal transduction of these receptors: the adaptor protein myeloid differentiation primary response 88 (MyD88), the apoptosis signal-regulating kinase (ASK)1 and ASK2, interleukin 1 receptor-associated kinase (IRAK)1 and IRAK4, caspase-8 (CASP8), Fas-associated protein with death domain (FADD), ubiquitin-binding protein SHARPIN and TRAF-interacting protein with forkhead-associated domain (TIFA). All these elements trigger the transcription of NF-κB, which promotes the transcription of NLRP3 and IL1B genes, habilitating the cell for responding to NLRP3 activators [38].
NLRP3 inflammasome activation in macrophages is a two-step process, thus it requires a priming signal. In the priming process, a non-activating stimulus causes the transcriptional expression of the main components of the inflammasome, this being the ‘first hit’. A second stimuli or ‘second hit’ aggravates the functional activity of the NLRP3 inflammasome [39]. Activation of the NLRP3 inflammasome can be produced by different stimuli, including ionic flux, K+ efflux, Ca2+ influx, Na+ influx and Cl- efflux, reactive oxygen species (ROS) and mitochondrial dysfunction or lysosomal damage. K+ efflux channels P2X purinoceptor 7 (P2X7R) participate in this type of inflammasome activation. Other plasma-membrane-resident Ca2+ channels, namely transient receptor potential melastatin 2 (TRPM2), TRPM7 and transient receptor potential vanilloid 2 (TRPV2), can lead to Ca2+ influx to the cytosol [21] (Figure 1c). Mitochondria regulate homeostasis and respond to changes in intracellular K+ and ROS, resulting in mitochondrial dysfunction and apoptosis. Additionally, mitochondrial apoptotic signaling stimulated by NF-κB cause the production of IL-1β. Oxidized mitochondrial DNA is released as a consequence of mitochondrial dysfunction and apoptosis, and it directly activates the NLRP3 inflammasome [40,41,42].
Apart from the NLR-ASC-caspase-1 canonical inflammasome activation, there is also a non-canonical inflammasome characterized by its activation via caspase-11 in mice with the human orthologs caspase-4/5. Caspase-11 recognizes lipopolysaccharide (LPS) transfected into the cytosol from Gram-negative bacteria, directly binding to its CARD domain. It initiates proteolytic maturation of IL-1β as well as pyroptotic cell death in a GSDMD-dependent manner [35,43]. Non-canonical inflammasome activation by a component of LPS was shown in a previous study where mice lacking caspase-11 were resistant to LPS-induced lethality, even in the presence of TLR4 [44]. However, from the canonical and non-canonical inflammasomes, an alternative pathway of inflammasome activation was observed. It does not require K+ efflux, induction of ASC speck formation, or leading to subsequent pyroptosis, and was spread by TLR4-TRIF-RIPK1-FADD-CASP8 signaling upstream of NLRP3 [45].
NLRP3 inflammasome can be regulated in a post-transcriptional and post-translational level. At the post-transcriptional level, epigenetic factors such as DNA methylation and histone acetylation can regulate NLRP3 mRNA expression in response to Mycobacterium tuberculosis infection [46]. Dysregulation of epigenetic mechanisms could contribute to the pathological development of autoinflammatory syndromes by upregulating the expression of inflammasome components. MicroRNAs are also studied as post-transcriptional regulators of NLRP3 inflammasomes (miR-223, miR-133a, miR-7, miR-30e…) [35,47]. NLRP3 inflammasome activation can also be regulated by post-translational modifications, mainly phosphorylation and ubiquitination. These modifications are often linked. They can provoke different fates on the NLRP3 protein, including the modification of interacting protein networks, trafficking, change in subcellular localization, activation/inhibition of enzymatic activity and proteasomal, lysosomal or autophagic degradation [48]. In fact, a recent study showed that NLRP3 phosphorylation in its LRR domain can regulate inflammasome assembly [49].

5. Inflammasome Effector Functions

As previously stated, inflammasomes play a crucial role in the innate immune system by their ability to control the activation of the proteolytic enzyme caspase-1, which leads to proteolytic maturation of the proinflammatory cytokines IL-1β and IL-18, as well as pyroptosis cell death [50]. Mature IL-1β binds to IL-1R, promoting the heterodimerization of the receptor and the subsequent recruitment of components such as MyD88 [51]. IL-1β leads the release of other cytokines such as IL-1α, IL-6 and TNF-α as well as other factors that control growth and differentiation of immune cells [52]. IL-18 participates in many physiological pathways. A higher level of IL-18 can cause metabolic syndromes. For instance, chronic inflammation generated in adipose tissues can lead to insulin resistance and type 2 diabetes mellitus [53].
Another important process carried out by inflammasomes is a lytic form of programmed cell death named pyroptosis. Both canonical inflammasome signaling, recruiting caspase-1, and noncanonical inflammasome, via caspase-4, caspase-5 (in humans) and caspase-11 (in mice), can trigger pyroptosis. It is characterized by cell swelling, membrane lysis, and release of inflammatory compounds into the extracellular space, such as IL-1β, IL-6 and IL-18. Previous studies have shown that gasdermins, a group of pore-forming effector proteins, can play inflammatory caspase-induced pyroptosis, being the N-terminal domain of gasdermin-D sufficient to trigger the process [35,54,55]. Additionally, caspase-8-dependent apoptosis is an additional pathway resulting from inflammasome activation. AIM2 and NLRP3 inflammasomes showed cleaved forms of apical caspase-8 and executioner caspase-3, in response to cytosolic DNA and nigericin, respectively. The process occurred independently from caspase-1 but depended on the inflammasome adapter ASC [56]. Interestingly, a recent study described the capacity of the Z-DNA binding protein 1 (ZBP1), an innate immune sensor capable of activating cell death in the form of pyroptosis, apoptosis and necroptosis (PANoptosis) together with the NLRP3 inflammasome [57].
Whereas the effector functions have been widely studied, there are several additional roles of the inflammasome complexes that have been less characterized. IL-1β is a leaderless cytoplasmic protein whose secretion mechanisms are poorly defined. An endoplasmic reticulum (ER)/Golgi-independent mechanism termed ‘unconventional protein secretion’ was shown, and it was dependent on caspase-1 activation. However, the specific mechanisms and molecular components involved in this process are unclear. Another emerging role of inflammasomes is the activation of eicosanoids, bioactive molecules derived from membrane lipids that play a role in homeostatic and pathological processes. Furthermore, a link between inflammasome activation and autophagy as well as regulation of phagosome maturation have been observed [58].

6. The Role of the Inflammasome in Adaptive Immunity and Autoimmunity

The production of proinflammatory cytokines is critical for an effective innate response, as well as a mechanism by which the innate immune system influences the subsequent development of an adaptive immune response [59]. As it is well known, inflammasomes are components of the innate immune system that produce the proinflammatory cytokines IL-1β and IL-18, and they drive the differentiation of specific lineages of helper T cells (Th1, Th2, Th17 and regulatory T cells), which are the main players in adaptive immunity [60]. On the other side, an aberrant inflammasome activation is responsible for the development of CAPS, as well as other common diseases such as metabolic disorders, crystal-related diseases and autoimmune diseases. Inflammation is also crucial in many renal diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD). Although the innate immune system is always involved, in these conditions, the adaptive immunity plays the main role [61].
Concerning autoimmune diseases, they are characterized by self-reactive cells and the overproduction of autoantibodies, produced because of a lack of immunological tolerance and aberrant autoreactive immune responses. The pathogenesis of autoimmune diseases remains to be clarified, but it has been demonstrated that aberration in innate and adaptive immunity is involved. NLRP3 inflammasome has been recently linked with innate immune signal recognition and induction of autoreactive immune responses, probably being a checkpoint in innate immunity to cause distorted adaptive immunity [62,63]. Therefore, how can the NLRP3 inflammasome function affect the development of autoimmune diseases? Cytokines released by the inflammasome, especially IL-1β, produce an inflammatory effect that promotes the development of most autoimmune diseases, including RA and inflammatory bowel disease [64,65]. Furthermore, the NLRP3 inflammasome is also responsible for autoimmune diseases due to an adaptive immune dysfunction. IL-1 mediates T cell proliferation, thus it can promote autoreactive T cells to cause b-cell death [63,66]. NLRP3 inflammasome promotes Th1 differentiation in RA, induced by IL-1β in a caspase-1-dependent manner, and it can also induce differentiation and polarization of Th2, Th17 and dendritic cells in other autoimmune diseases [63,67]. Th17 cells can produce proinflammatory cytokines namely IL-17A, IL-17F, IL-21 and IL-22, while Th1 cells secrete IFN-γ, induced by IL-18, and all of these factors contribute to autoimmunity development [60]. Additionally, autoimmune diseases can be promoted by pyroptosis, leading to the release of cellular debris and its reaction with immune cells, triggering inflammation [68].
Indeed, multiple polymorphisms in inflammasome genes have been associated with the susceptibility and development of autoimmune diseases. For instance, rare gain-of-function variants can be implicated in hereditary inflammatory diseases, characterized by uncontrolled production of IL-1β and/or IL-18, named inflammasomopathies. Mutations in NLRP3 are the prototypic inflammasomopathy, but they have also been described as autoinflammatory diseases associated with mutations that activate the NLRP1, NLRC4 and pyrin inflammasomes [69,70]. Moreover, single nucleotide polymorphisms (SNPs) play a crucial role in autoimmune diseases, and they can affect the priming of inflammasomes, some of their components or end products (IL-1β, IL-18) [71].

7. Inflammasome Involvement in Autoimmune Kidney Diseases

A summary of the main publications related to inflammasome investigations in glomerulonephritis is shown in Table 1.

