Inflammasomes are protein complexes that respond to a wide range of pathogens and cellular damage signals. Their activation prompts the caspase-1–mediated cleavage of the proinflammatory cytokines IL-1β and IL-18. Inflammasome dysregulation has been demonstrated to play a role in a range of diseases involving the adaptive immune system like multiple sclerosis, rheumatic diseases, and type 1 diabetes. Priming and activation of inflammasomes can be modulated by microRNAs (miRNAs), small noncoding RNAs that regulate gene expression posttranscriptionally. miRNAs, such as miR-223-3p, have been demonstrated to directly target the inflammasome components NLRP3, caspase-1, and caspase-8. Other miRNAs like miR-155-5p modulate TLR-, IL-1R–, TNFR-, and IFNAR-mediated signaling pathways upstream of the inflammasomes. In this study, we discuss how a more detailed elucidation of miRNA-driven inflammasome regulation helps in understanding the molecular processes underlying immune-mediated human diseases, holds potential for the identification of biomarkers and may offer novel targets for the development of future therapeutics.

Inflammasomes are endogenous multimeric protein complexes that mediate the cleavage of proinflammatory cytokines, especially IL-1β and IL-18, in response to a diverse set of stimuli such as infections, tissue damage, and metabolic imbalances (1, 2). Mutations in genes encoding the inflammasome sensor molecules NLRP3 and pyrin cause autoinflammatory disorders involving an excessive activation of innate immune responses, namely cryopyrin-associated periodic syndrome and familial Mediterranean fever (3). Moreover, changes in inflammasome activity and closely related dysregulation of IL-1β and IL-18 expression levels have been linked to cancer (4) and various human disorders involving autoimmune processes, such as multiple sclerosis (MS) (5) and rheumatic diseases (6).

MicroRNAs (miRNAs) are small noncoding RNAs of a length of ∼22 nt that act as posttranscriptional regulators of gene expression by binding complementary target sites in other transcripts (7). Several miRNAs can target mRNA transcripts of genes coding for components of inflammasome complexes or for regulators of the inflammasomes. Moreover, dysregulations of these miRNAs cause disease-like phenotypes in cellular and rodent models.

In the past 7 years, these mechanisms have been studied and important functional insights have been gained. This review aims to provide an overview of this emerging research field on miRNA–inflammasome interactions and to elucidate the potential of inflammasome-targeting miRNAs as biomarkers and for the development of novel therapeutic approaches for autoimmune diseases.

Several canonical inflammasomes named after their NLRP1, NLRP3, NLRC4, AIM2, and pyrin subunits and noncanonical inflammasomes named based on their caspases caspase-4, caspase-5, and caspase-8 have been described (1, 2). The common function of the canonical inflammasomes is the proteolytic activation of IL-1β and IL-18 and their secretion (2). Noncanonical inflammasomes have been proposed to realize additional functions such as the mediation of programmed cell death (pyroptosis) and the initiation of NLRP3 assembly (8, 9). Moreover, the inflammasomes control the microbiome in humans, both by hindering invasion of pathogens and by regulating the composition of commensal bacteria (10). In mouse models, deficiency of inflammasome components enhances the susceptibility to inflammatory bowel diseases (IBD) (10). Furthermore, several therapies that are currently in use to treat autoimmune disorders, such as IFN-β and glatiramer acetate in MS, glucocorticoids in systemic lupus erythematosus (SLE), and the TNF-α inhibitor infliximab in rheumatic diseases, partially affect the regulation of inflammasomes (1114).

The NLRP3 inflammasome, first described by Tschopp and colleagues (15) in 2002, is currently best understood. It is composed of the sensor molecule NLRP3, the adapter protein ASC that enhances complex formation, and of pro–caspase-1. Upon assembly, the activated inflammasome converts pro–caspase-1 into caspase-1, which cleaves pro–IL-1β and pro–IL-18 into mature proinflammatory cytokines IL-1β and IL-18 (15). The proinflammatory cytokines are then secreted (16).

The inflammasome components ASC and pro–caspase-1 are widely expressed in many tissues and in cell types of both the hematopoietic and the nonhematopoietic lineages (17). However, the inflammasome sensor molecules exhibit a more differentiated expression pattern, suggesting that in distinct microenvironments, various inflammatory stimuli are sensed and transduced, eliciting context-dependent responses (17). In the brain, and in particular the microglia, NLRP3 mRNA levels are among the highest throughout human tissues (18, 19). In resting cells, the expression of NLRP3 is not sufficient for inflammasome assembly, and a priming step leading to the upregulation of NLRP3 and pro–IL-1β expression is required. Once the cellular NLRP RNA level reaches an activating threshold, inflammasome assembly can be triggered (20). Pathways that prime NLRP3 and pro–IL-1β expression and activate inflammasome assembly have been excellently described in other reviews and are visualized in Fig. 1 (1, 2). One of the most effective mechanisms to mitigate the expression of NLRP3 is mediated by miRNAs, molecules that reduce NLRP3 protein expression through posttranscriptional downregulation of NLRP3 mRNA, as will be outlined in more detail in sections four and five.

FIGURE 1.

Regulation of the NLRP3 inflammasome and modulation by selected miRNAs. miRNAs (miRs) can modulate the expression of NLRP3 by downregulating NLRP3 mRNA directly and by targeting the transcripts of several proteins involved in regulatory pathways activated by TLR, IL-1R, TNFR, and IFNAR signaling. Other miRNAs target molecules involved in the cellular response to mitochondrial stress. Mitochondrial stress induces the production of ROS and TXNIP activation, both of which activate the assembly of the NLRP3 inflammasome. The maturation of pro–caspase-1 to caspase-1 allows for the cleavage of pro–IL-1β to proinflammatory cytokine IL-1β. Caspase-1 expression can be repressed by miR-214-3p. By these mechanisms, miRNAs constitute an extra layer of regulation that fine-tunes inflammasome priming in several cell types and over the course of inflammatory responses.

FIGURE 1.

Regulation of the NLRP3 inflammasome and modulation by selected miRNAs. miRNAs (miRs) can modulate the expression of NLRP3 by downregulating NLRP3 mRNA directly and by targeting the transcripts of several proteins involved in regulatory pathways activated by TLR, IL-1R, TNFR, and IFNAR signaling. Other miRNAs target molecules involved in the cellular response to mitochondrial stress. Mitochondrial stress induces the production of ROS and TXNIP activation, both of which activate the assembly of the NLRP3 inflammasome. The maturation of pro–caspase-1 to caspase-1 allows for the cleavage of pro–IL-1β to proinflammatory cytokine IL-1β. Caspase-1 expression can be repressed by miR-214-3p. By these mechanisms, miRNAs constitute an extra layer of regulation that fine-tunes inflammasome priming in several cell types and over the course of inflammatory responses.

Close modal

miRNAs are ∼22-nt long noncoding RNAs that posttranscriptionally regulate the expression of genes by binding to complementary sequences in target transcripts (7). In the current release 22 of the database miRBase, 2883 known human mature miRNAs are listed (21). Experimental disruptions of miRNA genes amount to diverse abnormal phenotypes, including defects in the thymus, hematopoietic lineages, and brain, altered susceptibility to infections, and autoimmune-mediated diseases (7). For example, the miRNAs miR-20b-5p, miR-30a-5p, and miR-146a-5p are dysregulated in immunological and neurologic pathways in MS (22), and the miRNA levels in peripheral blood cells of MS patients change in response to IFN-β treatment (23).

The first step of canonical miRNA biogenesis is transcription into primary miRNAs (pri-miRNAs) by RNA polymerase II (24, 25). Pri-miRNAs form double-stranded hairpin structures with a characteristic loop. In a two-step process, the pri-miRNA is first cleaved into a precursor miRNA by Drosha/DGCR8 and additional RNA-binding proteins, translocated into the cytoplasm by exportin-5, and subsequently processed by Dicer/TRBP to form a short miRNA duplex (2628). The resulting duplex is incorporated into an Argonaute protein and unwound so that only one strand (i.e., the mature miRNA) is kept in the RNA-induced silencing complex, which targets transcripts with matching sequences for degradation and prevents translation into proteins (25, 29, 30). Depending on which miRNA duplex strand is employed, the suffix “-3p” or “-5p” is used to designate the precise mature miRNA molecule (31, 32). Moreover, imprecise miRNA cleavage during their biogenesis as well as trimming, tailing, and editing have been described to produce isoforms of mature miRNAs with differing sequences (33).

Pairing of the miRNA seed region (nucleotides at position 2–7 from the 5′ end) and complementary sites preferably within 3′ untranslated regions (3′ UTRs) of target mRNAs mediates translational repression and destabilization of the bound transcripts (34). Additional matches of the transcript and the miRNA can augment the seed match as described by Bartel (7) in 2018. In humans, mRNA destabilization by deadenylation and subsequent exonucleolytic decay dominates the regulatory effect of miRNAs, whereas the contribution from direct translational repression is relatively small (3539). As of today, more than 400,000 regulatory interactions are listed in miRTarBase (release 7.0), a database collecting miRNA–target interactions from the literature (40). Other databases, such as miRWalk2.0, can be used to predict miRNA–target interactions based on different algorithms that, for example, employ sequence complementarity, binding energy, and sequence conservation (41).

The strongest evidence for miRNA–target interactions is usually provided by reporter assays, followed by Western blots and quantitative real-time PCR (qPCR) analyses. Reporter assays detect changes in the expression level of a hybrid construct consisting of a 3′ UTR of a putative target gene and a reporter (e.g., luciferase) in response to miRNA overexpression or inhibition (42). This method provides reliable evidence for direct interactions. Approximately 7600 of the 400,000 interactions listed in miRTarBase are supported by reporter assays. Analyzing changes in RNA and protein expression upon modulation of miRNA levels with qPCR and Western blots can be used to validate putative functional interactions as well, although this approach does not prove direct miRNA–target interactions as it also captures secondary effects (42). Novel highly parallel approaches for the identification of miRNA–target interactions include cross-linking and immunoprecipitation methods coupled with next-generation sequencing (4345) and posttranscriptional regulatory element sequencing (46). More than 350,000 of the 400,000 miRTarBase-listed interactions have only been identified by next-generation sequencing methods, indicating the need for further validation.

In 2012, two studies that were published in The Journal of Immunology identified miR-223-3p as the first human miRNA directly targeting NLRP3 (47, 48). A role for this interaction in the differentiation of monocytes to macrophages was proposed, as miR-223-3p levels decrease during differentiation, whereas NLRP3 protein levels increase (47). NLRP3 mRNA levels correlate negatively with miR-223-3p expression in myeloid cells (48). Interestingly, among human innate immune cells, miR-223-3p expression is highest in neutrophils and lowest in dendritic cells (48), hinting at a mechanism for why dendritic cells have a relatively low threshold for responding to NLRP3 stimuli (48). Moreover, a miRNA from EBV, a common virus that targets B cells and is considered essential for the development of the neurodegenerative disease MS (49), ebv-miR-BART15-3p, regulates NLRP3 expression (47). Infected B cells can secrete ebv-miR-BART15-3p via exosomes and inhibit NLRP3 activity in noninfected cells, which may be a mechanism for the virus to evade host defense (47). Interestingly, miR-223-3p and ebv-miR-BART15-3p hybridize with NLRP3 mRNA at the same site, and a common polymorphism, rs10802501, is present in the target site and could influence the binding of both miRNAs (47).

Meanwhile, several further miRNAs have been found to directly target transcripts of NLRP3 and thereby inhibit inflammasome priming in vitro and in vivo in human and animal studies (Table I). Among them are miR-7-5p and miR-30e-5p, and treatment with mimics of these miRNAs alleviates Parkinson disease (PD)-like phenotypes in mouse models involving accumulation of α-synuclein (5052). The NLRP3 inflammasome and IL-1β production can be activated by α-synuclein, which drives neurodegeneration (53). NLRP3 is also a direct target of miR-20b-5p, upregulation of which alleviates Mycobacterium tuberculosis–induced inflammation in murine lung tissue (54). Additionally, miR-22-3p downregulates NLRP3 expression, and this miRNA is decreased in injured coronary arterial endothelial cells in a rat model of high-fat diet–induced coronary heart disease, whereas upregulation of miR-22-3p exerts a protective effect on cell survival (55). The expression of miR-133b-3p, which also targets NLRP3, is reduced in the nasal mucosa of a mouse model of allergic rhinitis (56). Allergic symptoms of the mice, immune cell infiltration, cytokine levels, and NLRP3 inflammasome protein expression were reduced in the nasal mucosa after administration of modified miR-133b-3p mimics, indicating that miR-133b-3p downregulation may contribute to the pathogenesis of allergic rhinitis, whereas its upregulation shows therapeutic potential (56). In a mouse model of prosopalgia, miR-186-5p is downregulated, and injection of miRNA mimics could alleviate neuropathic pain behavior by downregulating NLRP3 expression and NLRP3 inflammasome activity (57). The levels of miR-495-3p are decreased in mice with myocardial ischemia/reperfusion injury, and by targeting NLRP3, treatment with miR-495-3p mimics alleviates inflammation (58). The predicted secondary structure of the NLRP3 mRNA 3′ UTR and the proposed binding sites of the nine miRNAs that so far have been shown to target NLRP3 are displayed in Fig. 2.

Table I.
Inflammasome component-targeting miRNAs
miRNATargetSpeciesCompartmentResearch ContextRef.
miR-7-5p NLRP3 Mouse ANSCs PD (50
miR-20b-5p NLRP3 Mouse SNpc, microglia M. tuberculosis infection (54
miR-22-3p NLRP3 Mouse Nasal mucosa Coronary heart disease (55
miR-30e-5p NLRP3 Mouse; human Microglia, midbrain; serum PD (50
miR-133b-3p NLRP3 Mouse Lung tissue Allergic rhinitis (56
miR-186-5p NLRP3 Mouse/rat Neuroblastoma-neuron hybrid Prosopalgia (57
miR-223-3p NLRP3 Human Monocytes, macrophages Myeloid cell development (47
miR-495-3p NLRP3 Mouse CMECs Myocardial ischemia reperfusion injury (58
ebv-miR-BART15-3p NLRP3 Human Monocytes, macrophages Myeloid cell development (47
miR-214-3p CASP1 Rat CAECs Cataractogenesis (59
miR-371a-5p CASP8 Human HCC cell line and HCC tissue Hepatocellular carcinoma (60
miR-373-3p CASP8 Human HCC cell line and HCC tissue Hepatocellular carcinoma (60
miR-543-3p CASP8 Human HCC cell line and HCC tissue Hepatocellular carcinoma (60
miRNATargetSpeciesCompartmentResearch ContextRef.
miR-7-5p NLRP3 Mouse ANSCs PD (50
miR-20b-5p NLRP3 Mouse SNpc, microglia M. tuberculosis infection (54
miR-22-3p NLRP3 Mouse Nasal mucosa Coronary heart disease (55
miR-30e-5p NLRP3 Mouse; human Microglia, midbrain; serum PD (50
miR-133b-3p NLRP3 Mouse Lung tissue Allergic rhinitis (56
miR-186-5p NLRP3 Mouse/rat Neuroblastoma-neuron hybrid Prosopalgia (57
miR-223-3p NLRP3 Human Monocytes, macrophages Myeloid cell development (47
miR-495-3p NLRP3 Mouse CMECs Myocardial ischemia reperfusion injury (58
ebv-miR-BART15-3p NLRP3 Human Monocytes, macrophages Myeloid cell development (47
miR-214-3p CASP1 Rat CAECs Cataractogenesis (59
miR-371a-5p CASP8 Human HCC cell line and HCC tissue Hepatocellular carcinoma (60
miR-373-3p CASP8 Human HCC cell line and HCC tissue Hepatocellular carcinoma (60
miR-543-3p CASP8 Human HCC cell line and HCC tissue Hepatocellular carcinoma (60

Several miRNAs have been found to downregulate the expression of components of the NLRP3 inflammasome (NLRP3, CASP1) and the noncanonical caspase-8 (CASP8) inflammasome in different research contexts. The investigations were performed using distinct compartments from distinct species. All of the miRNA–target interactions have been ascertained by luciferase reporter assays.

ANSC, adult neuronal stem cell; CAEC, coronary arterial endothelial cell; CMEC, cardiac microvascular endothelial cell; HCC, hepatocellular carcinoma; Ref., reference; SNpc, substantia nigra pars compacta.

FIGURE 2.

Predicted binding sites of miRNAs within the 3′ UTR of NLRP3 mRNA. Left, The predicted RNA structure of the 3′ UTR of NLRP3 (Ensembl database transcript identifier: ENST00000391828.7) as calculated by the RNAfold web server (version 2.4.9) using the “minimum free energy (mfe) and partition function” algorithm (135). Each base of the mRNA is displayed as a blue dot. The base pair probability is color-coded. The positions of three common SNPs in the 3′ UTR, rs10754558, rs10802501, and rs10802502, are highlighted in yellow. The binding sites of the nine NLRP3-targeting miRNAs from the literature were predicted by the RNAhybrid (version 2.2) webtool (136) with an energy threshold of −15 kcal/mol. Binding sites with no or one mismatch in the seed region are displayed in red and consecutively numbered beginning at the 5′ end of the 3′ UTR. Additionally, the binding sites of hsa-miR-223-3p (no. 15) and ebv-miR-BART15-3p (no. 16) as proposed by Haneklaus et al. (47) have been mapped. The 3′ ends of the miRNAs are indicated by arrowheads. Right, The miRNA, the minimum free energy (mfe) and the sequences at each binding site are listed. bp are displayed in color, and mismatching bases are in black. The miRNA seed regions and matching mRNA regions are highlighted in red. Of note, for five of the nine miRNAs, multiple binding sites within the NLRP3 3′ UTR have been determined, and at four sites, more than one of the miRNAs may bind to the 3′ UTR. Moreover, the three common SNPs in the NLRP3 3′ UTR are localized within miRNA binding sites. These SNPs might thus influence the hybridization kinetics of the miRNAs and the 3′ UTR sequence.

FIGURE 2.

Predicted binding sites of miRNAs within the 3′ UTR of NLRP3 mRNA. Left, The predicted RNA structure of the 3′ UTR of NLRP3 (Ensembl database transcript identifier: ENST00000391828.7) as calculated by the RNAfold web server (version 2.4.9) using the “minimum free energy (mfe) and partition function” algorithm (135). Each base of the mRNA is displayed as a blue dot. The base pair probability is color-coded. The positions of three common SNPs in the 3′ UTR, rs10754558, rs10802501, and rs10802502, are highlighted in yellow. The binding sites of the nine NLRP3-targeting miRNAs from the literature were predicted by the RNAhybrid (version 2.2) webtool (136) with an energy threshold of −15 kcal/mol. Binding sites with no or one mismatch in the seed region are displayed in red and consecutively numbered beginning at the 5′ end of the 3′ UTR. Additionally, the binding sites of hsa-miR-223-3p (no. 15) and ebv-miR-BART15-3p (no. 16) as proposed by Haneklaus et al. (47) have been mapped. The 3′ ends of the miRNAs are indicated by arrowheads. Right, The miRNA, the minimum free energy (mfe) and the sequences at each binding site are listed. bp are displayed in color, and mismatching bases are in black. The miRNA seed regions and matching mRNA regions are highlighted in red. Of note, for five of the nine miRNAs, multiple binding sites within the NLRP3 3′ UTR have been determined, and at four sites, more than one of the miRNAs may bind to the 3′ UTR. Moreover, the three common SNPs in the NLRP3 3′ UTR are localized within miRNA binding sites. These SNPs might thus influence the hybridization kinetics of the miRNAs and the 3′ UTR sequence.

Close modal

In regard to miRNA regulation, NLRP3 is the best investigated inflammasome component. Beyond that, two caspases are directly targeted by miRNAs. In human epithelial cells, overexpression of miR-214-3p downregulates caspase-1, and the miRNA is downregulated in cataract lens anterior capsular tissues (59). MiR-371a-5p, miR-373-3p, and miR-543-3p have been proposed to enhance TNF-α–induced cell death by targeting the noncanonical caspase-8 inflammasome, and they are overexpressed in hepatocellular carcinoma tissue (60). However, the miRNA-driven regulation of further human inflammasome components, such as other NLR sensor molecules, AIM2, caspase-4, and caspase-5, has so far not been investigated in specific experimental studies. Caspase-11, the murine ortholog of human caspase-4 and caspase-5, was shown to be indirectly regulated by miR-34a, miR-451, and miR-874 (61).

Inflammasomes are regulated by various upstream pathways. In several recent studies, it has been demonstrated that miRNAs that target these upstream pathways alter the expression levels of components of the NLRP3 inflammasome. As presented in Fig. 1 and listed in Table II, some miRNAs target more than one inflammasome regulator and likewise, a regulator can be downregulated by several miRNAs. The miRNA-mediated modulation of inflammasome regulation was investigated in very heterogeneous research contexts, such as gout, depression, and alcoholic liver disease.

Table II.
Selected miRNAs that target upstream regulators of the NLRP3 inflammasome
miRNATargetMethodEffectSpeciesCompartmentResearch ContextRef.
miR-145a-5p CD137 Luci Down Mouse VSMCs Atherosclerosis (82
miR-23a-3p CXCR4 Luci Down Mouse Spinal glial cells Neuropathic pain (80
miR-30c-5p FOXO3 Luci Down Human HAECs Atherosclerosis (81
miR-34c-5p IKBKG qPCR, WB n/a Human PBMCs DAMP-stimulated inflammation (66
miR-383-3p IL1R2 Luci Down Rat CAECs Atherosclerosis (68
miR-146a-5p IRAK1 qPCR, WB Down Mouse Macrophages Gouty arthritis (64
miR-302b-3p IRAK4 Luci Down Human Macrophages Gouty arthritis (63
miR-9-5p JAK1 Luci Down Human Macrophages, PBMCs Atherosclerosis (73
miR-155-5p MYD88 qPCR, WB Down Rat Prefrontal cortex Depression treatment (65
miR-145a-5p NFATC1 Luci Down Mouse VSMCs Atherosclerosis (82
miR-377-3p SOD1 WB Up Rat, mouse Kidney cortex, glomeruli, podocytes Fructose-induced kidney injury (75
miR-377-3p SOD2 WB Up Rat, mouse Kidney cortex, glomeruli, podocytes Fructose-induced kidney injury (75
miR-330-3p SPHK1 WB Down Human HK-2 cells Fructose-induced kidney injury (70
miR-146a-5p TRAF6 qPCR, WB Down Mouse Macrophages Gouty arthritis (64
miR-17-5p TXNIP Luci Down Rat Insulin-producing cells ER stress (76
miR-17-5p TXNIP qPCR, WB Down Rat Brain Ischemic brain injury (77
miR-20a-5p TXNIP Luci Down Rat Fibroblast-like synoviocytes RA (78
miR-148a-3p TXNIP Luci Down Mouse Hepatocytes Alcoholic liver disease (79
miRNATargetMethodEffectSpeciesCompartmentResearch ContextRef.
miR-145a-5p CD137 Luci Down Mouse VSMCs Atherosclerosis (82
miR-23a-3p CXCR4 Luci Down Mouse Spinal glial cells Neuropathic pain (80
miR-30c-5p FOXO3 Luci Down Human HAECs Atherosclerosis (81
miR-34c-5p IKBKG qPCR, WB n/a Human PBMCs DAMP-stimulated inflammation (66
miR-383-3p IL1R2 Luci Down Rat CAECs Atherosclerosis (68
miR-146a-5p IRAK1 qPCR, WB Down Mouse Macrophages Gouty arthritis (64
miR-302b-3p IRAK4 Luci Down Human Macrophages Gouty arthritis (63
miR-9-5p JAK1 Luci Down Human Macrophages, PBMCs Atherosclerosis (73
miR-155-5p MYD88 qPCR, WB Down Rat Prefrontal cortex Depression treatment (65
miR-145a-5p NFATC1 Luci Down Mouse VSMCs Atherosclerosis (82
miR-377-3p SOD1 WB Up Rat, mouse Kidney cortex, glomeruli, podocytes Fructose-induced kidney injury (75
miR-377-3p SOD2 WB Up Rat, mouse Kidney cortex, glomeruli, podocytes Fructose-induced kidney injury (75
miR-330-3p SPHK1 WB Down Human HK-2 cells Fructose-induced kidney injury (70
miR-146a-5p TRAF6 qPCR, WB Down Mouse Macrophages Gouty arthritis (64
miR-17-5p TXNIP Luci Down Rat Insulin-producing cells ER stress (76
miR-17-5p TXNIP qPCR, WB Down Rat Brain Ischemic brain injury (77
miR-20a-5p TXNIP Luci Down Rat Fibroblast-like synoviocytes RA (78
miR-148a-3p TXNIP Luci Down Mouse Hepatocytes Alcoholic liver disease (79

Several transcripts coding for proteins that act as regulators of the NLRP3 inflammasome are targeted by miRNAs. The expression of downstream inflammasome components has been shown to be up- or downregulated in response to increased levels of these miRNAs. The listed miRNA–mRNA interactions have been investigated in different research contexts.

CAEC, coronary arterial endothelial cell; DAMP, damage-associated molecular pattern; ER, endoplasmic reticulum; HAEC, human aortic endothelial cell; HK-2 cell, an immortalized proximal tubule epithelial cell line from normal adult human kidney; Luci, luciferase reporter assay; n/a, not available; Ref., reference; VSMC, vascular smooth muscle cell; WB, Western blot.

Several miRNAs negatively regulate the NF-κB–mediated priming of the NLRP3 inflammasome by targeting a wide range of transducers of TLR, IL-1R, and TNFR signaling. Signals from both TLR4 and IL-1R are transduced via MyD88, IRAK1, and IRAK4, TRAF6, IKK, and finally NF-κB, which then induces NLRP3 and pro–IL-1β transcription (2, 62). In a study on inflammation of gout, miR-302b-3p was upregulated after induction of THP-1–derived macrophages using monosodium urate (MSU). Overexpression of miR-302b-3p directly downregulates IRAK4 and thereby inhibits NLRP3 expression (63). Another research group investigated the function of miR-146a-5p in response to MSU stimulation (64). The mRNA and protein expression of TRAF6, IRAK1 and downstream NLRP3, ASC, and caspase-1 were upregulated in MSU-induced bone marrow–derived macrophages of miR-146a-5p knockout mice as compared with wild-type mice. The miR-146a-5p–deficient mice developed a more severe phenotype of gouty arthritis (64). Two further members of the TLR4 and IL-1R signaling pathways are targeted by miRNAs: MyD88 is downregulated by miR-155-5p overexpression in rat prefrontal cortex, and this alleviates depressive-like behaviors (65), and IKKγ (gene symbol: IKBKG) expression is reduced by miR-34c-5p. In response to stimulation with damage-associated molecular patterns, miR-34c-5p is highly expressed in PBMC (66). Through this mechanism, miR-34c-5p was proposed to limit aberrant sterile acute or chronic inflammation (66).

IL-1R2, a receptor that inhibits IL-1 signaling in a competitive manner (67), is downregulated by miR-383-3p (68). In myocardial tissues of a rat model of atherosclerosis, miR-383-3p levels are reduced, whereas IL-1R2 expression is increased. Overexpression of miR-383-3p decreases the protein levels of caspase-1 and the secretion of IL-1β and IL-18 in homocysteine-induced endothelial injury in rat coronary arteries (68).

Binding of TNF-α to TNFRSF1A followed by downstream TRAF2/SphK1/S1P signaling constitutes another NF-κB–activating and NLRP3 inflammasome-priming pathway (69). Administration of miR-330-3p mimics attenuates fructose-induced upregulation of SphK1, NF-κB, NLRP3, pro–IL-1β, and IL-1β protein levels, whereas miR-330-3p inhibition has the opposite effect in a proximal tubule epithelial cell line from adult human kidney (HK-2 cells) (70). By targeting SphK1, miR-330-3p thus inhibits inflammasome priming (70).

The receptor IFNAR responds to IFN-α and IFN-β and activates the JAK/STAT/ISGF3 pathway, which is able to drive NLRP3 expression (71). JAK1 is targeted by miR-9-5p, and this miRNA is increased in human primary peripheral blood monocytes stimulated with oxidized low density lipoproteins indicating its role as a regulator of inflammation (72, 73). Overexpression of miR-9-5p inhibits NLRP3 inflammasome priming in atherosclerosis inflammation models using human THP-1–derived macrophages and primary peripheral blood monocytes (72).

During mitochondrial stress, excessive reactive oxygen species (ROS), which can be reduced by superoxide dismutases (SOD), are generated, and they induce TXNIP, an NLRP3-activating factor (74). Fructose induces miR-377-3p expression in kidney cortex and glomeruli of rats as well as in a mouse podocyte cell line (75). Upregulation of miR-377-3p can inhibit SOD1 and SOD2 protein levels and thereby raise ROS levels and induce elevated NLRP3, ASC, caspase, and supernatant IL-1β protein levels in cultured mouse podocytes, implying a role for miR-377-3p in metabolic syndrome (75). The NLRP3-activating factor TXNIP is inhibited by several individual miRNAs. Expression levels of miR-17-5p in rat brains and rat insulin-producing cells with endoplasmic reticulum stress (76, 77), of miR-20a-5p in rat fibroblast-like synoviocytes in adjuvant-induced arthritis (78), and of miR-148a-3p in mouse hepatocytes in alcoholic hepatitis (79) are decreased and correlate with upregulation of TXNIP and activation of the NLRP3 inflammasome. Upregulation of the miRNAs with mimics reversed these effects. The chemokine receptor CXCR4 was proposed to stabilize TXNIP (80). In mice with sciatic nerve injury, overexpression of miR-23a-3p downregulates CXCR4, inhibits the activation of the NLRP3 inflammasome and subsequent IL-1β secretion, and alleviates neuropathic pain behavior (80).

Two further miRNAs are known to modulate inflammasome regulation. In an atherosclerosis model of human aortic endothelial cells treated with oxidized low density lipoproteins, miR-30c-5p is downregulated, leading to the upregulation of FOXO3 (81). The administration of miR-30c-5p mimics decreases FOXO3 expression, and the knockdown of FOXO3 downregulates NLRP3, caspase-1, and IL-1β mRNA and protein levels, indicating a protective effect of the miRNA (81). The promotor of miR-145a-5p can be hypermethylated during atherosclerotic plaque formation, which leads to a decreased expression of miR-145a-5p, an upregulation of its target genes CD137 and NFATc1, and this induces the activation of the NLRP3 inflammasome. An elevation of the miRNA levels has a protective effect on vascular smooth muscle cells (82).

Of the described miRNAs, miR-9-5p (83) and miR-155-5p (84) also directly target NFKB1 mRNA, and miR-7-5p (85) downregulates RELA expression. Both are subunits of NF-κB. However, for these interactions, the dysregulation of the inflammasomes was not investigated in response to miRNA dysregulation.

At least five of the above mentioned NLRP3 inflammasome-targeting miRNAs and seven miRNAs that modulate inflammasome regulation are associated with autoimmune diseases (Table III). In the following, miRNAs involved in MS, rheumatoid arthritis (RA), SLE, IBD, and type 1 diabetes (T1D) will be addressed. The most prominent roles for inflammasome-regulating miRNAs in autoimmune diseases have been demonstrated for miR-146a-5p, miR-155-5p, and miR-223-3p, all of which are dysregulated in RA, MS, and SLE.

Table III.
Inflammasome-modulating miRNAs dysregulated in autoimmune diseases
miRNAAIDCompartmentEffectComparisonRef.
miR-7-5p MS CD4+ T cells Up t versus ut (96
miR-9-5p MS Leukocytes Up MS versus hs (91
 RA Plasma Down RA versus hs (99
miR-17-5p MS CD4+ T cells Up rl versus rm (95
 MS CD4+ T cells Down t versus ut (95
 SLE PBMC Down SLE versus hs (114, 115
 SLE PBMC Up t versus ut (116
miR-20a-5p SLE Monocytes Down SLE versus hs (113
miR-20b-5p MS CD4+ T cells Up t versus ut (96
miR-22-3p MS Plasma Up MS versus hs (90
 RA Synovial tissue Down RA versus OA (97
 RA Serum Down res versus nr (98)a 
miR-23a-3p IBD Whole blood Up CD versus hs (120
 MS Serum Down PPMS versus hs (92)a 
miR-146a-5p IBD Colonic mucosa Up CD versus UC/hs (121)a 
 IBD Colonic tissue Up CD/UC versus hs (121)a 
 LN Glomeruli Up LN versus hs (117)a 
 MS PBMC Up RRMS versus hs (86, 87
 MS PBMC Down t versus ut (87
 MS PBMC Up rs2910164 C versus G (88)a 
 RA PBMC Up RA versus hs (104
 RA Regulatory T cells Down RA versus hs (106
 RA Synovial tissue Up RA versus OA (102)a 
 SLE Urine Up SLE versus hs (108
 SLE CD4+ T cells Up t versus ut (109
 T1D PBMC Down T1D versus hs (122
miR-148a-3p LN Serum, glomeruli Up LN versus hs (118
miR-155-5p IBD Colonic mucosa Up IBD versus hs (119
 IBD Neoplastic tissue Up +CRC versus −CRC (119
 MS CD14+ monocytes, active brain lesions Up MS versus hs (89
 MS PBMC Up RRMS versus hs (87
 RA Whole blood, PBMC, synoviocytes Up RA versus hs (103
 RA Synovial membrane, macrophages Up RA versus OA (101
 SLE PBMC Down SLE versus hs (114
 SLE Urine Up SLE versus hs (108
 SLE Urine Down t versus ut (108
miR-214 MS CD4+ T cells Down rl versus rm/hs (94)a 
miR-223-3p MS Serum Down MS versus hs (92
 RA T cells Up RA versus hs (105
 RA Synovial tissue Up RA versus OA (100)a 
 SLE Naive B cells Down SLE versus hs (112
miRNAAIDCompartmentEffectComparisonRef.
miR-7-5p MS CD4+ T cells Up t versus ut (96
miR-9-5p MS Leukocytes Up MS versus hs (91
 RA Plasma Down RA versus hs (99
miR-17-5p MS CD4+ T cells Up rl versus rm (95
 MS CD4+ T cells Down t versus ut (95
 SLE PBMC Down SLE versus hs (114, 115
 SLE PBMC Up t versus ut (116
miR-20a-5p SLE Monocytes Down SLE versus hs (113
miR-20b-5p MS CD4+ T cells Up t versus ut (96
miR-22-3p MS Plasma Up MS versus hs (90
 RA Synovial tissue Down RA versus OA (97
 RA Serum Down res versus nr (98)a 
miR-23a-3p IBD Whole blood Up CD versus hs (120
 MS Serum Down PPMS versus hs (92)a 
miR-146a-5p IBD Colonic mucosa Up CD versus UC/hs (121)a 
 IBD Colonic tissue Up CD/UC versus hs (121)a 
 LN Glomeruli Up LN versus hs (117)a 
 MS PBMC Up RRMS versus hs (86, 87
 MS PBMC Down t versus ut (87
 MS PBMC Up rs2910164 C versus G (88)a 
 RA PBMC Up RA versus hs (104
 RA Regulatory T cells Down RA versus hs (106
 RA Synovial tissue Up RA versus OA (102)a 
 SLE Urine Up SLE versus hs (108
 SLE CD4+ T cells Up t versus ut (109
 T1D PBMC Down T1D versus hs (122
miR-148a-3p LN Serum, glomeruli Up LN versus hs (118
miR-155-5p IBD Colonic mucosa Up IBD versus hs (119
 IBD Neoplastic tissue Up +CRC versus −CRC (119
 MS CD14+ monocytes, active brain lesions Up MS versus hs (89
 MS PBMC Up RRMS versus hs (87
 RA Whole blood, PBMC, synoviocytes Up RA versus hs (103
 RA Synovial membrane, macrophages Up RA versus OA (101
 SLE PBMC Down SLE versus hs (114
 SLE Urine Up SLE versus hs (108
 SLE Urine Down t versus ut (108
miR-214 MS CD4+ T cells Down rl versus rm/hs (94)a 
miR-223-3p MS Serum Down MS versus hs (92
 RA T cells Up RA versus hs (105
 RA Synovial tissue Up RA versus OA (100)a 
 SLE Naive B cells Down SLE versus hs (112

Of the inflammasome-modulating miRNAs described in sections 4 and 5, 12 miRNAs have been described to be dysregulated in patients with autoimmune diseases (AID). The miRNA expression in distinct compartments from patients with AID have been compared with various control groups, most often healthy subjects (hs), and the miRNA levels have been shown to be either downregulated or upregulated.

a

The investigated mature miRNA strand (-3p or -5p) was not specified.

CD, Crohn disease; +CRC, with colorectal carcinoma; −CRC, without colorectal carcinoma; hs, healthy subject; LN, lupus nephritis; nr, nonresponder; OA, osteoarthritis; PPMS, primary progressive MS; Ref., reference; res, responder; rl, relapse; rm, remission; t, treated; UC, ulcerative colitis; ut, untreated.

In PBMCs of patients with the immune-mediated neurodegenerative disease relapsing-remitting MS (RRMS), miR-146a-5p levels are increased compared with healthy controls, and treatment with glatiramer acetate partially restores miR-146a-5p expression to lower levels again (86, 87). Interestingly, the C allele of the single-nucleotide polymorphism (SNP) rs2910164 in the sequence encoding mir-146a is associated with an increased expression of this miRNA and release of proinflammatory cytokines in patients with RRMS, and in females, rs2910164 was proposed to alter MS susceptibility (88). Furthermore, miR-155-5p is expressed more strongly in peripheral blood monocytes and active brain lesions of MS patients than in healthy controls (87, 89). Enhanced miRNA levels have been demonstrated for miR-22-3p in plasma of MS patients (90) and for miR-9-5p in circulating leukocytes of RRMS patients (91). Other miRNAs are decreased in MS patient serum (e.g., miR-23a-3p and miR-223-3p in primary progressive MS) (92). The T allele of SNP rs1044165 located near the sequence encoding mir-223 is associated with a higher risk of developing MS (93). Furthermore, in RRMS, miR-214 levels are downregulated in T cells in patients in the relapsing phase compared with patients in the remitting phase and healthy controls (94). In response to treatment of RRMS patients with natalizumab, the expression of miR-17-5p in T cells is decreased in comparison with untreated patients, and during relapse, miR-17-5p is upregulated (95). In case of miR-7-5p and miR-20b-5p, expression is upregulated in T cells during natalizumab treatment (96).

In RA, miR-22-3p levels are reduced in synovial tissue from RA patients compared with osteoarthritis patients (97), and low expression of miR-22-3p is associated with a good response to adalimumab treatment (98). Furthermore, the expression of miR-9-5p is decreased in plasma of RA patients compared with healthy controls (99). The three miRNAs miR-223 (100), miR-155-5p (101), and miR-146a (102) are upregulated in synovial tissue from RA patients compared with osteoarthritis synovium, and two of them, miR-155-5p and miR-146a-5p, are elevated in PBMCs of RA patients (103, 104). Moreover, miR-223-3p is overexpressed in T cells, and miR-223-3p expression correlates with the titer of rheumatoid factor in patients with RA (105). Of interest, levels of miR-146a-5p are reduced in regulatory T cells of RA patients with active disease and correlate with joint inflammation (106). Furthermore, the rs2910164 genotype GG in the sequence encoding mir-146a is associated with an increased risk of developing RA in females, whereas the CC genotype is related to a decreased disease severity in females and an increased disease severity in males (107).

In patients with SLE, miR-155-5p expression is decreased in PBMCs compared with healthy controls (108). In SLE urine, miR-155-5p levels are significantly higher, treatment with calcitriol reduces miR-155-5p levels, and miR-155-5p levels correlate with proteinuria and disease activity, whereas the amount of miR-146a-5p correlates with the estimated glomerular filtration rate in SLE patients (108). In response to treatment with mycophenolic acid, miR-146a-5p levels are increased in T cells of patients versus untreated patients (109). Additionally, two SNPs upstream of the genome locus encoding mir-146a, rs57095329 and rs2431697, are associated with the miRNA expression level and with SLE development (110, 111). Several inflammasome-regulating miRNAs are downregulated in SLE: miR-223-3p in naive B cells of SLE patients (112), miR-20a-5p in monocytes (113), and miR-17-5p in PBMCs of adult (114) and pediatric SLE patients (115). Moreover, miR-17-5p levels are diminished in untreated patients compared with those on treatment (116). Regarding lupus nephritis, a common manifestation of SLE, higher levels of miR-146a in glomeruli and miR-148a-3p in serum and glomeruli of patients with lupus nephritis have been reported relative to healthy controls (117, 118).

In IBD patients, some inflammasome-modulating miRNAs are also dysregulated. For instance, miR-155-5p is overexpressed in colonic mucosa of IBD patients compared with non-IBD controls (119). The levels of miR-23a-3p are elevated in the blood of patients with Crohn disease in comparison with healthy controls (120). Furthermore, miR-146a expression is increased in colonic mucosa of Crohn disease patients compared with ulcerative colitis and healthy controls, and it is increased in colonic tissue in both Crohn disease and ulcerative colitis (121).

For T1D, miR-146a-5p is so far the only inflammasome-regulating miRNA reported to be downregulated in PBMCs of patients. Low miR-146a-5p expression correlates with ongoing islet autoimmunity in T1D (122).

Additionally, inflammasome-modulating miRNAs have been found to be dysregulated in PD, and although defined as a neurodegenerative disease, PD has been suggested to involve autoimmune (123) and inflammasome-mediated mechanisms as well (124). The expression level of miR-7-5p is decreased in the serum of patients with PD (52). In circulating lymphocytes of treated patients, miR-7-5p and miR-9-5p are upregulated compared with untreated controls (125). Moreover, miR-22-3p is downregulated in whole blood of newly diagnosed and in treated PD patients compared with healthy controls (126), and miR-34c-3p is downregulated in brain areas of PD patients with neuropathological lesions (127).

Since their first description in 1993 (128), miRNAs have emerged as important regulators of gene expression. Some miRNAs are key to human development; others can mediate diseases but the function of most miRNAs is still barely understood. A decade after the discovery of inflammasomes, the first miRNAs that control inflammasome components have been identified (47, 48). Since then, several miRNAs that target NLRP3, caspase-1, caspase-8, and upstream signaling cascades via IL-1R, TLRs, IFNAR, TNFR, and mitochondrial stress pathways have been demonstrated to modulate inflammasome activity and to be dysregulated in human immune-mediated diseases.

However, the network of possible miRNA–inflammasome interactions has only been touched upon. The miRNA-mediated posttranscriptional control of other inflammasome components such as ASC, NLRP1, NLRC4, and AIM2, noncanonical inflammasomes, and upstream pathways has not been thoroughly investigated so far. For many miRNAs, bioinformatics-based miRNA–target interaction predictions in the database miRWalk2.0 (41) point at a role in inflammasome regulation; for example, miR-520f-3p is predicted to target NLRP3 by 7 out of 12 algorithms. Valuable insights into autoimmune mechanisms may be gained by investigating such putative interactions experimentally at the RNA and protein level and with luciferase reporter assays that allow for the assessment of direct interactions. Not only human miRNAs may be interesting to study further, but so too could miRNAs from viruses, which have been associated with diseases, as was already shown for ebv-miR-BART15-3p from EBV (47). Moreover, for many miRNAs, which exact mature miRNA isoforms act as posttranscriptional regulators is poorly understood. Finally, several underexplored miRNAs may be expressed at high levels only under certain conditions (e.g., in specific immune cell subtypes or during inflammation) (129131). Investigating in more detail the expression and function of miRNAs in compartments that are especially relevant for diseases, like microglia for MS, may be valuable for a better understanding of the mechanisms underlying pathologic immune responses.

Several of the inflammasome-targeting miRNAs are associated with and relevant to the development of autoimmune diseases such as MS, RA, and SLE in humans. This suggests that miRNAs may also be useful as biomarkers in immune-mediated disorders. In particular, miR-146a-5p, miR-155-5p, and miR-223-3p are dysregulated in diverse autoimmune diseases, and SNPs close to or within the pri-miRNA–coding sequences are associated with disease risk and severity. Exploring these miRNAs and also other miRNAs and the respective SNP alleles may further increase our understanding of the genetics underlying complex autoimmune disorders. Likewise, SNPs in the target sequences in mRNA 3′ UTRs can alter their regulation by miRNAs and thereby also affect disease risk. Regarding the use of mature miRNA molecules as biomarkers, combinations of several miRNAs in a biomarker panel can yield better predictions, as was already shown for the combination of miR-142-3p, miR-146a-5p, and miR-155-5p for the stratification of RRMS (87, 132). Most of the studies described in this review found dysregulated miRNA levels in blood compartments and some in pathologic tissues of patients with autoimmune diseases. Serum and plasma are easily accessible biological materials, and exploiting miRNAs from these compartments as biomarkers is feasible. It may be interesting to investigate whether miRNAs with biomarker potential that have only been detected in certain cell types and tissues are differentially expressed in serum and plasma as well when employing sensitive methods. Still, for many miRNAs discussed in this review, studies with large patient cohorts are required to evaluate whether they can indeed serve as reliable biomarkers for disease diagnosis, as well as for prognosis and for monitoring the individual response to therapy and whether the accuracy can be improved by combining different miRNAs. Such efforts should also shed light on to the role of these miRNAs in the pathomechanisms underlying many autoimmune diseases but also autoinflammatory diseases such as cryopyrin-associated periodic syndrome and familial Mediterranean fever.

Finally, in animal disease models, the modulation of miRNA expression levels (e.g., miR-7-5p and miR-30e-5p in PD mouse models) (5052) already showed promising therapeutic effects, but this potential has yet to be translated into the clinical setting. Novel miRNA therapeutics have been developed in the past few years, but some therapeutic miRNA studies have already been terminated during the preclinical stage because of side effects involving liver damage and poor miRNA stability (133). However, miRNA drug delivery, efficacy, and safety issues are currently being addressed, and recently, the first miRNA-based therapeutic, miravirsen, an inhibitor of miR-122-5p, yielded positive results in a phase II clinical trial (134). Further optimization of miRNA therapeutics may yield a wider spectrum of successful miRNA drugs in the years to come, possibly including inflammasome-modulating miRNAs for the treatment of autoimmune disorders.

This work was supported by a scholarship from the Stiftung der Deutschen Wirtschaft (to N.B.).

Abbreviations used in this article:

IBD

inflammatory bowel disease

miRNA

microRNA

MS

multiple sclerosis

MSU

monosodium urate

PD

Parkinson disease

pri-miRNA

primary miRNA

qPCR

quantitative real-time PCR

RA

rheumatoid arthritis

ROS

reactive oxygen species

RRMS

relapsing-remitting MS

SLE

systemic lupus erythematosus

SNP

single-nucleotide polymorphism

SOD

superoxide dismutase

T1D

type 1 diabetes

3′ UTR

3′ untranslated region.

1
Broz
,
P.
,
V. M.
Dixit
.
2016
.
Inflammasomes: mechanism of assembly, regulation and signalling.
Nat. Rev. Immunol.
16
:
407
420
.
2
Latz
,
E.
,
T. S.
Xiao
,
A.
Stutz
.
2013
.
Activation and regulation of the inflammasomes.
Nat. Rev. Immunol.
13
:
397
411
.
3
Broderick
,
L.
,
D.
De Nardo
,
B. S.
Franklin
,
H. M.
Hoffman
,
E.
Latz
.
2015
.
The inflammasomes and autoinflammatory syndromes.
Annu. Rev. Pathol.
10
:
395
424
.
4
Lin
,
C.
,
J.
Zhang
.
2017
.
Inflammasomes in inflammation-induced cancer.
Front. Immunol.
8
:
271
.
5
Barclay
,
W.
,
M. L.
Shinohara
.
2017
.
Inflammasome activation in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE).
Brain Pathol.
27
:
213
219
.
6
Shen
,
H.-H.
,
Y.-X.
Yang
,
X.
Meng
,
X.-Y.
Luo
,
X.-M.
Li
,
Z.-W.
Shuai
,
D.-Q.
Ye
,
H.-F.
Pan
.
2018
.
NLRP3: a promising therapeutic target for autoimmune diseases.
Autoimmun. Rev.
17
:
694
702
.
7
Bartel
,
D. P.
2018
.
Metazoan microRNAs.
Cell
173
:
20
51
.
8
Baker
,
P. J.
,
D.
Boucher
,
D.
Bierschenk
,
C.
Tebartz
,
P. G.
Whitney
,
D. B.
D’Silva
,
M. C.
Tanzer
,
M.
Monteleone
,
A. A. B.
Robertson
,
M. A.
Cooper
, et al
.
2015
.
NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase-5.
Eur. J. Immunol.
45
:
2918
2926
.
9
Vince
,
J. E.
,
W. W.-L.
Wong
,
I.
Gentle
,
K. E.
Lawlor
,
R.
Allam
,
L.
O’Reilly
,
K.
Mason
,
O.
Gross
,
S.
Ma
,
G.
Guarda
, et al
.
2012
.
Inhibitor of apoptosis proteins limit RIP3 kinase-dependent interleukin-1 activation.
Immunity
36
:
215
227
.
10
Gagliani
,
N.
,
N. W.
Palm
,
M. R.
de Zoete
,
R. A.
Flavell
.
2014
.
Inflammasomes and intestinal homeostasis: regulating and connecting infection, inflammation and the microbiota.
Int. Immunol.
26
:
495
499
.
11
Malhotra
,
S.
,
J.
Río
,
E.
Urcelay
,
R.
Nurtdinov
,
M. F.
Bustamante
,
O.
Fernández
,
B.
Oliver
,
U.
Zettl
,
D.
Brassat
,
J.
Killestein
, et al
.
2015
.
NLRP3 inflammasome is associated with the response to IFN-β in patients with multiple sclerosis.
Brain
138
:
644
652
.
12
Thamilarasan
,
M.
,
M.
Hecker
,
R. H.
Goertsches
,
B. K.
Paap
,
I.
Schröder
,
D.
Koczan
,
H.-J.
Thiesen
,
U. K.
Zettl
.
2013
.
Glatiramer acetate treatment effects on gene expression in monocytes of multiple sclerosis patients.
J. Neuroinflammation
10
:
126
.
13
Smolen
,
J. S.
,
P.
Emery
.
2011
.
Infliximab: 12 years of experience.
Arthritis Res. Ther.
13
(
Suppl. 1
):
S2
.
14
Yang
,
Q.
,
C.
Yu
,
Z.
Yang
,
Q.
Wei
,
K.
Mu
,
Y.
Zhang
,
W.
Zhao
,
X.
Wang
,
W.
Huai
,
L.
Han
.
2014
.
Deregulated NLRP3 and NLRP1 inflammasomes and their correlations with disease activity in systemic lupus erythematosus.
J. Rheumatol.
41
:
444
452
.
15
Martinon
,
F.
,
K.
Burns
,
J.
Tschopp
.
2002
.
The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta.
Mol. Cell
10
:
417
426
.
16
Dinarello
,
C. A.
2009
.
Immunological and inflammatory functions of the interleukin-1 family.
Annu. Rev. Immunol.
27
:
519
550
.
17
Strowig
,
T.
,
J.
Henao-Mejia
,
E.
Elinav
,
R.
Flavell
.
2012
.
Inflammasomes in health and disease.
Nature
481
:
278
286
.
18
Lech
,
M.
,
A.
Avila-Ferrufino
,
V.
Skuginna
,
H. E.
Susanti
,
H.-J.
Anders
.
2010
.
Quantitative expression of RIG-like helicase, NOD-like receptor and inflammasome-related mRNAs in humans and mice.
Int. Immunol.
22
:
717
728
.
19
Halle
,
A.
,
V.
Hornung
,
G. C.
Petzold
,
C. R.
Stewart
,
B. G.
Monks
,
T.
Reinheckel
,
K. A.
Fitzgerald
,
E.
Latz
,
K. J.
Moore
,
D. T.
Golenbock
.
2008
.
The NALP3 inflammasome is involved in the innate immune response to amyloid-beta.
Nat. Immunol.
9
:
857
865
.
20
Herman
,
F. J.
,
G. M.
Pasinetti
.
2018
.
Principles of inflammasome priming and inhibition: implications for psychiatric disorders.
Brain Behav. Immun.
73
:
66
84
.
21
Kozomara
,
A.
,
S.
Griffiths-Jones
.
2014
.
miRBase: annotating high confidence microRNAs using deep sequencing data.
Nucleic Acids Res.
42
(
D1
):
D68
D73
.
22
Freiesleben
,
S.
,
M.
Hecker
,
U. K.
Zettl
,
G.
Fuellen
,
L.
Taher
.
2016
.
Analysis of microRNA and gene expression profiles in multiple sclerosis: integrating interaction data to uncover regulatory mechanisms.
Sci. Rep.
6
:
34512
.
23
Hecker
,
M.
,
M.
Thamilarasan
,
D.
Koczan
,
I.
Schröder
,
K.
Flechtner
,
S.
Freiesleben
,
G.
Füllen
,
H.-J.
Thiesen
,
U. K.
Zettl
.
2013
.
MicroRNA expression changes during interferon-beta treatment in the peripheral blood of multiple sclerosis patients.
Int. J. Mol. Sci.
14
:
16087
16110
.
24
Cai
,
X.
,
C. H.
Hagedorn
,
B. R.
Cullen
.
2004
.
Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs.
RNA
10
:
1957
1966
.
25
Creugny
,
A.
,
A.
Fender
,
S.
Pfeffer
.
2018
.
Regulation of primary microRNA processing.
FEBS Lett.
592
:
1980
1996
.
26
Lee
,
Y.
,
C.
Ahn
,
J.
Han
,
H.
Choi
,
J.
Kim
,
J.
Yim
,
J.
Lee
,
P.
Provost
,
O.
Rådmark
,
S.
Kim
,
V. N.
Kim
.
2003
.
The nuclear RNase III Drosha initiates microRNA processing.
Nature
425
:
415
419
.
27
Lund
,
E.
,
S.
Güttinger
,
A.
Calado
,
J. E.
Dahlberg
,
U.
Kutay
.
2004
.
Nuclear export of microRNA precursors.
Science
303
:
95
98
.
28
Chendrimada
,
T. P.
,
R. I.
Gregory
,
E.
Kumaraswamy
,
J.
Norman
,
N.
Cooch
,
K.
Nishikura
,
R.
Shiekhattar
.
2005
.
TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing.
Nature
436
:
740
744
.
29
Iwasaki
,
S.
,
M.
Kobayashi
,
M.
Yoda
,
Y.
Sakaguchi
,
S.
Katsuma
,
T.
Suzuki
,
Y.
Tomari
.
2010
.
Hsc70/Hsp90 chaperone machinery mediates ATP-dependent RISC loading of small RNA duplexes.
Mol. Cell
39
:
292
299
.
30
Kawamata
,
T.
,
Y.
Tomari
.
2010
.
Making RISC.
Trends Biochem. Sci.
35
:
368
376
.
31
Frank
,
F.
,
N.
Sonenberg
,
B.
Nagar
.
2010
.
Structural basis for 5′-nucleotide base-specific recognition of guide RNA by human AGO2.
Nature
465
:
818
822
.
32
Suzuki
,
H. I.
,
A.
Katsura
,
T.
Yasuda
,
T.
Ueno
,
H.
Mano
,
K.
Sugimoto
,
K.
Miyazono
.
2015
.
Small-RNA asymmetry is directly driven by mammalian Argonautes.
Nat. Struct. Mol. Biol.
22
:
512
521
.
33
Ameres
,
S. L.
,
P. D.
Zamore
.
2013
.
Diversifying microRNA sequence and function.
Nat. Rev. Mol. Cell Biol.
14
:
475
488
.
34
Bartel
,
D. P.
2009
.
MicroRNAs: target recognition and regulatory functions.
Cell
136
:
215
233
.
35
Chen
,
C.-Y. A.
,
A.-B.
Shyu
.
2011
.
Mechanisms of deadenylation-dependent decay.
Wiley Interdiscip. Rev. RNA
2
:
167
183
.
36
Chu
,
C.-Y.
,
T. M.
Rana
.
2006
.
Translation repression in human cells by microRNA-induced gene silencing requires RCK/p54.
PLoS Biol.
4
:
e210
.
37
Djuranovic
,
S.
,
A.
Nahvi
,
R.
Green
.
2012
.
miRNA-mediated gene silencing by translational repression followed by mRNA deadenylation and decay.
Science
336
:
237
240
.
38
Eichhorn
,
S. W.
,
H.
Guo
,
S. E.
McGeary
,
R. A.
Rodriguez-Mias
,
C.
Shin
,
D.
Baek
,
S.-H.
Hsu
,
K.
Ghoshal
,
J.
Villén
,
D. P.
Bartel
.
2014
.
mRNA destabilization is the dominant effect of mammalian microRNAs by the time substantial repression ensues.
Mol. Cell
56
:
104
115
.
39
Jonas
,
S.
,
E.
Izaurralde
.
2015
.
Towards a molecular understanding of microRNA-mediated gene silencing.
Nat. Rev. Genet.
16
:
421
433
.
40
Chou
,
C.-H.
,
S.
Shrestha
,
C.-D.
Yang
,
N.-W.
Chang
,
Y.-L.
Lin
,
K.-W.
Liao
,
W.-C.
Huang
,
T.-H.
Sun
,
S.-J.
Tu
,
W.-H.
Lee
, et al
.
2018
.
miRTarBase update 2018: a resource for experimentally validated microRNA-target interactions.
Nucleic Acids Res.
46
(
D1
):
D296
D302
.
41
Dweep
,
H.
,
N.
Gretz
.
2015
.
miRWalk2.0: a comprehensive atlas of microRNA-target interactions.
Nat. Methods
12
:
697
.
42
Kuhn
,
D. E.
,
M. M.
Martin
,
D. S.
Feldman
,
A. V.
Terry
Jr.
,
G. J.
Nuovo
,
T. S.
Elton
.
2008
.
Experimental validation of miRNA targets.
Methods
44
:
47
54
.
43
Chi
,
S. W.
,
J. B.
Zang
,
A.
Mele
,
R. B.
Darnell
.
2009
.
Argonaute HITS-CLIP decodes microRNA-mRNA interaction maps.
Nature
460
:
479
486
.
44
Hafner
,
M.
,
M.
Landthaler
,
L.
Burger
,
M.
Khorshid
,
J.
Hausser
,
P.
Berninger
,
A.
Rothballer
,
M.
Ascano
Jr.
,
A.-C.
Jungkamp
,
M.
Munschauer
, et al
.
2010
.
Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP.
Cell
141
:
129
141
.
45
Haecker
,
I.
,
R.
Renne
.
2014
.
HITS-CLIP and PAR-CLIP advance viral miRNA targetome analysis.
Crit. Rev. Eukaryot. Gene Expr.
24
:
101
116
.
46
Cottrell
,
K. A.
,
H. G.
Chaudhari
,
B. A.
Cohen
,
S.
Djuranovic
.
2018
.
PTRE-seq reveals mechanism and interactions of RNA binding proteins and miRNAs.
Nat. Commun.
9
:
301
.
47
Haneklaus
,
M.
,
M.
Gerlic
,
M.
Kurowska-Stolarska
,
A.-A.
Rainey
,
D.
Pich
,
I. B.
McInnes
,
W.
Hammerschmidt
,
L. A. J.
O’Neill
,
S. L.
Masters
.
2012
.
Cutting edge: miR-223 and EBV miR-BART15 regulate the NLRP3 inflammasome and IL-1β production.
J. Immunol.
189
:
3795
3799
.
48
Bauernfeind
,
F.
,
A.
Rieger
,
F. A.
Schildberg
,
P. A.
Knolle
,
J. L.
Schmid-Burgk
,
V.
Hornung
.
2012
.
NLRP3 inflammasome activity is negatively controlled by miR-223.
J. Immunol.
189
:
4175
4181
.
49
Pakpoor
,
J.
,
G.
Disanto
,
J. E.
Gerber
,
R.
Dobson
,
U. C.
Meier
,
G.
Giovannoni
,
S. V.
Ramagopalan
.
2013
.
The risk of developing multiple sclerosis in individuals seronegative for Epstein-Barr virus: a meta-analysis.
Mult. Scler.
19
:
162
166
.
50
Fan
,
Z.
,
M.
Lu
,
C.
Qiao
,
Y.
Zhou
,
J.-H.
Ding
,
G.
Hu
.
2016
.
MicroRNA-7 enhances subventricular zone neurogenesis by inhibiting NLRP3/caspase-1 axis in adult neural stem cells.
Mol. Neurobiol.
53
:
7057
7069
.
51
Li
,
D.
,
H.
Yang
,
J.
Ma
,
S.
Luo
,
S.
Chen
,
Q.
Gu
.
2018
.
MicroRNA-30e regulates neuroinflammation in MPTP model of Parkinson’s disease by targeting Nlrp3.
Hum. Cell
31
:
106
115
.
52
Zhou
,
Y.
,
M.
Lu
,
R.-H.
Du
,
C.
Qiao
,
C.-Y.
Jiang
,
K.-Z.
Zhang
,
J.-H.
Ding
,
G.
Hu
.
2016
.
MicroRNA-7 targets Nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson’s disease.
Mol. Neurodegener.
11
:
28
.
53
Codolo
,
G.
,
N.
Plotegher
,
T.
Pozzobon
,
M.
Brucale
,
I.
Tessari
,
L.
Bubacco
,
M.
de Bernard
.
2013
.
Triggering of inflammasome by aggregated α-synuclein, an inflammatory response in synucleinopathies.
PLoS One
8
:
e55375
.
54
Lou
,
J.
,
Y.
Wang
,
Z.
Zhang
,
W.
Qiu
.
2017
.
MiR-20b inhibits mycobacterium tuberculosis induced inflammation in the lung of mice through targeting NLRP3.
Exp. Cell Res.
358
:
120
128
.
55
Huang
,
W.-Q.
,
P.
Wei
,
R.-Q.
Lin
,
F.
Huang
.
2017
.
Protective effects of microrna-22 against endothelial cell injury by targeting NLRP3 through suppression of the inflammasome signaling pathway in a rat model of coronary heart disease.
Cell. Physiol. Biochem.
43
:
1346
1358
.
56
Xiao
,
L.
,
L.
Jiang
,
Q.
Hu
,
Y.
Li
.
2017
.
MicroRNA-133b ameliorates allergic inflammation and symptom in murine model of allergic rhinitis by targeting Nlrp3.
Cell. Physiol. Biochem.
42
:
901
912
.
57
Chen
,
M.-L.
,
K.
Lin
,
S.-K.
Lin
.
2018
.
NLRP3 inflammasome signaling as an early molecular response is negatively controlled by miR-186 in CFA-induced prosopalgia mice.
Braz. J. Med. Biol. Res.
51
:
e7602
.
58
Zhou
,
T.
,
D.-K.
Xiang
,
S.-N.
Li
,
L.-H.
Yang
,
L.-F.
Gao
,
C.
Feng
.
2018
.
MicroRNA-495 ameliorates cardiac microvascular endothelial cell injury and inflammatory reaction by suppressing the NLRP3 inflammasome signaling pathway.
Cell. Physiol. Biochem.
49
:
798
815
.
59
Jin
,
X.
,
H.
Jin
,
Y.
Shi
,
Y.
Guo
,
H.
Zhang
.
2017
.
Long non-coding RNA KCNQ1OT1 promotes cataractogenesis via miR-214 and activation of the caspase-1 pathway.
Cell. Physiol. Biochem.
42
:
295
305
.
60
Visalli
,
M.
,
M.
Bartolotta
,
F.
Polito
,
R.
Oteri
,
A.
Barbera
,
R.
Arrigo
,
R. M.
Di Giorgio
,
G.
Navarra
,
M.
Aguennouz
.
2018
.
miRNA expression profiling regulates necroptotic cell death in hepatocellular carcinoma.
Int. J. Oncol.
53
:
771
780
.
61
Truettner
,
J. S.
,
D.
Motti
,
W. D.
Dietrich
.
2013
.
MicroRNA overexpression increases cortical neuronal vulnerability to injury.
Brain Res.
1533
:
122
130
.
62
Jain
,
A.
,
S.
Kaczanowska
,
E.
Davila
.
2014
.
IL-1 receptor-associated kinase signaling and its role in inflammation, cancer progression, and therapy resistance.
Front. Immunol.
5
:
553
.
63
Ma
,
T.
,
X.
Liu
,
Z.
Cen
,
C.
Xin
,
M.
Guo
,
C.
Zou
,
W.
Song
,
R.
Xie
,
K.
Wang
,
H.
Zhou
, et al
.
2018
.
MicroRNA-302b negatively regulates IL-1β production in response to MSU crystals by targeting IRAK4 and EphA2.
Arthritis Res. Ther.
20
:
34
.
64
Zhang
,
Q.-B.
,
Y.-F.
Qing
,
C.-C.
Yin
,
L.
Zhou
,
X.-S.
Liu
,
Q.-S.
Mi
,
J.-G.
Zhou
.
2018
.
Mice with miR-146a deficiency develop severe gouty arthritis via dysregulation of TRAF 6, IRAK 1 and NALP3 inflammasome.
Arthritis Res. Ther.
20
:
45
.
65
Jia
,
K.-K.
,
S.-M.
Pan
,
H.
Ding
,
J.-H.
Liu
,
Y.-J.
Zheng
,
S.-J.
Wang
,
Y.
Pan
,
L.-D.
Kong
.
2018
.
Chaihu-shugan san inhibits inflammatory response to improve insulin signaling in liver and prefrontal cortex of CUMS rats with glucose intolerance.
Biomed. Pharmacother.
103
:
1415
1428
.
66
Unlu
,
S.
,
S.
Tang
,
E.
Wang
,
I.
Martinez
,
D.
Tang
,
M. E.
Bianchi
,
H. J.
Zeh
III
,
M. T.
Lotze
.
2012
.
Damage associated molecular pattern molecule-induced microRNAs (DAMPmiRs) in human peripheral blood mononuclear cells. [Published erratum appears in 2012 PLoS One 7.]
PLoS One
7
:
e38899
.
67
Lang
,
D.
,
J.
Knop
,
H.
Wesche
,
U.
Raffetseder
,
R.
Kurrle
,
D.
Boraschi
,
M. U.
Martin
.
1998
.
The type II IL-1 receptor interacts with the IL-1 receptor accessory protein: a novel mechanism of regulation of IL-1 responsiveness.
J. Immunol.
161
:
6871
6877
.
68
Lian
,
Z.
,
F.-F.
Lv
,
J.
Yu
,
J.-W.
Wang
.
2018
.
The anti-inflammatory effect of microRNA-383-3p interacting with IL1R2 against homocysteine-induced endothelial injury in rat coronary arteries.
J. Cell. Biochem.
119
:
6684
6694
.
69
Carr
,
J. M.
,
S.
Mahalingam
,
C. S.
Bonder
,
S. M.
Pitson
.
2013
.
Sphingosine kinase 1 in viral infections.
Rev. Med. Virol.
23
:
73
84
.
70
Gu
,
T.-T.
,
L.
Song
,
T.-Y.
Chen
,
X.
Wang
,
X.-J.
Zhao
,
X.-Q.
Ding
,
Y.-Z.
Yang
,
Y.
Pan
,
D.-M.
Zhang
,
L.-D.
Kong
.
2017
.
Fructose downregulates miR-330 to induce renal inflammatory response and insulin signaling impairment: attenuation by morin.
Mol. Nutr. Food Res.
61
:
1600760
.
71
Malireddi
,
R. K. S.
,
T.-D.
Kanneganti
.
2013
.
Role of type I interferons in inflammasome activation, cell death, and disease during microbial infection.
Front. Cell. Infect. Microbiol.
3
:
77
.
72
Wang
,
Y.
,
Z.
Han
,
Y.
Fan
,
J.
Zhang
,
K.
Chen
,
L.
Gao
,
H.
Zeng
,
J.
Cao
,
C.
Wang
.
2017
.
MicroRNA-9 inhibits NLRP3 inflammasome activation in human atherosclerosis inflammation cell models through the JAK1/STAT signaling pathway.
Cell. Physiol. Biochem.
41
:
1555
1571
.
73
Chen
,
T.
,
Z.
Huang
,
L.
Wang
,
Y.
Wang
,
F.
Wu
,
S.
Meng
,
C.
Wang
.
2009
.
MicroRNA-125a-5p partly regulates the inflammatory response, lipid uptake, and ORP9 expression in oxLDL-stimulated monocyte/macrophages.
Cardiovasc. Res.
83
:
131
139
.
74
Zhou
,
R.
,
A. S.
Yazdi
,
P.
Menu
,
J.
Tschopp
.
2011
.
A role for mitochondria in NLRP3 inflammasome activation. [Published erratum appears in 2011 Nature 475: 122.]
Nature
469
:
221
225
.
75
Wang
,
W.
,
X.-Q.
Ding
,
T.-T.
Gu
,
L.
Song
,
J.-M.
Li
,
Q.-C.
Xue
,
L.-D.
Kong
.
2015
.
Pterostilbene and allopurinol reduce fructose-induced podocyte oxidative stress and inflammation via microRNA-377.
Free Radic. Biol. Med.
83
:
214
226
.
76
Lerner
,
A. G.
,
J.-P.
Upton
,
P. V. K.
Praveen
,
R.
Ghosh
,
Y.
Nakagawa
,
A.
Igbaria
,
S.
Shen
,
V.
Nguyen
,
B. J.
Backes
,
M.
Heiman
, et al
.
2012
.
IRE1α induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress.
Cell Metab.
16
:
250
264
.
77
Chen
,
D.
,
B. J.
Dixon
,
D. M.
Doycheva
,
B.
Li
,
Y.
Zhang
,
Q.
Hu
,
Y.
He
,
Z.
Guo
,
D.
Nowrangi
,
J.
Flores
, et al
.
2018
.
IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic-ischemic brain injury in rats.
J. Neuroinflammation
15
:
32
.
78
Li
,
X.-F.
,
W.-W.
Shen
,
Y.-Y.
Sun
,
W.-X.
Li
,
Z.-H.
Sun
,
Y.-H.
Liu
,
L.
Zhang
,
C.
Huang
,
X.-M.
Meng
,
J.
Li
.
2016
.
MicroRNA-20a negatively regulates expression of NLRP3-inflammasome by targeting TXNIP in adjuvant-induced arthritis fibroblast-like synoviocytes.
Joint Bone Spine
83
:
695
700
.
79
Heo
,
M. J.
,
T. H.
Kim
,
J. S.
You
,
D.
Blaya
,
P.
Sancho-Bru
,
S. G.
Kim
.
2018
.
Alcohol dysregulates miR-148a in hepatocytes through FoxO1, facilitating pyroptosis via TXNIP overexpression.
Gut
. DOI: 10.1136/gutjnl-2017-315123.
80
Pan
,
Z.
,
Q.
Shan
,
P.
Gu
,
X. M.
Wang
,
L. W.
Tai
,
M.
Sun
,
X.
Luo
,
L.
Sun
,
C. W.
Cheung
.
2018
.
miRNA-23a/CXCR4 regulates neuropathic pain via directly targeting TXNIP/NLRP3 inflammasome axis.
J. Neuroinflammation
15
:
29
.
81
Li
,
P.
,
X.
Zhong
,
J.
Li
,
H.
Liu
,
X.
Ma
,
R.
He
,
Y.
Zhao
.
2018
.
MicroRNA-30c-5p inhibits NLRP3 inflammasome-mediated endothelial cell pyroptosis through FOXO3 down-regulation in atherosclerosis.
Biochem. Biophys. Res. Commun.
503
:
2833
2840
.
82
Zhong
,
W.
,
B.
Li
,
Y.
Xu
,
P.
Yang
,
R.
Chen
,
Z.
Wang
,
C.
Shao
,
J.
Song
,
J.
Yan
.
2018
.
Hypermethylation of the micro-RNA 145 promoter is the key regulator for NLRP3 inflammasome-induced activation and plaque formation.
JACC Basic Transl. Sci.
3
:
604
624
.
83
Guo
,
L.-M.
,
Y.
Pu
,
Z.
Han
,
T.
Liu
,
Y.-X.
Li
,
M.
Liu
,
X.
Li
,
H.
Tang
.
2009
.
MicroRNA-9 inhibits ovarian cancer cell growth through regulation of NF-kappaB1.
FEBS J.
276
:
5537
5546
.
84
Zhu
,
M.
,
M.
Wang
,
F.
Yang
,
Y.
Tian
,
J.
Cai
,
H.
Yang
,
H.
Fu
,
F.
Mao
,
W.
Zhu
,
H.
Qian
,
W.
Xu
.
2016
.
miR-155-5p inhibition promotes the transition of bone marrow mesenchymal stem cells to gastric cancer tissue derived MSC-like cells via NF-κB p65 activation.
Oncotarget
7
:
16567
16580
.
85
Zhao
,
X.-D.
,
Y.-Y.
Lu
,
H.
Guo
,
H.-H.
Xie
,
L.-J.
He
,
G.-F.
Shen
,
J.-F.
Zhou
,
T.
Li
,
S.-J.
Hu
,
L.
Zhou
, et al
.
2015
.
MicroRNA-7/NF-κB signaling regulatory feedback circuit regulates gastric carcinogenesis.
J. Cell Biol.
210
:
613
627
.
86
Fenoglio
,
C.
,
C.
Cantoni
,
M.
De Riz
,
E.
Ridolfi
,
F.
Cortini
,
M.
Serpente
,
C.
Villa
,
C.
Comi
,
F.
Monaco
,
L.
Mellesi
, et al
.
2011
.
Expression and genetic analysis of miRNAs involved in CD4+ cell activation in patients with multiple sclerosis.
Neurosci. Lett.
504
:
9
12
.
87
Waschbisch
,
A.
,
M.
Atiya
,
R. A.
Linker
,
S.
Potapov
,
S.
Schwab
,
T.
Derfuss
.
2011
.
Glatiramer acetate treatment normalizes deregulated microRNA expression in relapsing remitting multiple sclerosis.
PLoS One
6
:
e24604
.
88
Li
,
Y.
,
C.
Du
,
W.
Wang
,
G.
Ma
,
L.
Cui
,
H.
Zhou
,
H.
Tao
,
L.
Yao
,
B.
Zhao
,
K.
Li
.
2015
.
Genetic association of MiR-146a with multiple sclerosis susceptibility in the Chinese population.
Cell. Physiol. Biochem.
35
:
281
291
.
89
Moore
,
C. S.
,
V. T. S.
Rao
,
B. A.
Durafourt
,
B. J.
Bedell
,
S. K.
Ludwin
,
A.
Bar-Or
,
J. P.
Antel
.
2013
.
miR-155 as a multiple sclerosis-relevant regulator of myeloid cell polarization.
Ann. Neurol.
74
:
709
720
.
90
Siegel
,
S. R.
,
J.
Mackenzie
,
G.
Chaplin
,
N. G.
Jablonski
,
L.
Griffiths
.
2012
.
Circulating microRNAs involved in multiple sclerosis.
Mol. Biol. Rep.
39
:
6219
6225
.
91
Magner
,
W. J.
,
B.
Weinstock-Guttman
,
M.
Rho
,
D.
Hojnacki
,
R.
Ghazi
,
M.
Ramanathan
,
T. B.
Tomasi
.
2016
.
Dicer and microRNA expression in multiple sclerosis and response to interferon therapy.
J. Neuroimmunol.
292
:
68
78
.
92
Fenoglio
,
C.
,
E.
Ridolfi
,
C.
Cantoni
,
M.
De Riz
,
R.
Bonsi
,
M.
Serpente
,
C.
Villa
,
A. M.
Pietroboni
,
R. T.
Naismith
,
E.
Alvarez
, et al
.
2013
.
Decreased circulating miRNA levels in patients with primary progressive multiple sclerosis.
Mult. Scler.
19
:
1938
1942
.
93
Kiselev
,
I.
,
V.
Bashinskaya
,
O.
Kulakova
,
N.
Baulina
,
E.
Popova
,
A.
Boyko
,
O.
Favorova
.
2015
.
Variants of microRNA genes: gender-specific associations with multiple sclerosis risk and severity.
Int. J. Mol. Sci.
16
:
20067
20081
.
94
Ahmadian-Elmi
,
M.
,
A.
Bidmeshki Pour
,
R.
Naghavian
,
K.
Ghaedi
,
S.
Tanhaei
,
T.
Izadi
,
M. H.
Nasr-Esfahani
.
2016
.
miR-27a and miR-214 exert opposite regulatory roles in Th17 differentiation via mediating different signaling pathways in peripheral blood CD4+ T lymphocytes of patients with relapsing-remitting multiple sclerosis.
Immunogenetics
68
:
43
54
.
95
Meira
,
M.
,
C.
Sievers
,
F.
Hoffmann
,
M.
Rasenack
,
J.
Kuhle
,
T.
Derfuss
,
L.
Kappos
,
R. L. P.
Lindberg
.
2014
.
Unraveling natalizumab effects on deregulated miR-17 expression in CD4+ T cells of patients with relapsing-remitting multiple sclerosis.
J. Immunol. Res.
2014
:
897249
.
96
Ingwersen
,
J.
,
T.
Menge
,
B.
Wingerath
,
D.
Kaya
,
J.
Graf
,
T.
Prozorovski
,
A.
Keller
,
C.
Backes
,
M.
Beier
,
M.
Scheffler
, et al
.
2015
.
Natalizumab restores aberrant miRNA expression profile in multiple sclerosis and reveals a critical role for miR-20b.
Ann. Clin. Transl. Neurol.
2
:
43
55
.
97
Lin
,
J.
,
R.
Huo
,
L.
Xiao
,
X.
Zhu
,
J.
Xie
,
S.
Sun
,
Y.
He
,
J.
Zhang
,
Y.
Sun
,
Z.
Zhou
, et al
.
2014
.
A novel p53/microRNA-22/Cyr61 axis in synovial cells regulates inflammation in rheumatoid arthritis.
Arthritis Rheumatol.
66
:
49
59
.
98
Krintel
,
S. B.
,
C.
Dehlendorff
,
M. L.
Hetland
,
K.
Hørslev-Petersen
,
K. K.
Andersen
,
P.
Junker
,
J.
Pødenphant
,
T.
Ellingsen
,
P.
Ahlquist
,
H. M.
Lindegaard
, et al
.
2016
.
Prediction of treatment response to adalimumab: a double-blind placebo-controlled study of circulating microRNA in patients with early rheumatoid arthritis.
Pharmacogenomics J.
16
:
141
146
.
99
Wang
,
W.
,
Y.
Zhang
,
B.
Zhu
,
T.
Duan
,
Q.
Xu
,
R.
Wang
,
L.
Lu
,
Z.
Jiao
.
2015
.
Plasma microRNA expression profiles in Chinese patients with rheumatoid arthritis.
Oncotarget
6
:
42557
42568
.
100
Shibuya
,
H.
,
T.
Nakasa
,
N.
Adachi
,
Y.
Nagata
,
M.
Ishikawa
,
M.
Deie
,
O.
Suzuki
,
M.
Ochi
.
2013
.
Overexpression of microRNA-223 in rheumatoid arthritis synovium controls osteoclast differentiation.
Mod. Rheumatol.
23
:
674
685
.
101
Kurowska-Stolarska
,
M.
,
S.
Alivernini
,
L. E.
Ballantine
,
D. L.
Asquith
,
N. L.
Millar
,
D. S.
Gilchrist
,
J.
Reilly
,
M.
Ierna
,
A. R.
Fraser
,
B.
Stolarski
, et al
.
2011
.
MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis.
Proc. Natl. Acad. Sci. USA
108
:
11193
11198
.
102
Nakasa
,
T.
,
S.
Miyaki
,
A.
Okubo
,
M.
Hashimoto
,
K.
Nishida
,
M.
Ochi
,
H.
Asahara
.
2008
.
Expression of microRNA-146 in rheumatoid arthritis synovial tissue.
Arthritis Rheum.
58
:
1284
1292
.
103
Long
,
L.
,
P.
Yu
,
Y.
Liu
,
S.
Wang
,
R.
Li
,
J.
Shi
,
X.
Zhang
,
Y.
Li
,
X.
Sun
,
B.
Zhou
, et al
.
2013
.
Upregulated microRNA-155 expression in peripheral blood mononuclear cells and fibroblast-like synoviocytes in rheumatoid arthritis.
Clin. Dev. Immunol.
2013
:
296139
.
104
Pauley
,
K. M.
,
M.
Satoh
,
A. L.
Chan
,
M. R.
Bubb
,
W. H.
Reeves
,
E. K.
Chan
.
2008
.
Upregulated miR-146a expression in peripheral blood mononuclear cells from rheumatoid arthritis patients.
Arthritis Res. Ther.
10
:
R101
.
105
Lu
,
M.-C.
,
C.-L.
Yu
,
H.-C.
Chen
,
H.-C.
Yu
,
H.-B.
Huang
,
N.-S.
Lai
.
2014
.
Increased miR-223 expression in T cells from patients with rheumatoid arthritis leads to decreased insulin-like growth factor-1-mediated interleukin-10 production.
Clin. Exp. Immunol.
177
:
641
651
.
106
Zhou
,
Q.
,
S.
Haupt
,
J. T.
Kreuzer
,
A.
Hammitzsch
,
F.
Proft
,
C.
Neumann
,
J.
Leipe
,
M.
Witt
,
H.
Schulze-Koops
,
A.
Skapenko
.
2015
.
Decreased expression of miR-146a and miR-155 contributes to an abnormal Treg phenotype in patients with rheumatoid arthritis.
Ann. Rheum. Dis.
74
:
1265
1274
.
107
Zhou
,
X.
,
J.
Zhu
,
H.
Zhang
,
G.
Zhou
,
Y.
Huang
,
R.
Liu
.
2015
.
Is the microRNA-146a (rs2910164) polymorphism associated with rheumatoid arthritis? Association of microRNA-146a (rs2910164) polymorphism and rheumatoid arthritis could depend on gender.
Joint Bone Spine
82
:
166
171
.
108
Wang
,
G.
,
L.-S.
Tam
,
B. C.-H.
Kwan
,
E. K.-M.
Li
,
K.-M.
Chow
,
C. C.-W.
Luk
,
P. K.-T.
Li
,
C.-C.
Szeto
.
2012
.
Expression of miR-146a and miR-155 in the urinary sediment of systemic lupus erythematosus.
Clin. Rheumatol.
31
:
435
440
.
109
Tang
,
Q.
,
Y.
Yang
,
M.
Zhao
,
G.
Liang
,
H.
Wu
,
Q.
Liu
,
Y.
Xie
,
D.
Li
,
Y.
Dai
,
S.
Yung
, et al
.
2015
.
Mycophenolic acid upregulates miR-142-3P/5P and miR-146a in lupus CD4+T cells.
Lupus
24
:
935
942
.
110
Ji
,
J. D.
,
E. S.
Cha
,
W. J.
Lee
.
2014
.
Association of miR-146a polymorphisms with systemic lupus erythematosus: a meta-analysis.
Lupus
23
:
1023
1030
.
111
Löfgren
,
S. E.
,
J.
Frostegård
,
L.
Truedsson
,
B. A.
Pons-Estel
,
S.
D’Alfonso
,
T.
Witte
,
B. R.
Lauwerys
,
E.
Endreffy
,
L.
Kovács
,
C.
Vasconcelos
, et al
.
2012
.
Genetic association of miRNA-146a with systemic lupus erythematosus in Europeans through decreased expression of the gene.
Genes Immun.
13
:
268
274
.
112
Duroux-Richard
,
I.
,
J.
Cuenca
,
C.
Ponsolles
,
A. B.
Piñeiro
,
F.
Gonzalez
,
C.
Roubert
,
R.
Areny
,
R.
Chea
,
J.
Pefaur
,
Y.-M.
Pers
, et al
.
2015
.
MicroRNA profiling of B cell subsets from systemic lupus erythematosus patients reveals promising novel biomarkers.
Int. J. Mol. Sci.
16
:
16953
16965
.
113
Teruel
,
R.
,
C.
Pérez-Sánchez
,
J.
Corral
,
M. T.
Herranz
,
V.
Pérez-Andreu
,
E.
Saiz
,
N.
García-Barberá
,
I.
Martínez-Martínez
,
V.
Roldán
,
V.
Vicente
, et al
.
2011
.
Identification of miRNAs as potential modulators of tissue factor expression in patients with systemic lupus erythematosus and antiphospholipid syndrome.
J. Thromb. Haemost.
9
:
1985
1992
.
114
Kaga
,
H.
,
A.
Komatsuda
,
A.
Omokawa
,
M.
Ito
,
K.
Teshima
,
H.
Tagawa
,
K.
Sawada
,
H.
Wakui
.
2015
.
Downregulated expression of miR-155, miR-17, and miR-181b, and upregulated expression of activation-induced cytidine deaminase and interferon-α in PBMCs from patients with SLE.
Mod. Rheumatol.
25
:
865
870
.
115
Sarhan
,
R. A.
,
H. R. A.
Aboelenein
,
S. K. N.
Sourour
,
I. O.
Fawzy
,
S.
Salah
,
A. I.
Abdelaziz
.
2015
.
Targeting E2F1 and c-Myc expression by microRNA-17-5p represses interferon-stimulated gene MxA in peripheral blood mononuclear cells of pediatric systemic lupus erythematosus patients.
Discov. Med.
19
:
419
425
.
116
Aboelenein
,
H. R. A.
,
S.
Salah
,
Y. A.
Lashine
,
A. I.
Abdelaziz
.
2013
.
Dual downregulation of microRNA 17-5p and E2F1 transcriptional factor in pediatric systemic lupus erythematosus patients.
Rheumatol. Int.
33
:
1333
1338
.
117
Lu
,
J.
,
B. C.-H.
Kwan
,
F. M.-M.
Lai
,
L.-S.
Tam
,
E. K.-M.
Li
,
K.-M.
Chow
,
G.
Wang
,
P. K.-T.
Li
,
C.-C.
Szeto
.
2012
.
Glomerular and tubulointerstitial miR-638, miR-198 and miR-146a expression in lupus nephritis.
Nephrology (Carlton)
17
:
346
351
.
118
Qingjuan
,
L.
,
F.
Xiaojuan
,
Z.
Wei
,
W.
Chao
,
K.
Pengpeng
,
L.
Hongbo
,
Z.
Sanbing
,
H.
Jun
,
Y.
Min
,
L.
Shuxia
.
2016
.
miR-148a-3p overexpression contributes to glomerular cell proliferation by targeting PTEN in lupus nephritis.
Am. J. Physiol. Cell Physiol.
310
:
C470
C478
.
119
Svrcek
,
M.
,
N.
El-Murr
,
K.
Wanherdrick
,
S.
Dumont
,
L.
Beaugerie
,
J.
Cosnes
,
J.-F.
Colombel
,
E.
Tiret
,
J.-F.
Fléjou
,
T.
Lesuffleur
,
A.
Duval
.
2013
.
Overexpression of microRNAs-155 and 21 targeting mismatch repair proteins in inflammatory bowel diseases.
Carcinogenesis
34
:
828
834
.
120
Paraskevi
,
A.
,
G.
Theodoropoulos
,
I.
Papaconstantinou
,
G.
Mantzaris
,
N.
Nikiteas
,
M.
Gazouli
.
2012
.
Circulating MicroRNA in inflammatory bowel disease.
J. Crohn’s Colitis
6
:
900
904
.
121
Lin
,
J.
,
N. C.
Welker
,
Z.
Zhao
,
Y.
Li
,
J.
Zhang
,
S. A.
Reuss
,
X.
Zhang
,
H.
Lee
,
Y.
Liu
,
M. P.
Bronner
.
2014
.
Novel specific microRNA biomarkers in idiopathic inflammatory bowel disease unrelated to disease activity.
Mod. Pathol.
27
:
602
608
.
122
Yang
,
M.
,
L.
Ye
,
B.
Wang
,
J.
Gao
,
R.
Liu
,
J.
Hong
,
W.
Wang
,
W.
Gu
,
G.
Ning
.
2015
.
Decreased miR-146 expression in peripheral blood mononuclear cells is correlated with ongoing islet autoimmunity in type 1 diabetes patients 1miR-146.
J. Diabetes
7
:
158
165
.
123
Sulzer
,
D.
,
R. N.
Alcalay
,
F.
Garretti
,
L.
Cote
,
E.
Kanter
,
J.
Agin-Liebes
,
C.
Liong
,
C.
McMurtrey
,
W. H.
Hildebrand
,
X.
Mao
, et al
.
2017
.
T cells from patients with Parkinson’s disease recognize α-synuclein peptides.
Nature
546
:
656
661
.
124
Lang
,
Y.
,
F.
Chu
,
D.
Shen
,
W.
Zhang
,
C.
Zheng
,
J.
Zhu
,
L.
Cui
.
2018
.
Role of inflammasomes in neuroimmune and neurodegenerative diseases: a systematic review.
Mediators Inflamm.
2018
:
1549549
.
125
Alieva
,
A. Kh.
,
E. V.
Filatova
,
A. V.
Karabanov
,
S. N.
Illarioshkin
,
S. A.
Limborska
,
M. I.
Shadrina
,
P. A.
Slominsky
.
2015
.
miRNA expression is highly sensitive to a drug therapy in Parkinson’s disease.
Parkinsonism Relat. Disord.
21
:
72
74
.
126
Margis
,
R.
,
R.
Margis
,
C. R. M.
Rieder
.
2011
.
Identification of blood microRNAs associated to Parkinsonĭs disease.
J. Biotechnol.
152
:
96
101
.
127
Miñones-Moyano
,
E.
,
S.
Porta
,
G.
Escaramís
,
R.
Rabionet
,
S.
Iraola
,
B.
Kagerbauer
,
Y.
Espinosa-Parrilla
,
I.
Ferrer
,
X.
Estivill
,
E.
Martí
.
2011
.
MicroRNA profiling of Parkinson’s disease brains identifies early downregulation of miR-34b/c which modulate mitochondrial function.
Hum. Mol. Genet.
20
:
3067
3078
.
128
Lee
,
R. C.
,
R. L.
Feinbaum
,
V.
Ambros
.
1993
.
The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14.
Cell
75
:
843
854
.
129
de Rie
,
D.
,
I.
Abugessaisa
,
T.
Alam
,
E.
Arner
,
P.
Arner
,
H.
Ashoor
,
G.
Åström
,
M.
Babina
,
N.
Bertin
,
A. M.
Burroughs
, et al
.
2017
.
An integrated expression atlas of miRNAs and their promoters in human and mouse.
Nat. Biotechnol.
35
:
872
878
.
130
Contreras
,
J.
,
D. S.
Rao
.
2012
.
MicroRNAs in inflammation and immune responses.
Leukemia
26
:
404
413
.
131
Lee
,
Y. H.
,
H. S.
Na
,
S. Y.
Jeong
,
S. H.
Jeong
,
H. R.
Park
,
J.
Chung
.
2011
.
Comparison of inflammatory microRNA expression in healthy and periodontitis tissues.
Biocell
35
:
43
49
.
132
Gandhi
,
R.
2015
.
miRNA in multiple sclerosis: search for novel biomarkers.
Mult. Scler.
21
:
1095
1103
.
133
Chakraborty
,
C.
,
A. R.
Sharma
,
G.
Sharma
,
C. G. P.
Doss
,
S.-S.
Lee
.
2017
.
Therapeutic miRNA and siRNA: moving from bench to clinic as next generation medicine.
Mol. Ther. Nucleic Acids
8
:
132
143
.
134
Oberemok
,
V. V.
,
K. V.
Laikova
,
A. I.
Repetskaya
,
I. M.
Kenyo
,
M. V.
Gorlov
,
I. N.
Kasich
,
A. M.
Krasnodubets
,
N. V.
Gal’chinsky
,
I. I.
Fomochkina
,
A. S.
Zaitsev
, et al
.
2018
.
A half-century history of applications of antisense oligonucleotides in medicine, agriculture and forestry: we should continue the journey.
Molecules
23
:
1302
.
135
Lorenz
,
R.
,
S. H.
Bernhart
,
C.
Höner Zu Siederdissen
,
H.
Tafer
,
C.
Flamm
,
P. F.
Stadler
,
I. L.
Hofacker
.
2011
.
ViennaRNA package 2.0.
Algorithms Mol. Biol.
6
:
26
.
136
Rehmsmeier
,
M.
,
P.
Steffen
,
M.
Höchsmann
,
R.
Giegerich
.
2004
.
Fast and effective prediction of microRNA/target duplexes.
RNA
10
:
1507
1517
.

N.B. received speaking fees and travel funds from Novartis. M.H. received speaking fees and travel funds from Bayer HealthCare, Biogen, Novartis and Teva. U.K.Z. received research support as well as speaking fees and travel funds from Almirall, Bayer HealthCare, Biogen, Merck Serono, Novartis, Sanofi Genzyme, and Teva.