Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Mol Immunol ; 19(1): 79-91, 2022 01.
Article in English | MEDLINE | ID: mdl-34782759

ABSTRACT

OBJECTIVE: The interleukin (IL)-12 cytokine family is closely related to the development of T helper cells, which are responsible for autoimmune disease enhancement or suppression. IL-12 family members are generally heterodimers and share three α-subunits (p35, p19, and p28) and two ß-subunits (p40 and EBI3). However, a ß-sheet p40 homodimer has been shown to exist and antagonize IL-12 and IL-23 signaling 1. Therefore, we assumed the existence of a p40-EBI3 heterodimer in nature and sought to investigate its role in immune regulation. METHODS: The presence of the p40-EBI3 heterodimer was confirmed by ELISA, immunoprecipitation, and western blotting. A p40-EBI3 vector and p40-EBI3-Fc protein were synthesized to confirm the immunological role of this protein in mice with collagen-induced arthritis (CIA). The anti-inflammatory effects of p40-EBI3 were analyzed with regard to clinical, histological, and immune cell-regulating features in mice with CIA. RESULTS: Clinical arthritis scores and the expression levels of proinflammatory cytokines (e.g., IL-17, IL-1ß, IL-6, and TNF-α) were significantly attenuated in p40-EBI3-overexpressing and p40-EBI3-Fc-treated mice with CIA compared to vehicle-treated mice with CIA. Structural joint damage and vessel formation-related gene expression were also reduced by p40-EBI3 heterodimer treatment. In vitro, the p40-EBI3-Fc protein significantly suppressed the differentiation of Th17 cells and reciprocally induced CD4+CD25+Foxp3+ (regulatory T) cells. p40-EBI3 also inhibited osteoclast formation in a concentration-dependent manner. CONCLUSION: In this study, p40-EBI3 ameliorated proinflammatory conditions both in vivo and in vitro. We propose that p40-EBI3 is a novel anti-inflammatory cytokine involved in suppressing the immune response through the expansion of Treg cells and suppression of Th17 cells and osteoclastogenesis.


Subject(s)
Arthritis, Experimental , Autoimmune Diseases , Interleukin-12 , Animals , Cytokines/therapeutic use , Interleukin-12/chemistry , Interleukin-12/metabolism , Mice , Minor Histocompatibility Antigens , Receptors, Cytokine/genetics , Receptors, Cytokine/therapeutic use , T-Lymphocytes, Regulatory , Th17 Cells
2.
Sci Rep ; 9(1): 13706, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31548569

ABSTRACT

Human Oncostatin M (OSM), initially discovered as a tumour inhibitory factor secreted from U-937 cells, is a gp130 (IL-6/LIF) cytokine family member that exhibits pleiotropic effects in inflammation, haematopoiesis, skeletal tissue alteration, liver regeneration, cardiovascular and metabolic diseases. Cytoplasmic expression of OSM in Escherichia coli results in inclusion bodies, and complex solubilisation, refolding and purification is required to prepare bioactive protein. Herein, eight N-terminal fusion variants of OSM with hexahistidine (His6) tag and seven solubility-enhancing tags, including thioredoxin (Trx), small ubiquitin-related modifier (Sumo), glutathione S-transferase (GST), maltose-binding protein (MBP), N-utilisation substance protein A (Nusa), human protein disulphide isomerase (PDI) and the b'a' domain of PDI (PDIb'a'), were tested for soluble OSM expression in E. coli. The His6-OSM plasmid was also introduced into genetically engineered Origami 2 and SHuffle strains to test expression of the protein. At 18 °C, MBP-tagged OSM was highly expressed and solubility was dramatically enhanced. In addition, His6-OSM was more highly expressed and soluble in Origami 2 and SHuffle strains than in BL21(DE3). MBP-OSM and His6-OSM were purified more than 95% with yields of 11.02 mg and 3.27 mg from a 500 mL culture. Protein identity was confirmed by mass spectroscopy, and bioactivity was demonstrated by in vitro inhibition of Th17 cell differentiation.


Subject(s)
Oncostatin M/metabolism , Recombinant Fusion Proteins/metabolism , Escherichia coli , Gene Expression , Genetic Engineering , Histidine , Humans , Maltose-Binding Proteins/metabolism , Oligopeptides , Oncostatin M/genetics , Recombinant Fusion Proteins/genetics , Solubility
3.
Sci Rep ; 9(1): 5227, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30914691

ABSTRACT

Systemic lupus erythematosus (SLE) is mediated by a chronic and dysregulated inflammatory response. Interleukin (IL)-17, a proinflammatory cytokine, and T helper (Th)17 cells are associated with chronic autoimmune diseases. We hypothesized that inhibition of IL-17 would decrease the numbers of T cell subsets that function as B-cell helpers, as well as B-cell differentiation into plasma cells and autoantibody expression. The IL-17 level was increased markedly in Roquinsan/san mice. Loss of IL-17 in Roquinsan/san mice improved nephritis by downregulating immunoglobulin (Ig)G, IgG1, and IgG2a production. Formation of germinal centers (GCs), and follicular B- and T-cell differentiation was reduced, whereas the number of regulatory T (Treg) cells and immature B cells was increased, by IL-17 deficiency in Roquinsan/san mice. These results suggest that IL-17 inhibition can ameliorate SLE by inhibiting B-cell differentiation into GCs. Therefore, IL-17-producing Th17 cells show promise as a target for development of novel therapeutics for SLE.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Germinal Center/immunology , Interleukin-17/immunology , Lupus Nephritis/immunology , Plasma Cells/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , B-Lymphocytes, Regulatory/pathology , Germinal Center/pathology , Immunoglobulin G/immunology , Interleukin-17/genetics , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Mice , Mice, Knockout , Plasma Cells/pathology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
4.
J Transl Med ; 17(1): 18, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30630513

ABSTRACT

BACKGROUND: Spondyloarthritis (SpA) is chronic inflammatory arthritis, and interleukin (IL)-17 is crucial in SpA pathogenesis. Type 17 helper T (Th17) cells are one of major IL-17-secreting cells. Signal transducer and activator of transcription (STAT)-3 signaling induces Th17 differentiation. This study investigated the effects of protein inhibitor of activated STAT3 (PIAS3) on SpA pathogenesis. Curdlan was injected into SKG ZAP-70W163C mice for SpA induction. METHODS: The PIAS3 or Mock vector was inserted into mice for 10 weeks. Clinical and histologic scores of the paw, spine, and gut were evaluated. The expression of IL-17, tumor necrosis factor-α (TNF-α), STAT3, and bone morphogenic protein (BMP) was measured. Confocal microscopy and flow cytometry were used to assess Th cell differentiation. RESULTS: PIAS3 significantly diminished the histologic scores of the paw and gut. PIAS3-treated mice displayed decreased expression of IL-17, TNF-α, and STAT3 in the paw, spine, and gut. BMP-2/4 expression was lower in the spines of PIAS3-treated mice. Th cell differentiation was polarized toward the upregulation of regulatory T cells (Tregs) and the downregulation of Th17 in PIAS3-treated mice. CONCLUSION: PIAS3 had beneficial effects in mice with SpA by reducing peripheral arthritis and gut inflammation. Pro-inflammatory cytokines and Th17/Treg differentiation were controlled by PIAS3. In addition, BMPs were decreased in the spines of PIAS3-treated mice. These findings suggest that PIAS3 could have therapeutic benefits in patients with SpA.


Subject(s)
Gastrointestinal Tract/pathology , Inflammation/metabolism , Protein Inhibitors of Activated STAT/metabolism , Signal Transduction , Spondylarthritis/immunology , Spondylarthritis/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Mice, Inbred BALB C , STAT3 Transcription Factor/metabolism , Spleen/pathology
6.
Immunol Lett ; 201: 45-51, 2018 09.
Article in English | MEDLINE | ID: mdl-30395870

ABSTRACT

Inflammatory bowel disease (IBD) is caused by chronic inflammation of the gastrointestinal tract. The pathogenesis of IBD remains unclear. The inflammation is associated with activation of T helper (Th) lymphocytes and chronic production of inflammatory cytokines. Ro60 suppresses the expression of tumor necrosis factor α, interleukin (IL)-6, and interferon α by inhibiting Alu transcription; control of Ro60 mRNA expression may thus be therapeutically useful. However, few studies have evaluated the anti-inflammatory activity of Ro60. The Ro60 level is decreased in IBD patients; we thus hypothesized that Ro60 was involved in the development of this autoimmune disease. We subjected mice with dextran sodium sulfate (DSS)-induced colitis to gene therapy using a vector that overexpressed Ro60 threefold. We scored IBD progression by repeatedly weighing the mice. Ro60 ameliorated colitis severity and reduced the levels of tumor necrosis factor α, IL-6, IL-17, IL-8, and vascular endothelial growth factor. Ro60 overexpression decreased the levels of α-smooth muscle actin (a marker of activated myofibroblasts) and type I collagen. The anti-inflammatory and anti-fibrotic activities of Ro60 ameliorated the severity of DSS-induced colitis in mice by repressing inflammation, fibrosis, angiogenesis, and the production of reactive oxygen species.


Subject(s)
Autoantigens/metabolism , Colitis/immunology , Colon/pathology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , RNA, Small Cytoplasmic/metabolism , Ribonucleoproteins/metabolism , Actins/metabolism , Animals , Autoantigens/genetics , Colitis/therapy , Collagen Type I/metabolism , Colon/physiology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Fibrosis , Genetic Therapy , Genetic Vectors , Humans , Inflammation/therapy , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/therapy , Mice , Mice, Inbred C57BL , RNA, Small Cytoplasmic/genetics , Ribonucleoproteins/genetics
7.
Immunol Lett ; 203: 62-69, 2018 11.
Article in English | MEDLINE | ID: mdl-30240636

ABSTRACT

Cucurbitacin E (CuE) is a biochemical compound found in plants that are members of the family CuE has been studied for its roles in anti-inflammation and the inhibition of angiogenesis as well as for its properties as an antioxidant. CuE is a new agent that was identified as a selective inhibitor of the signal transducer and activator of transcription 3 (STAT3)-related pathway. STAT3, a pivotal transcription factor for Th17 differentiation, is critical for T cell alloactivation in acute graft-versus-host disease (aGvHD). We investigated whether CuE attenuates the development of aGvHD through the suppression of Th17 cells. The alloreactive proliferation of mouse and human T cells was reduced by CuE treatment. CuE also decreased pro-inflammatory cytokines, such as IL-17 and IFN-γ, in alloreactive T cells. STAT3-responsive and IL-17A-promoter activities were also suppressed by CuE treatment, confirming that activated STAT3 was decreased by CuE treatment. To construct an aGvHD-induced mouse line, splenocytes and bone marrow cells from C57BL/6 mice were transplanted into BALB/c mice with complete mis-matched major histocompatibility complex molecules. CuE was administered to aGvHD animals 3 days per week via intraperitoneal injection. CuE attenuated the severity of aGvHD disease-related scores compared to the vehicle group. CuE inhibited skin inflammation and fibrosis, as evidenced by the expression of α-Sma and Col-I in aGvHD mice compared to the vehicle group. Additionally, aGvHD mice treated with CuE showed improved histopathological features in the small and large intestines, whereas the vehicle group showed collapsed villi in the small intestine and cryptic structures in the large intestine. We also observed a marked reduction of pro-inflammatory cytokines in the intestinal tissue. Collectively, our data suggest that CuE could serve as a therapeutic agent for patients with aGvHD.


Subject(s)
Graft vs Host Disease , STAT3 Transcription Factor/immunology , Th17 Cells , Triterpenes/pharmacology , Acute Disease , Animals , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Humans , Mice , Mice, Inbred BALB C , Th17 Cells/immunology , Th17 Cells/pathology
8.
J Med Food ; 21(8): 745-754, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30110204

ABSTRACT

Notoginseng Radix and Rehmanniae Radix Preparata have been widely used traditionally for treating inflammatory diseases. This research studies the therapeutic effects of YH23537, the extracts of Notoginseng Radix and Rehmanniae Radix Preparata, on pain and cartilage degeneration in an experimental osteoarthritis (OA) model. Male Wistar rats were inoculated intra-articularly with 3 mg of monosodium iodoacetate (MIA) in the right intra-articular. Four days later, the animals were administrated orally with YH23537 daily for 24 days. Tactile allodynia and weight bearing were measured. Macroscopic and microscopic observations for articular cartilage were performed at the end of the experiment. Protein expression in the joint was determined by immunohistochemistry. The effects of YH23537 on mRNA levels in chondrocytes stimulated with interleukin (IL)-1ß were analyzed using random polymerase chain reaction. OA induction was confirmed by significant decrease of paw withdrawal latency, paw withdrawal threshold, and weight bearing compared with the normal group at 3 days after MIA injection. The YH23537-treated groups displayed significant increases in pain thresholds and weight bearing throughout the observation period. The damage to articular cartilage was significantly lessened visually and histopathologically by YH23537 treatment. YH23537 suppressed the expression of metalloproteinase-3, nitrotyrosine, IL-1ß and IL-6 increased in OA joints. YH23537 upregulated tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-3 in IL-1ß-stimulated human OA chondrocytes. The protein levels of the NF-κBp65 and HIF-2α in the joint tissues were reduced by YH23537. YH23537 exerted antinociceptive effects and cartilage protective effects in experimental OA rats by suppressing oxidative injury, inflammatory mediators, and inducing anabolic factors. We suggest that YH23537 may have efficacy for treating OA in humans.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cartilage Diseases/drug therapy , Osteoarthritis/drug therapy , Pain/drug therapy , Panax , Plant Extracts/pharmacology , Rehmannia , Administration, Oral , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/therapeutic use , Cartilage Diseases/chemically induced , Cartilage, Articular/drug effects , Disease Models, Animal , Iodoacetates , Male , Osteoarthritis/chemically induced , Pain Measurement , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Rats , Rats, Wistar
9.
Immunol Lett ; 197: 63-69, 2018 05.
Article in English | MEDLINE | ID: mdl-29545108

ABSTRACT

Yin Yang 1 (YY1) is a ubiquitously expressed transcription factor that functions in cooperation with various cofactors to regulate gene expression. In the immune system, YY1 enhances cytokine production and T helper (Th) 2 effector cell differentiation, resulting in the activation of inflammation. However, no studies have reported the role of YY1 in Th17 cell regulation, which is implicated in rheumatoid arthritis (RA). We investigated the expression of YY1 in Th17 cells in vitro and revealed increased levels of YY1 mRNA and protein. To elucidate the function of YY1 pathogenesis in RA, we used a collagen-induced arthritis (CIA) mouse model with YY1 deficiency. Deficiency of YY1 reduced the severity of arthritis and joint destruction. Moreover, Th17 cells were dramatically reduced in YY1-deficient mice. The cytokine interleukin (IL)-17 was decreased in YY1-deficient CD4+ T cells ex vivo and in vivo. Interestingly, the level of signal transducer and activator of transcription 3 (STAT3), tumor necrosis factor-α, IL-17, IL-6, and IL-1ß were markedly decreased in YY1-deficient mice with CIA. The cytokine-inducing function of YY1 was more specific to IL-17 than to interferon-γ. YY1 plays a role in Th17 cell differentiation and RA pathogenesis. Our findings suggest that future RA therapies should target the regulatory mechanism involved in Th17 cell differentiation, in which YY1 may cooperate with the STAT3 signaling pathway.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Inflammation/immunology , Joints/immunology , Th17 Cells/immunology , Th2 Cells/immunology , YY1 Transcription Factor/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Humans , Immunomodulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT3 Transcription Factor/metabolism , YY1 Transcription Factor/genetics
10.
J Med Food ; 21(1): 39-46, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29346060

ABSTRACT

Probiotic complex, zinc, and coenzyme Q10 (CoQ10) are recognized dietary supplements with an anti-inflammatory role. Although these supplementations are individually known to benefit rheumatoid arthritis (RA), there is no evidence suggesting any synergic effect. The primary goal of this study is to determine whether probiotic complex, zinc, and CoQ10 attenuate the development of collagen-induced arthritis (CIA). The combination of probiotic complex, zinc, and CoQ10 reduced CIA severity by downregulating the levels of IgG, IgG1, and IgG2a in serum. Joint inflammation, bone destruction, and cartilage damage were also improved by the complex. There was a decrease in the expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, IL-17, and vascular endothelial growth factor (VEGF) in the joint synovium. The balance between helper T 17 (Th17) cells and regulatory T (Treg) cells was shown to be controlled reciprocally by the complex. These findings suggest that the combination of probiotic complex, zinc, and CoQ10 can ameliorate the development of CIA by inhibiting the expression of proinflammatory cytokines, and is thus an important therapeutic candidate for RA treatment.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Probiotics/administration & dosage , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Ubiquinone/analogs & derivatives , Zinc/administration & dosage , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Disease Models, Animal , Drug Therapy, Combination , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Mice, Inbred DBA , Th17 Cells/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Ubiquinone/administration & dosage
12.
Sci Rep ; 7(1): 5506, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28710354

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) orchestrates the differentiation of several cell types, including interleukin-17 (IL-17)-releasing Th17 cells. Dysregulation of Th17 cells results in chronic inflammatory responses. Ssu72 is a C-terminal domain phosphatase required for transcriptional regulation. However, the mechanism by which Ssu72 affects STAT3 activation and Th17 cell differentiation is unclear. Here, we found that Ssu72 overexpression suppresses STAT3 activation and Th17 cell responses in vitro. A systemic infusion of Ssu72 attenuates experimental autoimmune arthritis by reducing STAT3 activity and the differentiation of Th17 cells. It also reduces joint destruction, serum immunoglobulin concentrations and osteoclastogenesis but increases the number of marginal zone B cells and B10 cells. These effects are associated with reduced p-STAT3 levels and the suppression of Th17 cell formation in vivo. Based on these data, Ssu72 is related to STAT3 activation and the inflammatory response; and Ssu72 overexpression in T-cell-mediated immunity has potential utility for the treatment of autoimmune arthritis.


Subject(s)
Arthritis, Rheumatoid/therapy , Genetic Vectors/administration & dosage , Phosphoprotein Phosphatases/genetics , STAT3 Transcription Factor/metabolism , Th17 Cells/immunology , Animals , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Cell Differentiation/drug effects , Collagen/adverse effects , Disease Models, Animal , Genetic Vectors/pharmacology , Lymphocyte Activation , Male , Mice , NIH 3T3 Cells , Phosphoprotein Phosphatases/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects
13.
Immunol Lett ; 190: 26-33, 2017 10.
Article in English | MEDLINE | ID: mdl-28728856

ABSTRACT

Achaete-scute complex homologue 2 (Ascl2) has been reported to induce the differentiation and activation of follicular helper T (TFH) cells, which are essential for development of Sjögren's syndrome (SS). This study examined whether Ascl2 plays a role in the development of SS. NOD/ShiLtJ mice were injected with an Ascl2-overexpression vector, and the infiltration of lymphocytes into salivary and lacrimal glands was assessed. The expression of inflammatory cytokines and chemoattractants for T or B cells was measured. The activation of TFH cells was assessed using a specific marker of TFH cells. Ascl2 level was also measured in SS patients. Overexpression of Ascl2 increased the expression of C-X-C chemokine receptor type 5 (CXCR5) in both salivary and lacrimal glands (p<0.0001). Overexpression of Ascl2 also increased the expression of proinflammatory cytokines and chemoattractants including interleukin 6 (IL-6), tumor necrosis factor-α, IL-8, programmed cell death 1 (PD-1), IL-21, and B-cell lymphoma 6 (Bcl-6). Overexpression of Ascl2 increased the populations of CD4+CXCR5+, CD4+ICOS+, and CD4+PD-1+ cells. The Ascl2 level was higher in peripheral blood mononuclear cells from SS patients compared with those from healthy controls. Our findings suggest that Ascl2 may play a role in the development and progression of SS and may be a therapeutic target in the treatment of SS.


Subject(s)
B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Lacrimal Apparatus/metabolism , Salivary Glands/metabolism , Sjogren's Syndrome/genetics , T-Lymphocytes, Helper-Inducer/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Germinal Center/immunology , Humans , Mice , Mice, Inbred NOD , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, CXCR5/metabolism
15.
J Immunol ; 198(7): 2661-2670, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28242651

ABSTRACT

Circulating autoantibodies and immune complex deposition are pathological hallmarks of systemic lupus erythematosus (SLE). B cell differentiation into plasma cells (PCs) and some T cell subsets that function as B cell helpers can be therapeutic targets of SLE. Mechanistic target of rapamycin (mTOR) signaling is implicated in the formation of B cells and germinal centers (GCs). We assessed the effect of metformin, which inhibits mTOR, on the development of autoimmunity using Roquinsan/san mice. Oral administration of metformin inhibited the formation of splenic follicles and inflammation in kidney and liver tissues. It also decreased serum levels of anti-dsDNA Abs without affecting serum glucose levels. Moreover, metformin inhibited CD21highCD23low marginal zone B cells, B220+GL7+ GC B cells, B220-CD138+ PCs, and GC formation. A significant reduction in ICOS+ follicular helper T cells was found in the spleens of the metformin-treated group compared with the vehicle-treated group. In addition, metformin inhibited Th17 cells and induced regulatory T cells. These alterations in B and T cell subsets by metformin were associated with enhanced AMPK expression and inhibition of mTOR-STAT3 signaling. Furthermore, metformin induced p53 and NF erythroid-2-related factor-2 activity in splenic CD4+ T cells. Taken together, metformin-induced alterations in AMPK-mTOR-STAT3 signaling may have therapeutic value in SLE by inhibiting B cell differentiation into PCs and GCs.


Subject(s)
Autoimmunity/drug effects , Cell Differentiation/drug effects , Hypoglycemic Agents/pharmacology , Lupus Erythematosus, Systemic/immunology , Metformin/pharmacology , Plasma Cells/drug effects , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/immunology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Blotting, Western , Cell Differentiation/immunology , Cell Separation , Disease Models, Animal , Flow Cytometry , Germinal Center/drug effects , Germinal Center/immunology , Immunohistochemistry , Male , Mice , Microscopy, Confocal , Plasma Cells/immunology , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/immunology , Ubiquitin-Protein Ligases/deficiency
16.
J Immunol ; 198(4): 1484-1491, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28093521

ABSTRACT

Oncostatin M (OSM) is a pleiotropic cytokine and a member of the IL-6 family. It has both proinflammatory and anti-inflammatory functions and is involved in the activation of STAT3 and STAT5. Rheumatoid arthritis is an autoimmune disease that causes chronic and excessive inflammation. Rheumatoid arthritis can lead to induction of Th17 cells, which express IL-17. The aim of this study was to measure the effects of OSM on the proliferation of regulatory T cells and Th17 cells from mice. IL-2 immune complex suppressed the development of collagen-induced arthritis in mice and altered the regulatory T/Th17 cell balance by increasing OSM expression. OSM mitigated the proliferation of Th17 cells and decreased the expression of IL-17 and IL-21. It promoted the activation of suppressor of cytokine signaling 3 (SOCS3), STAT3, and STAT5. Inhibition of SOCS3, STAT3, and STAT5 lessened the OSM-induced reduction in proliferation of Th17 cells. These observations suggest that OSM can inhibit Th17 differentiation by reciprocally controlling SOCS3, STAT3, and STAT5.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-17/genetics , Oncostatin M/physiology , Suppressor of Cytokine Signaling 3 Protein/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental , CD4-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Collagen/administration & dosage , Down-Regulation , Gene Expression Regulation , Interleukin-17/immunology , Interleukin-2/immunology , Interleukins/genetics , Interleukins/immunology , Mice , Oncostatin M/genetics , Oncostatin M/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein/immunology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects
17.
Front Immunol ; 8: 1793, 2017.
Article in English | MEDLINE | ID: mdl-29326694

ABSTRACT

Dysfunction of T helper 17 (Th17) cells leads to chronic inflammatory disorders. Signal transducer and activator of transcription 3 (STAT3) orchestrates the expression of proinflammatory cytokines and pathogenic cell differentiation from interleukin (IL)-17-producing Th17 cells. However, the pathways mediated by STAT3 signaling are not fully understood. Here, we observed that Fos-related antigen 1 (FRA1) and JUNB are directly involved in STAT3 binding to sites in the promoters of Fosl1 and Junb. Promoter binding increased expression of IL-17 and the development of Th17 cells. Overexpression of Fra1 and Junb in mice resulted in susceptibility to collagen-induced arthritis and an increase in Th17 cell numbers and inflammatory cytokine production. In patients with rheumatoid arthritis, FRA1 and JUNB were colocalized with STAT3 in the inflamed synovium. These observations suggest that FRA1 and JUNB are associated closely with STAT3 activation, and that this activation leads to Th17 cell differentiation in autoimmune diseases and inflammation.

18.
Sci Rep ; 6: 39393, 2016 12 23.
Article in English | MEDLINE | ID: mdl-28008946

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease that is related to the induction of T helper (Th)17 cells, which secrete interleukin-17, and activation of the signal transducer and activator of transcription (STAT) 3. The expression of high-temperature requirement protein A (HtrA) 2, a serine protease involved in apoptosis, was decreased in RA patients nonresponsive to drug treatment of RA. The aim of this study was to determine whether overexpression of HtrA2 has a therapeutic effect on RA. Th17 differentiation, osteoclastogenesis, and lymphocyte activation are increased in motor neuron degeneration (mnd)2 mice, which lack HtrA2 activity because of a missense mutation (Ser276Cys) in the protease domain of HtrA2. The inhibitor of HtrA2 also increased Th17 differentiation. On the other hand, HtrA2 induced cleavage of STAT3 and overexpression of HtrA2 attenuated CIA in a mouse model. HtrA2 overexpression inhibited plaque development as well as the differentiation of Th17 in ApoE-/- mice after immunization with proteoglycans to induce a hyperlipidemia-based RA animal model. The therapeutic function of HtrA2 in inflammatory diseases is linked with Th17 development and the STAT3 pathway in splenocytes. These results suggest that HtrA2 participates in immunomodulatory activity where the upregulation of HtrA2 may shed light on therapeutic approaches to RA and hyperlipidemia.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , High-Temperature Requirement A Serine Peptidase 2/genetics , STAT3 Transcription Factor/genetics , Animals , Autoimmune Diseases , Cell Differentiation/genetics , Disease Models, Animal , Hyperlipidemias/genetics , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Motor Neurons/metabolism , Mutation, Missense/genetics , Proteoglycans/genetics , Th17 Cells/metabolism , Up-Regulation/genetics
19.
Sci Rep ; 6: 34617, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708408

ABSTRACT

PTEN is a tyrosine phosphatase with significant function in inhibiting STAT3 activation. Recently, inactivation of STAT3 has been demonstrated as a therapeutic candidate for autoimmune arthritis. The expression of PTEN controlled by p53 regulates autoimmune arthritis through modulating the balance between Th17 and Treg. We hypothesized that PTEN regulated by p53 might reduce CIA severity and inflammatory response via inhibiting STAT3 activation. Our results revealed that PTEN could ameliorate experimental autoimmune arthritis by reducing STAT3 activity and Th17 differentiation. Systemic infusion of PTEN overexpression downregulated CIA severity. In addition, PTEN overexpression decreased the activation of T cells and modulated reciprocal differentiation of Th17 and Treg cells. We observed that PTEN expression downregulated by p53 deficiency induced the activation of STAT3. Loss of p53 exacerbated autoimmune arthritis and dysregulated the population of Th17 and Treg. These data suggest that induction of STAT3-modulatory activity of PTEN may be a therapeutic target for rheumatoid arthritis therapy.


Subject(s)
Arthritis, Experimental/genetics , PTEN Phosphohydrolase/genetics , STAT3 Transcription Factor/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Tumor Suppressor Protein p53/genetics , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cell Differentiation , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , PTEN Phosphohydrolase/immunology , Plasmids/administration & dosage , Plasmids/chemistry , Plasmids/metabolism , STAT3 Transcription Factor/immunology , Severity of Illness Index , Signal Transduction , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/immunology
20.
PLoS One ; 11(6): e0155853, 2016.
Article in English | MEDLINE | ID: mdl-27258062

ABSTRACT

DSS induced colitis is a chronic inflammatory disease characterized by inflammation in the gastrointestinal tract, which destabilizes the gut and induces an uncontrolled immune response. Although DSS induced colitis is generally thought to develop as a result of an abnormally active intestinal immune system, its pathogenesis remains unclear. Gene associated with retinoid interferon induced mortality (Grim) 19 is an endogenous specific inhibitor of STAT3, which regulates the expression of proinflammatory cytokines. In this study, we investigated the influence of GRIM19 in a DSS induced colitis mouse model. We hypothesized that Grim19 would ameliorate DSS induced colitis by altering STAT3 activity and intestinal inflammation. Grim19 ameliorated DSS induced colitis severity and protected intestinal tissue. The expression of STAT3 and proinflammatory cytokines such as IL-1ß and TNF-α in colon and lymph nodes was decreased significantly by Grim19. Moreover, DSS induced colitis progression in a Grim19 transgenic mouse line was inhibited in association with a reduction in STAT3 and IL-17 expression. These results suggest that Grim19 attenuates DSS induced colitis by suppressing the excessive inflammatory response mediated by STAT3 activation.


Subject(s)
Colitis/metabolism , Colon/metabolism , Lymph Nodes/metabolism , NADH, NADPH Oxidoreductases/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Inflammation/metabolism , Inflammation/pathology , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADH, NADPH Oxidoreductases/genetics , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...