7.1. Lupus Nephritis

SLE is a chronic disease that frequently affects the kidney. Lupus nephritis (LN) is the most common renal disease, involving approximately 50% of patients with SLE. This autoimmune disease mostly affects women of the reproductive age. In men, the disorder could be more aggressive. Patients usually have LN at an early age, and it usually presents itself in the initial stages of the disease. Patients with this renal impairment have an increased mortality rate. In total, 10–30% of patients with LN progress to renal failure requiring kidney replacement therapy [72,73].
Irregularities in innate and adaptive immunity contribute to the pathogenesis of SLE. LN occurs when the transcription of genes associated with neutrophils increases. The rise in IFN precedes the activation of neutrophils. The increment of IFN causes the differentiation of B cells into plasmablasts and produces inflammation of specific tissues through neutrophils and active myeloid cells. When these neutrophils die, extracellular neutrophil traps (NETs) appear [72]. NETs are meshing-chromatin fibers combined with granules derived from antimicrobial peptides and enzymes that play an important defense role [74]. These meshes help to maintain antigen-specific autoantibody production [72].
The formation of immune complexes that are deposited in the glomerulus is derived from the production of antibodies against nuclear and cellular antigens. Immune complexes can activate complement and cause kidney damage, especially through the alternative pathway. Plasma interstitial cells generated by B and T cells aggregate in the renal tubulointerstitium also generating the production of autoantibodies [73].
An increase in the inflammasome’s components was observed in biopsies of patients with LN as PYCARD (ASC), caspase-1 and IL-18, indicating their contribution to the disease [75]. Furthermore, the increased transcription of IL-18 in the tubulointerstitial and glomerular compartments [75] correlates with the severity of LN and the onset of proteinuria [76].

7.1.1. In Vitro Model

The activation of the inflammasome in cells of innate immunity could trigger or amplify an autoimmune response. After exposure to an inflammatory stimulus such as LPS, isolated fresh monocytes increase the activation of the inflammasome characterized by the rise in caspase-1, IL-1β and IL-18. A caspase-1 inhibitor added to in vitro cultures reduces IL-18 production [77].
As mentioned previously, the NETosis mechanism contributes to the death of neutrophils in SLE patients. Evidence from groups of researchers suggests that SLE patients are characterized by an imbalance between the development and clearance of NETs, which produces tissue damage [78,79]. Specifically, low-density proinflammatory granulocytes that occur in the bloodstream of SLE patients allow a much greater capacity to produce NETs [79].
Kahlenbertg and her coworkers [80] firstly demonstrated that NETs are, partly, activators of the inflammasome through the externalization of LL-37. NETs externalize various antimicrobial peptides. Specifically, cathelicidin LL-37 is a peptide synthesized by neutrophils, monocytes and macrophages, among others, with activity against several pathogens. LL-37 externalization in NETs has been identified in neutrophils from SLE patients [79]. In this study [80], authors purified and isolated human and murine macrophages. Results showed that LL-37 activated the NLRP3 inflammasome in macrophages and that SLE patients were more likely to activate the inflammasome in response to LL-37 and NETs, compared to macrophages from control patients. This stimulation perpetuates the increase in IL-1β and IL-18, which, in turn, will promote NETosis resulting in disease flares or organ damage, mainly kidneys, skin and brain. Furthermore, their data suggested that the NLRP3 inflammasome is required for caspase-1 activation by LL-37.

7.1.2. Animal Model

It is noteworthy to mention that in various murine studies, NLRP3 has been associated with LN. Kahlenberg et al. [81], studied the role of caspase-1 in the induction of murine lupus. Wild-type mouse models were exposed to pristane developed lupus-related autoantibodies and an active response to INF type I. Following pristane exposure, caspase-1 −/− mice did not have increased levels of IL-1β or IL-18, suggesting that caspase-1 played a role in the transcription of these cytokines. In addition, caspase-1 −/− mice showed less development of autoantibodies and immune complexes related to glomerulonephritis, contrasting with wild-type mice. P2X7, an extracellular ATP-gated ion channel receptor, has also been shown to play a role in NLRP3 activation and LN development. The use of a selective P2X7 antagonist brilliant blue G in MLR/lpr mice produced a downregulation of the NLRP3/ASC/Caspase-1 complex and therefore a suppression of IL-β. This reduced LN severity, proteinuria and blood urea nitrogen levels in mice. Likewise, P2X7/NLRP3 inhibition decreases the Th17:Treg ratio, decreasing anti-double-stranded DNA antibodies(anti-dsDNA). NZM2328 mice that were injected intravenously with adenovirus-expressing interferon-α particles confirmed these results [82]. Increased expression of P2X7 has also been observed in the kidney tissue from patients with SLE [83]. A neutralizing monoclonal antibody to high-mobility box 1 protein, a ubiquitin nuclear protein that binds to DNA, has also been shown to decrease IL-β, IL-6, IL-17 and IL-18 levels and caspase-1 in kidneys of BXSB mice. In addition, this model also attenuated proteinuria, glomerulonephritis, renal immune complex deposits and circulating anti-dsDNA [84].
Another inflammatory pathway that affects LN is NF-κB. NF-κB is a transcription factor that participates in innate and adaptive immunity [85]. In human studies, a correlation has been described between the activation of NF-κB and the histological and renal function impairment [86]. Zhao et al. [87] studied whether the inhibition of both pathways decreased LN progression in lupus-prone MRL / lpr mice. This interest was triggered by the renal pathophysiological role played by NF-κB. Inhibition of NF-κB and NLRP3 by Bay11-7082 prevented their formation and activation, respectively, resulting in an improvement in established kidney damage in LN. Moreover, Bay11-7082 decreased renal immune complex deposits and serum anti-dsDNA levels.
AIM2, has also been implicated in the pathogenesis of SLE. However, its inhibition has been shown to be two-edged in relation to the pathophysiology of LN. Zhang and coworkers analyzed the correlation between the severity of LN and AIM2 in SLE patients and lupus mice. AIM2 expression was elevated in PBMCs from SLE patients. In addition, AIM2 correlated with macrophage activation and was augmented in macrophages induced by lymphocyte-derived apoptotic DNA. The inhibition of AIM2 by siRNA decreased the infiltration of macrophages in renal tissue and produced an improvement in nephritis [88]. However, other researchers found that the inhibition of AIM2 generated susceptibility to developing SLE. p202 negatively regulates AIM2 in some mouse strains, increasing INF and predisposition to SLE [89].

7.1.3. Human Model

The role of the inflammasome in autoimmune diseases have been widely described. Studies with different SNPs related to inflammasome in patients with SLE have been reported in the literature. Pontillo et al. [90] analyzed 14 SNPs in 7 inflammasome genes, such as NLRP1, NLRP3, NLRC4, AIM2, CARD8, CASP1 and IL1B. The study showed, for the first time, an association between SNPs and SLE in a population from southern Brazil. The NLRP1 rs2670660 SNP, especially when combined with the NLRP1 variant rs12150220, confers an increased risk of SLE and developing nephritis, arthritis and rash. Other SNPs also described in autoimmune diseases, such as celiac disease [91] and diabetes [21], were not associated to SLE disease in their population [90]. They also found no association with SLE with respect to AIM2 or IL1B polymorphisms [90]. Furthermore, results from da Cruz et al. found a gain of function in the NLRP3 rs10754558 variant in patients with LN [92].

7.2. ANCA Glomerulonephritis

ANCA associated with vasculitis (AAV) is a life-threatening autoimmune disease characterized by an antibody-mediated glomerulonephritis and necrotizing vasculitis. AAV affects small and medium vessels, especially organs such as the kidney and lung. Pauci-immune and necrotizing glomerulonephritis are frequently associated in patients with vasculitis being more prevalent in men over 50 years of age. ANCA vasculitis is usually associated with ANCA-myeloperoxidase (MPO), ANCA-proteinase 3 (PR3) or ANCA-negative serotype positivity. This pathology is classified into different clinical variants such as microscopic polyangiitis, granulomatosis with polyangiitis (Wegener), Eosinophilic granulomatosis with polyangiitis (Churg-Strauss) or vasculitis limited to renal tissue [93]. Both innate and adaptive immunity participate in the development of AAV, although the exact mechanisms remain to be elucidated [93].
Neutrophils play a fundamental role in the pathogenesis of AAV inflicting tissue damage after degranulation induced by ANCA antibodies. Apart from antibodies, T cells are also involved in disease pathogenesis. Neutrophils secrete cytokines that recruit more neutrophils and other inflammatory cells. Infiltration of T cells is also part of the granulomas. The benefit of anti-T cell therapies demonstrates the involvement of this cell in AAV. The Th1 and Th17 phenotypes are involved in the acute phase. An increase in C5a and, therefore, a participation of the alternative complement pathway has been reported [94].

7.2.1. In Vitro Model

Inflammasome components such as cytokines are important mediators in AAV. Both Il-1β and Il-18 have been related to the pathogenesis of AAV, thus the implication of the inflammasome in the inflammation cascade of this disease is expected [61].
Il-18 plays a role in neutrophil chemoattraction independent of TNFα priming [95], contrary to what was reported in other studies [96]. Hewins et al. indicated that, in the presence of anti-TNFα antibody, ANCA-induced superoxide production was not decreased. This would explain the persistence of tissue damage in the presence of anti-TNFα treatment. Furthermore, Hewins and colleagues demonstrated renal Il-18 expression in patients with ANCA vasculitis [95].

