Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nat Immunol ; 17(5): 583-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26998763

ABSTRACT

Interleukin 1ß (IL-1ß) is critical for the in vivo survival, expansion and effector function of IL-17-producing helper T (T(H)17) cells during autoimmune responses, including experimental autoimmune encephalomyelitis (EAE). However, the spatiotemporal role and cellular source of IL-1ß during EAE pathogenesis are poorly defined. In the present study, we uncovered a T cell-intrinsic inflammasome that drives IL-1ß production during T(H)17-mediated EAE pathogenesis. Activation of T cell antigen receptors induced expression of pro-IL-1ß, whereas ATP stimulation triggered T cell production of IL-1ß via ASC-NLRP3-dependent caspase-8 activation. IL-1R was detected on T(H)17 cells but not on type 1 helper T (T(H)1) cells, and ATP-treated T(H)17 cells showed enhanced survival compared with ATP-treated T(H)1 cells, suggesting autocrine action of T(H)17-derived IL-1ß. Together these data reveal a critical role for IL-1ß produced by a T(H)17 cell-intrinsic ASC-NLRP3-caspase-8 inflammasome during inflammation of the central nervous system.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes/immunology , Th17 Cells/immunology , Adenosine Triphosphate/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Caspase 8/genetics , Caspase 8/immunology , Caspase 8/metabolism , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Flow Cytometry , Gene Expression/immunology , Immunoblotting , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism
2.
Nat Immunol ; 14(1): 72-81, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23202271

ABSTRACT

Act1 is an essential adaptor in interleukin 17 (IL-17)-mediated signaling and is recruited to the receptor for IL-17 after stimulation with IL-17. Here we found that Act1 was a 'client' protein of the molecular chaperone hsp90. The D10N variant of Act1 (Act1(D10N)) that is linked to susceptibility to psoriasis was defective in its interaction with hsp90, which resulted in a global loss of Act1 function. Act1-deficient mice modeled the mechanistic link between loss of Act1 function and susceptibility to psoriasis. Although Act1 was necessary for IL-17-mediated inflammation, Act1-deficient mice had a hyperactive response of the T(H)17 subset of helper T cells and developed spontaneous IL-22-dependent skin inflammation. In the absence of IL-17 signaling, IL-22 was the main contributor to skin inflammation, which provides a molecular mechanism for the association of Act1(D10N) with psoriasis susceptibility.


Subject(s)
Connexin 43/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Peptide Fragments/metabolism , Psoriasis/immunology , Th17 Cells/immunology , Animals , Cell Line , Connexin 43/genetics , Connexin 43/immunology , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Interleukin-17/metabolism , Mice , Mice, Knockout , Molecular Chaperones/genetics , Mutation/genetics , Peptide Fragments/genetics , Peptide Fragments/immunology , Polymorphism, Genetic , Protein Binding/genetics , Protein Binding/immunology , Psoriasis/genetics , Signal Transduction
3.
J Neuroinflammation ; 18(1): 98, 2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33879167

