Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 15(5): 449-56, 2014 May.
Article in English | MEDLINE | ID: mdl-24681564

ABSTRACT

The physiological functions of members of the tumor-necrosis factor (TNF) receptor (TNFR)-associated factor (TRAF) family in T cell immunity are not well understood. We found that in the presence of interleukin 6 (IL-6), naive TRAF5-deficient CD4(+) T cells showed an enhanced ability to differentiate into the TH17 subset of helper T cells. Accordingly, TH17 cell-associated experimental autoimmune encephalomyelitis (EAE) was greatly exaggerated in Traf5(-/-) mice. Although it is normally linked with TNFR signaling pathways, TRAF5 constitutively associated with a cytoplasmic region in the signal-transducing receptor gp130 that overlaps with the binding site for the transcription activator STAT3 and suppressed the recruitment and activation of STAT3 in response to IL-6. Our results identify TRAF5 as a negative regulator of the IL-6 receptor signaling pathway that limits the induction of proinflammatory CD4(+) T cells that require IL-6 for their development.


Subject(s)
Cytokine Receptor gp130/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocyte Subsets/immunology , TNF Receptor-Associated Factor 5/metabolism , Th17 Cells/immunology , Animals , CD4 Antigens/metabolism , Cell Differentiation/genetics , Cells, Cultured , Disease Progression , Interleukin-6/immunology , Mice , Mice, Inbred Strains , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , TNF Receptor-Associated Factor 5/genetics , Transcriptional Activation/genetics
2.
J Immunol ; 208(3): 642-650, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34996840

ABSTRACT

TNF receptor-associated factor 5 (TRAF5) restrains early signaling activity of the IL-6 receptor in naive CD4+ T cells by interacting with the shared gp130 chain, although TRAF5 was initially discovered as a cytoplasmic adaptor protein to activate signaling mediated by TNF receptor family molecules. This leads to the question of whether TRAF5 limits signaling via the receptor for IL-27, which is composed of gp130 and WSX-1. The aim of this study is to clarify the role of TRAF5 in IL-27 receptor signaling and to understand the differential role of TRAF5 on cytokine receptor signaling. We found that Traf5 -/- CD4+ T cells displayed significantly higher levels of phosphorylated STAT1 and STAT-regulated genes Socs3 and Tbx21, as early as 1 h after IL-27 exposure when compared with Traf5 +/+ CD4+ T cells. Upon IL-27 and TCR signals, the Traf5 deficiency significantly increased the induction of IL-10 and promoted the proliferation of CD4+ T cells. Traf5 -/- mice injected with IL-27 displayed significantly enhanced delayed-type hypersensitivity responses, demonstrating that TRAF5 works as a negative regulator for IL-27 receptor signaling. In contrast, IL-2 and proliferation mediated by glucocorticoid-induced TNF receptor-related protein (GITR) and TCR signals were significantly decreased in Traf5 -/- CD4+ T cells, confirming that TRAF5 works as a positive regulator for cosignaling via GITR. Collectively, our results demonstrate that TRAF5 reciprocally controls signals mediated by the IL-27 receptor and GITR in CD4+ T cells and suggest that the regulatory activity of TRAF5 in gp130 is distinct from that in TNF receptor family molecules in a T cell.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokine Receptor gp130/metabolism , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin/metabolism , TNF Receptor-Associated Factor 5/metabolism , Animals , Cell Proliferation , Hypersensitivity, Delayed/immunology , Interleukin-10/immunology , Interleukins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/immunology , Suppressor of Cytokine Signaling 3 Protein/metabolism , T-Box Domain Proteins/metabolism , TNF Receptor-Associated Factor 5/genetics
3.
Biochem Biophys Res Commun ; 637: 9-16, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36375254

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are resident cells and participate in innate and adaptive immunity. In the tumor microenvironment (TME), ILC2s contribute to both tumorigenesis and inhibition of tumor growth, but the true role of ILC2s in TME construction remains unclear. We show that IL-33 treatment induces an anti-tumor effect in vivo in a mouse model of melanoma in which ILC2s and CD8+ T cells infiltrate into tumor tissue. This anti-tumor effect is dependent on CD8+ T cells, however, IL-33 does not act directly on CD8+ T cells because the cells lack ST2, the receptor for IL-33. ILC2s and CD8+ T cells in tumors of IL-33-treated mice express OX40 ligand (OX40L) and OX40, respectively, and in vivo blockade of OX40L-OX40 interaction canceled the anti-tumor effect of IL-33. Co-culture of CD8+ T cells expressing OX40 with IL-33-stimulated ILC2 expressing OX40L promoted cell activation and proliferation of CD8+ T cells, which was significantly suppressed by administration of anti-OX40L blocking antibody. Thus, the IL-33-ILC2 axis promotes CD8+ T cell responses via OX40/OX40L interaction and exerts an anti-tumor effect.