7.2.2. Animal Model

Several studies establish ANCA necrotizing crescentic GN (NCGN) in animal models [97]. Dipeptidyl peptidase (DPPI) is a cysteine protease responsible for activating neutrophil serine proteases (NPS) such as cathepsin G (CG), neutrophil elastase (NE) and PR3. These enzymes, responsible for modulating inflammation, have been related to the pathophysiology of ANCA vasculitis. In an anti-MPO antibody-induced experimental model of NCGN, a protective role of DPPI in kidney disease was demonstrated with a local decrease in inflammatory cytokines, especially IL-1β [98]. In fact, the elevation and processing of Il-1β by PR3 and NE has been linked [99]. The group of Scheiber et al. [98] demonstrated that active PR3 or active PR3/NE causes an increase in cytokines and anti-MPO antibodies, generating NCGN. They produced NE-/PR3-mice that were protected from NCGN. This demonstrated the role of NSP in ANCA nephropathy. In addition, they noticed that treatment with anakinra, an IL-1 receptor antagonist, downregulates the inflammatory cascade and protects against NCGN.
Another different pathway that induces Il-1β production and causes NCGN is related to phagocyte NADPH oxidase (Phox). Phox is a heme protein heterodimer from pg91phox and p22phox responsible for generating ROS producing tissue damage [100]. Though, some studies explain that ROS is also involved in the knockdown of inflammation [101]. Another study from the German group of Schreiber et al. [100] discovered the role of Phox in limiting the inflammasome by downregulating its components such as caspase-1 and thus IL-1β. Those authors created an antibody-mediated anti-MPO model. Transplanted mice with gp91phox-deficient or p47phox-deficient bone marrow showed greater histological involvement with more inflammation and necrosis, compared to mice with wild-type bone marrow. Additionally, they also generated pg91phox/caspase-1-deficient bone marrow transplant mice. In this case, mice with double deficiency improved NCGN compared to mice with only pg91phox- deficiency. These results hypothesize that Phox limits the activity of caspase-1 and therefore the role of the inflammasome.

7.2.3. Human Model

Elevated serum levels of the cascade of the inflammasome components have also been found in patients with ANCA vasculitis. IL-18 levels have been seen in patients with ANCA vasculitis regardless of MPO or PR3 values [102]. It has been generally accepted that renal interstitial damage had to be associated with glomerular damage, but there are case reports of patients with ANCA vasculitis with only interstitial injury [103,104]. Tashiro et al. [105] demonstrated no correlation between glomerular damage and interstitial damage in biopsies from patients with ANCA vasculitis. They glimpsed the correlation of IL-1β levels with the severity of tubulointerstitial damage. Moreover, infiltrating macrophages showed positive staining for NLRP3 at the tubulointerstitium without detecting this positivity in the glomerulus. On the contrary, Hewins et al. [95] who found upregulated IL-18 in renal biopsies, reported that positivity in the glomerulus has been found in podocytes while in the tubulointerstitium IL-18-positive has been observed in infiltrating macrophages, myofibroblasts and tubular epithelial cells.
The activation of NOD-like receptors in patients with active stage of ANCA vasculitis is from Wang et al. [106]. The mean optical densities of NOD2, NLRP3 and NLRC5, both in the glomerulus and in the tubulointerstitium, were significantly higher in patients with ANCA vasculitis than in healthy controls, in patients with minimal change disease and in patients with type IV LN. NLRs were mainly expressed in podocytes and in infiltrating monocytes and macrophages, but hardly expressed in glomeruli, results similar to those of Tashiro et al. [105]. The expression of NOD2 and NLRC5 correlated with clinicopathological involvement, while NLRP3 did not [106]. Unlike these researchers, Tashiro and colleagues did correlate NLRP3 with the severity of kidney damage [105].

7.3. IgA Nephropathy

IgA nephropathy (IgAN) is the main cause of renal failure due to glomerulonephritis in the world [107]. Components of innate immunity are also involved in this nephropathy. The deposition of IgA aggregates or IgA immune complexes and subsequent activation of T cells causing inflammation is considered the main cause of the disease. The IgA subclass deposited in the glomerulus is the IgA1, which plays the central role in the pathophysiology of the disease. Mesangial cell proliferation is the typical histological finding of IgAN. Mesangial cells undergo proliferation under the action of IL-1, among other cytokines [108]. The contribution of cytokines involved in the inflammasome cascade suggests a role for this inflammatory component in IgAN.
The alternative complement pathway and lectin pathways are also involved in the development of the disease since C3, C4, C4d, properdin, C5b-C9 and mannose binding lectin are usually detected in renal biopsy [109].

7.3.1. Animal Model

Researchers have demonstrated IL-1 expression in kidney tissue affected with IgAN [110,111]. Chen et al. [112], using an animal model of IgAN with ddY mice found decreased mesangial proliferation in mice treated with IL-1 receptor antagonist (IL-1ra). These results suggested that IL-1 is enrolled in IgAN development, evidencing a potential role of the inflammasome cascade in IgAN.

7.3.2. Human/In Vitro Model

The role of NLRP3 in IgAN remains to be discovered. The Canadian team of Chun et al. was the first to evaluate in vivo and in vitro the expression of NLRP3 in the kidney of patients with IgAN and the progression of the disease. They found that NLRP3 was expressed mainly in the tubules with no staining in the normal glomerulus. However, in patients with IgAN, NLRP3 expression was detected in the glomerulus, although it was more increased in the tubules. Both in human kidney biopsies and in low passage human cells, they established a decrease in NLRP3 during tubular damage. Equally the immunostaining results and the NLRP3 mRNA expression corroborated the presence of NLRP3 and its subsequent loss after renal injury. These discoveries suggest that NLRP3 may be a biomarker of tubular integrity. In addition, NLRP3 plays its role in the early stages of kidney disease being implicated in chronic kidney disease. However, due to study limitations, the researchers were unable to report on the functional role of the inflammasome [113].
Other researchers were able to glimpse the role of NLRP3 in the pathophysiology of IgAN. IgA immune complexes elicited the activation of the NLRP3 inflammasome in macrophages. This, in turn, stimulated the production of ROS by the mitochondria. They performed a mouse model with IgAN knockout for NLRP3. The generation of IgA immune complexes was inhibited by knockout mice. A regaining in renal function was described in NLRP3 knockout and in the kidney-targeting delivery of shRNA of NLRP3. Finally, the researchers clarify that they cannot exclude the role of other inflammasomes in IgAN and that the use of shNLRP3 could be a treatment to improve or prevent the disease [114].

7.4. Anti-Glomerular Basement Membrane Glomerulonephritis

Anti-glomerular basement membrane (anti-GBM) is an infrequent autoimmune disease that affects the small vessels of the kidneys and lungs. Patients develop antibodies against the non-collagenous domain of the α3 chain of type IV collagen present in the basal membrane of those organs [115]. Although the humoral immunity plays a central role, the participation of cellular immunity has also been reported. Thus, the IgG1 and IgG3 subclasses have been clearly related to the severity of the disease. The deposition of antibodies in kidney vessels can origin inflammation by activating complement and the Fc receptor. On the other hand, the increase in CD4+ T cells has been correlated with the severity of the disease. Peripheral CD4+ progress in the presence of α3 (IV) NC1. In addition, in animal models, CD4+ has been shown to be a trigger for the development of anti-GBM antibodies [116].

Animal Model

The major cytokines derived from the inflammasome cascade, IL-1 and IL-18, have been shown to have a pathophysiological role in patients with anti-GBM disease. In a mouse model of anti-GBM, Il-1β −/− and IL1 type 1 receptor (IL-1R) −/− mice, the role of IL-1 isoforms, IL-1α and IL-1β, in anti-GMB GN was studied. IL-1β mice showed less development of crescentic formations, recruitment of macrophages and T cells, while IL-1R1 −/− mice appeared to have a role in the immune response, since they had fewer antibodies in serum [117]. Furthermore, other animal models have demonstrated the proinflammatory role of IL-18 in renal inflammation [118].
Glomerular infiltration by macrophages is probably one of the major sources of IL-1 cytokine production. Several studies have analyzed the chemoattractant role of this cytokine and have implemented treatment with antagonists IL-1ra in a rat model with anti-GBM. Both the group of Lan et al. [119] and Tang et al. [120], demonstrated that by using IL-1ra there was a decrease in the infiltration of glomerular macrophages and an improvement in proteinuria. Lan et al. [119] also revealed a stoppage of renal function worsening and prevented histological progression such as the formation of glomerular crescents. Tang et al. [120] obtained a decreased expression of ICAM-1 after treatment with IL-1ra, which was also associated with a decline in the infiltration of polymorphonuclear (PMN) cells and monocytes.
However, following the findings found by Timoshanko et al. [117] on the contribution of IL-1β in nephritis in anti-GBM, other authors concluded, using a murine model, that only dendritic cells that reside mainly in the tubulointerstitium express pro-IL-1β and therefore they activate NLRP3 and caspase-1 secreting mature IL-1β. They showed that the inflammasome axis does not contribute to glomerular inflammation since glomerular cells could not produce IL-1β during sterile inflammation [121].

8. Final Remarks

As comprehensively detailed, many studies have demonstrated the role of inflammasome in glomerulonephritis. However, further insights into the pathophysiological mechanism research focus on inflammasome and autoimmune diseases are needed.
On the other hand, the participation of the inflammasome in immunity encourages the need for new therapeutic weapons aimed at its modulation. Recently, antagonists of IL-1ra are already approved to treat non-renal diseases such as rheumatoid arthritis, CAPS, Familial Mediterranean fever and Still’s disease. Additionally, for now there is development for treating autoimmune diseases in patients that are non-responsive or over time are refractory to treatment with TNF-α antagonists and/or T-cell co-stimulation antagonists with an IL-18 antagonist. Given that the IL-1 blockade or IL-18 antagonist have been successful in non-human models of renal diseases modulating inflammasome activation. Perhaps, we are ready to introduce these targets in the nephrological clinics. However, the blockade of a single cytokine could be not enough to downregulate the activation of the inflammasome, then polytherapy could be considered. To our knowledge, while there are developed clinical trials about autoimmune and inflammatory diseases, there are no ongoing clinical trials involving glomerulonephritis and inflammasomes. Thus, further efforts in the exploration into how treatments affect the activity of the inflammasome axis could be a promising therapy.

