key: cord-0026538-at7p281k authors: Yu, Xueyuan; Lv, Jiajia; Wu, Jun; Chen, Yong; Chen, Fei; Wang, Li title: The autoimmune encephalitis-related cytokine TSLP in the brain primes neuroinflammation by activating the JAK2-NLRP3 axis date: 2021-11-27 journal: Clin Exp Immunol DOI: 10.1093/cei/uxab023 sha: b22381a2380a135d00a46a460df6db8cd23c8093 doc_id: 26538 cord_uid: at7p281k NLRP3 inflammasome hyperactivation contributes to neuroinflammation in autoimmune disorders, but the underlying regulatory mechanism remains to be elucidated. We demonstrate that compared with wild-type (WT) mice, mice lacking thymic stromal lymphopoietin (TSLP) receptor (TSLPR) (Tslpr(−/−) mice) exhibit a significantly decreased experimental autoimmune encephalomyelitis (EAE) score, reduced CD4(+) T cell infiltration, and restored myelin basic protein (MBP) expression in the brain after EAE induction by myelin oligodendrocyte glycoprotein(35–55) (MOG(35–55)). TSLPR signals through Janus kinase (JAK)2, but not JAK1 or JAK3, to induce NLRP3 expression, and Tslpr(−/−) mice with EAE show decreased JAK2 phosphorylation and NLRP3 expression in the brain. JAK2 inhibition by ruxolitinib mimicked loss of TSLPR function in vivo and further decreased TSLP expression in the EAE mouse brain. The NLRP3 inhibitor MCC950 decreased CD4(+) T cell infiltration, restored MBP expression, and decreased IL-1β and TSLP levels, verifying the pro-inflammatory role of NLRP3. In vitro experiments using BV-2 murine microglia revealed that TSLP directly induced NLRP3 expression, phosphorylation of JAK2 but not JAK1orJAK3, and IL-1β release, which were markedly inhibited by ruxolitinib. Furthermore, EAE induction led to an increase in the Th17 cell number, a decrease in the regulatory T (Treg) cell number in the blood, and an increase in the expression of the cytokine IL-17A in the WT mouse brain, which was drastically reversed in Tslpr(−/−) mice. In addition, ruxolitinib suppressed the increase in IL-17A expression in the EAE mouse brain. These findings identify TSLP as a prospective target for treating JAK2-NLRP3 axis-associated autoimmune inflammatory disorders. Experimental autoimmune encephalomyelitis (EAE), which is mediated by myelin-specific autoreactive T helper cells, is a classical animal model of autoimmune encephalitis, which includes multiple sclerosis (MS), a disease characterized by typical demyelination and neurodegeneration-associated symptoms [1] . Although drugs targeting several immunological pathways have shown beneficial effects in patients with demyelinating diseases, no cure is currently available [2] . Nucleotide-binding domain, leucine-rich repeat containing protein family, pyrin domain containing 3 (NLRP3), and the interleukin (IL)-1β pathway have been shown to be crucial for the development of EAE by participating in neuroinflammation [3, 4] . NLRP3 is activated in two ways: by stimulation of pathogen recognition receptors that activate nuclear factor κB (NF-κB) pathways and by stimuli such as lysosomal rupture, adenosine 5ʹ-triphosphate (ATP), and reactive oxygen species (ROS) [5] . Activation of the NLRP3 inflammasome leads to the production of cleaved caspase-1, which cleaves proIL-1β/pro-IL-18 into mature IL-1β/IL-18 and induces massive cell pyroptosis-related cytokine release and systemic inflammation [6] . The potential mechanisms by which NLRP3 causes neuroinflammation include mediation of Th1 and Th17 responses [7] and induction of chemotactic immune cell migration to the central nervous system (CNS) [8] . NLRP3-dependent IL-1β maturation has been detected in the lesions and cerebrospinal fluid (CSF) of MS patients [9] and correlates strongly with cortical lesion load [10] . Conversely, mice lacking IL-1 or with impaired IL-1 signalling show accelerated EAE development and worse EAE severity [11] . There are only a few available IL-1-targeting drugs for the treatment of autoimmune disorders [12, 13] . Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signalling is critical for T helper cell polarization and autoimmune neuroinflammation [14, 15] . In fact, several JAK inhibitors targeting a specific JAK or a wide range of JAKs have been used over the past decades as promising alternatives to traditional disease-modifying antirheumatic drugs for the treatment of several autoimmune diseases [16, 17] . Moreover, JAK mediates IL-4 receptor signals to prime Th2 responsedominant symptoms, such as itch in atopic dermatitis [18] , in which cytokine IL-4 production is closely regulated by the function of thymic stromal lymphopoietin (TSLP) [19] . Despite the high expression of TSLP and its role in JAK-STAT signalling, it is unclear whether TSLP functions via NLRP3 to induce autoimmune inflammation and if so, whether JAK is involved in this process. TSLP is an IL-7-related cytokine that acts on cells of various lineages, including macrophages, dendritic cells (DCs), and T cells [20] . By promoting the expression of major histocompatibility complex (MHC)-II and co-stimulatory molecules such as CD40, CD80, and CD86 and the production of chemokines, TSLP strongly enhances DC maturation and function [21] . In fact, MS and EAE have been reported to be associated with single-nucleotide polymorphisms (SNPs) in the IL-7Rα gene [22] . Binding of TSLP to the TSLP receptor (TSLPR) initiates intracellular JAK/STAT signalling to induce the production of IL-2, TNF, and IL-6 to potentiate inflammatory responses. Targeting these cytokines has been shown to be effective in alleviating EAE and other autoimmune diseases [23] [24] [25] [26] . It was reported that directly blocking JAK/ STAT signalling pathways with tofacitinib inhibits NLRP3 inflammasome and IL-1β production in neutrophils [27] . However, whether TSLPR signalling is capable of controlling the initiation of inflammation in EAE remains unclear. In this study, we show that Tslpr −/− mice presented reduced myelin oligodendrocyte glycoprotein peptide (MOG )induced EAE severity due to decreased phosphorylation of JAK2 and expression of NLRP3 and suppression of the Th17 response. Inhibition of JAK by ruxolitinib mimicked the effects of TSLPR deficiency. In addition, ruxolitinib and the NLRP3 inhibitor MCC950 both reduced inflammatory cell and CD4 + cell infiltration, decreased IL-1β and TSLP levels, and restored myelin expression in the brain tissues of EAE mice. Furthermore, an increased Th17 response was accompanied by a decrease in the number of regulatory T (Treg) cells in blood, while TSLPR deficiency and ruxolitinib reversed this phenomenon after EAE induction. These findings reveal that TSLP plays an essential role in the positive regulation of JAK2-NLRP3 axis-driven neuroinflammation in autoimmune disorders. Tslpr −/− mice and wild-type (WT) mice were bred under specific pathogen-free (SPF) conditions. Tslpr −/− mice were purchased from Shanghai Biomodel Organism Science & Technology Development Co., Ltd. Female Tslpr −/− and WT mice (10−12 weeks old) were immunized subcutaneously with MOG 35-55 (Beyotime, China) and 4 mg/ml heatinactivated Mycobacterium tuberculosis H37Ra (BD) on Day 1 to induce EAE as described previously [28] . Pertussis toxin (200 ng/mouse; List Biological Laboratories Inc.) was injected intraperitoneally on Day 0 and Day 2. The mice were sacrificed on Day 15, and brain tissues were collected for western blotting and immunohistochemistry. EAE-induced paralysis in mice was scored as follows: 0, no disease; 1, tail weakness; 2, paraparesis; 3, paraplegia; 4, paraplegia with forelimb weakness; and 5, morbidity or death. To evaluate the contribution of the NLRP3 inflammasome and JAK to CNS inflammation, 50 mg/kg MCC950 (dissolved in DMSO) was injected intraperitoneally [29] or ruxolitinib (90 mg/kg/day) was administered orally [30] after MOG immunization. To assess inflammatory infiltrates, brain tissues were harvested, fixed in 4% formalin, and stored at room temperature. The brain tissues were subjected to histological analysis, including HE staining, staining for NLRP3 and CD4 staining, and Luxol Fast Blue (LFB) (Servicebio, China) staining, to assess inflammatory cell infiltration, the number of CD4 + T cells, and the myelin sheath. Mouse blood was collected from the retro-orbital plexus in sodium citrate anticoagulant tubes. Peripheral blood mononuclear cells (PBMCs) were isolated with Mouse 1× Lymphocyte Separation Medium (DAKEWE, China) according to the manufacturer's instructions. The cells were collected, and the following monoclonal antibodies were used for flow cytometry analysis (Beckman Coulter): mouse CD4 (RM4-5, Biolegend) and CD25 (3C7, Biolegend). For intracellular staining, the cells were cultured in RPMI 1640 (Gibco) supplemented with 10% FBS and 1% penicillinstreptomycin (PS, Gibco) and then stimulated for 4 h with phorbol 12-myristate 13-acetate (50 ng/ml; Sigma-Aldrich), ionomycin (500 ng/ml; Sigma-Aldrich), and brefeldin A (1 μg/ ml, Merck). The cells were fixed with BD Cytofix/Cytoperm (BD), and the cell membrane was permeabilized with BD Perm/Wash buffer (BD) according to the manufacturer's instructions. For staining of the nuclear factor Foxp3, the cells were stained using a Foxp3 staining buffer set (eBioscience) according to the manufacturer's instructions. BV-2 mouse microglia (ATCC) were cultured in DMEM supplemented with 10% FBS and 1% PS. Confluent cells were pretreated for 30 min with 7.5 nM MCC950 (Selleck, USA) [29] or for 4 h with 10 nM ruxolitinib (MCE, USA) [31] before exposure to 10 ng/ml TSLP (Peprotech, USA) for an additional 24 h [32] . NLRP3 expression (CST, USA), JAK1 phosphorylation (ABSIN, China), JAK2 phosphorylation (CST), JAK3 phosphorylation (CST), basic myelin protein expression (MBP), β-actin expression, GAPDH expression, and β-tubulin expression (Servicebio, China) in mouse brain tissues and BV-2 cell lysates were assessed by western blotting as previously described [33] . ELISA IL-1β (Biolegend), TSLP (Biolegend), and IL-17A (Biolegend) levels in mouse brain tissue homogenates or the BV-2 cell supernatant were evaluated by ELISA according to the manufacturer's instructions. Unpaired two-tailed Student's t-test was used to statistically analyse all data (GraphPad Prism version 5.0; GraphPad Software). P < 0.05 was considered statistically significant. The data are expressed as the mean ± standard error of the mean (SEM). Mice treated with MOG 35-55 developed EAE, presenting with tail weakness, limb numbness, and paralysis, which resembled the symptoms of humans with MS [34] . Because androgens have been reported to be protective in EAE [35] , we administered MOG to female C57BL/6 mice to construct an EAE model (Fig. 1A) . To investigate the role of TSLPR in CNS autoimmunity, the clinical EAE scores of Tslpr −/− and Tslpr +/+ mice were determined. After EAE induction, Tslpr +/+ mice developed typical monophasic EAE symptoms, manifested by ascending paralysis, 10-12 days after MOG 35-55 immunization (Fig. 1B) . In contrast, Tslpr −/− mice showed delayed onset of paralysis at 11-13 days and less severe symptoms than Tslpr +/+ mice. However, the EAE scores of both groups peaked on Day 15 (Fig. 1B) . In addition, immunohistochemical analysis of brain tissues from Tslpr +/+ and Tslpr −/− mice showed that on Day 15 after EAE induction, there were significantly fewer infiltrating CD4 + lymphocytes in the brain tissues of Tslpr −/− mice than in those of Tslpr +/+ mice (Fig. 1C) . Importantly, the western blot results revealed that the expression of myelin basic protein (MBP) was reduced in Tslpr +/+ mice injected with MOG 35-55 compared to those not injected with MOG . However, compared with Tslpr +/+ mice with EAE, Tslpr −/− mice with EAE showed restored expression of MBP (Fig. 1D ). The development of EAE requires NLRP3 [8] . As presented above, TSLPR deficiency results in reduced CD4 + T lymphocyte infiltration during EAE development. Thus, we examined whether TSLPR signalling in EAE requires NLRP3 inflammasome activation. The results showed that the phosphorylation of JAK2 and expression of NLRP3 were increased in MOG 35-55 -treated mice compared to control mice ( Fig. 2A) . No significant difference was observed in the phosphorylation of JAK1 or JAK3 (Fig. 2B ). Immunohistochemical analysis of brain tissues from Tslpr +/+ and Tslpr −/− mice showed that the number of NLRP3 + cells in the brain was markedly increased in Tslpr +/+ mice injected with MOG 35-55 compared to those not injected with MOG , while a significant reduction in the number of NLRP3 + cells in the brain tissue was observed in Tslpr −/− mice compared to Tslpr +/+ mice after EAE induction (Fig. 2C) . Binding of TSLP to TSLPR activates JAK, specifically JAK1 and JAK2 but not JAK3, in primary T cells [33, 36] . To further explore the role of JAK2 in mediating neuroinflammation, we applied a JAK inhibitor, specifically a selective and orally bioavailable JAK1/2 inhibitor widely used for the treatment of myelofibrosis, to block JAK signalling. Oral administration of ruxolitinib to EAE mice resulted in significant reductions in inflammatory cell in filtration, as determined by HE staining (Fig. 3A) ; CD4 + T cell in filtration (Fig. 3B) ; restoration of the myelin sheath, as determined by LFB staining (Fig. 3C) ; and MBP expression, as indicated by western blotting (Fig. 3D) . The expression of NLRP3 in the brain was decreased in EAE mice treated with the JAK inhibitor ruxolitinib compared with those not treated with the JAK inhibitor (Fig. 3D) . ELISA showed that JAK inhibition reduced IL-1β levels in the brain, further confirming NLRP3 hyporeactivity (Fig. 3E) . ELISA also showed that TSLP levels in the brain were reduced after in JAK inhibitor-treated EAE mice compared to EAE mice not treated with the JAK inhibitor (Fig. 3F) . Together, our data demonstrate that JAK2 mediates neuroinflammation via NLRP3. To evaluate the role of NLRP3 in neuroinflammation in EAE, MCC950, a potent and selective small-molecule inhibitor of NLRP3, was used to block canonical and non-canonical NLRP3 activation [29] . Intraperitoneal injection of MCC950 into EAE mice resulted in significant reductions in inflammatory cell infiltration, as determined by HE staining (Fig. 4A ); CD4 + cell infiltration (Fig. 4B) ; restoration of the myelin sheath, as determined by LFB staining (Fig. 4C) ; and MBP expression, as determined by western blotting (Fig. 4D) . The levels of IL-1β (Fig. 4E) and TSLP (Fig. 4F) in the brain were also reduced in NLRP3 inhibitor-treated EAE mice compared to EAE mice not treated with the NLRP3 inhibitor. Microglia are the principal immune cells of the brain and have been identified as risk factors for neurodegenerative disease [37, 38] . To study whether TSLP induces NLRP3 expression and cytokine release, we used BV-2 cells, commonly employed mouse microglia, and found that NLRP3 expression was increased in BV-2 cells stimulated with TSLP compared to in BV-2 cells in the control group (Fig. 5A) . To verify whether the expression of NLRP3 depends on JAK signalling, as seen in the in vivo experiments, we first evaluated JAK1, JAK2, and JAK3 phosphorylation in BV-2 cells. The results showed that TSLP stimulation led to increased JAK2 phosphorylation but not JAK1 or JAK3 phosphorylation (Fig. 5B ). An increase in NLRP3 expression led to substantial release of IL-1β, which was significantly reduced when cells were pretreated with MCC950 to inhibit NLRP3 (Fig. 5C ). Further results revealed that ruxolitinib suppressed TSLP-induced NLRP3 expression (Fig. 5D ). This suppression of NLRP3 expression was accompanied by decreased release of IL-1β (Fig. 5E ). An extensive Th17 immune response contributes to neuroinflammation in EAE [39] , while induction of Treg cells ameliorates EAE [40] . We thus analysed Th17 cells and Treg cells in peripheral blood by flow cytometry. The number of Th17 (CD4 + IL-17A + ) cells was increased in mice immunized with MOG 35-55 compared to control mice (Fig. 6A ). In contrast, the number of Treg (CD4 + CD25 + Foxp3 + ) cells in the blood was deceased after EAE induction (Fig. 6B) . However, a significant decrease in the number of Th17 cells was observed in Tslpr −/− mice compared to WT mice after EAE induction. Notably, this decrease in the