Subject(s)
CD8-Positive T-Lymphocytes , Immunity, Innate , Interleukin-33 , Neoplasms , Receptors, OX40 , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Interleukin-33/immunology , Lymphocytes/immunology , OX40 Ligand/immunology , Receptors, OX40/immunology , Neoplasms/immunology
4.
Dig Dis Sci ; 67(4): 1252-1259, 2022 04.
Article in English | MEDLINE | ID: mdl-33818662

ABSTRACT

BACKGROUND: The Toll-like receptor signaling pathway contributes to the regulation of intestinal homeostasis through interactions with commensal bacteria. Although the transcriptional regulator IκB-ζ can be induced by Toll-like receptor signaling, its role in intestinal homeostasis is still unclear. AIMS: To investigate the role of IκB-ζ in gut homeostasis. METHODS: DSS-administration induced colitis in control and IκB-ζ-deficient mice. The level of immunoglobulins in feces was detected by ELISA. The immunological population in lamina propria (LP) was analyzed by FACS. RESULTS: IκB-ζ-deficient mice showed severe inflammatory diseases with DSS administration in the gut. The level of IgM in the feces after DSS administration was less in IκB-ζ-deficient mice compared to control mice. Upon administration of DSS, IκB-ζ-deficient mice showed exaggerated intestinal inflammation (more IFN-g-producing CD4+ T cells in LP), and antibiotic treatment canceled this inflammatory phenotype. CONCLUSION: IκB-ζ plays a crucial role in maintaining homeostasis in the gut.


Subject(s)
Colitis , Animals , Colitis/metabolism , Dextran Sulfate/toxicity , Homeostasis , Humans , Interferon-gamma , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
5.
Biol Pharm Bull ; 45(12): 1798-1804, 2022.
Article in English | MEDLINE | ID: mdl-36450532

ABSTRACT

OX40, a member of the tumor necrosis factor (TNF) receptor superfamily, is induced on activated T cells. Membrane-bound OX40 ligand (OX40L) expressed by activated antigen-presenting cells induces OX40 signaling, which promotes T cell immunity. OX40 agonism would be a potential target for immunotherapy, however, it remains unclear how the activity of OX40 can be successfully controlled by a designer OX40L protein. We prepared a soluble OX40L protein possessing a PA-peptide tag and a collagenous trimerization domain from mannose-binding lectin (MBL), and tested whether PA-MBL-OX40L fusion protein worked as an agonist for OX40. We found that the majority of recombinant PA-MBL-OX40L protein purified from culture supernatants displayed a trimer structure and bound to cell surface OX40 or OX40-Fc fusion protein in a dose-dependent manner. Upon stimulation of CD4+ T cells with TCR/CD3 without CD28, PA-MBL-OX40L displayed significantly increased proliferative and cytokine responses when compared with a benchmark agonistic monoclonal antibody for OX40. Both soluble and immobilized forms of PA-MBL-OX40L induced potent OX40 signaling in CD4+ T cells. Mice administered with PA-MBL-OX40L displayed significantly augmented T cell-mediated delayed-type hypersensitivity responses. Our results suggest that activity of OX40L could be engineered to elicit better T cell responses by rational design of its assembly and architecture.