Author Contributions

P.A.-P. and C.D.-R. performed a literature review of the topic and elaborated the manuscript. All authors have read and agreed. F.G.-P. elaborated the manuscript. A.V.-A., J.T., J.D., N.L., L.M.-V. and X.F. critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study has been funded by Instituto de Salud Carlos III through the grant CM21/00170 (co-funded by European Social Fund (ESF investing in your future). This work was supported by the Ministerio de Ciencia, Innovación y Universidades (Madrid, Spain) (Grants FIS-ISCIII PI20/00812, co-funded by FEDER funds/European Regional Development Fund—a way to build Europe and FI21/00067). We thank CERCA Programme/Generalitat de Catalunya for institutional support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rathinam, V.; Chan, F.K. Inflammasome, Inflammation, and Tissue Homeostasis. Trends Mol. Med. 2018, 24, 304–318. [Google Scholar] [CrossRef] [PubMed]
  2. Strowig, T.; Henao-Mejia, J.; Elinav, E.; Flavell, R. Inflammasomes in health and disease. Nature 2012, 481, 278–286. [Google Scholar] [CrossRef] [PubMed]
  3. Place, D.E.; Kanneganti, T.D. Recent advances in inflammasome biology. Curr. Opin. Immunol. 2018, 50, 32–38. [Google Scholar] [CrossRef]
  4. Zhang, K.-J.; Wu, Q.; Jiang, S.-M.; Ding, L.; Liu, C.-X.; Xu, M.; Wang, Y.; Zhou, Y.; Li, L. Pyroptosis: A New Frontier in Kidney Diseases. Oxidative Med. Cell. Longev. 2021, 2021, 6686617. [Google Scholar] [CrossRef]
  5. Chi, K.; Geng, X.; Liu, C.; Cai, G.; Hong, Q. Research Progress on the Role of Inflammasomes in Kidney Disease. Mediat. Inflamm. 2020, 2020, 8032797. [Google Scholar] [CrossRef] [Green Version]
  6. Broz, P.; Dixit, V. Inflammasomes: Mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 2016, 16, 407–420. [Google Scholar] [CrossRef]
  7. Platnich, J.M.; Muruve, D.A. NOD-like receptors and inflammasomes: A review of their canonical and non-canonical signaling pathways. Arch. Biochem. Biophys. 2019, 670, 4–14. [Google Scholar] [CrossRef]
  8. Sharma, M.; De Alba, E. Structure, activation and regulation of NLRP3 and AIM2 inflammasomes. Int. J. Mol. Sci. 2021, 22, 872. [Google Scholar] [CrossRef]
  9. Wang, Z.; Zhang, S.; Xiao, Y.; Zhang, W.; Wu, S.; Qin, T.; Yue, Y.; Qian, W.; Li, L. NLRP3 Inflammasome and Inflammatory Diseases. Oxidative Med. Cell. Longev. 2020, 2020, 4063562. [Google Scholar] [CrossRef]
  10. Kim, Y.K.; Shin, J.-S.; Nahm, M.H. NOD-Like Receptors in Infection, Immunity, and Diseases. Yonsei Med. J. 2016, 57, 5–14. [Google Scholar] [CrossRef] [Green Version]
  11. Wen, H.; Miao, E.A.; Ting, J.P.-Y. Mechanisms of NOD-like Receptor-Associated Inflammasome Activation. Immunity 2013, 39, 432–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Taabazuing, C.Y.; Griswold, A.R.; Bachovchin, D.A. The NLRP1 and CARD8 inflammasomes. Immunol. Rev. 2020, 297, 13–25. [Google Scholar] [CrossRef] [PubMed]
  13. Mitchell, P.S.; Sandstrom, A.; Vance, R.E. The NLRP1 inflammasome: New mechanistic insights and unresolved mysteries. Curr. Opin. Immunol. 2019, 60, 37–45. [Google Scholar] [CrossRef] [PubMed]
  14. Bauernfried, S.; Scherr, M.; Pichlmair, A.; Duderstadt, K.E.; Hornung, V. Human NLRP1 is a sensor for double-stranded RNA. Science 2021, 371, eabd0811. [Google Scholar] [CrossRef] [PubMed]
  15. Ciążyńska, M.; Olejniczak-Staruch, I.; Sobolewska-Sztychny, D.; Narbutt, J.; Skibińska, M.; Lesiak, A. The Role of NLRP1, NLRP3, and AIM2 Inflammasomes in Psoriasis: Review. Int. J. Mol. Sci. 2021, 22, 5898. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, T.; Zhu, C.; Wang, S.; Mo, L.; Yang, G.D.; Hu, J.; Zhang, F. Role of NLRP3 and NLRP1 inflammasomes signaling pathways in pathogenesis of rheumatoid arthritis. Asian Pac. J. Trop. Med. 2014, 7, 827–831. [Google Scholar] [CrossRef] [Green Version]
  17. Yang, Q.; Yu, C.; Yang, Z.; Wei, Q.; Mu, K.; Zhang, Y.; Zhao, W.; Wang, X.; Huai, W.; Han, L. Deregulated NLRP3 and NLRP1 Inflammasomes and Their Correlations with Disease Activity in Systemic Lupus Erythematosus. J. Rheumatol. 2013, 41, 444–452. [Google Scholar] [CrossRef]
  18. Haneklaus, M.; O’Neill, L.; Coll, R. Modulatory mechanisms controlling the NLRP3 inflammasome in inflammation: Recent developments. Curr. Opin. Immunol. 2013, 25, 40–45. [Google Scholar] [CrossRef]
  19. Wang, L.; Hauenstein, A.V. The NLRP3 inflammasome: Mechanism of action, role in disease and therapies. Mol. Asp. Med. 2020, 76, 100889. [Google Scholar] [CrossRef]
  20. De Torre-Minguela, C.; del Castillo, P.M.; Pelegrín, P. The NLRP3 and pyrin inflammasomes: Implications in the pathophysiology of autoinflammatory diseases. Front. Immunol. 2017, 8, 43. [Google Scholar] [CrossRef] [Green Version]
  21. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Zhao, J.; Wang, H.; Huang, Y.; Zhang, H.; Wang, S.; Gaskin, F.; Yang, N.; Fu, S.M. Lupus Nephritis: Glycogen Synthase Kinase 3β Promotion of Renal Damage Through Activation of the NLRP3 Inflammasome in Lupus-Prone Mice. Arthritis Rheumatol. 2014, 67, 1036–1044. [Google Scholar] [CrossRef] [PubMed]
  23. Duncan, J.A.; Canna, S.W. The NLRC4 Inflammasome. Immunol. Rev. 2018, 281, 115–123. [Google Scholar] [CrossRef] [PubMed]
  24. Bauer, R.; Rauch, I. The NAIP/NLRC4 inflammasome in infection and pathology. Mol. Asp. Med. 2020, 76, 100863. [Google Scholar] [CrossRef]
  25. Lee, B.L.; Mirrashidi, K.; Stowe, I.B.; Kummerfeld, S.K.; Watanabe, C.; Haley, B.; Cuellar, T.L.; Reichelt, M.; Kayagaki, N. ASC- A nd caspase-8-dependent apoptotic pathway diverges from the NLRC4 inflammasome in macrophages. Sci. Rep. 2018, 8, 3788. [Google Scholar] [CrossRef] [Green Version]
  26. Cho, S.X.; Vijayan, S.; Yoo, J.S.; Watanabe, T.; Ouda, R.; An, N.; Kobayashi, K. MHC class I transactivator NLRC5 in host immunity, cancer and beyond. Immunology 2021, 162, 252–261. [Google Scholar] [CrossRef]
  27. Davis, B.K.; Roberts, R.A.; Huang, M.T.; Willingham, S.B.; Conti, B.J.; Brickey, W.J.; Barker, B.R.; Kwan, M.; Taxman, D.J.; Accavitti-Loper, M.-A.; et al. Cutting Edge: NLRC5-Dependent Activation of the Inflammasome. J. Immunol. 2011, 186, 1333–1337. [Google Scholar] [CrossRef] [Green Version]
  28. Di Virgilio, F.; Walker, J. Innate Immune Receptors. NLR Proteins; Springer Nature: Hatfield, UK, 2016. [Google Scholar]
  29. Carneiro, L.; Magalhaes, J.; Tattoli, I.; Philpott, D.; Travassos, L. Nod-like proteins in inflammation and disease. J. Pathol. 2008, 214, 136–148. [Google Scholar] [CrossRef]
  30. Velloso, F.J.; Lima, M.T.; Anschau, V.; Sogayar, M.C.; Correa, R.G. NOD-like receptors: Major players (and targets) in the interface between innate immunity and cancer. Biosci. Rep. 2019, 39, BSR20181709. [Google Scholar] [CrossRef] [Green Version]
  31. Man, S.M.; Karki, R.; Kanneganti, T. AIM2 inflammasome in infection, cancer and autoimmunity: Role in DNA sensing, inflammation and innate immunity. Eur. J. Immunol. 2016, 46, 269–280. [Google Scholar] [CrossRef] [Green Version]
  32. Kumari, P.; Russo, A.J.; Shivcharan, S.; Rathinam, V.A. AIM2 in health and disease: Inflammasome and beyond. Immunol. Rev. 2020, 297, 83–95. [Google Scholar] [CrossRef] [PubMed]
  33. Nakaya, Y.; Lilue, J.; Stavrou, S.; Moran, E.A.; Ross, S.R. AIM2-Like Receptors Positively and Negatively Regulate the Interferon Response Induced by Cytosolic DNA. mBio 2017, 8, e00944-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sharma, B.R.; Karki, R.; Kanneganti, T. Role of AIM2 inflammasome in inflammatory diseases, cancer and infection. Eur. J. Immunol. 2019, 49, 1998–2011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 6, 36. [Google Scholar] [CrossRef] [PubMed]
  36. Man, S.M.; Kanneganti, T. Regulation of inflammasome activation. Immunol. Rev. 2015, 265, 6–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Bauernfeind, F.G.; Horvath, G.; Stutz, A.; Alnemri, E.; MacDonald, K.; Speert, D.; Fernandes-Alnemri, T.; Wu, J.; Monks, B.G.; Fitzgerald, K.A.; et al. Cutting Edge: NF-κB Activating Pattern Recognition and Cytokine Receptors License NLRP3 Inflammasome Activation by Regulating NLRP3 Expression. J. Immunol. 2009, 183, 787–791. [Google Scholar] [CrossRef]