number of Th17 cells was accompanied by an increase in the number of Treg cells in the blood (Fig. 6A and B) . The expression of the cytokine IL-17A in the mouse brain was evaluated. As anticipated, IL-17A expression was increased in mice with EAE compared to control WT mice, while TSLPR deficiency led to a decrease in the expression of IL-17A by nearly half (Fig. 6C) . Furthermore, we observed that inhibition of JAK2 by ruxolitinib significantly decreased IL-17A levels in the brains of WT mice after EAE induction (Fig. 6D ). Activation of NLRP3 is a tightly regulated process and a key step in autoimmunity in the CNS. In this study, we report three novel findings that broaden our understanding of NLRP3-associated neuroinflammation. First, by using Tslpr −/− mice, we demonstrated that TSLP signalling regulates neuroinflammation and paralysis in mice. Second, we showed that TSLPR signals via JAK2 to activate NLRP3associated neuroinflammation in the context of EAE. Third, we found that manipulation of the Th17/Treg balance is involved in TSLP-induced neuroinflammation. Taken together, the findings of the current study identify novel functions of TSLP-dominant JAK pathways upstream of NLRP3, which represent promising targets for the treatment of autoimmune disorders. . The data are presented as the mean ± SEM and were obtained from a high-power field (HPF) ( * P < 0.05, * * P < 0.01, * * * P < 0.001). Scale bar, 50 μm. Generally, the Th17 response is believed to cause neuronal death or an inflammatory response in autoimmune disorders [39, 41] . In addition, recent studies have shown that type 3 innate lymphoid cells (ILC3s) produce IL-17 in autoimmune disorders, such as ankylosing spondylitis [42] , and can maintain neuroinflammation by supporting T cell survival [43] . In this study, we found that BV-2 also produced IL-17A after TSLP stimulation (data not shown). Thus, both neuroinflammation and demyelination are largely believed to be mediated by the adaptive immune system via Th17 cells and by the innate immune system via ILC3-and even microglia-related responses. Based on the data of the current and previous studies showing a critical role for TSLP in DCs [21, 44] and ILC3 function [45] , we speculate that microglia and ILC3s, in addition to Th17 cells, play important roles in promoting autoimmune inflammation-associated demyelination. The precise role of TSLP in IL-17A + cell-mediated neuroinflammation and demyelination needs to be further investigated. Despite previous findings that Treg cell formation requires TSLP [46, 47] , we found that compared to EAE induction, which led to a decrease in the number of Treg cells, TSLPR deficiency led to an increase in the number of Treg cells in the blood, which is consistent with the reduction in Treg function observed in MS in a previous study [48] . Our data suggest that TSLP-producing cells are a more general means by which immune responses are facilitated, as they suppress the Treg cell response. In addition, these changes in the number of Treg cells in the blood after deletion of Tslpr correlate with alleviation of neuroinflammation and restoration of myelin expression, which is in agreement with a previous study showing that EAE symptoms are ameliorated in Tslp −/− mice [49] . In fact, the epithelial cell-derived cytokines TSLP, GM-CSF, and IL-25 have been shown to be master initiators of type 3 inflammation via their effects on a variety of cells, including Th17 cells, ILC3, and mast cells [50] [51] [52] . These cytokines are believed to rapidly bind to membrane receptors to generate innate immune responses and therefore prime adaptive immune cells. Strikingly, two recent studies have demonstrated that TSLP directly activates neurons [32, 53] . Our current data showing that TSLP is highly regulated in the brain in the context of EAE are consistent with the findings of a previous study [54] and further demonstrate that TSLPR signalling activates NLRP3-mediated inflammation through phosphorylation of JAK2 in response to MOG administration. Thus, we speculate that the cytokine TSLP may act as a master regulator of neuroinflammation in immune cells in the brain. . The data are presented as the mean ± SEM and were obtained from a HPF ( * P < 0.05, * * P < 0.01, * * * P < 0.001). Scale bar, 50 μm. In immune cells, cytokine signalling by the JAK-STAT pathway causes transcriptional changes to promote cellular activation. However, although JAK inhibitors have been reported as alternative immunotherapies in patients with autoimmune disorders such as neuromyelitis optica [55] , our data indicated that application of the JAK1/2 inhibitor ruxolitinib failed to induce typical neuroinflammation following MOG injection, as observed in the EAE mouse brain. Also, this decrease in neuroinflammation after treatment with ruxolitinib was accompanied by restored expression of MBP. Thus, we predict that alterations in classic JAK-mediated NLRP3 inflammasome activation, especially decreased IL-17A levels, as a reflection of the Th17 immune response, are sufficient to explain neuroinflammation and demyelination. One previous study showed that JAK1 mediates sensory neuronal responsiveness, which can be enhanced by cytokines such as IL-4 [18] . Our data are consistent with this study and further demonstrate that JAKs have novel functions in neurons and regulate the myelination/demyelination balance, at least through NLRP3-mediated pathways. However, we note that this alteration in MBP expression does not exclude the role of JAK2 or other pathways in modulating the transcription or posttranscriptional modification of MBP within the CNS. Future studies are required to better understand how changes in the JAK-STAT pathway impact myelin expression and neuroinflammation in autoimmune disorders. Clinical application of ruxolitinib, a non-selective JAK1/2 inhibitor, has been reported to alleviate neurologic disability in neuromyelitis optica [55] . In fact, other JAK inhibitors, such as tofacitinib and baricitinib, have shown significant clinical efficacy in autoimmune disorders in clinical trials [56, 57] . Previously, changes in neuroinflammation following JAK inhibition were attributed to the anti-inflammatory role of the Th17 response [41] . Recently, a study demonstrated that transient receptor potential (TRP) plays a critical role in EAE by mediating axonal and neuronal degeneration [58] and that the JAK-STAT pathway determines TRP expression [59, 60] , which indicates the involvement of TRP in JAK pathway-mediated neuroinflammation. Based on studies by our group and others, we speculate that the amelioration of neuroinflammation in EAE mice treated with ruxolitinib may be mediated, at least in part, by disruption of these signals in the CNS and that such treatments may alleviate neuroinflammation in autoimmune disorders. Strikingly, recent studies have provided experimental and clinical evidence for the efficacy of evobrutinib, the first Bruton's tyrosine kinase (BTK)-inhibiting molecule to be developed, and reported that impairment of encephalitogenic T cells reduces disease severity in clinical and mouse models of MS [61, 62] . Given that our current study demonstrates a direct role for JAK in neuroinflammation, whether blockade of a combination of TSLP, JAK, and BTK can synergistically alleviate neuroinflammation in autoimmune disorders such as MS needs to be further investigated. In conclusion, our data establish and highlight the capability of TSLPR-JAK signalling inhibition to control disease-driving neuroinflammation associated with NLRP3 in inflammatory demyelination in the CNS. This was specifically demonstrated by using ruxolitinib, a non-selective JAK inhibitor that has been tested in clinical trials of several autoimmune diseases, but the immunological effects of this inhibitor may be similar to those of other JAK inhibitors in clinical development. Thus, the mechanistic data provided here will be instrumental in facilitating how this molecule is integrated into current treatments for autoimmune disorders. . The data are presented as the mean ± SEM ( * P < 0.05, * * P < 0.01, * * * P < 0.001). Overlapping demyelinating syndromes and anti-N-methyl-D-aspartate receptor encephalitis Soelberg Sørensen P. Evolving concepts in the treatment of relapsing multiple sclerosis NLRP3 activation and mitosis are mutually exclusive events coordinated by NEK7, a new inflammasome component HIV-1 Tat primes and activates microglial NLRP3 inflammasome-mediated neuroinflammation Inflammasome complexes: emerging mechanisms and effector functions Caspase-1-induced pyroptotic cell death NLRP3 plays a critical role in the development of experimental autoimmune encephalomyelitis by mediating Th1 and Th17 responses NLRP3 inflammasome induces chemotactic immune cell migration to the CNS in experimental autoimmune encephalomyelitis The NALP3 inflammasome is involved in the innate immune response to amyloid-beta Cerebrospinal fluid IL-1β correlates with cortical pathology load in multiple sclerosis at clinical onset Interleukin-1 promotes autoimmune neuroinflammation by suppressing endothelial heme oxygenase-1 at the blood-brain barrier Natalizumab treatment in multiple sclerosis: marked decline of chemokines and cytokines in cerebrospinal fluid IFNβ and glatiramer acetate trigger different signaling pathways to regulate the IL-1 system in multiple sclerosis Defective sphingosine 1-phosphate receptor 1 (S1P1) phosphorylation exacerbates TH17-mediated autoimmune neuroinflammation OTUB1 inhibits CNS autoimmunity by preventing IFN-γ-induced hyperactivation of astrocytes The emerging role of Janus kinase inhibitors in the treatment of autoimmune and inflammatory diseases Novel therapies for immune-mediated inflammatory diseases: what can we learn from their use in rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, psoriasis, Crohn's disease and ulcerative colitis Sensory neurons co-opt classical immune signaling pathways to mediate chronic itch Basophil-derived IL-4 promotes epicutaneous antigen sensitization concomitant with the development of food allergy TSLP: from allergy to cancer Thymic stromal lymphopoietin epigenetically upregulates Fc receptor γ subunit-related receptors on antigen-presenting cells and induces TH2/TH17 polarization through dectin-2 Interleukin 7 receptor alpha chain (IL7R) shows allelic and functional association with multiple sclerosis IL-6-deficient mice are resistant to experimental autoimmune encephalomyelitis: roles of IL-6 in the activation and differentiation of autoreactive T cells Effect of anti-interferon-gamma and anti-interleukin-2 monoclonal antibody treatment on the development of actively and passively induced experimental allergic encephalomyelitis in the SJL/J mouse Control of established experimental allergic encephalomyelitis by inhibition of tumor necrosis factor (TNF) activity within the central nervous system using monoclonal antibodies and TNF receptor-immunoglobulin fusion proteins Critical proinflammatory role of thymic stromal lymphopoietin and its receptor in experimental autoimmune arthritis Tofacitinib inhibits granulocyte-macrophage colony-stimulating factor-induced NLRP3 inflammasome activation in human neutrophils Myeloid cell transmigration across the CNS vasculature triggers IL-1β-driven neuroinflammation during autoimmune encephalomyelitis in mice A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases The CSF3R T618I mutation causes a lethal neutrophilic neoplasia in mice that is responsive to therapeutic JAK inhibition Ruxolitinib reverses dysregulated T helper cell responses and controls autoimmunity caused by a novel signal transducer and activator of transcription 1 (STAT1) gain-of-function mutation Periostin activation of integrin receptors on sensory neurons induces allergic itch Heme oxygenase-1 directly binds STAT3 to control the generation of pathogenic Th17 cells during neutrophilic airway inflammation Th17 lymphocytes traffic to the central nervous system independently of α4 integrin expression during EAE Androgens are protective in experimental autoimmune encephalomyelitis: implications for multiple sclerosis IL-1β enables CNS access to CCR2hi monocytes and the generation of pathogenic cells through GM-CSF released by CNS endothelial cells New insights into microglia-neuron interactions: a neuron's perspective. Neuroscience Microglial genes regulating neuroinflammation in the progression of Alzheimer's disease PKM2 promotes Th17 cell differentiation and autoimmune inflammation by fine-tuning STAT3 activation In vivo induction of regulatory T cells via CTLA-4 signaling peptide to control autoimmune encephalomyelitis and prevent disease relapse The TGF-β superfamily cytokine Activin-A is induced during autoimmune neuroinflammation and drives pathogenic Th17 cell differentiation Type 3 innate lymphoid cells producing IL-17 and IL-22 are expanded in the gut, in the peripheral blood, synovial fluid and bone marrow of patients with ankylosing spondylitis Group 3 innate lymphoid cells accumulate and exhibit disease-induced activation in the meninges in EAE Airway epithelial TSLP production of TLR2 drives type 2 immunity in allergic airway inflammation Microbiota sensing by mincle-syk axis in dendritic cells regulates interleukin-17 and -22 production and promotes intestinal barrier integrity. Immunity Development of regulatory T cells requires IL-7Ralpha stimulation by IL-7 or TSLP Rhinovirus infection interferes with induction of tolerance to aeroantigens through OX40 ligand, thymic stromal lymphopoietin, and IL-33 The interleukin-7 receptor α chain contributes to altered homeostasis of regulatory T cells in multiple sclerosis Thymic stromal lymphopoietin deficiency attenuates experimental autoimmune encephalomyelitis Hepatitis C virus promotes T-helper (Th)17 responses through thymic stromal lymphopoietin production by infected hepatocytes Autoimmune Th17 cells induced synovial stromal and innate lymphoid cell secretion of the cytokine GM-CSF to initiate and augment autoimmune arthritis IL-25 enhances TH17 cell-mediated contact dermatitis by promoting IL-1β production by dermal dendritic cells The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch Thymic stromal lymphopoietin is expressed in the intact central nervous system and upregulated in the myelin-degenerative central nervous system Ruxolitinib treatment in a patient with neuromyelitis optica: a case report Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial Tofacitinib in patients with ankylosing spondylitis: a phase II, 16-week, randomised, placebo-controlled, dose-ranging study TRPM4 cation channel mediates axonal and neuronal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis Interleukin-6-mediated functional upregulation of TRPV1 receptors in dorsal root ganglion neurons through the activation of JAK/PI3K signaling pathway: roles in the development of bone cancer pain in a rat model Deletion of interleukin-6 signal transducer gp130 in small sensory neurons attenuates mechanonociception and down-regulates TRPA1 expression Inhibition of Bruton's tyrosine kinase interferes with pathogenic B-cell development in inflammatory CNS demyelinating disease Evobrutinib Phase 2 Study Group. Placebo-controlled trial of an oral BTK inhibitor in multiple sclerosis Grant in the Science and Technology Development Foundation of Nanjing Medical University and Grant in the National Natural Science Foundation of China, grant/award number: NMUB2019103 and 81900015, respectively. All authors declare that they have no competing interests. X.Y., J.L., J.W., and Y.C. performed the experiments, data acquisition, analysis and interpretation and drafting the article. F.C. analysed the data and critically revised the article. L.W. designed the project and critically revised the article for the important intellectual content and final approval of the version to be published. All authors provided important review of the manuscript. This study was approved by the institutional research ethics committee of Nanjing Medical University affiliated Nanjing Brain Hospital. The data sets used and/or analysed in the present study are available from the corresponding author on reasonable request.