Subject(s)
OX40 Ligand , T-Lymphocytes , Animals , Mice , CD4-Positive T-Lymphocytes , Immunologic Factors , Immunotherapy
6.
J Cell Physiol ; 236(11): 7554-7564, 2021 11.
Article in English | MEDLINE | ID: mdl-33973242

ABSTRACT

Growing evidence suggest the association between Moyamoya disease (MMD) and immune systems, such as antigen presenting cells in particular. Rnf213 gene, a susceptibility gene for MMD, is highly expressed in immune tissues, however, its function remains unclear. In addition, the physiological role of RNF213 gene polymorphism c.14576G > A (rs112735431), susceptibility variant for MMD, is also poorly understood. By studying Rnf213-knockout (Rnf213-KO) mice with deletion of largest exon32 and Rnf213-knockin (Rnf213-KI) mice with insertion of single-nucleotide polymorphism corresponding to c.14576G > A mutation in MMD patients, we aimed to investigate the role of RNF213 in dendritic cell development, and antigen processing and presentation. First, we found a high level of Rnf213 gene expression in conventional DCs and monocytes. Second, flow cytometric and confocal microscopic analysis revealed ovalbumin protein-pulsed Rnf213-KO and Rnf213-KI DCs showed impaired antigen uptake, proteolysis and reduced numbers of endosomes and lysosomes, and thereby failed to activate and proliferate antigen-specific T cells efficiently. In addition, Rnf213-KI DCs showed a similar phenotype to that of Rnf213-KO BMDCs. In conclusion, our findings suggest the critical role of RNF213 in antigen uptake, processing and presentation.


Subject(s)
Adenosine Triphosphatases/metabolism , Antigen Presentation , Antigens/metabolism , Dendritic Cells/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphatases/genetics , Animals , Antigens/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Mice, Knockout , Moyamoya Disease/genetics , Moyamoya Disease/immunology , Moyamoya Disease/metabolism , Phenotype , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/genetics
7.
Allergy ; 76(6): 1776-1788, 2021 06.
Article in English | MEDLINE | ID: mdl-33090507

ABSTRACT

BACKGROUND: Fatty acid-binding protein 3 (FABP3) is a cytosolic carrier protein of polyunsaturated fatty acids (PUFAs) and regulates cellular metabolism. However, the physiological functions of FABP3 in immune cells and how FABP3 regulates inflammatory responses remain unclear. METHODS: Contact hypersensitivity (CHS) induced by 2,4-dinitrofluorobenzene (DNFB) and fluorescein isothiocyanate was applied to the skin wild-type and Fabp3-/- mice. Skin inflammation was assessed using FACS, histological, and qPCR analyses. The development of γ/δ T cells was evaluated by a co-culture system with OP9/Dll1 cells in the presence or absence of transgene of FABP3. RESULTS: Fabp3-deficient mice exhibit a more severe phenotype of contact hypersensitivity (CHS) accompanied by infiltration of IL-17-producing Vγ4+ γ/δ T cells that critically control skin inflammation. In Fabp3-/- mice, we found a larger proportion of Vγ4+ γ/δ T cells in the skin, even though the percentage of total γ/δ T cells did not change at steady state. Similarly, juvenile Fabp3-/- mice also contained a higher amount of Vγ4+ γ/δ T cells not only in the skin but in the thymus when compared with wild-type mice. Furthermore, thymic double-negative (DN) cells expressed FABP3, and FABP3 negatively regulates the development of Vγ4+ γ/δ T cells in the thymus. CONCLUSIONS: These findings suggest that FABP3 functions as a negative regulator of skin inflammation through limiting pathogenic Vγ4+ γ/δ T-cell generation in the thymus.


Subject(s)
Dermatitis, Contact , T-Lymphocytes , Animals , Dermatitis, Contact/genetics , Disease Models, Animal , Fatty Acid Binding Protein 3 , Fatty Acid-Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/metabolism
8.
Int Immunol ; 32(4): 233-241, 2020 04 12.
Article in English | MEDLINE | ID: mdl-31819988

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) play critical roles in type 2 immunity and are crucial for pathogenesis of various types of inflammatory disease. IQ motif-containing GTPase-activating protein 1 (IQGAP1) is a ubiquitously expressed scaffold protein that is involved in multiple cellular functions such as cell survival and trafficking. While the roles for IQGAP1 in T and B lymphocytes have been uncovered, the physiological significance of IQGAP1 in innate lymphocytes remains to be elucidated. In the current study, we demonstrate that using bone marrow chimeras, the deficiency of IQGAP1 caused an impaired survival of lung ILC2s in a cell-intrinsic manner and that Iqgap1-/- mice displayed decreased accumulation of ILC2s after administration of papain and thereby reduced the pathology of the disease. Moreover, Iqgap1-/- ILC2s showed a significantly enhanced apoptosis as compared to wild-type ILC2s under both steady-state and inflammatory conditions. Together these results identify for the first time that IQGAP1 is essential for homeostasis of ILC2s in the lung.