  38. Xue, Y.; Tuipulotu, D.E.; Tan, W.H.; Kay, C.; Man, S.M. Emerging Activators and Regulators of Inflammasomes and Pyroptosis. Trends Immunol. 2019, 40, 1035–1052. [Google Scholar] [CrossRef]
  39. Patel, M.N.; Carroll, R.G.; Galván-Peña, S.; Mills, E.L.; Olden, R.; Triantafilou, M.; Wolf, A.I.; Bryant, C.E.; Triantafilou, K.; Masters, S.L. Inflammasome Priming in Sterile Inflammatory Disease. Trends Mol. Med. 2017, 23, 165–180. [Google Scholar] [CrossRef]
  40. Tschopp, J. Mitochondria: Sovereign of inflammation? Eur. J. Immunol. 2011, 41, 1196–1202. [Google Scholar] [CrossRef]
  41. Nakahira, K.; Haspel, J.A.; Rathinam, V.A.; Lee, S.J.; Dolinay, T.; Lam, H.C.; Englert, J.A.; Rabinovitch, M.; Cernadas, M.; Kim, H.P.; et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. Immunol. 2011, 12, 222–230. [Google Scholar] [CrossRef] [Green Version]
  42. Shimada, K.; Crother, T.R.; Karlin, J.; Dagvadorj, J.; Chiba, N.; Chen, S.; Ramanujan, V.K.; Wolf, A.J.; Vergnes, L.; Ojcius, D.M.; et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 2012, 36, 401–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Russo, A.J.; Behl, B.; Banerjee, I.; Rathinam, V.A. Emerging Insights into Noncanonical Inflammasome Recognition of Microbes. J. Mol. Biol. 2017, 430, 207–216. [Google Scholar] [CrossRef] [PubMed]
  44. Kayagaki, N.; Wong, M.; Stowe, I.B.; Ramani, S.; Gonzalez, L.C.; Akashi-takamura, S.; Miyake, K.; Zhang, J.; Lee, W.P.; Forsberg, L.S.; et al. Noncanonical Inflammasome Activation by Intracellular LPS Independent of TLR4. Science 2013, 341, 1246–1249. [Google Scholar] [CrossRef] [PubMed]
  45. Gaidt, M.M.; Ebert, T.; Chauhan, D.; Schmidt, T.; Schmid-Burgk, J.L.; Rapino, F.; Robertson, A.A.B.; Cooper, M.A.; Graf, T.; Hornung, V. Human Monocytes Engage an Alternative Inflammasome Pathway. Immunity 2016, 44, 833–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Wei, M.; Wang, L.; Wu, T.; Xi, J.; Han, Y.; Yang, X.; Zhang, D.; Fang, Q.; Tang, B. NLRP3 Activation Was Regulated by DNA Methylation Modification during Mycobacterium tuberculosis Infection. BioMed. Res. Int. 2016, 2016, 4323281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Poli, G.; Fabi, C.; Bellet, M.M.; Costantini, C.; Nunziangeli, L.; Romani, L.; Brancorsini, S. Epigenetic Mechanisms of Inflammasome Regulation. Int. J. Mol. Sci. 2020, 21, 5758. [Google Scholar] [CrossRef]
  48. Moretti, J.; Blander, J.M. Increasing complexity of NLRP3 inflammasome regulation. J. Leukoc. Biol. 2021, 109, 561–571. [Google Scholar] [CrossRef]
  49. Niu, T.; De Rosny, C.; Chautard, S.; Rey, A.; Patoli, D.; Groslambert, M.; Cosson, C.; Lagrange, B.; Zhang, Z.; Visvikis, O.; et al. NLRP3 phosphorylation in its LRR domain critically regulates inflammasome assembly. Nat. Commun. 2021, 12, 5862. [Google Scholar] [CrossRef]
  50. Rathinam, V.A.K.; Vanaja, S.K.; Fitzgerald, K.A. Regulation of inflammasome signaling. Nat. Immunol. 2012, 13, 333–342. [Google Scholar] [CrossRef] [Green Version]
  51. Chauhan, D.; Walle, L.V.; Lamkanfi, M. Therapeutic modulation of inflammasome pathways. Immunol. Rev. 2020, 297, 123–138. [Google Scholar] [CrossRef]
  52. Dinarello, C.A. A clinical perspective of IL-1β as the gatekeeper of inflammation. Eur. J. Immunol. 2011, 41, 1203–1217. [Google Scholar] [CrossRef] [PubMed]
  53. Yasuda, K.; Nakanishi, K.; Tsutsui, H. Interleukin-18 in Health and Disease. Int. J. Mol. Sci. 2019, 20, 649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kesavardhana, S.; Kanneganti, T. Mechanisms governing inflammasome activation, assembly and pyroptosis induction. Int. Immunol. 2017, 29, 201–210. [Google Scholar] [CrossRef]
  55. Liu, X.; Zhang, Z.; Ruan, J.; Pan, Y.; Magupalli, V.G.; Wu, H.; Lieberman, J. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 2016, 535, 153–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Sagulenko, V.; Thygesen, S.J.; Sester, D.P.; Idris, A.; Cridland, J.A.; Vajjhala, P.R.; Roberts, T.L.; Schroder, K.; Vince, J.E.; Hill, J.M.; et al. AIM2 and NLRP3 inflammasomes activate both apoptotic and pyroptotic death pathways via ASC. Cell Death Differ. 2013, 20, 1149–1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Zheng, M.; Kanneganti, T. The regulation of the ZBP1-NLRP3 inflammasome and its implications in pyroptosis, apoptosis, and necroptosis (PANoptosis). Immunol. Rev. 2020, 297, 26–38. [Google Scholar] [CrossRef]
  58. Rathinam, V.A.K.; Fitzgerald, K.A. Inflammasome complexes: Emerging mechanisms and effector functions Inflammasome complex formation and canonical functions. Cell 2016, 165, 792–800. [Google Scholar] [CrossRef] [Green Version]
  59. Ciraci, C.; Janczy, J.R.; Sutterwala, F.S.; Cassel, S. Control of innate and adaptive immunity by the inflammasome. Microbes Infect. 2012, 14, 1263–1270. [Google Scholar] [CrossRef] [Green Version]
  60. Chen, M.; Wang, H.; Chen, W.; Meng, G. Regulation of adaptive immunity by the NLRP3 inflammasome. Int. Immunopharmacol. 2011, 11, 549–554. [Google Scholar] [CrossRef]
  61. Hutton, H.L.; Ooi, J.; Holdsworth, S.R.; Kitching, A.R. The NLRP3 inflammasome in kidney disease and autoimmunity. Nephrology 2016, 21, 736–744. [Google Scholar] [CrossRef]
  62. Papp, G.; Boros, P.; Nakken, B.; Szodoray, P.; Zeher, M. Regulatory immune cells and functions in autoimmunity and transplantation immunology. Autoimmun. Rev. 2017, 16, 435–444. [Google Scholar] [CrossRef] [Green Version]
  63. Zhang, Y.; Yang, W.; Li, W.; Zhao, Y. NLRP3 Inflammasome: Checkpoint Connecting Innate and Adaptive Immunity in Autoimmune Diseases. Front. Immunol. 2021, 12, 4166. [Google Scholar] [CrossRef] [PubMed]
  64. Shin, J.I.; Lee, K.H.; Joo, Y.H.; Lee, J.M.; Jeon, J.; Jung, H.J.; Shin, M.; Cho, S.; Kim, T.H.; Park, S.; et al. Inflammasomes and autoimmune and rheumatic diseases: A comprehensive review. J. Autoimmun. 2019, 103, 102299. [Google Scholar] [CrossRef] [PubMed]
  65. Zhen, Y.; Zhang, H. NLRP3 inflammasome and inflammatory bowel disease. Front. Immunol. 2019, 10, 276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zheng, Y.; Wang, Z.; Zhou, Z. miRNAs: Novel regulators of autoimmunity-mediated pancreatic β-cell destruction in type 1 diabetes. Cell. Mol. Immunol. 2017, 14, 488–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Xue, G.; Jin, G.; Fang, J.; Lu, Y. IL-4 together with IL-1β induces antitumor Th9 cell differentiation in the absence of TGF-β signaling. Nat. Commun. 2019, 10, 1376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Lewis, K.L.; Reizis, B. Dendritic cells: Arbiters of immunity and immunological tolerance. Cold Spring Harb. Perspect. Biol. 2012, 4, a007401. [Google Scholar] [CrossRef] [Green Version]
  69. Fernandes, F.P.; Leal, V.N.C.; De Lima, D.S.; Reis, E.C.; Pontillo, A. Inflammasome genetics and complex diseases: A comprehensive review. Eur. J. Hum. Genet. 2020, 28, 1307–1321. [Google Scholar] [CrossRef]
  70. Davidson, S.; Steiner, A.; Harapas, C.R.; Masters, S.L. An Update on Autoinflammatory Diseases: Interferonopathies. Curr. Rheumatol. Rep. 2018, 20, 38. [Google Scholar] [CrossRef]
  71. Yang, C.A.; Chiang, B.L. Inflammasomes and human autoimmunity: A comprehensive review. J. Autoimmun. 2015, 61, 1–8. [Google Scholar] [CrossRef]
  72. Almaani, S.; Meara, A.; Rovin, B.H. Update on Lupus Nephritis. Clin. J. Am. Soc. Nephrol. 2017, 12, 825–835. [Google Scholar] [CrossRef] [PubMed]
  73. Parikh, S.V.; Almaani, S.; Brodsky, S.; Rovin, B.H. Update on Lupus Nephritis: Core Curriculum 2020. Am. J. Kidney Dis. 2020, 76, 265–281. [Google Scholar] [CrossRef] [PubMed]
  74. Brinkmann, V.; Zychlinsky, A. Beneficial suicide: Why neutrophils die to make NETs. Nat. Rev. Microbiol. 2007, 5, 577–582. [Google Scholar] [CrossRef] [PubMed]
  75. Kahlenberg, J.M.; Thacker, S.G.; Berthier, C.C.; Cohen, C.D.; Kretzler, M.; Kaplan, M. Inflammasome Activation of IL-18 Results in Endothelial Progenitor Cell Dysfunction in Systemic Lupus Erythematosus. J. Immunol. 2011, 187, 6143–6156. [Google Scholar] [CrossRef] [Green Version]