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is a debilitating neurological disease caused by autoimmune destruction of the myelin sheath. Experimental autoimmune encephalomyelitis (EAE) is a widely used animal model for the pathogenesis of MS. We and others have previously demonstrated that IL-17 is critical for the pathogenesis of EAE. The concentration of IL-17 is significantly higher in the sera of MS patients than in healthy controls and correlates with disease activity. Moreover, anti-IL-17 neutralizing antibody demonstrated promising efficacy in a phase II trial in MS patients, further substantiating a key pathogenic role for IL-17 in MS. While Th17 and IL-17 are emerging as a bona fide drivers for neuroinflammation, it remains unclear what effector molecule executes the inflammatory tissue destruction in Th17-driven EAE. METHODS: By microarray analysis, we found STEAP4 is a downstream molecule of IL-17 signaling in EAE. We then used STEAP4 global knockout mice and STEAP4 conditional knockout mice to test its role in the pathogenesis of EAE. RESULTS: Here, we report that the metalloreductase, STEAP4, is a key effector molecule that participates and contributes to the pathogenesis of Th17-mediated neuroinflammation in experimental autoimmune encephalomyelitis. STEAP4 knockout mice displayed delayed onset and reduced severity of EAE induced by active immunization. The reduced disease phenotype was not due to any impact of STEAP4 deficiency on myelin reactive T cells. In contrast, STEAP4 knockout mice were resistant to passively induced EAE, pointing to a role for STEAP4 in the effector stage of EAE. Notably, STEAP4 was only induced the spinal cord of EAE mice that received Th17 cells but not Th1 cells. Consistently, STEAP4 deficiency protected from only Th17 but not Th1-induced EAE. Finally, using Nestin-Cre STEAP4fl/fl mice, we showed that ablation of STEAP4 expression in the resident cells in the central nervous system attenuated disease severity in both active immunization and passive Th17 transfer-induced EAE. CONCLUSION: In this study, we identified STEAP4 as a Th17-specific effector molecule that participates and contributes to the pathogenesis of neuroinflammation, thus potentially provide a novel target for MS therapy.


Subject(s)
Central Nervous System/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Th17 Cells/immunology , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/physiopathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/physiopathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/metabolism
4.
Immunity ; 36(5): 821-33, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22608496

ABSTRACT

Interleukin-25 (IL-25 or IL-17E), a member of the structurally related IL-17 family, functions as an important mediator of T helper 2 cell-type (type 2) responses. We examined the cell type-specific role of IL-25-induced Act1-mediated signaling in protective immunity against helminth infection. Targeted Act1 deficiency in epithelial cells resulted in a marked delay in worm expulsion and abolished the expansion of the Lin(-)c-Kit(+) innate cell population in the mesenteric lymph node, lung, and liver. Th2 cell-inducing cytokine (IL-25 and IL-33) expression were reduced in the intestinal epithelial cells from the infected and IL-25-injected epithelial-specific Act1-deficient mice. Adoptive transfer of Lin(-)c-Kit(+) cells or combined injection of IL-25 and IL-33 restored the type 2 responses in these mice. Taken together, these results suggest that epithelial-specific Act1 mediates the expansion of the Lin(-)c-Kit(+) innate cell population through the positive-feedback loop of IL-25, initiating the type 2 immunity against helminth infection.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Epithelial Cells/immunology , Helminthiasis/immunology , Helminths/immunology , Interleukins/immunology , Th2 Cells/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Lineage , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Helminthiasis/metabolism , Helminths/metabolism , Immunity, Innate/immunology , Interleukins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Liver/cytology , Liver/immunology , Liver/metabolism , Lung/cytology , Lung/immunology , Lung/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Th2 Cells/metabolism
5.
Immunity ; 34(1): 3-5, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21272781

ABSTRACT

In this issue of Immunity, Shaw et al. (2011) report that the NOD-RICK signaling axis is required for the activation of dendritic cells infiltrating the central nervous system, leading to reactivation of antigen-specific T cells and autoimmune inflammation.

6.
J Immunol ; 200(5): 1889-1900, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29352002

ABSTRACT

Dysregulation of the immune barrier function of the intestinal epithelium can often result in dysbiosis. In this study we report a novel role of intestinal epithelial cell (IEC)-derived liver kinase B1 (LKB1) in suppressing colitogenic microbiota. IEC-specific deletion of LKB1 (LKB1ΔIEC) resulted in an increased susceptibility to dextran sodium sulfate (DSS)-induced colitis and a definitive shift in the composition of the microbial population in the mouse intestine. Importantly, transfer of the microbiota from LKB1ΔIEC mice was sufficient to confer increased susceptibility to DSS-induced colitis in wild-type recipient mice. Collectively, the data indicate that LKB1 deficiency in intestinal epithelial cells nurtures the outgrowth of colitogenic bacteria in the commensal community. In addition, LKB1 deficiency in the intestinal epithelium reduced the production of IL-18 and antimicrobial peptides in the colon. Administration of exogenous IL-18 restored the expression of antimicrobial peptides, corrected the outgrowth of several bacterial genera, and rescued the LKB1ΔIEC mice from increased sensitivity to DSS challenge. Taken together, our study reveals an important function of LKB1 in IECs for suppressing colitogenic microbiota by IL-18 expression.