Subject(s)
Lung/immunology , Lymphocytes/immunology , ras GTPase-Activating Proteins/immunology , Animals , Homeostasis/immunology , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , ras GTPase-Activating Proteins/deficiency
9.
Int Immunol ; 32(4): 283-292, 2020 04 12.
Article in English | MEDLINE | ID: mdl-31954058

ABSTRACT

Nephrotic syndrome (NS) is a renal disease characterized by severe proteinuria and hypoproteinemia. Although several single-gene mutations have been associated with steroid-resistant NS, causative genes for steroid-sensitive NS (SSNS) have not been clarified. While seeking to identify causative genes associated with SSNS by whole-exome sequencing, we found compound heterozygous variants/mutations (c.524T>C; p.I175T and c.662G>A; p.R221H) of the interleukin-1 receptor accessory protein (IL1RAP) gene in two siblings with SSNS. The siblings' parents are healthy, and each parent carries a different heterozygous IL1RAP variant/mutation. Since IL1RAP is a critical subunit of the functional interleukin-1 receptor (IL-1R), we investigated the effect of these variants on IL-1R subunit function. When stimulated with IL-1ß, peripheral blood mononuclear cells from the siblings with SSNS produced markedly lower levels of cytokines compared with cells from healthy family members. Moreover, IL-1R with a variant IL1RAP subunit, reconstituted on a hematopoietic cell line, had impaired binding ability and low reactivity to IL-1ß. Thus, the amino acid substitutions in IL1RAP found in these NS patients are dysfunctional variants/mutations. Furthermore, in the kidney of Il1rap-/- mice, the number of myeloid-derived suppressor cells, which require IL-1ß for their differentiation, was markedly reduced although these mice did not show significantly increased proteinuria in acute nephrotic injury with lipopolysaccharide treatment. Together, these results identify two IL1RAP variants/mutations in humans for the first time and suggest that IL1RAP might be a causative gene for familial NS.


Subject(s)
Interleukin-1 Receptor Accessory Protein/genetics , Nephrotic Syndrome/genetics , Steroids/adverse effects , Animals , Child, Preschool , Female , Genetic Variation , Humans , Infant, Newborn , Interleukin-1 Receptor Accessory Protein/blood , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mutation , Nephrotic Syndrome/chemically induced , Nephrotic Syndrome/drug therapy , Siblings , Steroids/therapeutic use
10.
FASEB J ; 34(1): 540-554, 2020 01.
Article in English | MEDLINE | ID: mdl-31914585

ABSTRACT

A costimulatory signal from the tumor necrosis factor receptor (TNFR) family molecule OX40 (CD134), which is induced on activated T cells, is important for T-cell immunity. Aberrant OX40 cosignaling has been implicated in autoimmune and inflammatory disorders. However, the molecular mechanism by which the OX40 cosignaling regulates the T-cell response remains obscure. We found that OX40 associated with a scaffold protein, IQ motif-containing GTPase-activating protein 1 (IQGAP1) after ligation by its ligand OX40L. Naïve CD4+ T cells from Iqgap1-/- mice displayed enhanced proliferation and cytokine secretion upon receiving OX40 cosignaling. A C-terminal IQGAP1 region was responsible for its association with OX40, and TNFR-associated factor 2 (TRAF2) bridged these two proteins. The enhanced cytokine response in Iqgap1-/- T cells was restored by the expression of the C-terminal IQGAP1. Thus, the IQGAP1 binding limits the OX40 cosignaling. Disease severity of experimental autoimmune encephalomyelitis (EAE) was significantly exacerbated in Iqgap1-/- mice as compared to wild-type mice. Additionally, recipient mice with Iqgap1-/- donor CD4+ T cells exhibited significantly higher EAE scores than those with their wild-type counterparts, and OX40 blockade led to a significant reduction in the EAE severity. Thus, our study defines an important component of the OX40 cosignaling that restricts inflammation driven by antigen-activated T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunologic Memory/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Receptors, OX40/metabolism , ras GTPase-Activating Proteins/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, OX40/genetics , Signal Transduction
11.
FASEB J ; 34(11): 14820-14831, 2020 11.
Article in English | MEDLINE | ID: mdl-32910505