  76. Hatef, M.R.; Sahebari, M.; Rezaieyazdi, Z.; Nakhjavani, M.R.; Mahmoudi, M. Stronger Correlation between Interleukin 18 and Soluble Fas in Lupus Nephritis Compared with Mild Lupus. ISRN Rheumatol. 2013, 2013, 1–6. [Google Scholar] [CrossRef] [Green Version]
  77. Perez-Alamino, R.; Cuchacovich, R.; Espinoza, L.R.; Porretta, C.P.; Zea, A. Role of Inflammasome Activation in Systemic Lupus Erythematosus: Are Innate Immune Cells Activated? Reum. Clin. 2021, 17, 187–191. [Google Scholar] [CrossRef]
  78. Hakkim, A.; Fürnrohr, B.G.; Amann, K.; Laube, B.; Abed, U.A.; Brinkmann, V.; Herrmann, M.; Voll, R.E.; Zychlinsky, A. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc. Natl. Acad. Sci. USA 2010, 107, 9813–9818. [Google Scholar] [CrossRef] [Green Version]
  79. Villanueva, E.; Yalavarthi, S.; Berthier, C.C.; Hodgin, J.B.; Khandpur, R.; Lin, A.M.; Rubin, C.J.; Zhao, W.; Olsen, S.H.; Klinder, M.; et al. Netting neutrophils induce endothelial damage, infiltrate tissues and expose immunostimulatory molecules in systemic lupus erythematosus. J. Immunol. 2011, 187, 538–552. [Google Scholar] [CrossRef] [Green Version]
  80. Kahlenberg, J.M.; Carmona-Rivera, C.; Smith, C.K.; Kaplan, M. Neutrophil Extracelullar Trap-associated Protein Activation of the NLRP3 Inflammasome Is Enhanced in Lupus Macrophages. J. Immunol. 2013, 190, 1217–1226. [Google Scholar] [CrossRef] [Green Version]
  81. Kahlenberg, J.M.; Yalavarthi, S.; Zhao, W.; Hodgin, J.B.; Reed, T.J.; Tsuji, N.M.; Kaplan, M. An essential role for caspase-1 in the induction of murine lupus and its associated vascular damage. Arthritis Rheumatol. 2014, 66, 152–162. [Google Scholar] [CrossRef] [Green Version]
  82. Zhao, J.; Wang, H.; Dai, C.; Wang, H.; Zhang, H.; Huang, Y.; Wang, S.; Gaskin, F.; Yang, N.; Fu, S.M. P2X7Blockade Attenuates Murine Lupus Nephritis by Inhibiting Activation of the NLRP3/ASC/Caspase 1 Pathway. Arthritis Care Res. 2013, 65, 3176–3185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Turner, C.M.; Tam, F.W.K.; Lai, P.-C.; Tarzi, R.M.; Burnstock, G.; Pusey, C.D.; Cook, H.T.; Unwin, R.J. Increased expression of the pro-apoptotic ATP-sensitive P2X7 receptor in experimental and human glomerulonephritis. Nephrol. Dial. Transplant. 2006, 22, 386–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Zhang, C.; Li, C.; Jia, S.; Yao, P.; Yang, Q.; Zhang, Y. High-Mobility Group Box 1 Inhibition Alleviates Lupus-Like Disease in BXSB Mice. Scand. J. Immunol. 2014, 79, 333–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Tak, P.P.; Firestein, G. NF-κB: A key role in inflammatory diseases. J. Clin. Investig. 2001, 107, 7–11. [Google Scholar] [CrossRef]
  86. Zheng, L.; Sinniah, R.; Hsu, S. Pathogenic role of NF-κB activation in tubulointerstitial inflammatory lesions in human lupus nephritis. J. Histochem. Cytochem. 2008, 56, 517–529. [Google Scholar] [CrossRef] [Green Version]
  87. Zhao, J.; Zhang, H.; Huang, Y.; Wang, H.; Wang, S.; Zhao, C.; Liang, Y.; Yang, N. Bay11-7082 attenuates murine lupus nephritis via inhibiting NLRP3 inflammasome and NF-κB activation. Int. Immunopharmacol. 2013, 17, 116–122. [Google Scholar] [CrossRef]
  88. Zhang, W.; Cai, Y.; Xu, W.; Yin, Z.; Gao, X.; Xiong, S. AIM2 facilitates the apoptotic DNA-induced systemic lupus erythematosus via arbitrating macrophage functional maturation. J. Clin. Immunol. 2013, 33, 925–937. [Google Scholar] [CrossRef]
  89. Yin, Q.; Sester, D.P.; Tian, Y.; Hsiao, Y.-S.; Lu, A.; Cridland, J.A.; Sagulenko, V.; Thygesen, S.J.; Choubey, D.; Hornung, V.; et al. Molecular Mechanism for p202-Mediated Specific Inhibition of AIM2 Inflammasome Activation. Cell Rep. 2013, 4, 327–339. [Google Scholar] [CrossRef] [Green Version]
  90. Pontillo, A.; Girardelli, M.; Kamada, A.J.; Pancotto, J.A.T.; Donadi, E.A.; Crovella, S.; Sandrin-Garcia, P. Polimorphisms in inflammasome genes are involved in the predisposition to systemic lupus erythematosus. Autoimmunity 2012, 45, 271–278. [Google Scholar] [CrossRef]
  91. Pontillo, A.; Vendramin, A.; Catamo, E.; Fabris, A.; Crovella, S. The missense variation Q705K in CIAS1/NALP3/NLRP3 gene and an NLRP1 haplotype are associated with celiac disease. Off. J. Am. Coll. Gastroenterol. ACG 2011, 106, 539–544. [Google Scholar] [CrossRef]
  92. Da Cruz, H.L.A.; Cavalcanti, C.A.J.; Silva, J.D.A.; De Lima, C.A.D.; Fragoso, T.S.; Barbosa, A.D.; Dantas, A.T.; Mariz, H.D.A.; Duarte, A.L.B.P.; Pontillo, A.; et al. Differential expression of the inflammasome complex genes in systemic lupus erythematosus. Immunogenetics 2020, 72, 217–224. [Google Scholar] [CrossRef] [PubMed]
  93. Jennette, J.C.; Nachman, P. ANCA glomerulonephritis and vasculitis. Clin. J. Am. Soc. Nephrol. 2017, 12, 1680–1691. [Google Scholar] [CrossRef] [PubMed]
  94. Villacorta, J.; Martínez-Valenzuela, L.; Martin-Capon, I.; Bordignon-Draibe, J. Antineutrophil Cytoplasmic Antibody-Associated Vasculitis: Toward an Individualized Approach. Nephron 2022, 146, 121–137. [Google Scholar] [CrossRef] [PubMed]
  95. Hewins, P.; Morgan, M.D.; Holden, N.; Neil, D.; Williams, J.M.; Savage, C.; Harper, L. IL-18 is upregulated in the kidney and primes neutrophil responsiveness in ANCA-associated vasculitis. Kidney Int. 2006, 69, 605–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Cannetti, C.A.; Leung, B.P.; Culshaw, S.; McInnes, I.B.; Cunha, F.Q.; Liew, F. IL-18 Enhances Collagen-Induced Arthritis by Recruiting Neutrophils Via TNF-α and Leukotriene B 4. J. Immunol. 2003, 171, 1009–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Shochet, L.; Holdsworth, S.; Kitching, A.R. Animal Models of ANCA Associated Vasculitis. Front. Immunol. 2020, 11, 525. [Google Scholar] [CrossRef] [Green Version]
  98. Schreiber, A.; Pham, C.T.N.; Hu, Y.; Schneider, W.; Luft, F.C.; Kettritz, R. Neutrophil serine proteases promote IL-1β generation and injury in necrotizing crescentic glomerulonephritis. J. Am. Soc. Nephrol. 2012, 23, 470–482. [Google Scholar] [CrossRef] [Green Version]
  99. Adkison, A.M.; Raptis, S.Z.; Kelley, D.G.; Pham, C. Dipeptidyl peptidase I activates neutrophil-derived serine proteases and regulates the development of acute experimental arthritis. J. Clin. Investig. 2002, 109, 363–371. [Google Scholar] [CrossRef]
  100. Schreiber, A.; Luft, F.C.; Kettritz, R. Phagocyte NADPH Oxidase Restrains the Inflammasome in ANCA-Induced GN. J. Am. Soc. Nephrol. 2014, 26, 411–424. [Google Scholar] [CrossRef] [Green Version]
  101. Gerderman, K.A.; Hultqvist, M.; Pizzola, A.; Zhao, M.; Nandakumar, K.S.; Mattsson, R.; Holmdahl, R. Macrophages suppress T cell responses and arthritis development in mice by producing reactive oxygen species. J. Clin. Investig. 2007, 117, 3020–3028. [Google Scholar] [CrossRef]
  102. Hultgren, O.; Andersson, B.; Hahn-Zoric, M.; Almroth, G. Serum concentration of interleukin-18 is up-regulated in patients with ANCA-associated vasculitis. Autoimmunity 2007, 40, 529–531. [Google Scholar] [CrossRef] [PubMed]
  103. Nakabayashi, K.; Sumiishi, A.; Sano, K.; Fujioka, Y.; Yamada, A.; Karube, M.; Koji, H.; Arimura, Y.; Nagasawa, T. Tubulointerstitial nephritis without glomerular lesions in three patients with myeloperoxidase-ANCA-associated vasculitis. Clin. Exp. Nephrol. 2009, 13, 605–613. [Google Scholar] [CrossRef] [PubMed]
  104. Plafkin, C.; Zhong, W.; Singh, T. ANCA vasculitis presenting with acute interstitial nephritis without glomerular involvement. Clin. Nephrol.—Case Stud. 2019, 7, 46–50. [Google Scholar] [CrossRef] [PubMed]
  105. Tashiro, M.; Sasatomi, Y.; Watanabe, R.; Watanabe, M.; Miyake, K.; Abe, Y.; Yasuno, T.; Ito, K.; Ueki, N.; Hamauchi, A.; et al. IL-1ß promotes tubulointerstitial injury in MPO-ANCA-associated glomerulonephritis. Clin. Nephrol. 2016, 86, 190–199. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, L.-Y.; Sun, X.-J.; Chen, M.; Zhao, M.-H. The expression of NOD2, NLRP3 and NLRC5 and renal injury in anti-neutrophil cytoplasmic antibody-associated vasculitis. J. Transl. Med. 2019, 17, 197. [Google Scholar] [CrossRef] [PubMed]