Subject(s)
Epithelial Cells/immunology , Intestinal Mucosa/immunology , Intestines/immunology , Microbiota/immunology , Protein Serine-Threonine Kinases/immunology , AMP-Activated Protein Kinases , Animals , Colitis/chemically induced , Colitis/immunology , Colon/drug effects , Colon/immunology , Dextran Sulfate/pharmacology , Dysbiosis/immunology , Interleukin-18/immunology , Intestines/drug effects , Mice , Mice, Inbred C57BL
7.
J Transl Med ; 17(1): 37, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670047

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease characterized by the presence of pathogenic autoantibodies associated with polyclonal B cell hyperreactivity. Previous study reported that autophagy-related gene Leucine-rich repeat kinase 2 (LRRK2) was likely a susceptible gene for SLE. However, the pathogenic function of LRRK2 in SLE is undefined. METHODS: Using quantitative PCR, we compared the expression levels of LRRK2 in B cells between SLE patients and healthy controls. The expression levels of LRRK2 in in vitro induced CD19hi B cells and naïve B cells were compared as well based on RNA-seq assay. A pristane-induced lupus-like mouse model was used to explore the effects of LRRK2 on the development of SLE. IgG level, B cell subsets in the spleens and bone marrows and pathological features in the kidneys were compared between wildtype (WT) and Lrrk2-/- littermates. RESULTS: It was obvious that LRRK2 expression was dramatically up-regulated in primary B cells from SLE patients compared to those from healthy controls, as well as in activated CD19hi B cells. More significantly, LRRK2 expression in B cells was positively correlated with system lupus erythematosus disease activity index (SLEDAI), an indicator for disease severity, and serum IgG levels in SLE patients. Negative correlations were observed between LRRK2 expression and serum C3 or C4 levels, two clinical features associated with SLE-related nephritis. LRRK2 deficiency reduced the death rate of pristane treated mice. Decreased levels of total IgG and autoantibody were detected in the serum with less deposition of immune complexes and attenuated pathological symptoms in the kidneys of Lrrk2-/- mice. Consistent with the reduction in IgG production, the percentages of germinal center B cells and plasma cells decreased significantly as well with LRRK2 deficiency. CONCLUSIONS: Our study demonstrates that LRRK2 expression is upregulated in B cells from SLE patients with strong correlations to disease severity. LRRK2 deficiency largely attenuates the pathogenic progress of lupus-like features in pristane-induced mice. This is probably achieved through affecting B cell terminal differentiation and subsequent immunoglobulin production.


Subject(s)
Antibody Formation/immunology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/immunology , Animals , B-Lymphocytes/immunology , Case-Control Studies , Female , Germinal Center/immunology , Kidney/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/deficiency , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Plasma Cells/immunology , Severity of Illness Index , Terpenes , Up-Regulation
8.
Immunity ; 32(3): 414-25, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20303295

ABSTRACT

Interleukin-17 (IL-17) secreted by T helper 17 (Th17) cells is essential in the development of experimental autoimmune encephalomyelitis (EAE). However, it remains unclear how IL-17-mediated signaling in different cellular compartments participates in the central nervous system (CNS) inflammatory process. We examined CNS inflammation in mice with specific deletion of Act1, a critical component required for IL-17 signaling, in endothelial cells, macrophages and microglia, and neuroectoderm (neurons, astrocytes, and oligodendrocytes). In Act1-deficient mice, Th17 cells showed normal infiltration into the CNS but failed to recruit lymphocytes, neutrophils, and macrophages. Act1 deficiency in endothelial cells or in macrophages and microglia did not substantially impact the development of EAE. However, targeted Act1 deficiency in neuroectoderm-derived CNS-resident cells resulted in markedly reduced severity in EAE. Specifically, Act1-deficient astrocytes showed impaired IL-17-mediated inflammatory gene induction. Thus, astroctyes are critical in IL-17-Act1-mediated leukocyte recruitment during autoimmune-induced inflammation of the CNS.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Astrocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/immunology , Signal Transduction , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Astrocytes/metabolism , Cell Survival , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology
9.
Immunity ; 32(1): 54-66, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20060329

ABSTRACT

Interleukin-1 (IL-1)-mediated signaling in T cells is essential for T helper 17 (Th17) cell differentiation. We showed here that SIGIRR, a negative regulator of IL-1 receptor and Toll-like receptor signaling, was induced during Th17 cell lineage commitment and governed Th17 cell differentiation and expansion through its inhibitory effects on IL-1 signaling. The absence of SIGIRR in T cells resulted in increased Th17 cell polarization in vivo upon myelin oligodendrocyte glycoprotein (MOG(35-55)) peptide immunization. Recombinant IL-1 promoted a marked increase in the proliferation of SIGIRR-deficient T cells under an in vitro Th17 cell-polarization condition. Importantly, we detected increased IL-1-induced phosphorylation of JNK and mTOR kinase in SIGIRR-deficient Th17 cells compared to wild-type Th17 cells. IL-1-induced proliferation was abolished in mTOR-deficient Th17 cells, indicating the essential role of mTOR activation. Our results demonstrate an important mechanism by which SIGIRR controls Th17 cell expansion and effector function through the IL-1-induced mTOR signaling pathway.


Subject(s)
Cell Differentiation/immunology , Interleukin-17/immunology , Intracellular Signaling Peptides and Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Receptors, Interleukin-1/immunology , T-Lymphocytes, Helper-Inducer/cytology , Animals , Cell Lineage/immunology , Cell Proliferation , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Immunoprecipitation , Interleukin-17/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases , Transfection
10.
J Immunol ; 189(1): 33-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22649194

ABSTRACT

The effector T cell subset, Th17, plays a significant role in the pathogenesis of multiple sclerosis and of other autoimmune diseases. The signature cytokine, IL-17, engages the IL-17R and recruits the E3-ligase NF-κB activator 1 (Act1) upon stimulation. In this study, we examined the role of TNFR-associated factor (TRAF)4 in IL-17 signaling and Th17-mediated autoimmune encephalomyelitis. Primary cells from TRAF4-deficient mice displayed markedly enhanced IL-17-activated signaling pathways and induction of chemokine mRNA. Adoptive transfer of MOG35-55 specific wild-type Th17 cells into TRAF4-deficient recipient mice induced an earlier onset of disease. Mechanistically, we found that TRAF4 and TRAF6 used the same TRAF binding sites on Act1, allowing the competition of TRAF4 with TRAF6 for the interaction with Act1. Taken together, the results of this study reveal the necessity of a unique role of TRAF4 in restricting the effects of IL-17 signaling and Th17-mediated disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-17/antagonists & inhibitors , Interleukin-17/physiology , Signal Transduction/immunology , TNF Receptor-Associated Factor 4/physiology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , HEK293 Cells , HeLa Cells , Humans , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , Signal Transduction/genetics , TNF Receptor-Associated Factor 4/deficiency , TNF Receptor-Associated Factor 4/genetics , Th17 Cells/immunology , Th17 Cells/pathology
11.
Acta Crystallogr Sect E Struct Rep Online ; 70(Pt 11): o1199, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25484827

ABSTRACT

In the title compound, C11H11N3O3S·3H2O, the non-H atoms of the main mol-ecule are approximately planar, with an r.m.s. deviation of 0.030 Å. There is a bifurcated intra-molecular N-H⋯(O,S) hydrogen bond present forming S(6) and S(5) ring motifs. In the crystal, O-H⋯O and N-H⋯O hydrogen bonds link the molecules into a three-dimensional network.