ABSTRACT

Glucocorticoid-induced TNFR family related gene (GITR) is a member of the TNFR superfamily that is expressed on cells of the immune system. Although the protective and pathogenic roles of GITR in T cell immunity are well characterized, the role of GITR in innate immunity in the intestinal tissues has not been well clarified. In this study, using a dextran sulfate sodium (DSS)-induced colitis model in mice, we found that GITR-deficiency rendered mice more susceptible to acute intestinal inflammation and that a significantly higher number of activated natural killer (NK) cells was accumulated in the colonic lamina propria of Gitr-/- mice as compared to wild-type mice. Additionally, Rag2-/- Gitr-/- mice, which lack T cells but have NK cells, also displayed more severe colonic inflammation than Rag2-/- mice. In contrast, an anti-GITR agonistic antibody significantly alleviated colitis in Rag2-/- mice. Engagement of GITR inhibited IL-15-mediated activating signaling events in NK cells, which include cell activation and proliferation, and production of cytokines and cytotoxic granules. Taken together, our results provide the first evidence that GITR negatively controls intestinal inflammation through NK cell functions.


Subject(s)
Colitis, Ulcerative/immunology , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Intestinal Mucosa/immunology , Killer Cells, Natural/immunology , Animals , Cells, Cultured , Colitis, Ulcerative/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucocorticoid-Induced TNFR-Related Protein/genetics , Interleukin-15/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL
12.
J Immunol ; 203(6): 1447-1456, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31420465

ABSTRACT

The physiological functions of TNF receptor-associated factor 5 (TRAF5) in the skin inflammation and wound healing process are not well characterized. We found that Traf5 -/- mice exhibited an accelerated skin wound healing as compared with wild-type counterparts. The augmented wound closure in Traf5 -/- mice was associated with a massive accumulation of plasmacytoid dendritic cells (pDCs) into skin wounds and an enhanced expression of genes related to wound repair at skin sites. In accordance with this result, adoptive transfer of Traf5 -/- pDCs, but not wild-type pDCs, into the injured skin area in wild-type recipient mice significantly promoted skin wound healing. The expression of skin-tropic chemokine receptor CXCR3 was significantly upregulated in Traf5-/- pDCs, and treatment with a CXCR3 inhibitor cancelled the promoted wound healing in Traf5-/- mice, suggesting a pivotal role of CXCR3 in pDC-dependent wound healing. Traf5 -/- pDCs displayed significantly higher expression of IFN regulatory factor 5 (IRF5), which correlated with greater induction of proinflammatory cytokine genes and CXCR3 protein after stimulation with TLR ligands. Consistently, transduction of exogeneous TRAF5 in Traf5-/- pDCs normalized the levels of abnormally elevated proinflammatory molecules, including IRF5 and CXCR3. Furthermore, knockdown of IRF5 also rescued the abnormal phenotypes of Traf5-/- pDCs. Therefore, the higher expression and induction of IRF5 in Traf5-/- pDCs causes proinflammatory and skin-tropic characteristics of the pDCs, which may accelerate skin wound healing responses. Collectively, our results uncover a novel role of TRAF5 in skin wound healing that is mediated by IRF5-dependent function of pDCs.


Subject(s)
Dendritic Cells/metabolism , Interferon Regulatory Factors/metabolism , TNF Receptor-Associated Factor 5/metabolism , Animals , Cytokines/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Receptors, CXCR3/metabolism , Signal Transduction/physiology , Skin/metabolism , Up-Regulation/physiology , Wound Healing/physiology
13.
J Epidemiol ; 31(1): 65-76, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-31932529

ABSTRACT

BACKGROUND: We established a community-based cohort study to assess the long-term impact of the Great East Japan Earthquake on disaster victims and gene-environment interactions on the incidence of major diseases, such as cancer and cardiovascular diseases. METHODS: We asked participants to join our cohort in the health check-up settings and assessment center based settings. Inclusion criteria were aged 20 years or over and living in Miyagi or Iwate Prefecture. We obtained information on lifestyle, effect of disaster, blood, and urine information (Type 1 survey), and some detailed measurements (Type 2 survey), such as carotid echography and calcaneal ultrasound bone mineral density. All participants agreed to measure genome information and to distribute their information widely. RESULTS: As a result, 87,865 gave their informed consent to join our study. Participation rate at health check-up site was about 70%. The participants in the Type 1 survey were more likely to have psychological distress than those in the Type 2 survey, and women were more likely to have psychological distress than men. Additionally, coastal residents were more likely to have higher degrees of psychological distress than inland residents, regardless of sex. CONCLUSION: This cohort comprised a large sample size and it contains information on the natural disaster, genome information, and metabolome information. This cohort also had several detailed measurements. Using this cohort enabled us to clarify the long-term effect of the disaster and also to establish personalized prevention based on genome, metabolome, and other omics information.


