Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cytokine ; 176: 156540, 2024 04.
Article in English | MEDLINE | ID: mdl-38359559

ABSTRACT

Death Receptor 3 (DR3) is a cytokine receptor of the Tumor Necrosis Factor receptor superfamily that plays a multifaceted role in both innate and adaptive immunity. Based on the death domain motif in its cytosolic tail, DR3 had been proposed and functionally affirmed as a trigger of apoptosis. Further studies, however, also revealed roles of DR3 in other cellular pathways, including inflammation, survival, and proliferation. DR3 is expressed in various cell types, including T cells, B cells, innate lymphocytes, myeloid cells, fibroblasts, and even outside the immune system. Because DR3 is mainly expressed on T cells, DR3-mediated immune perturbations leading to autoimmunity and other diseases were mostly attributed to DR3 activation of T cells. However, which T cell subset and what T effector functions are controlled by DR3 to drive these processes remain incompletely understood. DR3 engagement was previously found to alter CD4 T helper subset differentiation, expand the Foxp3+ Treg cell pool, and maintain intraepithelial γδ T cells in the gut. Recent studies further unveiled a previously unacknowledged aspect of DR3 in regulating innate-like invariant NKT (iNKT) cell activation, expanding the scope of DR3-mediated immunity in T lineage cells. Importantly, in the context of iNKT cells, DR3 ligation exerted costimulatory effects in agonistic TCR signaling, unveiling a new regulatory framework in T cell activation and proliferation. The current review is aimed at summarizing such recent findings on the role of DR3 on conventional T cells and innate-like T cells and discussing them in the context of immunopathogenesis.


Subject(s)
Receptors, Cytokine , Receptors, Tumor Necrosis Factor, Member 25 , Humans , Tumor Necrosis Factor Ligand Superfamily Member 15 , Inflammation/metabolism , T-Lymphocyte Subsets/metabolism
3.
Nat Immunol ; 24(6): 1036-1048, 2023 06.
Article in English | MEDLINE | ID: mdl-37106040

ABSTRACT

Allergic diseases are a major global health issue. Interleukin (IL)-9-producing helper T (TH9) cells promote allergic inflammation, yet TH9 cell effector functions are incompletely understood because their lineage instability makes them challenging to study. Here we found that resting TH9 cells produced IL-9 independently of T cell receptor (TCR) restimulation, due to STAT5- and STAT6-dependent bystander activation. This mechanism was seen in circulating cells from allergic patients and was restricted to recently activated cells. STAT5-dependent Il9/IL9 regulatory elements underwent remodeling over time, inactivating the locus. A broader 'allergic TH9' transcriptomic and epigenomic program was also unstable. In vivo, TH9 cells induced airway inflammation via TCR-independent, STAT-dependent mechanisms. In allergic patients, TH9 cell expansion was associated with responsiveness to JAK inhibitors. These findings suggest that TH9 cell instability is a negative checkpoint on bystander activation that breaks down in allergy and that JAK inhibitors should be considered for allergic patients with TH9 cell expansion.


Subject(s)
Hypersensitivity , Janus Kinase Inhibitors , Humans , Interleukin-9/genetics , T-Lymphocytes, Helper-Inducer , STAT5 Transcription Factor/genetics , Chromatin/genetics , Inflammation , Hypersensitivity/genetics , Cell Differentiation , STAT6 Transcription Factor
4.
Cell Mol Life Sci ; 80(3): 76, 2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36847849

ABSTRACT

Invariant natural killer T (iNKT) cells correspond to a population of thymus-generated T cells with innate-like characteristics and effector functions. Among the various iNKT subsets, NKT17 is the only subset that produces the proinflammatory cytokine IL-17. But, how NKT17 cells acquire this ability and what would selectively trigger their activation remain incompletely understood. Here, we identified the cytokine receptor DR3 being specifically expressed on thymic NKT17 cells and mostly absent on other thymic iNKT subsets. Moreover, DR3 ligation promoted the in vivo activation of thymic NKT17 cells and provided costimulatory effects upon agonistic α-GalCer stimulation. Thus, we identified a specific surface marker for thymic NKT17 cells that triggers their activation and augments their effector functions both in vivo and in vitro. These findings provide new insights for deciphering the role and function of murine NKT17 cells and for understanding the development and activation mechanisms of iNKT cells in general.


Subject(s)
Natural Killer T-Cells , Receptors, Tumor Necrosis Factor, Member 25 , Thymus Gland , Animals , Mice , Cytokines , Receptors, Cytokine , Receptors, Tumor Necrosis Factor, Member 25/metabolism
5.
J Allergy Clin Immunol ; 148(4): 911-925, 2021 10.
Article in English | MEDLINE | ID: mdl-34625141

ABSTRACT

Since its discovery, the Janus kinase-signal transduction and activation of transcription (JAK-STAT) pathway has become recognized as a central mediator of widespread and varied human physiological processes. The field of JAK-STAT biology, particularly its clinical relevance, continues to be shaped by 2 important advances. First, the increased use of genomic sequencing has led to the discovery of novel clinical syndromes caused by mutations in JAK and STAT genes. This has provided insights regarding the consequences of aberrant JAK-STAT signaling for immunity, lymphoproliferation, and malignancy. In addition, since the approval of ruxolitinib and tofacitinib, the therapeutic use of JAK inhibitors (jakinibs) has expanded to include a large spectrum of diseases. Efficacy and safety data from over a decade of clinical studies have provided additional mechanistic insights while improving the care of patients with inflammatory and neoplastic conditions. This review discusses major advances in the field, focusing on updates in genetic diseases and in studies of clinical jakinibs in human disease.


Subject(s)
Genetic Diseases, Inborn/drug therapy , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/immunology , STAT Transcription Factors/immunology , Animals , Cytokines/immunology , Genetic Diseases, Inborn/immunology , Humans , Janus Kinases/genetics , Mutation , STAT Transcription Factors/genetics , Signal Transduction
6.
Eur J Immunol ; 51(7): 1615-1627, 2021 07.
Article in English | MEDLINE | ID: mdl-33930196

ABSTRACT

The European Journal of Immunology was launched 50 years ago, coinciding with the discovery of many cytokines and growth factors and the emergence of an entirely new field of research. Ultimately, our knowledge about the biological activity of these factors allowed us to better understand how the immune system functions in the context of inflammatory and autoimmune diseases leading to the development of targeted biologic therapies. The study of cytokine signal transduction led to the discovery of Janus kinases (JAK), and the consideration of therapeutically targeting JAKs to treat immune and inflammatory diseases. This year also marks the tenth anniversary of the approval of the first JAK inhibitor (jakinib) and now there are a total of nine approved jakinibs for treatment of rheumatologic, dermatologic, gastrointestinal, and neoplastic indications and most recently COVID-19. Here, we summarized the discoveries that led to development of first-generation jakinibs, discussed some of the newer, possibly more selective jakinibs, as well as jakinibs that also target other kinases. We also illustrated the rationale behind the application of these drugs in the treatment of COVID-19 cytokine storm. In this review, we will discuss the clinical success of jakinibs, the gaps in our understanding of their biological activities as well as challenges in regard to their clinical application.


Subject(s)
Autoimmune Diseases/drug therapy , COVID-19 Drug Treatment , Cytokine Release Syndrome/drug therapy , Hypersensitivity/drug therapy , Janus Kinase Inhibitors/therapeutic use , Cytokine Release Syndrome/pathology , Cytokines/biosynthesis , Cytokines/immunology , Humans , Janus Kinases/antagonists & inhibitors , SARS-CoV-2/drug effects , Signal Transduction/immunology
7.
Immunity ; 54(3): 514-525.e6, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33657395

ABSTRACT

MicroRNAs are important regulators of immune responses. Here, we show miR-221 and miR-222 modulate the intestinal Th17 cell response. Expression of miR-221 and miR-222 was induced by proinflammatory cytokines and repressed by the cytokine TGF-ß. Molecular targets of miR-221 and miR-222 included Maf and Il23r, and loss of miR-221 and miR-222 expression shifted the transcriptomic spectrum of intestinal Th17 cells to a proinflammatory signature. Although the loss of miR-221 and miR-222 was tolerated for maintaining intestinal Th17 cell homeostasis in healthy mice, Th17 cells lacking miR-221 and miR-222 expanded more efficiently in response to IL-23. Both global and T cell-specific deletion of miR-221 and miR-222 rendered mice prone to mucosal barrier damage. Collectively, these findings demonstrate that miR-221 and miR-222 are an integral part of intestinal Th17 cell response that are induced after IL-23 stimulation to constrain the magnitude of proinflammatory response.


Subject(s)
Inflammation/immunology , Interleukin-23/metabolism , Intestinal Mucosa/immunology , MicroRNAs/genetics , Th17 Cells/immunology , Animals , Feedback, Physiological , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-maf/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
8.
Microorganisms ; 10(1)2021 Dec 30.
Article in English | MEDLINE | ID: mdl-35056521

ABSTRACT

Very little is known about disease transmission via the gut microbiome. We hypothesized that certain inflammatory features could be transmitted via the gut microbiome and tested this hypothesis using an animal model of inflammatory diseases. Twelve-week-old healthy C57 Bl/6 and Germ-Free (GF) female and male mice were fecal matter transplanted (FMT) under anaerobic conditions with TNFΔARE-/+ donors exhibiting spontaneous Rheumatoid Arthritis (RA) and Inflammatory Bowel Disease (IBD) or with conventional healthy mice control donors. The gut microbiome analysis was performed using 16S rRNA sequencing amplification and bioinformatics analysis with the HIVE bioinformatics platform. Histology, immunohistochemistry, ELISA Multiplex analysis, and flow cytometry were conducted to confirm the inflammatory transmission status. We observed RA and IBD features transmitted in the GF mice cohort, with gut tissue disruption, cartilage alteration, elevated inflammatory mediators in the tissues, activation of CD4/CD8+ T cells, and colonization and transmission of the gut microbiome similar to the donors' profile. We did not observe a change or transmission when conventional healthy mice were FMT with TNFΔARE-/+ donors, suggesting that a healthy microbiome might withstand an unhealthy transplant. These findings show the potential involvement of the gut microbiome in inflammatory diseases. We identified a cluster of bacteria playing a role in this mechanism.

10.
Arthritis Res Ther ; 22(1): 106, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32381123

ABSTRACT

BACKGROUND: The tumor necrosis factor (TNF) superfamily cytokine TNF-like protein 1A (TL1A) and its receptor DR3 are essential for diverse animal models of autoimmune disease and may be pathogenic in rheumatoid arthritis (RA). However, the relationship of TL1A to disease duration, activity, and response to anti-TNF and other therapies in RA is not clear. METHODS: We measured soluble TL1A in synovial fluid (SF), serum, or plasma from RA first-degree relatives (FDRs) and in early RA and established disease. We measured the effects of anti-TNF and methotrexate (MTX) therapy on circulating TL1A from multiple independent RA treatment trials. We also determined the ability of a blocking anti-TL1A antibody to inhibit clinical disease and articular bone destruction in the murine collagen-induced arthritis (CIA) model of human RA. RESULTS: Soluble TL1A was specifically elevated in the blood and SF of patients with RA compared to patients with other diseases and was elevated early in disease and in at-risk anti-cyclic citrullinated peptide (CCP) (+) first-degree relatives (FDRs). Therapeutic TNF inhibition reduced serum TL1A in both responders and non-responders, whereas TL1A declined following MTX treatment only in responders. In murine CIA, TL1A blockade was clinically efficacious and reduced bone erosions. CONCLUSIONS: TL1A is specifically elevated in RA from early in the disease course and in at-risk FDRs. The decline in TL1A after TNF blockade suggests that TL1A levels may be a useful biomarker for TNF activity in RA. These results support the further investigation of the relationship between TL1A and TNF and TL1A blockade as a potential therapeutic strategy in RA.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Tumor Necrosis Factor Ligand Superfamily Member 15/blood , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/genetics , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Humans , Methotrexate/therapeutic use , Mice , Synovial Fluid , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor Ligand Superfamily Member 15/antagonists & inhibitors , Tumor Necrosis Factor-alpha
11.
Immunity ; 51(4): 682-695.e6, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31353223

ABSTRACT

Innate lymphocytes maintain tissue homeostasis at mucosal barriers, with group 2 innate lymphoid cells (ILC2s) producing type 2 cytokines and controlling helminth infection. While the molecular understanding of ILC2 responses has advanced, the complexity of microenvironmental factors impacting ILC2s is becoming increasingly apparent. Herein, we used single-cell analysis to explore the diversity of gene expression among lung lymphocytes during helminth infection. Following infection, we identified a subset of ILC2s that preferentially expressed Il5-encoding interleukin (IL)-5, together with Calca-encoding calcitonin gene-related peptide (CGRP) and its cognate receptor components. CGRP in concert with IL-33 and neuromedin U (NMU) supported IL-5 but constrained IL-13 expression and ILC2 proliferation. Without CGRP signaling, ILC2 responses and worm expulsion were enhanced. Collectively, these data point to CGRP as a context-dependent negative regulatory factor that shapes innate lymphocyte responses to alarmins and neuropeptides during type 2 innate immune responses.


Subject(s)
Inflammation/immunology , Lymphocytes/immunology , Nippostrongylus/physiology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Strongylida Infections/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Immunity, Innate , Interleukin-33/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/metabolism , Receptors, Calcitonin Gene-Related Peptide/genetics , Single-Cell Analysis , Th2 Cells/immunology , Transplantation Chimera
12.
Immunity ; 50(1): 106-120.e10, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30650370

ABSTRACT

CD4+ T helper (Th) differentiation is regulated by diverse inputs, including the vitamin A metabolite retinoic acid (RA). RA acts through its receptor RARα to repress transcription of inflammatory cytokines, but is also essential for Th-mediated immunity, indicating complex effects of RA on Th specification and the outcome of the immune response. We examined the impact of RA on the genome-wide transcriptional response during Th differentiation to multiple subsets. RA effects were subset-selective and were most significant in Th9 cells. RA globally antagonized Th9-promoting transcription factors and inhibited Th9 differentiation. RA directly targeted the extended Il9 locus and broadly modified the Th9 epigenome through RARα. RA-RARα activity limited murine Th9-associated pulmonary inflammation, and human allergic inflammation was associated with reduced expression of RA target genes. Thus, repression of the Th9 program is a major function of RA-RARα signaling in Th differentiation, arguing for a role for RA in interleukin 9 (IL-9) related diseases.


Subject(s)
Hypersensitivity/immunology , Lung/physiology , Pneumonia/immunology , Retinoic Acid Receptor alpha/metabolism , T-Lymphocytes, Helper-Inducer/physiology , Animals , Epigenetic Repression , HEK293 Cells , Humans , Hypersensitivity/genetics , Interleukin-9/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/genetics , Retinoic Acid Receptor alpha/genetics , Signal Transduction , Transcription, Genetic , Tretinoin/metabolism
13.
PLoS Genet ; 14(9): e1007458, 2018 09.
Article in English | MEDLINE | ID: mdl-30199539

ABSTRACT

Chronic inflammation in inflammatory bowel disease (IBD) results from a breakdown of intestinal immune homeostasis and compromise of the intestinal barrier. Genome-wide association studies have identified over 200 genetic loci associated with risk for IBD, but the functional mechanisms of most of these genetic variants remain unknown. Polymorphisms at the TNFSF15 locus, which encodes the TNF superfamily cytokine commonly known as TL1A, are associated with susceptibility to IBD in multiple ethnic groups. In a wide variety of murine models of inflammation including models of IBD, TNFSF15 promotes immunopathology by signaling through its receptor DR3. Such evidence has led to the hypothesis that expression of this lymphocyte costimulatory cytokine increases risk for IBD. In contrast, here we show that the IBD-risk haplotype at TNFSF15 is associated with decreased expression of the gene by peripheral blood monocytes in both healthy volunteers and IBD patients. This association persists under various stimulation conditions at both the RNA and protein levels and is maintained after macrophage differentiation. Utilizing a "recall-by-genotype" bioresource for allele-specific expression measurements in a functional fine-mapping assay, we localize the polymorphism controlling TNFSF15 expression to the regulatory region upstream of the gene. Through a T cell costimulation assay, we demonstrate that genetically regulated TNFSF15 has functional relevance. These findings indicate that genetically enhanced expression of TNFSF15 in specific cell types may confer protection against the development of IBD.


Subject(s)
Colitis, Ulcerative/genetics , Crohn Disease/genetics , Genetic Predisposition to Disease , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Adult , Alleles , Cells, Cultured , Colitis, Ulcerative/blood , Colitis, Ulcerative/immunology , Crohn Disease/blood , Crohn Disease/immunology , Female , Haplotypes/genetics , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Polymorphism, Single Nucleotide/genetics , Primary Cell Culture , Quantitative Trait Loci/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/blood , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Young Adult
14.
Nat Commun ; 9(1): 1390, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29643386

ABSTRACT

Melorheostosis is a sporadic disease of uncertain etiology characterized by asymmetric bone overgrowth and functional impairment. Using whole exome sequencing, we identify somatic mosaic MAP2K1 mutations in affected, but not unaffected, bone of eight unrelated patients with melorheostosis. The activating mutations (Q56P, K57E and K57N) cluster tightly in the MEK1 negative regulatory domain. Affected bone displays a mosaic pattern of increased p-ERK1/2 in osteoblast immunohistochemistry. Osteoblasts cultured from affected bone comprise two populations with distinct p-ERK1/2 levels by flow cytometry, enhanced ERK1/2 activation, and increased cell proliferation. However, these MAP2K1 mutations inhibit BMP2-mediated osteoblast mineralization and differentiation in vitro, underlying the markedly increased osteoid detected in affected bone histology. Mosaicism is also detected in the skin overlying bone lesions in four of five patients tested. Our data show that the MAP2K1 oncogene is important in human bone formation and implicate MEK1 inhibition as a potential treatment avenue for melorheostosis.


Subject(s)
Bone and Bones/metabolism , MAP Kinase Kinase 1/genetics , Melorheostosis/genetics , Mutation , Osteoblasts/metabolism , Osteogenesis/genetics , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone and Bones/pathology , Calcification, Physiologic , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Humans , MAP Kinase Kinase 1/metabolism , Melorheostosis/metabolism , Melorheostosis/pathology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mosaicism , Osteoblasts/pathology , Primary Cell Culture , Signal Transduction , Skin/metabolism , Skin/pathology , Exome Sequencing
15.
Sci Immunol ; 3(21)2018 03 02.
Article in English | MEDLINE | ID: mdl-29500225

ABSTRACT

Atopic dermatitis is an allergic inflammatory skin disease characterized by the production of the type 2 cytokines in the skin by type 2 innate lymphoid cells (ILC2s) and T helper 2 (TH2) cells, and tissue eosinophilia. Using two distinct mouse models of atopic dermatitis, we show that expression of retinoid-related orphan receptor α (RORα) in skin-resident T regulatory cells (Tregs) is important for restraining allergic skin inflammation. In both models, targeted deletion of RORα in mouse Tregs led to exaggerated eosinophilia driven by interleukin-5 (IL-5) production by ILC2s and TH2 cells. Expression of RORα in skin-resident Tregs suppressed IL-4 expression and enhanced expression of death receptor 3 (DR3), which is the receptor for tumor necrosis factor (TNF) family cytokine, TNF ligand-related molecule 1 (TL1A), which promotes Treg functions. DR3 is expressed on both ILC2s and skin-resident Tregs Upon deletion of RORα in skin-resident Tregs, we found that Tregs were no longer able to sequester TL1A, resulting in enhanced ILC2 activation. We also documented higher expression of RORα in skin-resident Tregs than in peripheral blood circulating Tregs in humans, suggesting that RORα and the TL1A-DR3 circuit could be therapeutically targeted in atopic dermatitis.


Subject(s)
Dermatitis, Atopic/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Humans , Immunity, Innate , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Member 25/immunology , Skin/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology
16.
J Immunol ; 200(4): 1360-1369, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29335258

ABSTRACT

TNF superfamily cytokines play major roles in the regulation of adaptive and innate immunity. The TNF superfamily cytokine TL1A (TNFSF15), through its cognate receptor DR3 (TNFRSF25), promotes T cell immunity to pathogens and directly costimulates group 2 and 3 innate lymphoid cells. Polymorphisms in the TNFSF15 gene are associated with the risk for various human diseases, including inflammatory bowel disease. Like other cytokines in the TNF superfamily, TL1A is synthesized as a type II transmembrane protein and cleaved from the plasma membrane by metalloproteinases. Membrane cleavage has been shown to alter or abrogate certain activities of other TNF family cytokines; however, the functional capabilities of membrane-bound and soluble forms TL1A are not known. Constitutive expression of TL1A in transgenic mice results in expansion of activated T cells and promotes intestinal hyperplasia and inflammation through stimulation of group 2 innate lymphoid cells. Through the generation of membrane-restricted TL1A-transgenic mice, we demonstrate that membrane TL1A promotes expression of inflammatory cytokines in the lung, dependent upon DR3 expression on T cells. Soluble TL1A alone was unable to produce this phenotype but was still able to induce intestinal type 2 inflammation independently of T cells. These data suggest differential roles for membrane and soluble TL1A on adaptive and innate immune cells and have implications for the consequences of blocking these two forms of TL1A.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Lymphocyte Activation/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Animals , Mice , Mice, Transgenic , T-Lymphocytes/immunology
17.
Methods Mol Biol ; 1585: 59-71, 2017.
Article in English | MEDLINE | ID: mdl-28477187

ABSTRACT

In vitro polarization of naïve CD4+ T cells toward distinct T helper lineages is crucial for establishing the factors and features that determine the differentiation, stability, and effector function for each T helper subsets. In this regard, the recently defined Th9 subset has been reported with two essential cytokines requirement for their generation. Generating Th9 cells in vitro from naïve CD4+ T cells requires the combination of TGF-ß and IL-4. However, the amount of IL-9 producing under these minimal conditions is often small. The intent of this chapter is to provide examples to increase the generation of IL-9 producing T cells in vitro by modulating TCR strength and co-stimulation through the TNF family member TL1A. We hope that these methods to efficiently differentiate naïve CD4+ T cells toward IL-9 producing cells will facilitate understanding the differentiation and function of Th9 cells and their pathogenesis in various inflammatory and autoimmune diseases.


Subject(s)
Interleukin-9/metabolism , Receptors, Antigen, T-Cell/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Humans , Interleukin-4/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Transforming Growth Factor beta/metabolism
18.
Semin Immunopathol ; 39(1): 21-28, 2017 01.
Article in English | MEDLINE | ID: mdl-27896636

ABSTRACT

The tumor necrosis factor (TNF) receptors and their corresponding cytokine ligands have been implicated in many aspects of the biology of immune functions. TNF receptors have key roles during various stages of T cell homeostasis. Many of them can co-stimulate lymphocyte proliferation and cytokine production. Additionally, several TNF cytokines can regulate T cell differentiation, including promoting Th1, Th2, Th17, and more recently the newly described Th9 subset. Four TNF family cytokines have been identified as regulators for IL-9 production by T cells. OX40L, TL1A, and GITRL can promote Th9 formation but can also divert iTreg into Th9, while 4-1BBL seems to inhibit IL-9 production from iTreg and has not been studied for its ability to promote Th9 generation. Regulation of IL-9 production by TNF family cytokines has repercussions in vivo, including enhancement of anti-tumor immunity and immunopathology in allergic lung and ocular inflammation. Regulating T cell production of IL-9 through blockade or agonism of TNF family cytokine receptors may be a therapeutic strategy for autoimmune and allergic diseases and in tumor.


Subject(s)
Cell Differentiation , Interleukin-9/metabolism , Multigene Family , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Tumor Necrosis Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Disease Susceptibility , Drug Discovery , Humans , Interleukin-9/genetics , Lymphocyte Activation , Protein Binding , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Helper-Inducer/cytology , Tumor Necrosis Factors/chemistry , Tumor Necrosis Factors/genetics
19.
Nat Commun ; 7: 13895, 2016 12 23.
Article in English | MEDLINE | ID: mdl-28008916

ABSTRACT

Mutations affecting the apoptosis-inducing function of the Fas/CD95 TNF-family receptor result in autoimmune and lymphoproliferative disease. However, Fas can also costimulate T-cell activation and promote tumour cell growth and metastasis. Palmitoylation at a membrane proximal cysteine residue enables Fas to localize to lipid raft microdomains and induce apoptosis in cell lines. Here, we show that a palmitoylation-defective Fas C194V mutant is defective in inducing apoptosis in primary mouse T cells, B cells and dendritic cells, while retaining the ability to enhance naive T-cell differentiation. Despite inability to efficiently induce cell death, the Fas C194V receptor prevents the lymphoaccumulation and autoimmunity that develops in Fas-deficient mice. These findings indicate that induction of apoptosis through Fas is dependent on receptor palmitoylation in primary immune cells, and Fas may prevent autoimmunity by mechanisms other than inducing apoptosis.


Subject(s)
Apoptosis/immunology , Autoimmunity/immunology , Membrane Microdomains/immunology , Mice, Transgenic , fas Receptor/immunology , Animals , Apoptosis/genetics , Autoimmunity/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , HEK293 Cells , Humans , Lipoylation/immunology , Membrane Microdomains/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mutation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , fas Receptor/genetics , fas Receptor/metabolism
20.
Nat Commun ; 7: 10857, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26936133

ABSTRACT

Th9 cells produce interleukin (IL)-9, a cytokine implicated in allergic asthma and autoimmunity. Here we show that Itk, a mediator of T cell receptor signalling required for Th2 immune responses and the development of asthma, is a positive regulator of Th9 differentiation. In a model of allergic lung disease, Itk-deficient mice show reduced pulmonary inflammation and IL-9 production by T cells and innate lymphoid type 2 cells (ILC2), despite normal early induction of ILC2s. In vitro, Itk(-/-) CD4(+) T cells do not produce IL-9 and have reduced levels of IRF4 (Interferon Regulator Factor 4), a critical transcription factor for effector T cell function. Both IL-9 and IRF4 expression are rescued by either IL-2 or constitutively active STAT5, but not NFATc1. STAT5 binds the Irf4 promoter, demonstrating one mechanism by which IL-2 rescues weakly activated T cells. Itk inhibition also reduces IL-9 expression by human T cells, implicating ITK as a key regulator of Th9 induction.


Subject(s)
Cell Differentiation/physiology , Interferon Regulatory Factors/metabolism , Interleukin-2/metabolism , Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes , Female , Gene Expression Regulation/physiology , Interferon Regulatory Factors/genetics , Interleukin-2/genetics , Lung Diseases/chemically induced , Male , Mice , Mice, Knockout , Papain/toxicity , Protein Kinases/genetics , Protein-Tyrosine Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...