12.
Cells ; 13(7)2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38607004

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) is a serine-threonine protein kinase belonging to the ROCO protein family. Within the kinase domain of LRRK2, a point mutation known as LRRK2 G2019S has emerged as the most prevalent variant associated with Parkinson's disease. Recent clinical studies have indicated that G2019S carriers have an elevated risk of cancers, including colon cancer. Despite this observation, the underlying mechanisms linking LRRK2 G2019S to colon cancer remain elusive. In this study, employing a colitis-associated cancer (CAC) model and LRRK2 G2019S knock-in (KI) mouse model, we demonstrate that LRRK2 G2019S promotes the pathogenesis of colon cancer, characterized by increased tumor number and size in KI mice. Furthermore, LRRK2 G2019S enhances intestinal epithelial cell proliferation and inflammation within the tumor microenvironment. Mechanistically, KI mice exhibit heightened susceptibility to DSS-induced colitis, with inhibition of LRRK2 kinase activity ameliorating colitis severity and CAC progression. Our investigation also reveals that LRRK2 G2019S promotes inflammasome activation and exacerbates gut epithelium necrosis in the colitis model. Notably, GSDMD inhibitors attenuate colitis in LRRK2 G2019S KI mice. Taken together, our findings offer experimental evidence indicating that the gain-of-kinase activity in LRRK2 promotes colorectal tumorigenesis, suggesting LRRK2 as a potential therapeutic target in colon cancer patients exhibiting hyper LRRK2 kinase activity.


Subject(s)
Colitis , Colonic Neoplasms , Gasdermins , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Animals , Mice , Colitis/chemically induced , Colitis/complications , Colitis/genetics , Colonic Neoplasms/genetics , Inflammation/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Tumor Microenvironment
13.
J Neurosci ; 32(24): 8284-92, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22699909

ABSTRACT

Cuprizone inhibits mitochondrial function and induces demyelination in the corpus callosum, which resembles pattern III lesions in multiple sclerosis patients. However, the molecular and cellular mechanism by which cuprizone induces demyelination remains unclear. Interleukin-17 (IL-17) secreted by T helper 17 cells and γδT cells are essential in the development of experimental autoimmune encephalomyelitis. In this study, we examined the importance of IL-17 signaling in cuprizone-induced demyelination. We found that mice deficient in IL-17A, IL-17 receptor C (IL-17RC), and adaptor protein Act1 (of IL-17R) all had reduced demyelination accompanied by lessened microglial and polydendrocyte cellular reactivity compared with that in wild-type mice in response to cuprizone feeding, demonstrating the essential role of IL-17-induced Act1-mediated signaling in cuprizone-induced demyelination. Importantly, specific deletion of Act1 in astrocytes reduced the severity of tissue injury in this model, indicating the critical role of CNS resident cells in the pathogenesis of cuprizone-induced demyelination. In cuprizone-fed mice, IL-17 was produced by CNS CD3(+) T cells, suggesting a source of IL-17 in CNS upon cuprizone treatment.


Subject(s)
Connexin 43/physiology , Cuprizone/toxicity , Demyelinating Diseases/physiopathology , Interleukin-17/physiology , Multiple Sclerosis/chemically induced , Multiple Sclerosis/physiopathology , Peptide Fragments/physiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , CD3 Complex/immunology , Chelating Agents , Connexin 43/genetics , Connexin 43/metabolism , Demyelinating Diseases/chemically induced , Disease Models, Animal , Female , Gene Knockdown Techniques , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
J Immunol ; 187(6): 3155-64, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21856933

ABSTRACT

The cellular and molecular mechanisms driven by IL-25 and its cognate receptor IL-17RB necessary for the promotion of Th2-mediating pathogenic pulmonary inflammation remains to be defined. We have previously reported the critical role of the U-box-type E3 ubiquitin ligase Act1 (1) for the downstream signaling of the IL-17 cytokine family including the Th2-promoting cytokine IL-25 (IL-17E) (2). In this study, we report that IL-25-driven but not conventional IL-4-driven Th2 polarization and cytokine production is impaired in Act1-deficient T cells. Also, Act1 deficiency in the T cell compartment results in the abrogation of eosinophilic airway infiltration as well as airway hyperresponsiveness in mouse models of Ag-induced airway inflammation. The in vivo generation of Ag-specific Th2 cytokine-producing cells is defective in the absence of Act1 expression in T cells after OVA/aluminum hydroxide immunization. Notably, the production of OVA-specific IgG(1) but not IgG(2a) or IgE is also impaired. At the molecular level, we report that IL-25-mediated induction of Th2 master regulator GATA-3 and the transcription factor GFI-1 is attenuated in Act1-deficient T cells. Taken together, our findings indicate that Act1 expression in T cells is required for cellular and humoral Th2-mediated allergic responses and the development of airway hyperresponsiveness, in part, through Act1's function in IL-25-induced development of Th2 T cells.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Hypersensitivity/immunology , Interleukins/immunology , Pneumonia/immunology , Th2 Cells/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hypersensitivity/metabolism , Immunohistochemistry , Interleukins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Pneumonia/metabolism , Signal Transduction/immunology , Th2 Cells/metabolism
15.
J Immunol ; 186(5): 3006-14, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270393

ABSTRACT

Bone marrow-derived plasmacytoid dendritic cells (pDCs) from IL-1R-associated kinase (IRAK)2-deficient mice produced more IFNs than did wild-type pDCs upon stimulation with the TLR9 ligand CpG. Furthermore, in CpG-stimulated IRAK2-deficient pDCs there was increased nuclear translocation of IFN regulatory factor 7, the key transcription factor for IFN gene transcription in these cells. In IRAK2-deficient macrophages, enhanced NF-κB activation and increased expression of CpG-induced genes were detected within 2 h after treatment. However, at later times, NF-κB activation was decreased and, in contrast to the results with IFN, there was less secretion of other proinflammatory cytokines (such as TNF-α) and chemokines in CpG-stimulated IRAK2-deficient pDCs and macrophages. Therefore, although IRAK2 is a negative regulator of TLR9-mediated IFN production through its modulation of the transcriptional activity of IFN regulatory factor 7, it is also a positive regulator of TLR9-mediated proinflammatory cytokine and chemokine production at some level subsequent to transcription.


Subject(s)
Cytokines/biosynthesis , Inflammation Mediators/metabolism , Interferon Type I/biosynthesis , Interferon-beta/biosynthesis , Interleukin-1 Receptor-Associated Kinases/physiology , Toll-Like Receptor 9/physiology , Animals , Cells, Cultured , CpG Islands/immunology , Cytokines/antagonists & inhibitors , Cytokines/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Down-Regulation/immunology , Herpesviridae Infections/enzymology , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Interferon Regulatory Factor-7/antagonists & inhibitors , Interferon Regulatory Factor-7/physiology , Interferon Type I/antagonists & inhibitors , Interferon-alpha , Interferon-beta/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/deficiency , Ligands , Mice , Mice, Knockout , Recombinant Proteins , Rhadinovirus/immunology , Toll-Like Receptor 9/metabolism , Tumor Virus Infections/enzymology , Tumor Virus Infections/immunology , Tumor Virus Infections/pathology
16.
bioRxiv ; 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37425755

ABSTRACT

LRRK2 G2019S is the most prevalent variant associated with Parkinson's disease (PD), found in 1-3% of sporadic and 4-8% of familial PD cases. Intriguingly, emerging clinical studies have suggested that LRRK2 G2019S carriers have an increased risk of cancers including colorectal cancer. However, the underlying mechanisms of the positive correlation between LRRK2-G2019S and colorectal cancer remain unknown. Using a mouse model of colitis-associated cancer (CAC) and LRRK2 G2019S knockin (KI) mice, here we report that LRRK2 G2019S promotes the pathogenesis of colon cancer as evidenced by increased tumor number and tumor size in LRRK2 G2019S KI mice. LRRK2 G2019S promoted intestinal epithelial cell proliferation and inflammation within the tumor microenvironment. Mechanistically, we found that LRRK2 G2019S KI mice are more susceptible to dextran sulfate sodium (DSS)-induced colitis. Suppressing the kinase activity of LRRK2 ameliorated the severity of colitis in both LRRK2 G2019S KI and WT mice. At the molecular level, our investigation unveiled that LRRK2 G2019S promotes the production of reactive oxygen species, triggers inflammasome activation, and induces cell necrosis in the gut epithelium in a mouse model of colitis. Collectively, our data provide direct evidence that gain-of-kinase activity in LRRK2 promotes colorectal tumorigenesis, implicating LRRK2 as a potential target in colon cancer patients with hyper LRRK2 kinase activity.

17.
Nat Commun ; 13(1): 2748, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35585086

ABSTRACT

Toll-like receptors/Interleukin-1 receptor signaling plays an important role in high-fat diet-induced adipose tissue dysfunction contributing to obesity-associated metabolic syndromes. Here, we show an unconventional IL-1R-IRAKM-Slc25a1 signaling axis in adipocytes that reprograms lipogenesis to promote diet-induced obesity. Adipocyte-specific deficiency of IRAKM reduces high-fat diet-induced body weight gain, increases whole body energy expenditure and improves insulin resistance, associated with decreased lipid accumulation and adipocyte cell sizes. IL-1ß stimulation induces the translocation of IRAKM Myddosome to mitochondria to promote de novo lipogenesis in adipocytes. Mechanistically, IRAKM interacts with and phosphorylates mitochondrial citrate carrier Slc25a1 to promote IL-1ß-induced mitochondrial citrate transport to cytosol and de novo lipogenesis. Moreover, IRAKM-Slc25a1 axis mediates IL-1ß induced Pgc1a acetylation to regulate thermogenic gene expression in adipocytes. IRAKM kinase-inactivation also attenuates high-fat diet-induced obesity. Taken together, our study suggests that the IL-1R-IRAKM-Slc25a1 signaling axis tightly links inflammation and adipocyte metabolism, indicating a potential therapeutic target for obesity.


Subject(s)
Insulin Resistance , Lipogenesis , Adipocytes/metabolism , Animals , Diet, High-Fat , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Obesity/etiology , Obesity/metabolism , Organic Anion Transporters/metabolism , Receptors, Interleukin-1/metabolism , Thermogenesis
18.
Nat Commun ; 13(1): 2406, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35504893

ABSTRACT

The C-type lectin receptor Mincle is known for its important role in innate immune cells in recognizing pathogen and damage associated molecular patterns. Here we report a T cell-intrinsic role for Mincle in the pathogenesis of experimental autoimmune encephalomyelitis (EAE). Genomic deletion of Mincle in T cells impairs TH17, but not TH1 cell-mediated EAE, in alignment with significantly higher expression of Mincle in TH17 cells than in TH1 cells. Mechanistically, dying cells release ß-glucosylceramide during inflammation, which serves as natural ligand for Mincle. Ligand engagement induces activation of the ASC-NLRP3 inflammasome, which leads to Caspase8-dependent IL-1ß production and consequentially TH17 cell proliferation via an autocrine regulatory loop. Chemical inhibition of ß-glucosylceramide synthesis greatly reduces inflammatory CD4+ T cells in the central nervous system and inhibits EAE progression in mice. Taken together, this study indicates that sensing of danger signals by Mincle on TH17 cells plays a critical role in promoting CNS inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Th17 Cells , Animals , Central Nervous System/metabolism , Glucosylceramides/metabolism , Inflammation/metabolism , Ligands , Mice
19.
J Immunol ; 182(3): 1631-40, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155512

ABSTRACT

IL-25 initiates, promotes, and augments Th2 immune responses. In this study, we report that Act1, a key component in IL-17-mediated signaling, is an essential signaling molecule for IL-25 signaling. Although Act1-deficient mice showed reduced expression of KC (CXCL1) and neutrophil recruitment to the airway compared with wild-type mice in response to IL-17 stimulation, Act1 deficiency abolished IL-25-induced expression of IL-4, IL-5, IL-13, eotaxin-1 (CCL11), and pulmonary eosinophilia. Using a mouse model of allergic pulmonary inflammation, we observed diminished Th2 responses and lung inflammation in Act1-deficient mice compared with wild-type mice. Importantly, Act1 deficiency in epithelial cells reduced the phenotype of allergic pulmonary inflammation due to loss of IL-17-induced neutrophilia and IL-25-induced eosinophilia, respectively. These results demonstrate the essential role of epithelial-derived Act1 in allergic pulmonary inflammation through the distinct impact of the IL-17R-Act1 and IL-25R-Act1 axes. Such findings are crucial for the understanding of pathobiology of atopic diseases, including allergic asthma, which identifies Act1 as a potential therapeutic target.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Inflammation Mediators/physiology , Interleukin-17/physiology , Interleukins/physiology , Lung/pathology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Respiratory Mucosa/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Eosinophilia/immunology , Eosinophilia/metabolism , Eosinophilia/prevention & control , Female , HeLa Cells , Humans , Interleukin-17/administration & dosage , Interleukins/administration & dosage , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Respiratory Hypersensitivity/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
20.
J Immunol ; 183(1): 568-77, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542468

ABSTRACT

Both IL-23- and IL-1-mediated signaling pathways play important roles in Th17 cell differentiation, cytokine production, and autoimmune diseases. The IL-1R-associated kinase 4 (IRAK4) is critical for IL-1/TLR signaling. We show here that inactivation of IRAK4 kinase in mice (IRAK4 KI) results in significant resistance to experimental autoimmune encephalomyelitis due to a reduction in infiltrating inflammatory cells into the CNS and reduced Ag-specific CD4(+) T cell-mediated IL-17 production. Adoptive transfer of myelin oligodendrocyte glycoprotein 35-55-specific IRAK4 KI Th17 cells failed to induce experimental autoimmune encephalomyelitis in either wild-type or IRAK4 KI recipient mice, indicating the lack of autoantigen-specific Th17 cell activities in the absence of IRAK4 kinase activity. Furthermore, the absence of IRAK4 kinase activity blocked induction of IL-23R expression, STAT3 activation by IL-23, and Th17 cytokine expression in differentiated Th17 cells. Importantly, blockade of IL-1 signaling by IL-1RA inhibited Th17 differentiation and IL-23-induced cytokine expression in differentiated Th17 cells. The results of these studies demonstrate that IL-1-mediated IRAK4 kinase activity in T cells is essential for induction of IL-23R expression, Th17 differentiation, and autoimmune disease.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/physiology , Interleukin-17/physiology , T-Lymphocytes, Helper-Inducer/enzymology , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Cell Migration Inhibition/genetics , Cell Migration Inhibition/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Enzyme Activation/genetics , Enzyme Activation/immunology , Female , Gene Knock-In Techniques , Glycoproteins/administration & dosage , Glycoproteins/antagonists & inhibitors , Immunity, Innate/genetics , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-17/antagonists & inhibitors , Interleukin-17/biosynthesis , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Mice , Molecular Sequence Data , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/administration & dosage , Peptide Fragments/antagonists & inhibitors , Signal Transduction/genetics , Signal Transduction/immunology , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes, Helper-Inducer/pathology
SELECTION OF CITATIONS
SEARCH DETAIL