Subject(s)
Earthquakes/statistics & numerical data , Gene-Environment Interaction , Psychological Distress , Adult , Cardiovascular Diseases/epidemiology , Cohort Studies , Community-Based Participatory Research , Disasters , Female , Genome , Humans , Incidence , Japan/epidemiology , Life Style , Male , Metabolome , Middle Aged , Neoplasms/epidemiology , Surveys and Questionnaires , Young Adult
14.
Biochem Biophys Res Commun ; 521(2): 353-359, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31668809

ABSTRACT

The conventional dendritic cells (cDCs) and plasmacytoid DCs (pDCs) originate from the same common dendritic cell precursor cells in the bone marrow. The pDCs produce large amounts of type 1 interferon in response to foreign nucleic acid and crucially contribute to host defense against viral infection. Tumor necrosis factor (TNF) receptor-associated factor 5 (TRAF5) is a pivotal component of various TNF receptor signaling pathways in the immune system. Although the functions of TRAF5 in T and B lymphocytes have been well studied, its roles in pDCs remains to be fully elucidated. In this study, we show that the expression of TRAF5 supports the generation of pDCs in the bone marrow and also critically contributes to the homeostasis of the pDC subset in the periphery in a cell-intrinsic manner. Furthermore, we provide evidence that TRAF5 promotes the commitment of DC precursor cells toward pDC versus cDC subsets, which is regulated by the balance of transcription factors TCF4 and ID2. Together our findings reveal that TRAF5 acts as a positive regulator of pDC differentiation from bone marrow progenitors.


Subject(s)
Bone Marrow Cells/cytology , Dendritic Cells/cytology , Stem Cells/cytology , TNF Receptor-Associated Factor 5/physiology , Animals , Bone Marrow , Cell Differentiation , Cells, Cultured , Humans , Inhibitor of Differentiation Protein 2/physiology , Transcription Factor 4/physiology , Transcription Factors/physiology
15.
J Immunol ; 201(8): 2264-2272, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30209188

ABSTRACT

Bmi1 is a polycomb group protein and regulator that stabilizes the ubiquitination complex PRC1 in the nucleus with no evidently direct link to the NF-κB pathway. In this study, we report a novel function of Bmi1: its regulation of IκBα ubiquitination in the cytoplasm. A deficiency of Bmi1 inhibited NF-κB-mediated gene expression in vitro and a NF-κB-mediated mouse model of arthritis in vivo. Mechanistic analysis showed that Bmi1 associated with the SCF ubiquitination complex via its N terminus and with phosphorylation by an IKKα/ß-dependent pathway, leading to the ubiquitination of IκBα. These effects on NF-κB-related inflammation suggest Bmi1 in the SCF complex is a potential therapeutic target for various diseases and disorders, including autoimmune diseases.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Cytoplasm/metabolism , Endothelial Cells/physiology , Multiprotein Complexes/metabolism , NF-KappaB Inhibitor alpha/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Multiprotein Complexes/genetics , NF-kappa B/metabolism , Polycomb Repressive Complex 1/genetics , Protein Binding , Protein Stability , Proteolysis , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , SKP Cullin F-Box Protein Ligases/genetics , Transcriptional Activation , Ubiquitination
16.
Int Immunol ; 30(7): 291-299, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29668931

ABSTRACT

Tumor necrosis factor receptor-associated factor 2 (TRAF2) and TRAF5 constitutively bind to glycoprotein 130 kDa (gp130) and inhibit IL-6-driven activation of signal transducer and activator of transcription 3 (STAT3) in CD4+ T cells, which limits the differentiation of pro-inflammatory IL-17-producing helper T cells that require IL-6-receptor (IL-6R) signals for their development. However, it is not known how the interaction between TRAF and gp130 negatively regulates STAT3 activity in the IL-6R complex. We hypothesized that TRAF proteins associated with gp130 might limit the activation of Janus kinase that is needed for the activation of STAT3. To test this, we transfected HEK293T cells to express gp130 and TRAF2 or TRAF5 together with two chimeric JAK1 proteins combined with either the N-terminal or the C-terminal protein fragment of firefly luciferase. Using this luciferase fragment complementation system, we found that the recovery of luciferase enzyme activity was coincident with proximal JAK1-JAK1 interaction and phosphorylation of JAK1 in the IL-6R complex and that the expression of TRAF protein significantly inhibited the recovery of luciferase activity. The binding of TRAF to gp130 via the C-terminal TRAF domain was essential for the inhibition. In accordance with this, upon stimulation of endogenous gp130 with a complex of IL-6 and IL-6R, Traf5-/- CD4+ T cells displayed significantly higher amounts of phosphorylated JAK1 than did their wild-type counterparts. Therefore, our results demonstrate that gp130-associated TRAF2 and TRAF5 inhibit the interaction between two JAK proteins in the IL-6R complex that is essential for initiating the JAK-STAT signaling pathway.


Subject(s)
Cytokine Receptor gp130/metabolism , Interleukin-6/metabolism , Janus Kinase 1/metabolism , Signal Transduction , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 5/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Cytokine Receptor gp130/immunology , Interleukin-6/immunology , Janus Kinase 1/immunology , Mice , Phosphorylation , Protein Binding , TNF Receptor-Associated Factor 2/immunology , TNF Receptor-Associated Factor 5/immunology
17.
Adv Exp Med Biol ; 1189: 53-84, 2019.
Article in English | MEDLINE | ID: mdl-31758531

ABSTRACT

Costimulatory signals initiated by the interaction between the tumor necrosis factor (TNF) ligand and cognate TNF receptor (TNFR) superfamilies promote clonal expansion, differentiation, and survival of antigen-primed CD4+ and CD8+ T cells and have a pivotal role in T-cell-mediated adaptive immunity and diseases. Accumulating evidence in recent years indicates that costimulatory signals via the subset of the TNFR superfamily molecules, OX40 (TNFRSF4), 4-1BB (TNFRSF9), CD27, DR3 (TNFRSF25), CD30 (TNFRSF8), GITR (TNFRSF18), TNFR2 (TNFRSF1B), and HVEM (TNFRSF14), which are constitutive or inducible on T cells, play important roles in protective immunity, inflammatory and autoimmune diseases, and tumor immunotherapy. In this chapter, we will summarize the findings of recent studies on these TNFR family of co-signaling molecules regarding their function at various stages of the T-cell response in the context of infection, inflammation, and cancer. We will also discuss how these TNFR co-signals are critical for immune regulation and have therapeutic potential for the treatment of T-cell-mediated diseases.


Subject(s)
Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , T-Lymphocytes/cytology , Tumor Necrosis Factor-alpha/metabolism , Humans , Immunotherapy , Lymphocyte Activation , Neoplasms
18.
J Biol Chem ; 292(44): 18098-18112, 2017 11 03.
Article in English | MEDLINE | ID: mdl-28916727

ABSTRACT

Pulmonary alveolar proteinosis (PAP) is a severe respiratory disease characterized by dyspnea caused by accumulation of surfactant protein. Dysfunction of alveolar macrophages (AMs), which regulate the homeostasis of surfactant protein, leads to the development of PAP; for example, in mice lacking BTB and CNC homology 2 (Bach2). However, how Bach2 helps prevent PAP is unknown, and the cell-specific effects of Bach2 are undefined. Using mice lacking Bach2 in specific cell types, we found that the PAP phenotype of Bach2-deficient mice is due to Bach2 deficiency in more than two types of immune cells. Depletion of hyperactivated T cells in Bach2-deficient mice restored normal function of AMs and ameliorated PAP. We also found that, in Bach2-deficient mice, hyperactivated T cells induced gene expression patterns that are specific to other tissue-resident macrophages and dendritic cells. Moreover, Bach2-deficient AMs exhibited a reduction in cell cycle progression. IFN-γ released from T cells induced Bach2 expression in AMs, in which Bach2 then bound to regulatory regions of inflammation-associated genes in myeloid cells. Of note, in AMs, Bach2 restricted aberrant responses to excessive T cell-induced inflammation, whereas, in T cells, Bach2 puts a brake on T cell activation. Moreover, Bach2 stimulated the expression of multiple histone genes in AMs, suggesting a role of Bach2 in proper histone expression. We conclude that Bach2 is critical for the maintenance of AM identity and self-renewal in inflammatory environments. Treatments targeting T cells may offer new therapeutic strategies for managing secondary PAP.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Dendritic Cells/immunology , Gene Expression Regulation , Immunity, Innate , Lung/immunology , Macrophages, Alveolar/immunology , Pulmonary Alveolar Proteinosis/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Biomarkers/metabolism , Cell Lineage , Cells, Cultured , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Gene Expression Profiling , Heterozygote , Lung/metabolism , Lung/pathology , Lymphocyte Activation , Lymphocyte Depletion , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Mice, Knockout , Mice, Transgenic , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Alveolar Proteinosis/pathology , Specific Pathogen-Free Organisms , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
19.
Eur J Immunol ; 47(2): 305-313, 2017 02.
Article in English | MEDLINE | ID: mdl-27861804

ABSTRACT

Dendritic cells (DCs) in lymphoid and non-lymphoid tissues are professional antigen-presenting cells that are essential for effective immunity and tolerance. However, the presence and characteristics of DCs in steady-state salivary glands (SGs) currently remain unknown. We herein identified CD64- CD11c+ classical DCs (cDCs) as well as CD64+ macrophages among CD45+ MHC class II+ antigen-presenting cells in steady-state murine SGs. SG cDCs were divided into CD103+ CD11b- and CD103- CD11b+ cDCs. CD103+ CD11b- cDCs expressed XCR1, CLEC9A, and interferon regulatory factor 8, whereas CD103- CD11b+ cDCs strongly expressed CD172a. Both cDC subsets in SGs markedly expanded in response to the Flt3 ligand (Flt3L), were replenished by bone marrow-derived precursors, and differentiated from common DC precursors, but not monocytes. Furthermore, ovalbumin-pulsed SG CD103+ CD11b- cDCs induced the proliferation of naïve ovalbumin-specific CD8+ T cells and production of interferon-γ from proliferating T cells. SG CD103+ CD11b- cDCs expanded by Flt3L in vivo exhibited the same properties. These results indicate that bona fide cDCs reside in steady-state murine SGs and cDCs with the CD103+ CD11b- phenotype possess antigen cross-presenting capacity. Moreover, Flt3L enhances protective immunity by expanding cDCs. Taken together, SG cDCs might play an important role in maintaining immune homeostasis in the tissues.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/physiology , Macrophages/physiology , Salivary Glands/cytology , Animals , Antigens, CD/metabolism , CD11b Antigen/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cross-Priming , Female , Integrin alpha Chains/metabolism , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Receptors, IgG/metabolism , fms-Like Tyrosine Kinase 3/immunology
20.
Biochem Biophys Res Commun ; 499(3): 544-550, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29596835

ABSTRACT

Oncostatin M (OSM) is involved in pathogenesis of several human inflammatory diseases including lung inflammation and fibrosis. Although accumulating evidence indicates that OSM mediates lung inflammation, the precise mechanism for OSM on lung inflammation still remains unclear. In this study, we found that OSM receptor was abundantly expressed on endothelial and stromal/fibroblast cells in the lung of mice. In vitro stimulation with OSM upregulated vascular cell adhesion molecule-1 (VCAM-1), which promotes eosinophil infiltration in the lung tissues, on freshly-isolated lung stromal/fibroblast cells from wild-type mice. However, these cells from TNF receptor associated factor 5 (TRAF5)-deficient mice failed to show the increase in VCAM-1 expression after OSM stimulation. Furthermore, Traf5-/- mice showed markedly attenuated lung inflammation in terms of eosinophil infiltration upon intranasal administration with OSM as compared to wild-type mice. These results indicate that TRAF5 is crucially involved in OSM-mediated lung inflammation probably by inducing lung stromal/fibroblast cell activation.


Subject(s)
Oncostatin M/metabolism , Pneumonia/metabolism , Pneumonia/pathology , TNF Receptor-Associated Factor 5/metabolism , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Eosinophils/metabolism , Eosinophils/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Interleukin-6/metabolism , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL