Skip to main content

Molecular biology of autoinflammatory diseases

Abstract

The long battle between humans and various physical, chemical, and biological insults that cause cell injury (e.g., products of tissue damage, metabolites, and/or infections) have led to the evolution of various adaptive responses. These responses are triggered by recognition of damage-associated molecular patterns (DAMPs) and/or pathogen-associated molecular patterns (PAMPs), usually by cells of the innate immune system. DAMPs and PAMPs are recognized by pattern recognition receptors (PRRs) expressed by innate immune cells; this recognition triggers inflammation. Autoinflammatory diseases are strongly associated with dysregulation of PRR interactomes, which include inflammasomes, NF-κB-activating signalosomes, type I interferon-inducing signalosomes, and immuno-proteasome; disruptions of regulation of these interactomes leads to inflammasomopathies, relopathies, interferonopathies, and proteasome-associated autoinflammatory syndromes, respectively. In this review, we discuss the currently accepted molecular mechanisms underlying several autoinflammatory diseases.

Background

The human body has evolved various adaptive responses that protect against cell and tissue damage caused by physical, chemical, and biological factors. Such factors include molecules released by damaged tissues, metabolites, and/or infection (e.g., by bacteria, viruses, and parasites) [1,2,3,4]. Inflammation, an adaptive response to cell injury, generates damage-associated molecular patterns (DAMPs) and/or pathogen-associated molecular patterns (PAMPs), which are then recognized by pattern recognition receptors (PRRs) expressed mainly by innate immune cells [5]. PRRs include Toll-like receptors (TLRs), Nod-like receptors (NLRs), C-type lectin receptors (CLRs), and RIG-I-like receptors (RLRs) that recognize DAMPs and PAMPs to initiate immune responses. These receptors are also called innate immune receptors [6] (Fig. 1).

Fig. 1
figure 1

Pattern-recognition receptors in innate immune cells. PRRs include Toll-like receptors (TLRs), Nod-like receptors (NLRs), and RIG-I-like receptors (RLRs) that recognize various damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) to initiate immune responses. These receptors are also called innate immune receptors. Gain-of-function mutations of the innate immune receptors or loss-of-function mutations of their inhibitors are related to autoinflammatory diseases. Red characters indicate DAMPs and PAMPs. Red characters indicate classified autoinflammatory diseases

Autoinflammatory diseases are strongly associated with dysregulation of these PRR-containing interactomes, which include inflammasomes, nuclear factor (NF)-κB-activating signalosomes, type I interferon-inducing signalosomes, and immuno-proteasomes; dysfunction of these interactomes results in inflammasomopathies, relopathies, interferonopathies, and proteasome-associated autoinflammatory syndromes (PRAAS), respectively [7,8,9,10,11]. This explains the pathogenesis of autoinflammatory diseases involving recurrent inflammatory flare-ups in the absence of autoantibodies or antigen-specific T lymphocytes [12]. Knowledge of the molecular mechanism(s) underlying the functions of these innate immune receptors is useful for the treatment and management of individuals with autoinflammatory diseases (Fig. 1).

Interleukin-1β-mediated autoinflammatory diseases (inflammasomopathies)

When NOD-like receptors harboring a PYRIN domain (PYD) (e.g., NLRP1, NLRP2, NLRP3, NLRP6, NLRP9, and NLRP12) and other pyrin domain-containing PRRs (e.g., pyrin, AIM2, and IFI-16) sense DAMPs, PAMPs, or intracellular microenvironmental changes (e.g., potassium efflux), they interact with an adaptor protein apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) via PYD, and pro-caspase-1 via a caspase-recruitment domain (CARD). This interaction activates caspase-1, a process accompanied by pyroptotic cell death [13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29]. NOD-like receptors carrying a CARD domain or CARD including proteins alternatively interact with caspase-1 via CARD with ASC and pro-caspase-1 such as NLRP1, NLRC4, CARD8, and caspase-11 [30,31,32]. The resulting complexes act as a sensor of cell injury; this sensor is referred to as the inflammasome, an interleukin (IL)-1β- and IL-18-processing platform that plays a crucial role in the maturation and secretion of these cytokines from cells. The process is accompanied by a type of cell death, named pyroptosis, which is triggered by cleavage of gasdermin D (GSDMD) [33, 34] (Fig. 2). Below, we discuss specific inflammasomopathies.

Fig. 2
figure 2

Cryopyrin-associated periodic fever syndrome (CAPS), TNF receptor-associated periodic syndrome (TRAPS), and autoinflammation and phospholipase Cγ2 (PLCγ2)-associated antibody deficiency and immune dysregulation (APLAID) are related to NLRP3 inflammasome. Gain-of-function mutations of NLRP3 (e.g., R260W) leads to prolonged activation of NLRP3 inflammasome. Autoinflammatory syndrome caused by the gain of function of NLRP1, NLRP12, or other NLRP mutations is thought to be basically caused by the same mechanisms. Mutated TNFRSF1A (TNFR) in patients with TRAPS is misfolded and accumulated in the endoplasmic reticulum (ER), causing ER stress and increased generation of mitochondrial reactive oxygen species (ROS) that activates the NLRP3 inflammasome. PLCγ2 mutation in patients with APLAID (e.g., S707Y) leads to calcium influx from the ER and increased cytoplasmic Ca2+ levels promote activation of NLRP3 inflammasome

Cryopyrin-associated periodic syndrome

Cryopyrin is the same protein as NLRP3 which was named by the nomenclature committee. Gain-of-function mutations in NLRP3 lead to cryopyrin-associated periodic syndrome (CAPS), a spectrum of diseases that includes familial cold autoinflammatory syndrome (FCAS, formerly termed familial cold urticaria (FCU)), Muckle–Wells syndrome (MWS), and neonatal-onset multisystem inflammatory disease (NOMID; also called chronic infantile neurologic cutaneous and articular syndrome (CINCA)). Currently, 248 variants of the CIAS1 gene have been reported by “INFEVERS” (https://infevers.umai-montpellier.fr/web/search.php?n=4) [35]. The NLRP3 mutations in CAPS result in constitutive activation of the NLRP3 inflammasome (i.e., the threshold for stimulation is extremely low). Activation of the inflammasome leads to excess pyroptosis of cells expressing components of the NLRP3 inflammasome; these cells secrete excessive amounts of activated IL-1β upon autoinflammatory attack [36,37,38,39,40,41] (Fig. 2). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Table 1 The corresponding diseases caused by the similar signaling of autoinflammatory diseases

NLRP1-associated autoinflammation with arthritis and dyskeratosis

The NLRP1 inflammasome was the first “inflammasome” to be identified [14]. NLRP1 interacts with ASC through its PYD domain. ASC then interacts with pro-caspase-1 via its CARD domain, resulting in activation of IL-1β secretion; also, NLRP1 interacts with caspase-1 through its CARD domain to activate IL-1β secretion [96]. Currently, several mutations (A54T, A59P, A66V, M77T, R726W, T755N, F787_R843del, and P1214R) in the gene encoding NLRP1 have been identified (https://infevers.umai-montpellier.fr/web/search.php?n=31). Patients harboring these mutations exhibit dyskeratosis, oligo/polyarthritis, and recurrent fever, along with immunological dysfunction and vitamin A deficiency [97,98,99]. The mutations may trigger proteasome-dependent functional degradation of NLRP1, and degraded CARD-FIIND-containing-NLRP1 fragments act as a scaffold like ASC for inflammasome activation [100] (Fig. 2). Corresponding common diseases caused by the similar signaling are shown in Table 1.

NLRP12 autoinflammatory syndrome

NLRP12 inhibits the activation of NF-κB. Mutations in NLRP12 are found in patients with hereditary periodic fever syndrome, the clinical signs of which are consistent with a diagnosis of CAPS [101]. Currently, 79 variants of the gene encoding NLRP12 have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=9). Since some patients with gain-of-function mutations in NLRP12 exhibit symptoms similar to those of CAPS, the disease was named FCAS2 [102] and patients with NALP12 periodic fever syndrome respond to canakinumab (an anti-human IL-1β monoclonal antibody) and/or etanercept (a tumor necrosis factor (TNF) receptor-IgG heavy chain chimeric protein that acts as a bivalent antagonist of TNF activity) [102], the pathogenesis of NLRP12 autoinflammatory syndrome (NLRP12-AD) may explain the gain of function of the NLRP12 inflammasome by a similar mechanism of the NLRP3 inflammasome (Fig. 2). Corresponding common diseases caused by the similar signaling are shown in Table 1.

TNF receptor-associated periodic fever syndrome

The causative gene product of TNF receptor-associated periodic fever syndrome (TRAPS) is TNF receptor superfamily member 1A (TNFRSF1A) [12]. So far, 180 variations of the TNFRSF1A gene have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=2). The cysteine-to-cysteine disulfide bonds in the extracellular domain of TNFRSF1A for ER stress are thought to be important for disease pathogenesis. More than one-third of patients with TRAPS harbor the R92Q and P46L mutations [103]. In TRAPS, misfolding of mutated TNFRSF1A leads to accumulation of the protein in the endoplasmic reticulum (ER), which causes ER stress and increased generation of mitochondrial reactive oxygen species; this in turn activates inflammasomes [104, 105] (Fig. 2). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Autoinflammation and phospholipase Cγ2-associated antibody deficiency and immune dysregulation

Autoinflammation and phospholipase Cγ2 (PLCγ2)-associated antibody deficiency and immune dysregulation (APLAID) responds to PLCγ2 which encodes for a constitutively repressed phospholipase. The S707Y PLCγ2 mutation disrupts the autoinhibition of PLCγ2, thereby increasing PLCγ2 activity and calcium influx from the ER in the leukocytes of patients with APLAID [106, 107]. Increased cytoplasmic Ca2+ levels promote the assembly of the NLRP3 inflammasome [108] (Fig. 2). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Familial Mediterranean fever

The causative gene of familial Mediterranean fever (FMF), MEFV, encodes pyrin (also named marenostrin) [109, 110]. Currently, 389 variants of MEFV have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=1). FMF was reported to be autosomal recessive; mutations in pyrin are thought to result in loss of its ability to inhibit inflammasomes. Nowadays, pyrin assembles with ASC and pro-caspase-1 to form the pyrin inflammasome, as well as the NLRP3 inflammasome [111]. Usually, pyrin is phosphorylated by serine/threonine-protein kinases PKN1 and PKN2, and inhibited by 14-3-3 proteins. When virulence factors expressed or secreted by bacteria and/or viruses inhibit RhoA GTPase, the pyrin inflammasome triggers activation and secretion of IL-1β [112] (Fig. 3). Yersinia pestis-like bacteria have a YopM protein which interacts with pyrin to inhibit inflammatory responses for avoiding further anti-bacterial responses [113]. In patients with FMF, pyrin harboring mutant human B30.2 domains defect such kind of ability, thereby preventing binding to ASC; this makes prolonged inflammasome activation and IL-1β secretion [114] (Fig. 3). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 3
figure 3

Prolonged activation of pyrin inflammasome in patients with familial Mediterranean fever (FMF). Upon Yersinia pestis-like bacterial infection, Yersinia outer protein (Yop)M interacts with pyrin to inhibit pyrin inflammasome to avoid further anti-bacterial responses. In patients with FMF, pyrin-harboring mutant human B30.2 domains enable to interact with YopM, resulting in prolonged pyrin inflammasome activation and IL-1β secretion

Periodic fever immunodeficiency and thrombocytopenia

The causative gene product of periodic fever immunodeficiency and thrombocytopenia (PFIT) is WDR1 [115, 116], which interacts with cofilin to promote cleavage and depolymerization of F-actin [117, 118]. The L293F mutation in WDR1 disrupts intramolecular hydrophobic interactions, which are important for maintaining actin protein structure. This disruption leads to actin accumulation and aggregates with pyrin resulting in pyrin activation and release of IL-18 [119] (Fig. 4). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 4
figure 4

Activation of pyrin inflammasome is inhibited by phosphorylation by PKN1/2. Upon bacterial infection, bacterial toxin can inhibit PKN1/2 activity, resulting in pyrin inflammasome activation. The L293F mutation in WDR1 leads to actin accumulation and aggregates with pyrin, resulting in pyrin activation

Pyrin-associated autoinflammation with neutrophilic dermatosis

The MEFV mutations in patients with pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND) harbor S242R and E244K mutations in pyrin; these mutations are located in the 14-3-3 binding motif, which interferes with binding of pyrin to 14-3-3, thereby allowing assembly of the pyrin inflammasome and excessive release of IL-1β [120,121,122,123] (Fig. 5). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 5
figure 5

Activation of pyrin inflammasome in patients with pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND). Pyrin mutations in patients with PAAND (e.g., S242R and E244K) mutations are located in the 14-3-3 binding motif, which interferes with the binding of pyrin to 14-3-3, thereby allowing activation of the pyrin inflammasome

Pyogenic arthritis, pyoderma gangrenosum, and acne syndrome

The causative gene product of pyogenic arthritis, pyoderma gangrenosum, and acne (PAPA) syndrome is proline-serine-threonine phosphatase-interacting protein 1 (PSTPIP1) (also called CD2-binding protein 1 (CD2BP1)) [124, 125]. Currently, 66 variants of the PSTPIP1 gene have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=5). In patients with PAPA syndrome, mutations in PSTPIP1 result in hyperphosphorylation of PSTPIP1, which strengthens its interaction with pyrin via the B-box domain to activate the pyrin inflammasome. This leads to increased secretion of IL-1β [125] (Fig. 6). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 6
figure 6

Activation of pyrin inflammasome in patients with pyogenic arthritis, pyoderma gangrenosum, and acne (PAPA) syndrome. Mutations of PSTPIP1 in patients with PAPA (e.g., A230T and E250Q) result in hyperphosphorylation of PSTPIP1, which strengthens its interaction with pyrin via the B-box domain to activate the pyrin inflammasome

Mevalonate kinase deficiency/hyper-IgD syndrome

The causative gene product of mevalonate kinase deficiency/hyper-IgD syndrome (MKD) (also known as hyper-IgD syndrome (HIDS)) is mevalonate kinase (MVK) [126]. Currently, 264 variants of this gene have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=3). Geranylgeranyl pyrophosphate, the substrate of geranylgeranylation, is a product of the mevalonate pathway. Deficiency of MVK leads to depletion of geranylgeranyl pyrophosphate, resulting in the inactivation of RhoA [127, 128]. Since the inactivation of RhoA activates the pyrin inflammasome, MKD leads to an inflammasomopathy. Indeed, canakinumab, an anti-IL-1β monoclonal antibody, is an effective treatment for MKD, suggesting that IL-1β is a common mediator of these diseases [129] (Fig. 7). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 7
figure 7

Activation of pyrin inflammasome in patients with hyper-IgD syndrome (HIDS). Deficiency of MVK activity by loss-of-function mutations leads to depletion of geranylgeranyl pyrophosphate, resulting in the inactivation of RhoA and following activation of PKN1/2. This causes activation of pyrin inflammasome

NLRC4 inflammasomopathies

Gain-of-function mutations in NLRC4 result in early-onset recurrent fever and macrophage activation syndrome (MAS), neonatal-onset enterocolitis with periodic fever, fatal or near-fatal episodes of autoinflammation, or symptoms resembling those of FCAS [68, 130, 131]. So far, more than 31 genetic variants of NLRC4 have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=25). The NLRC4 inflammasome activates caspase-1 either with or without an adaptor ASC, which in turn activates IL-1β and IL-18. NLRC4 inflammasomopathies are linked more closely with hypersecretion of IL-18 rather than of IL-1β; however, the precise mechanism remains to be elucidated [132] (Fig. 8). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 8
figure 8

NLRC4 inflammasomopathies are related to gain-of-function mutations of NLRC4 inflammasome. Gain-of-function of mutations in NLRC4 in patients with NLRC4 inflammasomopathies constitutively actives for NLRC4 inflammasome. The NLRC4 inflammasome activates caspase-1 either with or without an adaptor ASC leading to hypersecretion of IL-18 and IL-1β

NF-κB-related autoinflammatory diseases (relopathies)

Dysregulations of NF-κB signaling are closely linking to the ubiquitination system. In addition to constitutive activation of NF-κB, loss-of-function mutations in the ubiquitin-mediated NF-κB regulatory system cause autoinflammatory diseases [10] (Fig. 9).

Fig. 9
figure 9

Autoinflammatory disease-related NF-κB activation pathway. In normal, upon recognition of DAMPs, PAMPs, or appropriate ligands, TLR, TNFR, Nod1, and Nod2 activate NF-κB. Dysregulations of the NF-κB signaling are closely linking to autoinflammatory diseases called relopathies. The NF-κB activation pathway is regulated by polyubiquitination chains

Blau syndrome/early-onset sarcoidosis

The gene responsible for Blau syndrome (BS)/early-onset sarcoidosis (EOS) is IBD1, and its causative gene product is NOD2 [133]. Usually, NOD2 recognizes muramyl dipeptide (MDP), leading to activation of NF-κB. Currently, 185 variants of NOD2 have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=6). Gain-of-function mutations in NOD2 increase signaling via NOD2-RIPK2-associated activation of NF-κB [134, 135] (Fig. 10). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 10
figure 10

NF-κB activation pathway in patients with Blau syndrome (BS)/early-onset sarcoidosis (EOS). NOD2 recognizes muramyl dipeptide (MDP), a minimum component of peptidoglycan, leading to activation of NF-κB. Gain-of-function mutations in NOD2 (e.g., R334W) increase signaling via NOD2-RIPK2-associated activation of NF-κB

A20 protein haploinsufficiency

A20 (also called TNF-α-induced protein (TNFAIP) 3, is an intracellular deubiquitinase. A20 plays a role in deubiquitination of several proteins, including NF-κB. A20 protein haploinsufficiency (HA20) is caused by heterozygous mutation or deletion of A20, resulting in insufficient deubiquitination of TRAF6 downstream of the TNF-α pathway, RIPK1 downstream of the TLR pathway, and RIPK2 downstream of the NOD1 or NOD2 pathways. Loss of A20 function leads to constitutive activation of NF-κB signaling [136, 137]. A20 also regulates the activity of the NLRP3 inflammasome in macrophages [138]. So far, 55 variants of A20 have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=26). Haplodeficient mutations severely reduce A20 function, leading to prolonged activation of NF-κB [139] (Fig. 11). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 11
figure 11

NF-κB activation pathway in patients with A20 protein haploinsufficiency (HA20). Loss-of-function mutations of A20 (e.g., L227X) reduce the deubiquitination activity of A20 leading to prolonged activation of NF-κB

Immunodeficiency, autoinflammation, and amylopectinosis with inherited linear ubiquitin chain assembly complex deficiency

Loss-of-function mutation in linear ubiquitin chain assembly complex (LUBAC), a protein complex comprising heme-oxidized IRP2 ubiquitin ligase 1 (HOIL-1) (also called RBCK1), HOIL-1 interaction protein (HOIP, also called RNF31), and SHANK-associated RH domain-interacting protein (SHARPIN) is associated with autoinflammation [140,141,142,143,144,145]. The L72P mutation in the HOIP protein affects its interaction with OTU deubiquitinase with linear linkage specificity (OTULIN) and lysine 63 deubiquitinase (CYLD); however, the most common disease-causing phenomenon is loss of expression of the L72P allele of HOIP. Combined heteromutations comprise L41fsX7 and Q185X, which result in deficient HOIL-1 expression. Lack of HOIL-1 expression by fibroblasts impairs phosphorylation of IKK kinase, slower degradation of IκBα, and decreased ubiquitination of NEMO in response to stimulation with either TNF-α or IL-1β. LUBAC deficiency in fibroblasts downregulates NF-κB activation in response to IL-1β or TNF-α, whereas deficient monocytes release more IL-6 but less IL-10 in response to IL-1β [146,147,148] (Fig. 12). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 12
figure 12

Loss-of-function mutation in linear ubiquitin chain assembly complex (LUBAC) is associated with autoinflammation. Loss-of-function mutations in the HOIP (e.g., L72P) affects its interaction with OTU deubiquitinase with linear linkage specificity (OTULIN) and lysine 63 deubiquitinase (CYLD), leading to prolonged NF-κB activation

OTULIN-related autoinflammatory syndrome

OTULIN is a deubiquitination enzyme that hydrolyzes methionine-1 (M1), which links to liner ubiquitin chains to regulate the activity of NF-κB [149]. Homozygous loss-of-function mutations in OTULIN cause OTULIN-related autoinflammatory syndrome (ORAS) [150]. The L272P mutation is located in a helix of the catalytic OTU domain, which forms part of the binding pocket for M1-linked distal ubiquitin; this mutation disrupts the binding of OTULIN and ubiquitin to its substrate [151, 152] (Fig. 13). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 13
figure 13

Homozygous loss-of-function mutations in OTULIN in patients with OTULIN-related autoinflammatory syndrome (ORAS). OTULIN is a deubiquitination enzyme that hydrolyzes methionine-1 (M1), which links to liner ubiquitin chains to regulate the activity of NF-κB. The L272P mutation of OTULIN disrupts the binding of OTULIN and ubiquitin to its substrate, leading to prolonged NF-κB activation

IL-1 receptor-related autoinflammatory diseases

IL-1 receptor-related autosomal recessive autoinflammatory diseases are caused by mutations in IL1RN (interleukin-1 receptor antagonist), resulting in a condition called deficiency of interleukin-1 receptor antagonist (DIRA) [153,154,155]. So far, 22 variants of this gene have been reported (https://infevers.umai-montpellier.fr/web/search.php?n=10). IL-1RA deficiency results in uncontrolled IL-1α, IL-1β and NF-κB signaling [156] (Fig. 9). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Interferonopathies

Anti-viral first-line defense is dependent on innate immune receptors (e.g., cGAS, MDA5, and RIG-I) that are detecting intracellular viral, bacterial, or own nucleic acid, linking to type I interferon signaling. Interferonopathies are closely linked to dysfunction of these innate immune receptors and type I interferon signaling, Immunoproteasome dysfunction is also linked to the interferonopathies [157] (Fig. 14).

Fig. 14
figure 14

Type I interferon signaling in interferonopathies. Type I interferon singling is closely linked to innate immune receptors such as cGAS, MDA5, and RIG-I that are sensing viral, bacterial, or own DNA or RNA. Interferon-stimulated genes (ISGs) are induced by interferon regulatory factors (IRFs) downstream of cGAS, MDA5, and RIG-I. Immunoproteasome dysfunction is also linked to interferonopathies

Aicardi–Goutières syndrome

Aicardi–Goutières syndrome (AGS) is an inherited encephalopathy that affects newborn infants and usually results in severe neuro-physical disability. AGS is caused by loss-of-function mutations in the genes encoding the three prime repair exonuclease 1 (TREX1), the ribonuclease H2 subunit (RNASEH2)A, RNASEH2B, RNASEH2C, the phosphohydrolase SAM domain and HD domain-containing protein 1 (SAMHD1), or the dsRNA-specific adenosine deaminases acting on RNA1 (ADAR1) [158, 159]. In addition, gain-of-function mutations in the dsRNA sensor MDA5 (also called IFIH1) have been identified in AGS patients [157]. AGS pathology seems to be caused by the accumulation of nucleic acids, which can cause neurological and liver abnormalities that resemble congenital viral infection (Fig. 15). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 15
figure 15

Loss-of-function mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, or ADAR1 in patients with Aicardi–Goutières syndrome (AGS). Loss-of-function mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, or ADAR1 results in the accumulation of nucleic acids which leads to the induction of IRF transcription

Stimulator of interferon gene-associated vasculopathy with onset in infancy

Stimulator of interferon gene (STING)-associated vasculopathy with onset in infancy (SAVI) is caused by gain-of-function mutations in STING (also called TMEM173). Mutation of the STING amplifies the function of STING, which is an adaptor molecule involved in signal transduction through cGAS, leading to hyperactivation of type I IFN pathways [160]. Corresponding common diseases caused by the similar signaling are shown in Table 1.

Coatomer protein alpha syndromes

Coatomer protein alpha (COPA) syndrome, characterized by high-titer autoantibodies, interstitial lung disease, and inflammatory arthritis, was found to be deleterious mutations in the COPA gene (encoding coatomer subunit α). Mutant COPA causes defective intracellular transport via coat protein complex I which leads to ER stress and the upregulation of the levels of transcripts encoding IL-1β, IL-6, and IL-23 [161]. COPA is a critical regulator of STING transport ER and retrieval of STING from the Golgi. Mutant COPA retention of STING on the Golgi resulting in STING activation leads to prolonged type I interferon signaling [86] (Fig. 16). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 16
figure 16

Mutations in αCOP in patients with COPA syndrome. Mutations in αCOP (e.g., K230N) impair ER-Golgi transport resulting in ER stress-induced NF-κB activation and the mutant αCOP retention of STING on the Golgi resulting in STING activation leads to prolonged type I interferon signaling

Proteasome-associated autoinflammatory syndromes

Nakajo–Nishimura syndrome (NNS) and chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature syndrome (CANDLE) were the first PRAAS to be described. Loss-of-function mutation in immunoproteasome components such as proteasome subunit beta type (PSMB)8, PSMB4, PSMA3, PSMB9, or proteasome maturation protein (POMP) leads to increased secretion of type I IFN by immune cells [162, 163] (Fig. 17). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 17
figure 17

Loss-of-function mutations in immunoproteasome components PSMB8, PSMB4, PSMA3, PSMB9, or proteasome maturation protein (POMP) in patients with PRAAS/NNS/CANDLE. Loss-of-function mutation in immunoproteasome components such as PSMB8, PSMB4, PSMA3, PSMB9, or proteasome maturation protein (POMP) leads to increased secretion of type I IFN by immune cells

Singleton–Merten syndrome

Singleton–Merten syndrome (SMS) is caused by gain-of-function mutations in the RNA sensor MDA5 or RIG-I. Typical SMS is caused by a mutation in MDA5, whereas atypical SMS is caused by a mutation in RIG-I; both mutations cause constitutive activation of IFN signaling pathways [164, 165]. Notably, mutations in the MDA5 are also associated with AGS, so that both SMS and AGS share a common molecular mechanism [164] (Fig. 18). Corresponding common diseases caused by the similar signaling are shown in Table 1.

Fig. 18
figure 18

Gain-of-function mutations in MDA5 or RIG-I in patients with Singleton-Merten syndrome (SMS). Typical SMS is caused by gain-of-function mutations in MDA5. Atypical SMS is caused by gain-of-function mutations in RIG-I. Gain-of-function mutations of MDA5 and RIG-I lead to constitutive activation of type I IFN signaling pathways

Conclusions

Here, we describe briefly the molecular mechanisms underlying autoinflammatory diseases caused by dysregulation of IL-1β or IL-18 processing, NF-κB activation, and IFN secretion. Disruption of the fine balance within these signaling pathways contributes to the pathogenesis of autoinflammatory diseases. Increasing our knowledge of the molecular biology underlying autoinflammatory diseases will facilitate the development of disease-targeting biologics. Therefore, future studies should elucidate the autoinflammatory disease-specific signalosome in detail.

Availability of data and materials

Not applicable.

Abbreviations

DAMP:

Damage-associated molecular pattern

PAMP:

Pathogen-associated molecular pattern

PRR:

Pattern recognition receptor

TLR:

Toll-like receptor

NLR:

NOD-like receptor

CLR:

C-type lectin receptor

RLR:

RIG-I-like receptor

NF:

Nuclear factor

PRAAS:

Proteasome-associated autoinflammatory syndromes

IL:

Interleukin

PYD:

Pyrin domain

AIM2:

Absent in melanoma 2

ASC:

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

CARD:

Caspase-recruitment domain

GSDMD:

Gasdermin D

CAPS:

Cryopyrin-associated periodic syndrome

FCAS:

Familial cold autoinflammatory syndrome

FCU:

Familial cold urticaria

MWS:

Muckle–Wells syndrome

NOMID:

Neonatal-onset multisystem inflammatory disease

CINCA:

Chronic infantile neurologic, cutaneous, and arthritis

NAIAD:

NLRP1-associated autoinflammation with arthritis and dyskeratosis

NLRP12-AD:

NLRP12 autoinflammatory syndrome

TNF:

Tumor necrosis factor

TRAPS:

TNF receptor-associated periodic fever syndrome

TNFRSF:

TNF receptor superfamily member

APLAID:

Autoinflammation and PLCγ2-associated antibody deficiency and immune dysregulation

FMF:

Familial Mediterranean fever

PFIT:

Periodic fever immunodeficiency and thrombocytopenia

PAAND:

Pyrin-associated autoinflammation with neutrophilic dermatosis

PAPA:

Pyogenic arthritis, pyoderma gangrenosum, and acne

PSTPIP1:

Proline-serine-threonine phosphatase-interacting protein 1

CD2BP1:

CD2-binding protein 1

MKD:

Mevalonate kinase deficiency

HIDS:

Hyperimmunoglobulinemia D and periodic fever syndrome

MVK:

Mevalonate kinase

MAS:

Macrophage activation syndrome

BS:

Blau syndrome

EOS:

Early-onset sarcoidosis

MDP:

Muramyl dipeptide

HA20:

A20 protein haploinsufficiency

TNFAIP:

Tumor necrosis factor alpha-induced protein

IAALUCD:

Immunodeficiency, autoinflammation, and amylopectinosis with inherited linear ubiquitin chain assembly complex deficiency

LUBAC:

Linear ubiquitin chain assembly complex

HOIL-1:

Heme-oxidized IRP2 ubiquitin ligase 1

HOIP:

HOIL-1 interaction protein

SHARPIN:

SHANK-associated RH domain-interacting protein

OTULIN:

OTU deubiquitinase with linear linkage specificity

CYLD:

CYLD lysine 63 deubiquitinase

ORAS:

OTULIN-related autoinflammatory syndrome

DIRA:

Deficiency of the IL-1-receptor antagonist

AGS:

Aicardi–Goutières syndrome

TREX:

Three prime repair exonuclease

RNASEH2:

Ribonuclease H2 subunit

SAMHD:

SAM domain and HD domain-containing protein

ADAR1:

dsRNA-specific adenosine deaminases acting on RNA 1

STING:

Stimulator of interferon genes

SAVI:

STING-associated vasculopathy with onset in infancy

COPA:

Coatomer protein alpha

NNS:

Nakajo–Nishimura syndrome

CANDLE:

Chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature syndrome

PSMB:

Proteasome subunit beta type

POMP:

Proteasome maturation protein

SMS:

Singleton–Merten syndrome

References

  1. Kumar V, Abbas AK, Aster JC. Robbins & Cotran pathologic basis of disease. 9th ed. Amsterdam: Elsevier B.V; 2015.

    Google Scholar 

  2. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–7. https://doi.org/10.1038/nature05485.

    Article  CAS  PubMed  Google Scholar 

  3. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 2010;140(6):900–17. https://doi.org/10.1016/j.cell.2010.02.034.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011;29(1):415–45. https://doi.org/10.1146/annurev-immunol-031210-101322.

    Article  CAS  PubMed  Google Scholar 

  5. Janeway CA Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol. 1989;54(Pt 1):1–13. https://doi.org/10.1101/sqb.1989.054.01.003.

    Article  CAS  PubMed  Google Scholar 

  6. Tang T, Gong T, Jiang W, Zhou R. GPCRs in NLRP3 Inflammasome activation, regulation, and therapeutics. Trends Pharmacol Sci. 2018;39(9):798–811. https://doi.org/10.1016/j.tips.2018.07.002.

    Article  CAS  PubMed  Google Scholar 

  7. Masters SL, Simon A, Aksentijevich I, Kastner DL. Horror autoinflammaticus: the molecular pathophysiology of autoinflammatory disease (*). Annu Rev Immunol. 2009;27(1):621–68. https://doi.org/10.1146/annurev.immunol.25.022106.141627.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jones JF, Minnich LM, Lucas DO, Langford MP, Stanton GJ. Does an interferonopathy underlie a severe combined immunodeficiency disease (SCID) syndrome? Birth Defects Orig Artic Ser. 1983;19(3):109–14.

    CAS  PubMed  Google Scholar 

  9. Crow YJ. Type I interferonopathies: a novel set of inborn errors of immunity. Ann N Y Acad Sci. 2011;1238(1):91–8. https://doi.org/10.1111/j.1749-6632.2011.06220.x.

    Article  CAS  PubMed  Google Scholar 

  10. Moghaddas F, Masters SL. Monogenic autoinflammatory diseases: Cytokinopathies. Cytokine. 2015;74(2):237–46. https://doi.org/10.1016/j.cyto.2015.02.012.

    Article  CAS  PubMed  Google Scholar 

  11. McDermott A, Jacks J, Kessler M, Emanuel PD, Gao L. Proteasome-associated autoinflammatory syndromes: advances in pathogeneses, clinical presentations, diagnosis, and management. Int J Dermatol. 2015;54(2):121–9. https://doi.org/10.1111/ijd.12695.

    Article  PubMed  Google Scholar 

  12. McDermott MF, Aksentijevich I, Galon J, McDermott EM, Ogunkolade BW, Centola M, et al. Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell. 1999;97:133–44. https://doi.org/10.1016/s0092-8674(00)80721-7.

    Article  CAS  PubMed  Google Scholar 

  13. Poyet JL, Razmara M, Datta P, Zhang Z, Alnemri ES. The PYRIN-CARD protein ASC is an activating adaptor for caspase-1. Srinivasula SM, J Biol Chem. 2002;277:21119-21122. doi: https://doi.org/10.1074/jbc.C200179200, 24.

    Article  CAS  PubMed  Google Scholar 

  14. Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol Cell. 2002;10(2):417–26. https://doi.org/10.1016/S1097-2765(02)00599-3.

    Article  CAS  PubMed  Google Scholar 

  15. Bruey JM, Bruey-Sedano N, Newman R, Chandler S, Stehlik C, Reed JC. PAN1/NALP2/PYPAF2, an inducible inflammatory mediator that regulates NF-κB and caspase-1 activation in macrophages. J Biol Chem. 2004;279(50):51897–907. https://doi.org/10.1074/jbc.M406741200.

    Article  CAS  PubMed  Google Scholar 

  16. Manji GA, Wang L, Geddes BJ, Brown M, Merriam S, Al-Garawi A, et al. PYPAF1, a PYRIN-containing Apaf1-like protein that assembles with ASC and regulates activation of NF-kappa B. J Biol Chem. 2002;277(13):11570–5. https://doi.org/10.1074/jbc.M112208200.

    Article  PubMed  Google Scholar 

  17. Agostini L, Martinon F, Burns K, McDermott MF, Hawkins PN, Tschopp J. NALP3 forms an IL-1β-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity. 2004;20(3):319–25. https://doi.org/10.1016/s1074-7613(04)00046-9.

    Article  CAS  PubMed  Google Scholar 

  18. Grenier JM, Wang L, Manji GA, Huang WJ, Al-Garawi A, Kelly R, et al. Functional screening of five PYPAF family members identifies PYPAF5 as a novel regulator of NF-kappaB and caspase-1. FEBS Lett. 2002;530(1-3):73–8. https://doi.org/10.1016/s0014-5793(02)03416-6.

    Article  CAS  PubMed  Google Scholar 

  19. Wang L, Manji GA, Grenier JM, Al-Garawi A, Merriam S, Lora JM, et al. PYPAF7, a novel PYRIN-containing Apaf1-like protein that regulates activation of NF-κB and caspase-1-dependent cytokine processing. J Biol Chem. 2002;277(33):29874–80. https://doi.org/10.1074/jbc.M203915200.

    Article  CAS  PubMed  Google Scholar 

  20. Ngo C, Man SM. NLRP9b: a novel RNA-sensing inflammasome complex. Cell Res. 2017;27(11):1302–3. https://doi.org/10.1038/cr.2017.93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhu S, Ding S, Wang P, Wei Z, Pan W, Palm NW, et al. Nlrp9b inflammasome restricts rotavirus infection in intestinal epithelial cells. Nature. 2017;546(7660):667–70. https://doi.org/10.1038/nature22967.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Antushevich H. Interplays between inflammasomes and viruses, bacteria (pathogenic and probiotic), yeasts and parasites. Immunol Lett. 2020;228:1–14. https://doi.org/10.1016/j.imlet.2020.09.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. DeYoung KL, Ray ME, Su YA, Anzick SL, Johnstone RW, Trapani JA, et al. Cloning a novel member of the human interferon-inducible gene family associated with control of tumorigenicity in a model of human melanoma. Oncogene. 1997;15(4):453–7. https://doi.org/10.1038/sj.onc.1201206.

    Article  CAS  PubMed  Google Scholar 

  24. Bürckstümmer T, Baumann C, Blüml S, Dixit E, Dürnberger G, Jahn H, et al. An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat Immunol. 2009;10(3):266–72. https://doi.org/10.1038/ni.1702.

    Article  CAS  PubMed  Google Scholar 

  25. Fernandes-Alnemri T, Yu JW, Datta P, Wu J, Alnemri ES. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature. 2009;458(7237):509–13. https://doi.org/10.1038/nature07710.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature. 2009;458(7237):514–8. https://doi.org/10.1038/nature07725.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schroder K, Muruve DA, Tschopp J. Innate immunity: cytoplasmic DNA sensing by the AIM2 inflammasome. Curr Biol. 2009;19(6):R262–5. https://doi.org/10.1016/j.cub.2009.02.011.

    Article  CAS  PubMed  Google Scholar 

  28. Choubey D, Duan X, Dickerson E, Ponomareva L, Panchanathan R, Shen H, et al. Interferon-inducible p200-family proteins as novel sensors of cytoplasmic DNA: role in inflammation and autoimmunity. J Interf Cytokine Res. 2010;30(6):371–80. https://doi.org/10.1089/jir.2009.0096.

    Article  CAS  Google Scholar 

  29. Kerur N, Veettil MV, Sharma-Walia N, Bottero V, Sadagopan S, Otageri P, et al. IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection. Cell Host Microbe. 2011;9(5):363–75. https://doi.org/10.1016/j.chom.2011.04.008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Poyet JL, Srinivasula SM, Tnani M, Razmara M, Fernandes-Alnemri T, Alnemri ES. Identification of Ipaf, a human caspase-1-activating protein related to Apaf-1. J Biol Chem. 2001;276(30):28309–13. https://doi.org/10.1074/jbc.C100250200.

    Article  CAS  PubMed  Google Scholar 

  31. Razmara M, Srinivasula SM, Wang L, Poyet JL, Geddes BJ, DiStefano PS, et al. CARD-8 protein, a new CARD family member that regulates caspase-1 activation and apoptosis. J Biol Chem. 2002;277(16):13952–8. https://doi.org/10.1074/jbc.M107811200.

    Article  CAS  PubMed  Google Scholar 

  32. Taabazuing CY, Griswold AR, Bachovchin DA. The NLRP1 and CARD8 inflammasomes. Immunol Rev. 2020;297(1):13–25. https://doi.org/10.1111/imr.12884.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660–5. https://doi.org/10.1038/nature15514.

    Article  CAS  PubMed  Google Scholar 

  34. Kayagaki N, Stowe IB, Lee BL, O'Rourke K, Anderson K, Warming S, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015;526(7575):666–71. https://doi.org/10.1038/nature15541.

    Article  CAS  PubMed  Google Scholar 

  35. Sarrauste de Menthière C, Terrière S, Pugnère D, Ruiz M, Demaille J, Touitou I. INFEVERS: the registry for FMF and hereditary inflammatory disorders mutations. Nucleic Acids Res. 2003;31:282–5. https://doi.org/10.1093/nar/gkg031.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner RD. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet. 2001;29(3):301–5. https://doi.org/10.1038/ng756.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Neven B, Callebaut I, Prieur AM, Feldmann J, Bodemer C, Lepore L, et al. Molecular basis of the spectral expression of CIAS1 mutations associated with phagocytic cell-mediated autoinflammatory disorders CINCA/NOMID, MWS, and FCU. Blood. 2004;103(7):2809–15. https://doi.org/10.1182/blood-2003-07-2531.

    Article  CAS  PubMed  Google Scholar 

  38. Dowds TA, Masumoto J, Zhu L, Inohara N, Núñez G. Cryopyrin-induced interleukin 1β secretion in monocytic cells: enhanced activity of disease-associated mutants and requirement for ASC. J Biol Chem. 2004;279(21):21924–8. https://doi.org/10.1074/jbc.M401178200.

    Article  CAS  PubMed  Google Scholar 

  39. Fujisawa A, Kambe N, Saito M, Nishikomori R, Tanizaki H, Kanazawa N, et al. Disease-associated mutations in CIAS1 induce cathepsin B-dependent rapid cell death of human THP-1 monocytic cells. Blood. 2007;109(7):2903–11. https://doi.org/10.1182/blood-2006-07-033597.

    Article  CAS  PubMed  Google Scholar 

  40. Saito M, Nishikomori R, Kambe N, Fujisawa A, Tanizaki H, Takeichi K, et al. Disease-associated CIAS1 mutations induce monocyte death, revealing low-level mosaicism in mutation-negative cryopyrin-associated periodic syndrome patients. Blood. 2008;111(4):2132–41. https://doi.org/10.1182/blood-2007-06-094201.

    Article  CAS  PubMed  Google Scholar 

  41. Yamazaki T, Masumoto J, Agematsu K, Sawai N, Kobayashi S, Shigemura T, et al. Anakinra improves sensory deafness in a Japanese patient with Muckle-Wells syndrome, possibly by inhibiting the cryopyrin inflammasome. Arthritis Rheum. 2008;58(3):864–8. https://doi.org/10.1002/art.23261.

    Article  CAS  PubMed  Google Scholar 

  42. Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–41. https://doi.org/10.1038/nature04516.

    Article  CAS  PubMed  Google Scholar 

  43. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010;464(7293):1357–61. https://doi.org/10.1038/nature08938.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Masters SL, Dunne A, Subramanian SL, Hull RL, Tannahill GM, Sharp FA, et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1beta in type 2 diabetes. Nat Immunol. 2010;11(10):897–904. https://doi.org/10.1038/ni.1935.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–8. https://doi.org/10.1038/nature11729.

    Article  CAS  PubMed  Google Scholar 

  46. Saresella M, La Rosa F, Piancone F, Zoppis M, Marventano I, Calabrese E, et al. The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer’s disease. Mol Neurodegener. 2016;11(1):23. https://doi.org/10.1186/s13024-016-0088-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Codolo G, Plotegher N, Pozzobon T, Brucale M, Tessari I, Bubacco L, et al. Triggering of inflammasome by aggregated α-synuclein, an inflammatory response in synucleinopathies. PLoS One. 2013;8(1):e55375. https://doi.org/10.1371/journal.pone.0055375.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Johann S, Heitzer M, Kanagaratnam M, Goswami A, Rizo T, Weis J, et al. NLRP3 inflammasome is expressed by astrocytes in the SOD1 mouse model of ALS and in human sporadic ALS patients. Glia. 2015;63(12):2260–73. https://doi.org/10.1002/glia.22891.

    Article  PubMed  Google Scholar 

  49. Inoue M, Shinohara ML. NLRP3 Inflammasome and MS/EAE. Autoimmune Dis. 2013;2013:859145–8. https://doi.org/10.1155/2013/859145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kayagaki N, Wong MT, Stowe IB, Ramani SR, Gonzalez LC, Akashi-Takamura S, et al. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science. 2013;341(6151):1246–9. https://doi.org/10.1126/science.1240248.

    Article  CAS  PubMed  Google Scholar 

  51. Hagar JA, Powell DA, Aachoui Y, Ernst RK, Miao EA. Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science. 2013;341(6151):1250–3. https://doi.org/10.1126/science.1240988.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Sandanger Ø, Ranheim T, Vinge LE, Bliksøen M, Alfsnes K, Finsen AV, et al. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury. Cardiovasc Res. 2013;99(1):164–74. https://doi.org/10.1093/cvr/cvt091.

    Article  CAS  PubMed  Google Scholar 

  53. Fann DY, Lee SY, Manzanero S, Tang SC, Gelderblom M, Chunduri P, et al. Intravenous immunoglobulin suppresses NLRP1 and NLRP3 inflammasome-mediated neuronal death in ischemic stroke. Cell Death Dis. 2013;4(9):e790. https://doi.org/10.1038/cddis.2013.326.

    Article  CAS  PubMed  Google Scholar 

  54. Averette KM, Pratt MR, Yang Y, Bassilian S, Whitelegge JP, Loo JA, et al. Anthrax lethal toxin induced lysosomal membrane permeabilization and cytosolic cathepsin release is Nlrp1b/Nalp1b-dependent. PLoS One. 2009;4(11):e7913. https://doi.org/10.1371/journal.pone.0007913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Vladimer GI, Weng D, Paquette SW, Vanaja SK, Rathinam VA, Aune MH, et al. The NLRP12 inflammasome recognizes Yersinia pestis. Immunity. 2012 Jul 27;37(1):96–107. https://doi.org/10.1016/j.immuni.2012.07.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ataide MA, Andrade WA, Zamboni DS, Wang D, Souza Mdo C, Franklin BS, et al. Malaria-induced NLRP12/NLRP3-dependent caspase-1 activation mediates inflammation and hypersensitivity to bacterial superinfection. PLoS Pathog. 2014;10(1):e1003885. https://doi.org/10.1371/journal.ppat.1003885.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zaki MH, Man SM, Vogel P, Lamkanfi M, Kanneganti TD. Salmonella exploits NLRP12-dependent innate immune signaling to suppress host defenses during infection. Proc Natl Acad Sci U S A. 2014;111(1):385–90. https://doi.org/10.1073/pnas.1317643111.

    Article  CAS  PubMed  Google Scholar 

  58. Silveira TN, Gomes MT, Oliveira LS, Campos PC, Machado GG, Oliveira SC. NLRP12 negatively regulates proinflammatory cytokine production and host defense against Brucella abortus. Eur J Immunol. 2017;47(1):51–9. https://doi.org/10.1002/eji.201646502.

    Article  CAS  PubMed  Google Scholar 

  59. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci U S A. 1975;72(9):3666–70. https://doi.org/10.1073/pnas.72.9.3666.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kutukculer N, Topyildiz E, Berdeli A, Guven Bilgin B, Aykut A, Durmaz A, et al. Four diseases, PLAID, APLAID, FCAS3 and CVID and one gene (PHOSPHOLIPASE C, GAMMA-2; PLCG2): Striking clinical phenotypic overlap and difference. Clin Case Rep. 2021;9(4):2023–31. https://doi.org/10.1002/ccr3.3934.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet. 2017;49:1373–84. https://doi.org/10.1038/ng.3916.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. van der Lee SJ, Conway OJ, Jansen I, Carrasquillo MM, Kleineidam L, van den Akker E, et al. A nonsynonymous mutation in PLCG2 reduces the risk of Alzheimer's disease, dementia with Lewy bodies and frontotemporal dementia, and increases the likelihood of longevity. Acta Neuropathol. 2019;138:237–50. https://doi.org/10.1007/s00401-019-02026-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chung LK, Park YH, Zheng Y, Brodsky IE, Hearing P, Kastner DL, et al. The Yersinia virulence factor YopM hijacks host kinases to inhibit type III effector-triggered activation of the pyrin inflammasome. Cell Host Microbe. 2016;20(3):296–306. https://doi.org/10.1016/j.chom.2016.07.018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Talman AM, Chong R, Chia J, Svitkina T, Agaisse H. Actin network disassembly powers dissemination of Listeria monocytogenes. J Cell Sci. 2014;127:240–9. https://doi.org/10.1242/jcs.140038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Janssen WJM, Grobarova V, Leleux J, Jongeneel L, van Gijn M, van Montfrans JM, et al. Proline-serine-threonine phosphatase interacting protein 1 (PSTPIP1) controls immune synapse stability in human T cells. J Allergy Clin Immunol. 2018;142(6):1947–55. https://doi.org/10.1016/j.jaci.2018.01.030.

    Article  CAS  PubMed  Google Scholar 

  66. Yvan-Charvet L, Bonacina F, Guinamard RR, Norata GD. Immunometabolic function of cholesterol in cardiovascular disease and beyond. Cardiovasc Res. 2019;115(9):1393–407. https://doi.org/10.1093/cvr/cvz127.

    Article  CAS  PubMed  Google Scholar 

  67. Sutterwala FS, Mijares LA, Li L, Ogura Y, Kazmierczak BI, Flavell RA. Immune recognition of Pseudomonas aeruginosa mediated by the IPAF/NLRC4 inflammasome. J Exp Med. 2007;204(13):3235–45. https://doi.org/10.1084/jem.20071239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Canna SW, de Jesus AA, Gouni S, Brooks SR, Marrero B, Liu Y, et al. An activating NLRC4 inflammasome mutation causes autoinflammation with recurrent macrophage activation syndrome. Nat Genet. 2014;46(10):1140–6. https://doi.org/10.1038/ng.3089.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gandotra S, Jang S, Murray PJ, Salgame P, Ehrt S. Nucleotide-binding oligomerization domain protein 2-deficient mice control infection with Mycobacterium tuberculosis. Infect Immun. 2007;75(11):5127–34. https://doi.org/10.1128/IAI.00458-07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Adrianto I, Wen F, Templeton A, Wiley G, King JB, Lessard CJ, et al. Association of a functional variant downstream of TNFAIP3 with systemic lupus erythematosus. Nat Genet. 2011;43:253–8. https://doi.org/10.1038/ng.766.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Moaaz M, Mohannad N. Association of the polymorphisms of TRAF1 (rs10818488) and TNFAIP3 (rs2230926) with rheumatoid arthritis and systemic lupus erythematosus and their relationship to disease activity among Egyptian patients. Cent Eur J Immunol. 2016;41(2):165–75. https://doi.org/10.5114/ceji.2016.60991.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Vozandychova V, Stojkova P, Hercik K, Rehulka P, Stulik J. The ubiquitination system within bacterial host-pathogen interactions. Microorganisms. 2021;9(3):638. https://doi.org/10.3390/microorganisms9030638.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. van Wijk SJL, Fricke F, Herhaus L, Gupta J, Hötte K, Pampaloni F, et al. Linear ubiquitination of cytosolic Salmonella Typhimurium activates NF-κB and restricts bacterial proliferation. Nat Microbiol. 2017;2(7):17066. https://doi.org/10.1038/nmicrobiol.2017.66.

    Article  CAS  PubMed  Google Scholar 

  74. Kaneko N, Kurata M, Yamamoto T, Morikawa S, Masumoto J. The role of interleukin-1 in general pathology. Inflamm Regen. 2019;39(1):12. https://doi.org/10.1186/s41232-019-0101-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Pendergraft WF 3rd, Means TK. AGS, SLE, and RNASEH2 mutations: translating insights into therapeutic advances. J Clin Invest. 2015;125(1):102–4. https://doi.org/10.1172/JCI78533.

    Article  PubMed  Google Scholar 

  76. Kim H, Sanchez GA, Goldbach-Mansky R. Insights from Mendelian interferonopathies: comparison of CANDLE, SAVI with AGS, monogenic lupus. J Mol Med (Berl). 2016;94(10):1111–27. https://doi.org/10.1007/s00109-016-1465-5.

    Article  CAS  Google Scholar 

  77. Xu J, Liu H, Yang Y, Wang X, Liu P, Li Y, et al. Genome-wide profiling of cervical RNA-binding proteins identifies human papillomavirus regulation of RNASEH2A expression by viral E7 and E2F1. mBio. 2019;10:e02687–18. https://doi.org/10.1128/mBio.02687-18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Saiada F, Zhang K, Li R. PIAS1 potentiates the anti-EBV activity of SAMHD1 through SUMOylation. Cell Biosci. 2021;11(1):127. https://doi.org/10.1186/s13578-021-00636-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Martinat C, Cormier A, Tobaly-Tapiero J, Palmic N, Casartelli N, Mahboubi B, et al. SUMOylation of SAMHD1 at Lysine 595 is required for HIV-1 restriction in non-cycling cells. Nat Commun. 2021;12(1):4582. https://doi.org/10.1038/s41467-021-24802-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wang L, Sun Y, Song X, Wang Z, Zhang Y, Zhao Y, et al. Hepatitis B virus evades immune recognition via RNA adenosine deaminase ADAR1-mediated viral RNA editing in hepatocytes. Cell Mol Immunol. 2021;18(8):1871–82. https://doi.org/10.1038/s41423-021-00729-1.

    Article  CAS  PubMed  Google Scholar 

  81. Khadka S, Williams CG, Sweeney-Gibbons J, Basler CF. 3’ untranslated regions of Marburg and Ebola virus mRNAs possess negative regulators of translation that are modulated by ADAR1 editing. J Virol. 2021:JVI0065221. https://doi.org/10.1128/JVI.00652-21 Online ahead of print.

  82. Andrejeva J, Childs KS, Young DF, Carlos TS, Stock N, Goodbourn S, et al. The V proteins of paramyxoviruses bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of the IFN-beta promoter. Proc Natl Acad Sci U S A. 2004;101(49):17264–9. https://doi.org/10.1073/pnas.0407639101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, et al. Reis e Sousa C, Matsuura Y, Fujita T, Akira S. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441(7089):101–5. https://doi.org/10.1038/nature04734.

    Article  CAS  PubMed  Google Scholar 

  84. Staels F, Betrains A, Doubel P, Willemsen M, Cleemput V, Vanderschueren S, et al. Adult-onset ANCA-associated vasculitis in SAVI: extension of the phenotypic spectrum, case report and review of the literature. Front Immunol. 2020;11:575219. https://doi.org/10.3389/fimmu.2020.575219.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol. 2015;15(12):760–70. https://doi.org/10.1038/nri3921.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Deng Z, Chong Z, Law CS, Mukai K, Ho FO, Martinu T, et al. A defect in COPI-mediated transport of STING causes immune dysregulation in COPA syndrome. J Exp Med. 2020;217(11):e20201045. https://doi.org/10.1084/jem.20201045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Ichikawa HT, Conley T, Muchamuel T, Jiang J, Lee S, Owen T, et al. Beneficial effect of novel proteasome inhibitors in murine lupus via dual inhibition of type I interferon and autoantibody-secreting cells. Arthritis Rheum. 2012;64(2):493–503. https://doi.org/10.1002/art.33333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Kasahara M. Role of immunoproteasomes and thymoproteasomes in health and disease. Pathol Int. 2021;71(6):371–82. https://doi.org/10.1111/pin.13088.

    Article  CAS  PubMed  Google Scholar 

  89. Hutchinson S, Sims S, O'Hara G, Silk J, Gileadi U, Cerundolo V, et al. A dominant role for the immunoproteasome in CD8+ T cell responses to murine cytomegalovirus. PLoS One. 2011;6(2):e14646. https://doi.org/10.1371/journal.pone.0014646.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Robek MD, Garcia ML, Boyd BS, Chisari FV. Role of immunoproteasome catalytic subunits in the immune response to hepatitis B virus. J Virol. 2007;81(2):483–91. https://doi.org/10.1128/JVI.01779-06.

    Article  CAS  PubMed  Google Scholar 

  91. Van Kaer L, Ashton-Rickardt PG, Eichelberger M, Gaczynska M, Nagashima K, Rock KL, et al. Altered peptidase and viral-specific T cell response in LMP2 mutant mice. Immunity. 1994;1(7):533–41. https://doi.org/10.1016/1074-7613(94)90043-4.

    Article  PubMed  Google Scholar 

  92. Zieba BA, Henry L, Lacroix M, Jemaà M, Lavabre-Bertrand T, Meunier L, et al. The proteasome maturation protein POMP increases proteasome assembly and activity in psoriatic lesional skin. J Dermatol Sci. 2017;88(1):10–9. https://doi.org/10.1016/j.jdermsci.2017.04.009.

    Article  CAS  PubMed  Google Scholar 

  93. Boner W, Morgan IM. Novel cellular interacting partners of the human papillomavirus 16 transcription/replication factor E2. Virus Res. 2002;90(1-2):113–8. https://doi.org/10.1016/s0168-1702(02)00145-4.

    Article  CAS  PubMed  Google Scholar 

  94. Oliveira L, Sinicato NA, Postal M, Appenzeller S, Niewold TB. Dysregulation of antiviral helicase pathways in systemic lupus erythematosus. Front Genet. 2014;5:418. https://doi.org/10.3389/fgene.2014.00418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Kato H, Fujita T. RIG-I-like receptors and autoimmune diseases. Curr Opin Immunol. 2015;37:40–5. https://doi.org/10.1016/j.coi.2015.10.002.

    Article  CAS  PubMed  Google Scholar 

  96. Masters SL, Gerlic M, Metcalf D, Preston S, Pellegrini M, O’Donnell JA, et al. NLRP1 inflammasome activation induces pyroptosis of hematopoietic progenitor cells. Immunity. 2012;37(6):1009–23. https://doi.org/10.1016/j.immuni.2012.08.027.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Grandemange S, Sanchez E, Louis-Plence P, Tran Mau-Them F, Bessis D, Coubes C, et al. A new autoinflammatory and autoimmune syndrome associated with NLRP1 mutations: NAIAD (NLRP1-associated autoinflammation with arthritis and dyskeratosis). Ann Rheum Dis. 2017;76(7):1191–8. https://doi.org/10.1136/annrheumdis-2016-210021.

    Article  CAS  PubMed  Google Scholar 

  98. Soler VJ, Tran-Viet KN, Galiacy SD, Limviphuvadh V, Klemm TP, St Germain E, et al. Whole exome sequencing identifies a mutation for a novel form of corneal intraepithelial dyskeratosis. J Med Genet. 2013;50(4):246–54. https://doi.org/10.1136/jmedgenet-2012-101325.

    Article  CAS  PubMed  Google Scholar 

  99. Herlin T, Jørgensen SE, Høst C, Mitchell PS, Christensen MH, Laustsen M, et al. Autoinflammatory disease with corneal and mucosal dyskeratosis caused by a novel NLRP1 variant. Rheumatology (Oxford). 2020;59:2334–9. https://doi.org/10.1093/rheumatology/kez612.

    Article  Google Scholar 

  100. Sandstrom A, Mitchell PS, Goers L, Mu EW, Lesser CF, Vance RE. Functional degradation: a mechanism of NLRP1 inflammasome activation by diverse pathogen enzymes. Science. 2019;364:eaau1330. https://doi.org/10.1126/science.aau1330.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Jéru I, Duquesnoy P, Fernandes-Alnemri T, Cochet E, Yu JW, Lackmy-Port-Lis M, et al. Mutations in NALP12 cause hereditary periodic fever syndromes. Proc Natl Acad Sci U S A. 2008;105(5):1614–9. https://doi.org/10.1073/pnas.0708616105.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Del Porto F, Cifani N, Proietta M, Verrecchia E, Di Rosa R, Manna R, et al. NLRP12 gene mutations and auto-inflammatory diseases: ever-changing evidence. Rheumatology (Oxford). 2020;59(11):3129–36. https://doi.org/10.1093/rheumatology/keaa304.

    Article  Google Scholar 

  103. Lachmann HJ, Papa R, Gerhold K, Obici L, Touitou I, Cantarini L, et al. Paediatric Rheumatology International Trials Organisation (PRINTO), the EUROTRAPS and the Eurofever Project. The phenotype of TNF receptor-associated autoinflammatory syndrome (TRAPS) at presentation: a series of 158 cases from the Eurofever/EUROTRAPS international registry. Ann Rheum Dis. 2014;73:2160–7. https://doi.org/10.1136/annrheumdis-2013-204184.

    Article  CAS  PubMed  Google Scholar 

  104. Bulua AC, Simon A, Maddipati R, Pelletier M, Park H, Kim KY, et al. Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). J Exp Med. 2011;208(3):519–33. https://doi.org/10.1084/jem.20102049.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Simon A, Park H, Maddipati R, Lobito AA, Bulua AC, Jackson AJ, et al. Concerted action of wild-type and mutant TNF receptors enhances inflammation in TNF receptor 1-associated periodic fever syndrome. Proc Natl Acad Sci U S A. 2010;107(21):9801–6. https://doi.org/10.1073/pnas.0914118107.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Zhou Q, Lee GS, Brady J, Datta S, Katan M, Sheikh A, et al. A hypermorphic missense mutation in PLCG2, encoding phospholipase Cγ2, causes a dominantly inherited autoinflammatory disease with immunodeficiency. Am J Hum Genet. 2012;91(4):713–20. https://doi.org/10.1016/j.ajhg.2012.08.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Chae JJ, Park YH, Park C, Hwang IY, Hoffmann P, Kehrl JH, et al. Connecting two pathways through Ca 2+ signaling: NLRP3 inflammasome activation induced by a hypermorphic PLCG2 mutation. Arthritis Rheum. 2015;67(2):563–7. https://doi.org/10.1002/art.38961.

    Article  CAS  Google Scholar 

  108. Lee GS, Subramanian N, Kim AI, Aksentijevich I, Goldbach-Mansky R, Sacks DB, et al. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature. 2012;492(7427):123–7. https://doi.org/10.1038/nature11588.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. The International FMF Consortium. Ancient missense mutations in a new member of the RoRet gene family are likely to cause familial Mediterranean fever. Cell. 1997;90(4):797–807. https://doi.org/10.1016/s0092-8674(00)80539-5.

    Article  Google Scholar 

  110. French FMF Consortium. A candidate gene for familial Mediterranean fever. Nat Genet. 1997;17(1):25–31. https://doi.org/10.1038/ng0997-25.

    Article  Google Scholar 

  111. Xu H, Yang J, Gao W, Li L, Li P, Zhang L, et al. Innate immune sensing of bacterial modifications of Rho GTPases by the Pyrin inflammasome. Nature. 2014;513(7517):237–41. https://doi.org/10.1038/nature13449.

    Article  CAS  PubMed  Google Scholar 

  112. Park YH, Wood G, Kastner DL, Chae JJ. Pyrin inflammasome activation and RhoA signaling in the autoinflammatory diseases FMF and HIDS. Nat Immunol. 2016;17(8):914–21. https://doi.org/10.1038/ni.3457.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Park YH, Remmers EF, Lee W, Ombrello AK, Chung LK, Shilei Z, et al. Ancient familial Mediterranean fever mutations in human pyrin and resistance to Yersinia pestis. Nat Immunol. 2020;21(8):857–67. https://doi.org/10.1038/s41590-020-0705-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Chae JJ, Cho YH, Lee GS, Cheng J, Liu PP, Feigenbaum L, et al. Gain-of-function Pyrin mutations induce NLRP3 protein-independent interleukin-1β activation and severe autoinflammation in mice. Immunity. 2011;34(5):755–68. https://doi.org/10.1016/j.immuni.2011.02.020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Kile BT, Panopoulos AD, Stirzaker RA, Hacking DF, Tahtamouni LH, Willson TA, et al. Mutations in the cofilin partner Aip1/Wdr1 cause autoinflammatory disease and macrothrombocytopenia. Blood. 2007;110(7):2371–80. https://doi.org/10.1182/blood-2006-10-055087.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Kim ML, Chae JJ, Park YH, De Nardo D, Stirzaker RA, Ko HJ, et al. Aberrant actin depolymerization triggers the pyrin inflammasome and autoinflammatory disease that is dependent on IL-18, not IL-1β. J Exp Med. 2015;212(6):927–38. https://doi.org/10.1084/jem.20142384.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Rodal AA, Tetreault JW, Lappalainen P, Drubin DG, Amberg DC. Aip1p interacts with cofilin to disassemble actin filaments. J Cell Biol. 1999;145(6):1251–64. https://doi.org/10.1083/jcb.145.6.1251.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Okada K, Obinata T, Abe H. XAIP1: a Xenopus homologue of yeast actin interacting protein 1 (AIP1), which induces disassembly of actin filaments cooperatively with ADF/cofilin family proteins. J Cell Sci. 1999;112(Pt 10):1553–65. https://doi.org/10.1242/jcs.112.10.1553.

    Article  CAS  PubMed  Google Scholar 

  119. Standing AS, Malinova D, Hong Y, Record J, Moulding D, Blundell MP, et al. Autoinflammatory periodic fever, immunodeficiency, and thrombocytopenia (PFIT) caused by mutation in actin-regulatory gene WDR1. J Exp Med. 2017;214(1):59–71. https://doi.org/10.1084/jem.20161228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Jéru I, Papin S, L'hoste S, Duquesnoy P, Cazeneuve C, Camonis J, et al. Interaction of pyrin with 14.3.3 in an isoform-specific and phosphorylation-dependent manner regulates its translocation to the nucleus. Arthritis Rheum. 2005;52(6):1848–57. https://doi.org/10.1002/art.21050.

    Article  CAS  PubMed  Google Scholar 

  121. Masters SL, Lagou V, Jéru I, Baker PJ, Van Eyck L, Parry DA, et al. Familial autoinflammation with neutrophilic dermatosis reveals a regulatory mechanism of pyrin activation. Sci Transl Med. 2016;8:332ra45. https://doi.org/10.1126/scitranslmed.aaf1471.

    Article  CAS  PubMed  Google Scholar 

  122. Moghaddas F, Llamas R, De Nardo D, Martinez-Banaclocha H, Martinez-Garcia JJ, Mesa-Del-Castillo P, et al. A novel pyrin-associated autoinflammation with neutrophilic dermatosis mutation further defines 14-3-3 binding of pyrin and distinction to familial Mediterranean fever. Ann Rheum Dis. 2017;76(12):2085–94. https://doi.org/10.1136/annrheumdis-2017-211473.

    Article  CAS  PubMed  Google Scholar 

  123. Schnappauf O, Chae JJ, Kastner DL, Aksentijevich I. The pyrin inflammasome in health and disease. Front Immunol. 2019;10:1745. https://doi.org/10.3389/fimmu.2019.01745.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Wise CA, Gillum JD, Seidman CE, Lindor NM, Veile R, Bashiardes S, et al. Mutations in CD2BP1 disrupt binding to PTP PEST and are responsible for PAPA syndrome, an autoinflammatory disorder. Hum Mol Genet. 2002;11(8):961–9. https://doi.org/10.1093/hmg/11.8.961.

    Article  CAS  PubMed  Google Scholar 

  125. Shoham NG, Centola M, Mansfield E, Hull KM, Wood G, Wise CA, et al. Pyrin binds the PSTPIP1/CD2BP1 protein, defining familial Mediterranean fever and PAPA syndrome as disorders in the same pathway. Proc Natl Acad Sci U S A. 2003;100(23):13501–6. https://doi.org/10.1073/pnas.2135380100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Drenth JP, Cuisset L, Grateau G, Vasseur C, van de Velde-Visser SD, de Jong JG, et al. Mutations in the gene encoding mevalonate kinase cause hyper-IgD and periodic fever syndrome. International Hyper-IgD Study Group. Nat Genet. 1999;22(2):178–81. https://doi.org/10.1038/9696.

    Article  CAS  PubMed  Google Scholar 

  127. Mandey SH, Kuijk LM, Frenkel J, Waterham HR. A role for geranylgeranylation in interleukin-1β secretion. Arthritis Rheum. 2006;54(11):3690–5. https://doi.org/10.1002/art.22194.

    Article  CAS  PubMed  Google Scholar 

  128. Normand S, Massonnet B, Delwail A, Favot L, Cuisset L, Grateau G, et al. Specific increase in caspase-1 activity and secretion of IL-1 family cytokines: a putative link between mevalonate kinase deficiency and inflammation. Eur Cytokine Netw. 2009;20(3):101–7. https://doi.org/10.1684/ecn.2009.0163.

    Article  CAS  PubMed  Google Scholar 

  129. De Benedetti F, Gattorno M, Anton J, Ben-Chetrit E, Frenkel J, Hoffman HM, et al. Canakinumab for the Treatment of autoinflammatory recurrent fever syndromes. N Engl J Med. 2018;378(20):1908–19. https://doi.org/10.1056/NEJMoa1706314.

    Article  PubMed  Google Scholar 

  130. Romberg N, Al Moussawi K, Nelson-Williams C, Stiegler AL, Loring E, Choi M, et al. Mutation of NLRC4 causes a syndrome of enterocolitis and autoinflammation. Nat Genet. 2014;46(10):1135–9. https://doi.org/10.1038/ng.3066.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Kitamura A, Sasaki Y, Abe T, Kano H, Yasutomo K. An inherited mutation in NLRC4 causes autoinflammation in human and mice. J Exp Med. 2014;211(12):2385–96. https://doi.org/10.1084/jem.20141091.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Sasaki Y, Otsuka K, Arimochi H, Tsukumo SI, Yasutomo K. Distinct roles of IL-1β and IL-18 in NLRC4-induced autoinflammation. Front Immunol. 2020;11:591713. https://doi.org/10.3389/fimmu.2020.591713.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Miceli-Richard C, Lesage S, Rybojad M, Prieur AM, Manouvrier-Hanu S, Häfner R, et al. CARD15 mutations in Blau syndrome. Nat Genet. 2001;29(1):19–20. https://doi.org/10.1038/ng720.

    Article  CAS  PubMed  Google Scholar 

  134. Tanabe T, Chamaillard M, Ogura Y, Zhu L, Qiu S, Masumoto J, et al. Regulatory regions and critical residues of NOD2 involved in muramyl dipeptide recognition. EMBO J. 2004;23(7):1587–97. https://doi.org/10.1038/sj.emboj.7600175.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Kanazawa N, Okafuji I, Kambe N, Nishikomori R, Nakata-Hizume M, Nagai S, et al. Early-onset sarcoidosis and CARD15 mutations with constitutive nuclear factor-κB activation: common genetic etiology with Blau syndrome. Blood. 2005;105(3):1195–7. https://doi.org/10.1182/blood-2004-07-2972.

    Article  CAS  PubMed  Google Scholar 

  136. Zhou Q, Wang H, Schwartz DM, Stoffels M, Park YH, Zhang Y, et al. Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet. 2016;48:67–73. https://doi.org/10.1038/ng.3459.

    Article  CAS  PubMed  Google Scholar 

  137. Shigemura T, Kaneko N, Kobayashi N, Kobayashi K, Takeuchi Y, Nakano N, et al. Novel heterozygous C243Y A20/TNFAIP3 gene mutation is responsible for chronic inflammation in autosomal-dominant Behçet’s disease. RMD Open. 2016;2(1):e000223. https://doi.org/10.1136/rmdopen-2015-000223.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Duong BH, Onizawa M, Oses-Prieto JA, Advincula R, Burlingame A, Malynn BA, et al. A20 restricts ubiquitination of pro-interleukin-1β protein complexes and suppresses NLRP3 inflammasome activity. Immunity. 2015;42(1):55–67. https://doi.org/10.1016/j.immuni.2014.12.031.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Das T, Chen Z, Hendriks RW, Kool M. A20/tumor necrosis factor α-induced protein 3 in immune cells controls development of autoinflammation and autoimmunity: lessons from mouse models. Front Immunol. 2018;9:104. https://doi.org/10.3389/fimmu.2018.00104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Tokunaga F, Sakata S, Saeki Y, Satomi Y, Kirisako T, Kamei K, et al. Involvement of linear polyubiquitylation of NEMO in NF-κB activation. Nat Cell Biol. 2009;11(2):123–32. https://doi.org/10.1038/ncb1821.

    Article  CAS  PubMed  Google Scholar 

  141. Iwai K, Tokunaga F. Linear polyubiquitination: a new regulator of NF-kappaB activation. EMBO Rep. 2009;10(7):706–13. https://doi.org/10.1038/embor.2009.144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Kirisako T, Kamei K, Murata S, Kato M, Fukumoto H, Kanie M, et al. A ubiquitin ligase complex assembles linear polyubiquitin chains. EMBO J. 2006;25(20):4877–87. https://doi.org/10.1038/sj.emboj.7601360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Ikeda F, Deribe YL, Skånland SS, Stieglitz B, Grabbe C, Franz-Wachtel M, et al. SHARPIN forms a linear ubiquitin ligase complex regulating NF-κB activity and apoptosis. Nature. 2011;471(7340):637–41. https://doi.org/10.1038/nature09814.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Gerlach B, Cordier SM, Schmukle AC, Emmerich CH, Rieser E, Haas TL, et al. Linear ubiquitination prevents inflammation and regulates immune signalling. Nature. 2011;471(7340):591–6. https://doi.org/10.1038/nature09816.

    Article  CAS  PubMed  Google Scholar 

  145. Tokunaga F, Nakagawa T, Nakahara M, Saeki Y, Taniguchi M, Sakata S, et al. SHARPIN is a component of the NF-κB-activating linear ubiquitin chain assembly complex. Nature. 2011;471(7340):633–6. https://doi.org/10.1038/nature09815.

    Article  CAS  PubMed  Google Scholar 

  146. Boisson B, Laplantine E, Prando C, Giliani S, Israelsson E, Xu Z, et al. Immunodeficiency, autoinflammation and amylopectinosis in humans with inherited HOIL-1 and LUBAC deficiency. Nat Immunol. 2012;13:1178–86. https://doi.org/10.1038/ni.2457.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Schaeffer V, Akutsu M, Olma MH, Gomes LC, Kawasaki M, Dikic I. Binding of OTULIN to the PUB domain of HOIP controls NF-κB signaling. Mol Cell. 2014;54(3):349–61. https://doi.org/10.1016/j.molcel.2014.03.016.

    Article  CAS  PubMed  Google Scholar 

  148. Boisson B, Laplantine E, Dobbs K, Cobat A, Tarantino N, Hazen M, et al. Human HOIP and LUBAC deficiency underlies autoinflammation, immunodeficiency, amylopectinosis, and lymphangiectasia. J Exp Med. 212:939–51. https://doi.org/10.1084/jem.20141130.

  149. Damgaard RB, Walker JA, Marco-Casanova P, Morgan NV, Titheradge HL, Elliott PR, et al. The Deubiquitinase OTULIN is an essential negative regulator of inflammation and autoimmunity. Cell. 2016;166:1215-1230.e20. https://doi.org/10.1016/j.cell.2016.07.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Zhou Q, Yu X, Demirkaya E, Deuitch N, Stone D, Tsai WL, et al. Biallelic hypomorphic mutations in a linear deubiquitinase define otulipenia, an early-onset autoinflammatory disease. Proc Natl Acad Sci U S A. 2016;113(36):10127–32. https://doi.org/10.1073/pnas.1612594113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Damgaard RB, Elliott PR, Swatek KN, Maher ER, Stepensky P, Elpeleg O, et al. OTULIN deficiency in ORAS causes cell type-specific LUBAC degradation, dysregulated TNF signalling and cell death. EMBO Mol Med. 2019;11:e9324. https://doi.org/10.15252/emmm.201809324.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Chen D, Li Z, Liu Y, Sampaio N, Yang D, Aksentijevich I, et al. Human induced pluripotent stem cells generated from a patient with a homozygous L272P mutation in the OTULIN gene (NIHTVBi014-A). Stem Cell Res. 2020;47:101921. https://doi.org/10.1016/j.scr.2020.101921.

    Article  CAS  PubMed  Google Scholar 

  153. Aksentijevich I, Masters SL, Ferguson PJ, Dancey P, Frenkel J, van Royen-Kerkhoff A, et al. An autoinflammatory disease with deficiency of the interleukin-1-receptor antagonist. N Engl J Med. 2009;360:2426–37. https://doi.org/10.1056/NEJMoa0807865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Reddy S, Jia S, Geoffrey R, Lorier R, Suchi M, Broeckel U, et al. An autoinflammatory disease due to homozygous deletion of the IL1RN locus. N Engl J Med. 2009;360(23):2438–44. https://doi.org/10.1056/NEJMoa0809568.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Jesus AA, Osman M, Silva CA, Kim PW, Pham TH, Gadina M, et al. A novel mutation of IL1RN in the deficiency of interleukin-1 receptor antagonist syndrome: description of two unrelated cases from Brazil. Arthritis Rheum. 2011;63(12):4007–17. https://doi.org/10.1002/art.30588.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Jesus AA, Goldbach-Mansky R. IL-1 blockade in autoinflammatory syndromes. Annu Rev Med. 2014;65(1):223–44. https://doi.org/10.1146/annurev-med-061512-150641.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Rodero MP, Crow YJ. Type I interferon-mediated monogenic autoinflammation: The type I interferonopathies, a conceptual overview. J Exp Med. 2016;213(12):2527–38. https://doi.org/10.1084/jem.20161596.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Crow YJ, Rehwinkel J. Aicardi-Goutières syndrome and related phenotypes: linking nucleic acid metabolism with autoimmunity. Hum Mol Genet. 2009;18(R2):R130–6. https://doi.org/10.1093/hmg/ddp293.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Crow YJ, Chase DS, Lowenstein Schmidt J, Szynkiewicz M, Forte GM, Gornall HL, et al. Characterization of human disease phenotypes associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR, and IFIH1. Am J Med Genet A. 2015;167A:296–312. https://doi.org/10.1002/ajmg.a.36887.

    Article  CAS  PubMed  Google Scholar 

  160. Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, GAM S, et al. Activated STING in a vascular and pulmonary syndrome. N Engl J Med. 2014;371:507–18. https://doi.org/10.1056/NEJMoa1312625.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Watkin LB, Jessen B, Wiszniewski W, Vece TJ, Jan M, Sha Y, et al. COPA mutations impair ER-Golgi transport and cause hereditary autoimmune-mediated lung disease and arthritis. Nat Genet. 2015;47:654–60. https://doi.org/10.1038/ng.3279.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Arima K, Kinoshita A, Mishima H, Kanazawa N, Kaneko T, Mizushima T, et al. Proteasome assembly defect due to a proteasome subunit beta type 8 (PSMB8) mutation causes the autoinflammatory disorder, Nakajo-Nishimura syndrome. Proc Natl Acad Sci U S A. 2011;108:14914–9. https://doi.org/10.1073/pnas.1106015108.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Brehm A, Liu Y, Sheikh A, Marrero B, Omoyinmi E, Zhou Q, et al. Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production. J Clin Invest. 2015;125:4196–211. https://doi.org/10.1172/JCI81260.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Feigenbaum A, Müller C, Yale C, Kleinheinz J, Jezewski P, Kehl HG, et al. Singleton-Merten syndrome: an autosomal dominant disorder with variable expression. Am J Med Genet A. 2013;161A(2):360–70. https://doi.org/10.1002/ajmg.a.35732.

    Article  PubMed  Google Scholar 

  165. Jang MA, Kim EK, Now H, Nguyen NT, Kim WJ, Yoo JY, et al. Mutations in DDX58, which encodes RIG-I, cause atypical Singleton-Merten syndrome. Am J Hum Genet. 2015;96(2):266–74. https://doi.org/10.1016/j.ajhg.2014.11.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We greatly appreciate Dr. Isabelle Touitou, chief editor of “INFEVERS” database, and its contributors.

Funding

This work was supported by Grants-in-Aid for Scientific Research (B) (JSPS KAKENHI Grant Number: 20H03719 to JM) and Grant-in-Aid for Encouragement of Scientists (JSPS KAKENHI Grant Number: 20H01085 to NK).

Author information

Authors and Affiliations

Authors

Contributions

The manuscript was written by NK and JM. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Junya Masumoto.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Masumoto, J., Zhou, W., Morikawa, S. et al. Molecular biology of autoinflammatory diseases. Inflamm Regener 41, 33 (2021). https://doi.org/10.1186/s41232-021-00181-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41232-021-00181-8

Keywords