  107. Rodrigues, J.C.; Haas, M.; Reich, H. IgA nephropathy. Clin. J. Am. Soc. Nephrol. 2017, 12, 677–686. [Google Scholar] [CrossRef]
  108. Wardle, E. Cytokine growth factors and glomerulonephritis. Nephron 1991, 57, 257–261. [Google Scholar] [CrossRef]
  109. Rajasekaran, A.; Julian, B.A.; Rizk, D.V. IgA Nephropathy: An Interesting Autoimmune Kidney Disease. Am. J. Med. Sci. 2020, 361, 176–194. [Google Scholar] [CrossRef]
  110. Yoshioka, K.; Takemura, T.; Murakami, K.; Okada, M.; Yagi, K.; Miyazato, H.; Matsushima, K.; Maki, S. In situ expression of cytokines in IgA nephritis. Kidney Int. 1993, 44, 825–833. [Google Scholar] [CrossRef] [Green Version]
  111. Zou, J.-N.; Xiao, J.; Hu, S.-S.; Fu, C.-S.; Zhang, X.-L.; Zhang, Z.-X.; Lu, Y.-J.; Chen, W.-J.; Ye, Z.-B. Toll-like Receptor 4 Signaling Pathway in the Protective Effect of Pioglitazone on Experimental Immunoglobulin A Nephropathy. Chin. Med. J. 2017, 130, 906–913. [Google Scholar] [CrossRef]
  112. Chen, A.; Sheu, L.F.; Chou, W.Y.; Tsai, S.C.; Chang, D.M.; Liang, S.C.; Lin, F.G.; Lee, W. Interleukin-1 receptor antagonist modulates the progression of a spontaneously occurring IgA nephropathy in mice. Am. J. Kidney Dis. 1997, 30, 693–702. [Google Scholar] [CrossRef]
  113. Chun, J.; Chung, H.; Wang, X.; Barry, R.; Taheri, Z.M.; Platnich, J.; Ahmed, S.B.; Trpkov, K.; Hemmelgarn, B.; Benediktsson, H.; et al. NLRP3 Localizes to the Tubular Epithelium in Human Kidney and Correlates With Outcome in IgA Nephropathy. Sci. Rep. 2016, 6, 24667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Tsai, Y.-L.; Hua, K.-F.; Chen, A.; Wei, C.-W.; Chen, W.-S.; Wu, C.-Y.; Chu, C.-L.; Yu, Y.-L.; Lo, C.-W.; Ka, S.-M. NLRP3 inflammasome: Pathogenic role and potential therapeutic target for IgA nephropathy. Sci. Rep. 2017, 7, srep41123. [Google Scholar] [CrossRef] [PubMed]
  115. McAdoo, S.P.; Pusey, C. Anti-glomerular basement membrane disease. Clin. J. Am. Soc. Nephrol. 2017, 12, 1162–1172. [Google Scholar] [CrossRef] [Green Version]
  116. Gulati, K.; McAdoo, S.P. Anti–Glomerular Basement Membrane Disease. Rheum. Dis. Clin. N.Am. 2018, 44, 651–673. [Google Scholar] [CrossRef]
  117. Timoshanko, J.R.; Kitching, A.R.; Iwakura, Y.; Holdsworth, S.R.; Tipping, P. Contributions of IL-1β and IL-1α to Crescentic Glomerulonephritis in Mice. J. Am. Soc. Nephrol. 2004, 15, 910–918. [Google Scholar] [CrossRef] [Green Version]
  118. Kitching, A.R.; Turner, A.L.; Wilson, G.R.A.; Semple, T.; Odobasic, D.; Timoshanko, J.R.; O’Sullivan, K.M.; Tipping, P.G.; Takeda, K.; Akira, S.; et al. IL-12p40 and IL-18 in crescentic glomerulonephritis: IL-12p40 is the key Th1-defining cytokine chain, whereas IL-18 promotes local inflammation and leukocyte recruitment. J. Am. Soc. Nephrol. 2005, 16, 2023–2033. [Google Scholar] [CrossRef]
  119. Lan, H.Y.; Nikolic-Paterson, D.J.; Zarama, M.; Vannice, J.L.; Atkins, R. Suppression of experimental crescentic glomerulonephritis by deoxyspergualin. J. Am. Soc. Nephrol. 1993, 3, 1765–1774. [Google Scholar] [CrossRef]
  120. Tang, W.W.; Feng, L.; Vannice, J.L.; Wilson, C.B. Interleukin-1 receptor antagonist ameliorates experimental anti-glomerular basement membrane antibody-associated glomerulonephritis. J. Clin. Investig. 1994, 93, 273–279. [Google Scholar] [CrossRef] [Green Version]
  121. Lichtnekert, J.; Kulkarni, O.P.; Mulay, S.R.; Rupanagudi, K.V.; Ryu, M.; Allam, R.; Vielhauer, V.; Muruve, D.; Lindenmeyer, M.T.; Cohen, C.D.; et al. Anti-gbm glomerulonephritis involves il-1 but is independent of nlrp3/asc inflammasome-mediated activation of caspase-1. PLoS ONE 2011, 6, e26778. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Inflammasome structure and mechanism of activation. (a) Schematic representation of NLRP3 inflammasome assembly and detailed conformation of NLRP3 scaffold, an adaptor apoptosis speck-like protein (ASC) and the effector procaspase-1. (b) Structure of NLRP1, NLRP3, NLRC4 and AIM2 which participate in the formation of the main inflammasomes. NLR family members (NLRP1, NLRP3, NLRC4) contain leucine-rich repeats (LRR) and central nucleotide binding domain (NBD). The N-terminal PYD domain is present in NLRP subfamily members, whereas NLRC4 presents a CARD domain. NLRP1 also contains a C-terminal extension containing a function-to-find domain (FIIND) and a CARD domain. AIM2 is composed of a N-terminal PYD domain and a C-terminal HIN (hematopoietic, interferon-inducible and nuclear localization) domain. (c) NLRP3 activation pathways and effector functions. NLRP3 inflammasome assembly can be triggered by several ways: PAMPs and DAMPs detection via PRRs, by cytokine stimulation via IL-1 receptor (IL-1R) or through TNF link to tumor necrosis factor (TNF) receptors TNFR1 and TNFR2. These elements trigger the transcription of NF-κB, which promotes the transcription of NLRP3 and IL1B genes; this is the first signal or priming. The second signal or activation can be produced by ionic flux, K+ efflux, Ca2+ influx, Na+ influx and Cl- efflux, reactive oxygen species (ROS) and mitochondrial dysfunction or lysosomal damage. NLRP3 inflammasome assembly provokes IL-1β and IL-18 cytokines’ proteolytic maturation, which also participate in autoimmunity development and pyroptosis by the action of gasdermin-D. Protein myeloid differentiation primary response 88, MyD88; apoptosis signal-regulating kinase, ASK; kinases interleukin 1 receptor-associated kinase, IRAK; caspase-8, CASP8; Fas-associated protein with death domain, FADD; P2X purinoceptor 7, P2X7R; transient receptor potential melastatin, TRPM; transient receptor potential vanilloid, TRPV. Figure 1 has been created with BioRender.com.
Figure 1. Inflammasome structure and mechanism of activation. (a) Schematic representation of NLRP3 inflammasome assembly and detailed conformation of NLRP3 scaffold, an adaptor apoptosis speck-like protein (ASC) and the effector procaspase-1. (b) Structure of NLRP1, NLRP3, NLRC4 and AIM2 which participate in the formation of the main inflammasomes. NLR family members (NLRP1, NLRP3, NLRC4) contain leucine-rich repeats (LRR) and central nucleotide binding domain (NBD). The N-terminal PYD domain is present in NLRP subfamily members, whereas NLRC4 presents a CARD domain. NLRP1 also contains a C-terminal extension containing a function-to-find domain (FIIND) and a CARD domain. AIM2 is composed of a N-terminal PYD domain and a C-terminal HIN (hematopoietic, interferon-inducible and nuclear localization) domain. (c) NLRP3 activation pathways and effector functions. NLRP3 inflammasome assembly can be triggered by several ways: PAMPs and DAMPs detection via PRRs, by cytokine stimulation via IL-1 receptor (IL-1R) or through TNF link to tumor necrosis factor (TNF) receptors TNFR1 and TNFR2. These elements trigger the transcription of NF-κB, which promotes the transcription of NLRP3 and IL1B genes; this is the first signal or priming. The second signal or activation can be produced by ionic flux, K+ efflux, Ca2+ influx, Na+ influx and Cl- efflux, reactive oxygen species (ROS) and mitochondrial dysfunction or lysosomal damage. NLRP3 inflammasome assembly provokes IL-1β and IL-18 cytokines’ proteolytic maturation, which also participate in autoimmunity development and pyroptosis by the action of gasdermin-D. Protein myeloid differentiation primary response 88, MyD88; apoptosis signal-regulating kinase, ASK; kinases interleukin 1 receptor-associated kinase, IRAK; caspase-8, CASP8; Fas-associated protein with death domain, FADD; P2X purinoceptor 7, P2X7R; transient receptor potential melastatin, TRPM; transient receptor potential vanilloid, TRPV. Figure 1 has been created with BioRender.com.
Ijms 23 04208 g001
Table 1. Summary table: studies about the inflammasome’s role in glomerulonephritis.
Table 1. Summary table: studies about the inflammasome’s role in glomerulonephritis.
LUPUS NEPRHITIS
Significant FindingsReferences
IN VITRO MODEL
LL-37:
LDGs have the capacity to produce NETs which increase the externalization of immunostimulatory proteins and autoantigens as LL-37, IL-17 and dsDNA. Kidneys from SLE patients are infiltrated by netting neutrophils which show LL-37 and dsDNA explaining the role of aberrant lupus neutrophils the pathogenic role of NETs.Villanueva et al., Journal of Immunology, 2011.
NLRP3 is activated by NETs and the expression of the NETs-associated protein LL-37. This stimulus contributes to the production of IL1β and IL-18 causing NETosis.Kahlenberg et al., Journal of immunology, 2013.
Expression of axis’s inflammasome:
Isolated fresh monocytes from SLE patients increased inflammasome activation described by the elevated expression of Caspase-1, IL-1β and IL-18. Perez-Alamino et al., Reumatologia Clinica, 2021.
ANIMAL MODEL
P2X7:
Increased expression of P2X7 has been observed in kidney biopsies from patients with SLE.TTurner et al., Nephrology, dialysis, transplantation, 2007.
Upregulation of P2X7/NLRP3 in kidneys of MRL/lpr mice associates an increase in IL-1β and renal damage developing LN.P2X7 inhibition decreases autoantibodies and immune complexes deposited in the kidneys.Zhao et al., Arthritis Rheumatology, 2013.
NFκB and NLPR3:
The inhibition of NFκB and NLPR3 by Bay11-7082 in MRL/lpr mice reduces nephritis, the levels of IL-1β, TNF-α and anti-dsDNA and the deposition of immune complexes.Zhao et al., International Immunopharmacology, 2013.
AIM2:
AIM2 is augmented in macrophages induced by lymphocyte-derived apoptotic DNA. Its knock-down by siRNA ameliorates infiltration of macrophages in tissues.Zhang et al., Journal of Clinical Immunology, 2013.
p202 limits AIM2. This increases INF causing susceptibility to murine lupus.Yin et al., Cell reports, 2014.
Caspasa-1:
The caspase-1 −/− mouse model exposed to pristane protected against the development of autoantibodies related to SLE, nephritis and the action of type I INF.Kahlenberg et al., Arthritis and Rheumatology, 2014.
HMGB1:
Blocking HMGB1 in BXSB mice reduces the machinery of NLPR3 and improves renal inflammation.Zhang et al., Scandinavian Journal of immunology, 2014.
HUMAN MODEL
NLRP1:
The NLRP1 rs2670660 and NLRP1 rs12150220-rs2670660 A-G haplotype polymorphisms were associated with SLE and the event of nephritis, arthritis and rash.Pontillo et al., Autoimmunity, 2012.
NLPR3, NLRP1, Caspasa-1, AIM2:
The variant rs10754558 NLRP3 was more common in SLE patients with nephritis. The stimulus with LPS+ATP generated the expression of NLRP1, AIM2, CASP1 and IL1β genes, indicating that NLRP1 is responsible for the IL-β production reflected in monocytes.da Cruz et al., Immunogenetics, 2020.
ANCA GLOMERULONEPHRITIS
Significant FindingsReferences
IN VITRO MODEL
IL-18:
Il-18 expression is upregulated in patients with ANCA vasculitis.Hewins et al., Kidney International, 2006.
ANIMAL MODEL
NPS:
NE-/PR3- mice in anti-MPO antibody-induced model reduce local cytokines and induction of NCGN.Schreiber et al., Journal of the American Society of Nephrology, 2012.
NADPH oxidase:
An antibody-mediated anti-MPO model, gp91phox-deficient or p47phox-deficient mice had worsening NCGN. Gp91phox-deficient/caspase-1 double-deficient mice improved NCGN, suggesting that Phox limits the activity of caspase-1 and thus of the inflammasome.Schreiber et al., Journal of the American Nephrology, 2015.
HUMAN MODEL
IL-18:
IL-18 is elevated in the serum from patients diagnosed with ANCA vasculitis compared to healthy controls. The increase in IL-18 is regardless of MPO/PR3 levels. Hultren et al., Autoimmunity, 2007.
NLRP3, NOD2, NLRC5:
The investigators glimpsed the role of NLRP3 in the tubulointerstitial compartment and the correlation of IL-1β levels with the severity of tubulointerstitial injury in the glomerulus.Tashiro et al., Clinical Nephrology, 2016.
NOD2, NLRP3 and NLRC5 were mostly expressed in podocytes and in infiltrating monocytes and macrophages, but barely expressed in glomeruli. Wang et al., Journal of Translational Medicine, 2019.
IGA NEPHROPATHY
Significant FindingsReferences
ANIMAL MODEL
IL-1ra:
The use of Il-1 receptor antagonist in a IgAN’s mouse model (ddY mice) ceases the exacerbation of the disease.Chen et al., American Journal of Kidney Diseases, 1997.
HUMAN/IN VITRO MODEL
NLRP3 was mostly expressed in the tubules with no staining in the glomerulus of
normal kidneys. Nevertheless, in patients with IgAN, NLRP3 expression was
detected in the glomerulus, though it was more increased in the tubules. In
human kidney biopsies and in low passage human cells, they established
that NLRP3 was decreased during tubular damage. Equally the immunostaining
results and the NLRP3 mRNA expression confirmed the presence of NLRP3
and its subsequent loss after renal injury.
Chun et al., Scientific reports, 2016.
IgAN knockout NLRP3 mice model was generated. The production of IgA
immune complexes was inhibited by knockout mice. NLRP3 knockout mice and
the kidney-targeting delivery of shRNA of NLRP3 improve renal function.
Tsai et al., scientific reports, 2017.
ANTIGLOMERULAR BASEMENT MEMBRANE GLOMERULONEPHRITIS
Significant FindingsReferences
ANIMAL MODEL
IL-1ra:
IL-1ra protects against clinical and histological worsening in a rat anti-GBM
model.
Lan et al., Kidney International, 1993.
In a rat anti-GBM model, IL-1ra diminishs proteinuria and the expression of
adhesion molecules of PMN, such as ICAM-1.
Tang et al., The Journal of Clinical Investigation, 1994.
IL-18 and IL-12p40:
Anti-GBM mice model with IL-12p40−/−, IL-18−/− and both IL-12p40−/− and
IL-18 demonstrate IL-12p40 as a crucial cytokine chain in nephritogenic Th1
responses and IL-18 as a proinflammatory local (renal) cytokine.
Kitching et al., Journal of the American Society of Nephrology, 2005.
IL-1βand IL-1RI:
An anti-GBM IL-1β −/− and IL-1RI −/− mouse model was formed. IL-1β −/− mice
demonstrated a reduction in crescent formation and cell recruitment. IL-1RI −/−
mice presented less serum titers antibodies, less proteinuria and reduced serum
creatinine.
Timoshanko et al., Journal of the American Society of Nephrology, 2004.
Anti-GBM nephritis develops independently of the NLRP3-caspase-1 axis due
to the inability of glomerular cells to generate IL-1β.
Lichtnekert et al., Plos One, 2011.
LDG, low density granulocytes; NETs, neutrophils extracellular traps; dsDNA, anti-double-stranded DNA; SLE, systemic lupus erythematosus; LN, lupus nephritis; NFκB, nuclear factor kappa B; TNF-α, tumor necrosis factor α; siRNA, small interfering RNA; INF, interferon; HMGB1, high-mobility group box 1 protein; LPS, Lipopolysaccharide; ATP, adenosine triphosphate; CASP1, caspase-1; ANCA, Anti-Neutrophilic Cytoplasmic Autoantibody; NE, neutrophil elastase; PR3, proteinase 3; MPO, myeloperoxidase; NCGN, necrotizing crescentic glomerulonephritis; Phox, NADPH oxidase; PR3, proteinase 3; IgAN, IgA nephropathy; mRNA, messenger RNA; shRNA, short hairpin RNA; Il-1ra, interleukin-1 receptor antagonist; anti-GBM, anti-glomerular basement membrane; PMN, polymorphonuclear; ICAM-1, intercellular adhesion molecule-1; IL-1RI, IL1 type 1 receptor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Anton-Pampols, P.; Diaz-Requena, C.; Martinez-Valenzuela, L.; Gomez-Preciado, F.; Fulladosa, X.; Vidal-Alabro, A.; Torras, J.; Lloberas, N.; Draibe, J. The Role of Inflammasomes in Glomerulonephritis. Int. J. Mol. Sci. 2022, 23, 4208. https://doi.org/10.3390/ijms23084208

AMA Style

Anton-Pampols P, Diaz-Requena C, Martinez-Valenzuela L, Gomez-Preciado F, Fulladosa X, Vidal-Alabro A, Torras J, Lloberas N, Draibe J. The Role of Inflammasomes in Glomerulonephritis. International Journal of Molecular Sciences. 2022; 23(8):4208. https://doi.org/10.3390/ijms23084208

Chicago/Turabian Style

Anton-Pampols, Paula, Clara Diaz-Requena, Laura Martinez-Valenzuela, Francisco Gomez-Preciado, Xavier Fulladosa, Anna Vidal-Alabro, Joan Torras, Núria Lloberas, and Juliana Draibe. 2022. "The Role of Inflammasomes in Glomerulonephritis" International Journal of Molecular Sciences 23, no. 8: 4208. https://doi.org/10.3390/ijms23084208

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop