Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 198(7): 2661-2670, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28242651

RESUMO

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.


Assuntos
Autoimunidade/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Lúpus Eritematoso Sistêmico/imunologia , Metformina/farmacologia , Plasmócitos/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/imunologia , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Western Blotting , Diferenciação Celular/imunologia , Separação Celular , Modelos Animais de Doenças , Citometria de Fluxo , Centro Germinativo/efeitos dos fármacos , Centro Germinativo/imunologia , Imuno-Histoquímica , Masculino , Camundongos , Microscopia Confocal , Plasmócitos/imunologia , Fator de Transcrição STAT3/efeitos dos fármacos , Fator de Transcrição STAT3/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/efeitos dos fármacos , Serina-Treonina Quinases TOR/imunologia , Ubiquitina-Proteína Ligases/deficiência
2.
J Immunol ; 198(4): 1484-1491, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28093521

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interleucina-17/genética , Oncostatina M/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas/genética , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Artrite Experimental , Linfócitos T CD4-Positivos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Colágeno/administração & dosagem , Regulação para Baixo , Regulação da Expressão Gênica , Interleucina-17/imunologia , Interleucina-2/imunologia , Interleucinas/genética , Interleucinas/imunologia , Camundongos , Oncostatina M/genética , Oncostatina M/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína 3 Supressora da Sinalização de Citocinas/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th17/efeitos dos fármacos
3.
Biol Blood Marrow Transplant ; 20(8): 1112-20, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24813170

RESUMO

Acute graft-versus-host disease (aGVHD) is a major cause of mortality in allogeneic bone marrow transplantation. Here, the diminishing effect of activator protein 1 (AP-1) blocking with a synthetic retinoid (SR11302) on the severity of aGVHD in a murine model was investigated. MHC-mismatched strain combinations were used in vivo: C57BL/6 (H-2k(b)) donors into lethally irradiated BALB/c (H-2k(d)) recipients. SR11302 inhibited alloreactive T cell response in a dose-dependent manner and negatively regulated signal transducer and activator of transcription 3 (STAT3) activation. AP-1 blocking in T cells inhibited the differentiation of Th1 and Th17. Conversely, Foxp3(+) regulatory T cells (Treg) population dramatically expanded. Transfer of SR11302-treated donor splenocytes into lethally irradiated recipients diminished the lethality and clinical severity of aGVHD. In line with these results, AP-1 blocking in donor splenocytes exhibited reduced Th17/Th1 population and enhanced in vivo Treg population. Beneficial Treg expanding property of SR11302 was associated with the induction of Foxp3 and STAT5 transcription factor, where the inhibiting property of Th17 was achieved by suppressing the phosphorylated form of STAT3 and enhancing SOCS3. In conclusion, the preventive potential of AP-1 inhibitor in aGVHD may be accomplished by altering CD4(+) T cell differentiation through modulating transcription factors.


Assuntos
Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas/métodos , Linfócitos T Reguladores/transplante , Fator de Transcrição AP-1/metabolismo , Condicionamento Pré-Transplante/métodos , Doença Aguda , Animais , Diferenciação Celular , Proliferação de Células , Citocinas , Humanos , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante Homólogo
5.
Mol Cells ; 25(2): 172-7, 2008 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-18414002

RESUMO

Eukaryotic translation initiation factor 5B (eIF5B) plays a role in recognition of the AUG codon in conjunction with translation factor eIF2, and promotes joining of the 60S ribosomal subunit. To see whether the eIF5B proteins of other organisms function in Saccharomyces cerevisiae, we cloned the corresponding genes from Oryza sativa, Arabidopsis thaliana, Aspergillus nidulans and Candida albican and expressed them under the control of the galactose-inducible GAL promoter in the fun12Delta strain of Saccharomyces cerevisiae. Expression of Candida albicans eIF5B complemented the slow-growth phenotype of the fun12Delta strain, but that of Aspergillus nidulance did not, despite the fact that its protein was expressed better than that of Candida albicans. The Arabidopsis thaliana protein was also not functional in Saccharomyces. These results reveal that the eIF5B in Candida albicans has a close functional relationship with that of Sacharomyces cerevisiae, as also shown by a phylogenetic analysis based on the amino acid sequences of the eIF5Bs.


Assuntos
Candida albicans/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Saccharomyces cerevisiae/metabolismo , Sequência de Aminoácidos , Candida albicans/citologia , Fatores de Iniciação em Eucariotos/química , Teste de Complementação Genética , Immunoblotting , Dados de Sequência Molecular , Fenótipo , Filogenia , Saccharomyces cerevisiae/citologia , Alinhamento de Sequência
6.
J Med Food ; 20(9): 821-829, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28816577

RESUMO

Coenzyme Q10 (CoQ10) is a powerful antioxidant substance synthesized in the body. The current study aimed to determine whether CoQ10 suppresses inflammation and inhibits p-STAT3 expression in an experimental colitis mouse model. The mice were orally fed with CoQ10 once a day for 13 days. Histological analysis of the colons was performed by immunohistochemistry. Expression of IL-17, FOXP3, p53, AMPK, and mTOR and activation of p-STAT3 and p-STAT5 in lymph node and spleen tissues were detected by confocal microscopy of stained tissue sections. The relative mRNA expression was measured with real-time PCR, and protein levels were examined by western blot. CoQ10 reduced the disease activity index score and the colon histological score. It also reduced inflammatory mediators in the colon and increased the colon length. The expression of IL-17 and p-STAT3 was decreased with CoQ10 treatment. In contrast, CoQ10 treatment increased the expression of p-AMPK and FOXP3. Expression of anti-inflammatory cytokines was shown to increase in colitis mice treated with CoQ10. These results suggested that CoQ10 may reduce the severity of colitis and suppress inflammation through the inhibition of p-STAT3 and IL-17. These results support the use of CoQ10 as a potential targeted therapy for the treatment of colitis.


Assuntos
Anti-Inflamatórios/administração & dosagem , Colite/tratamento farmacológico , Colite/imunologia , Interleucina-17/imunologia , Fator de Transcrição STAT3/imunologia , Células Th17/imunologia , Ubiquinona/análogos & derivados , Animais , Colite/genética , Modelos Animais de Doenças , Humanos , Interleucina-17/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT3/genética , Ubiquinona/administração & dosagem
7.
Tissue Eng Regen Med ; 13(2): 162-170, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30603396

RESUMO

Human palatine tonsil-derived mesenchymal stem cells (TMSCs) are known to be a new source of progenitor cells. Using waste tissue after tonsillectomy as a cell provider can be the biggest benefit of TMSCs, compared with other stem cells. The purpose of this study was to investigate tenogenic differentiation of TMSCs and to access the differential effects of transforming growth factor beta 3 (TGF-ß3) on the tenogenesis of TMSCs. Human tonsil was obtained after tonsillectomy. Using a cytometric analysis, we were able to find that the TMSCs had typical mesenchymal stem cell markers: positive for CD73, CD90, and CD105, and negative for CD14, CD34, and CD45. Using TGF-ß3, the expressions of tenocyte-specific genes and proteins, such as collagen type 1 (COL1), tenomodulin (TNMD), and scleraxis (SCX), were measured by a quantitative polymerase chain reaction (PCR), immunofluorescence staining, immunohistochemistry and Western blot analyses. Quantitative PCR assay showed that TGF-ß3 significantly increased the expressions of tenocyte lineage marker genes, including COL1, TNMD, and SCX, at a 3-day treatment, compared with control. However, these increases were not found at long-term exposures (7 or 10 days), except that TNMD expression was maintained at 50 ng/mL at a 7-day exposure to TGF-ß3. Like genes, the protein expression levels of COL1, TNMD, and SCX were also induced in TGF-ß3-treated TMSCs in a 3-day treatment, which were maintained for 10 days, as evidenced by immunofluorescence staining, immunohistochemistry and Western blot analyses. This study demonstrated that TMSCs in tenogenic stimulation with TGF-ß3 have a high tenogenic differentiation potential.

8.
PLoS One ; 10(9): e0135858, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26360050

RESUMO

OBJECTIVE: Metformin is used to treat type 2 diabetes. We sought to determine whether metformin reduces inflammation, by regulating p-signal transducer and activator of transcription 3 (STAT3) expression and T-helper 17 (Th17) cell proliferation, in a mouse model of inflammatory bowel disease (IBD). METHODS: IBD mice were administered metformin for 16 days and their tissues were analyzed. AMP-activated protein kinase (AMPK), the mammalian target of rapamycin (mTOR), p-STAT3 and p-STAT5 in the spleen and lymph nodes were detected using immunohistochemistry and confocal microscopy. Gene expression was determined using quantitative PCR assays, and protein expression levels were measured using western blotting and enzyme-linked immunosorbent assays. Human HT-29 cell proliferation was evaluated using MTT assays. RESULTS: Metformin reduced disease activity index scores and inhibited weight loss. Metformin also decreased the colonic histological score and inflammatory mediators and increased colon lengths increased. Treatment with metformin inhibited the expression of interleukin (IL)-17, p-STAT3, and p-mTOR. In contrast, metformin treatment increased expression levels of p-AMPK and Foxp3. In addition, expression of inflammatory cytokines decreased in a dose-dependent manner in inflamed human HT-29 cells cultured with metformin at various concentrations. CONCLUSIONS: Metformin attenuates IBD severity and reduces inflammation through the inhibition of p-STAT3 and IL-17 expression. Our results have increased our understanding of this chronic inflammatory disease, and support the strategy of using p-STAT3 inhibitors to treat IBD.


Assuntos
Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Metformina/farmacologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Células Th17/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , Fosforilação , RNA Mensageiro/genética , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Serina-Treonina Quinases TOR/metabolismo , Células Th17/citologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
9.
PLoS One ; 10(6): e0130882, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26083352

RESUMO

Increases in oxidative stress are thought to be associated with the development of osteoarthritis (OA). Eupatilin, one of the major compounds present in artemisia species, was shown to have both anti-oxidative and anti-inflammatory properties. Here, we investigated the in vivo effects of eupatilin on pain severity and cartilage degradation in an experimental rat model of OA, along with the mechanisms of action underlying these effects. Experimental OA was induced via an intra-articular injection of monosodium iodoacetate (MIA), with oral administration of eupatilin initiated on the day of MIA injection. Pain was assessed by measuring the paw withdrawal latency and threshold. Cartilage destruction was analyzed macroscopically and histomorphologically. The effects of eupatilin on mRNA expression were investigated in interleukin-1ß (IL-1ß)-stimulated human OA chondrocytes. Eupatilin treatment exhibited clear antinociceptive effects, along with an attenuation of cartilage degradation in OA rats. Additionally, the number of osteoclasts present in the subchondral bone region was significantly decreased following eupatilin treatment. Eupatilin reduced the expression of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), nitrotyrosine and inducible nitric oxide synthase (iNOS) in cartilage. mRNA levels of matrix metalloproteinase-3 (MMP-3), MMP13, and a disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5) were reduced in IL-1ß-stimulated human OA chondrocytes, while tissue inhibitor of metalloproteinases-1 (TIMP-1) was induced. Phosphorylated protein levels of the c-jun N-terminal kinase (JNK) was reduced by eupatilin. Taken together, these results suggest that eupatilin suppresses oxidative damage and reciprocally enhances extracellular matrix production in articular chondrocytes, making eupatilin a promising therapeutic option for the treatment of OA.


Assuntos
Analgésicos/farmacologia , Artrite Experimental/prevenção & controle , Condrócitos/efeitos dos fármacos , Flavonoides/farmacologia , Osteoartrite/prevenção & controle , Dor/tratamento farmacológico , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/patologia , Biomarcadores/metabolismo , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Condrócitos/patologia , Humanos , Técnicas Imunoenzimáticas , Masculino , Osteoartrite/induzido quimicamente , Osteoartrite/patologia , Ratos , Ratos Wistar
10.
PLoS One ; 9(2): e86062, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24558360

RESUMO

Epigallocatechin-3-gallate (EGCG) is a green tea polyphenol exerting potent anti-oxidant and anti-inflammatory effects by inhibiting signaling and gene expression. The objective of the study was to evaluate the effect of EGCG on interleukin (IL)-1 receptor antagonist knockout (IL-1RaKO) autoimmune arthritis models. IL-1RaKO arthritis models were injected intraperitoneally with EGCG three times per week after the first immunization. EGCG decreased the arthritis index and showed protective effects against joint destruction in the IL-1RaKO arthritis models. The expression of pro-inflammatory cytokines, oxidative stress proteins, and p-STAT3 (Y705) and p-STAT3 (S727), mTOR and HIF-1α were significantly lower in mice treated with EGCG. EGCG reduced osteoclast markers in vivo and in vitro along with anti-osteoclastic activity was observed in EGCG-treated IL-1RaKO mice. The proportion of Foxp3(+) Treg cells increased in the spleens of mice treated with EGCG, whereas the proportion of Th17 cells reduced. In vitro, p-STAT3 (Y705) and p-STAT3 (S727), HIF1α and glycolytic pathway molecules were decreased by EGCG. EGCG suppressed the activation of mTOR and subsequently HIF-1α, which is considered as a metabolic check point of Th17/Treg differentiation supporting the therapeutic potential of EGCG in autoimmune arthritis.


Assuntos
Antioxidantes/química , Catequina/análogos & derivados , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator de Transcrição STAT3/metabolismo , Células Th17/imunologia , Animais , Artrite/genética , Artrite/metabolismo , Doenças Autoimunes/genética , Catequina/química , Diferenciação Celular , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Osteoclastos/citologia , Estresse Oxidativo , Receptores de Interleucina-1/metabolismo , Linfócitos T Reguladores/imunologia
11.
Immune Netw ; 14(1): 45-53, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24605080

RESUMO

Osteoarthritis (OA) is a degenerative joint disease characterized by a progressive loss of cartilage. And, increased oxidative stress plays a relevant role in the pathogenesis of OA. Ursodeoxycholic acid (UDCA) is a used drug for liver diseases known for its free radical-scavenging property. The objectives of this study were to investigate the in vivo effects of UDCA on pain severity and cartilage degeneration using an experimental OA model and to explore its mode of actions. OA was induced in rats by intra-articular injection of monosodium iodoacetate (MIA) to the knee. Oral administration UDCA was initiated on the day of MIA injection. Limb nociception was assessed by measuring the paw withdrawal latency and threshold. Samples were analyzed macroscopically and histologically. Immunohistochemistry was used to investigate the expression of interleukin-1ß (IL-1ß), IL-6, nitrotyrosine and inducible nitric oxide synthase (iNOS) in knee joints. UDCA showed an antinociceptive property and attenuated cartilage degeneration. OA rats given oral UDCA significantly exhibited a decreased number of osteoclasts in subchondral bone legion compared with the vehicle-treated OA group. UDCA reduced the expression of IL-1ß, IL-6, nitrotyrosine and iNOS in articular cartilage. UDCA treatment significantly attenuated the mRNA expression of matrix metalloproteinase-3 (MMP-3), -13, and ADAMTS5 in IL-1ß-stimulated human OA chondrocytes. These results show the inhibitory effects of UDCA on pain production and cartilage degeneration in experimentally induced OA. The chondroprotective properties of UDCA were achieved by suppressing oxidative damage and inhibiting catabolic factors that are implicated in the pathogenesis of cartilage damage in OA.

12.
Arthritis Rheumatol ; 66(7): 1768-78, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24644005

RESUMO

OBJECTIVE: Intravenous immunoglobulin (IVIG) is used as a therapeutic agent in various autoimmune diseases. The aims of this study were to investigate the therapeutic effects of IVIG on collagen-induced arthritis (CIA) and identify the mechanism responsible for any therapeutic effects. METHODS: IVIG was administered to mice with CIA, and the in vivo effects were determined. Th17 and Treg cell frequencies were analyzed by flow cytometry, and cytokine levels in the supernatant were measured by enzyme-linked immunosorbent assay. Subpopulations of T cells and B cells in the spleen were assessed by confocal microscopy. RESULTS: The arthritis severity score and incidence of arthritis were lower in mice treated with IVIG compared with untreated mice. Histopathologic analysis showed less joint damage in mice treated with IVIG. The expression of proinflammatory cytokines, specific type II collagen antibodies, and osteoclast markers was significantly reduced in mice treated with IVIG. Administration of IVIG induced increased FoxP3 expression and inhibited Th17 cell development. The number of FoxP3+ Treg cells was increased, and the number of Th17 cells was decreased in the spleens of mice treated with IVIG. The number of FoxP3+ follicular helper T cells was increased, and subsequent maturation of germinal center B cells was inhibited by IVIG. In addition, IVIG up-regulated interleukin-10 (IL-10) and Fcγ receptor IIB expression. The treatment effects of IVIG on arthritis were lost in IL-10-knockout mice. CONCLUSION: These results showed that IVIG has therapeutic effects by modulating CD4+ T cell differentiation. The therapeutic effects of IVIG are dependent on IL-10.


Assuntos
Artrite Experimental/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Imunoglobulinas Intravenosas/farmacologia , Interleucina-10/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Artrite Experimental/imunologia , Doenças Autoimunes/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Receptores de IgG/genética , Receptores de IgG/imunologia , Índice de Gravidade de Doença , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Células Th17/citologia , Células Th17/imunologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
13.
Arthritis Rheumatol ; 66(5): 1195-207, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24782183

RESUMO

OBJECTIVE: The small molecule halofuginone has been shown to inhibit fibrosis, angiogenesis, and tumor progression. This study was undertaken to evaluate the effects of halofuginone in preventing autoimmune arthritis in mice. METHODS: The effects of halofuginone on joint diseases were assessed by clinical scoring and histologic analysis. Protein expression levels were confirmed by immunohistochemistry, enzyme-linked immunosorbent assay, flow cytometry, and/or Western blotting. The expression levels of messenger RNA (mRNA) for various molecules were determined by real-time polymerase chain reaction (PCR). Proliferation of osteoclast precursors was assessed by bromodeoxyuridine uptake. Osteoclast differentiation and activity were determined by quantifying tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells and area of resorbed bone. RESULTS: Treatment with halofuginone suppressed the development of autoimmune arthritis and reciprocally regulated Th17 cells and FoxP3+ Treg cells. These effects of halofuginone on Th17 differentiation involved increased signaling of ERK and reduction of STAT-3 and NF-ATc1 expression. Furthermore, halofuginone induced the expression of indoleamine 2,3-dioxygenase (IDO) in dendritic cells, leading to reduced production of Th17 cells. In addition, halofuginone prevented the formation and activity of osteoclasts through suppression of transcription factors, such as activator protein 1 and NF-ATc1, and inhibited cell cycle arrest by the committed osteoclast precursors via expression of Ccnd1 encoding cyclin D1. CONCLUSION: Taken together, our results suggest that halofuginone is a promising therapeutic agent for the treatment of Th17 cell-mediated inflammatory diseases and bone diseases.


Assuntos
Artrite/prevenção & controle , Doenças Autoimunes/prevenção & controle , Diferenciação Celular/fisiologia , Osteoclastos/patologia , Piperidinas/uso terapêutico , Inibidores da Síntese de Proteínas/uso terapêutico , Quinazolinonas/uso terapêutico , Linfócitos T Reguladores/patologia , Células Th17/patologia , Animais , Artrite/patologia , Artrite/fisiopatologia , Doenças Autoimunes/patologia , Doenças Autoimunes/fisiopatologia , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Fatores de Transcrição Forkhead/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoprotegerina/farmacologia , Osteoprotegerina/uso terapêutico , Piperidinas/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Quinazolinonas/farmacologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th17/efeitos dos fármacos
14.
Arch Pharm Res ; 36(1): 116-24, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23338062

RESUMO

Osteoarthritis (OA) is a degenerative joint disease characterized by the progressive loss of articular cartilage and chronic pain. Although cyclooxygenase-2 (COX-2) inhibitors such as celecoxib are recommended to patients at high risk of gastrointestinal (GI) adverse events, COX-2 inhibitors do not completely prevent GI adverse events. Rebamipide, a gastroprotective agent, has anti-inflammatory properties and acts as an oxygen radical scavenger. The aim of this study was to investigate the in vivo effects of coadministration of rebamipide and celecoxib in an OA rat model. OA was induced by intra-articular injection of monosodium iodoacetate. Oral administration of rebamipide was initiated on the day of OA induction. In this study, rebamipide showed antinociceptive properties and attenuated cartilage degeneration. Rebamipide reduced the expression of matrix metalloproteinase 13, interleukin-1ß, inducible nitric oxide synthase, and nitrotyrosine in OA cartilage. OA rats treated with celecoxib in combination with rebamipide demonstrated a higher pain threshold than those treated with monotherapy. Histological examination also showed that the joints from OA animals treated with combination therapy demonstrated less cartilage damage than those of animals treated with monotherapy. We showed that the potential benefit of combination therapy with celecoxib and rebamipide on pain and cartilage degeneration in OA.


Assuntos
Alanina/análogos & derivados , Artrite Experimental/tratamento farmacológico , Cartilagem/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Osteoartrite/tratamento farmacológico , Pirazóis/uso terapêutico , Quinolonas/uso terapêutico , Sulfonamidas/uso terapêutico , Administração Oral , Alanina/administração & dosagem , Alanina/efeitos adversos , Alanina/uso terapêutico , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/patologia , Comportamento Animal/efeitos dos fármacos , Cartilagem/patologia , Celecoxib , Inibidores de Ciclo-Oxigenase 2/administração & dosagem , Inibidores de Ciclo-Oxigenase 2/efeitos adversos , Quimioterapia Combinada , Injeções Intra-Articulares , Ácido Iodoacético/farmacologia , Masculino , Osteoartrite/induzido quimicamente , Osteoartrite/patologia , Dor/prevenção & controle , Pirazóis/administração & dosagem , Pirazóis/efeitos adversos , Quinolonas/administração & dosagem , Quinolonas/efeitos adversos , Ratos , Ratos Wistar , Sulfonamidas/administração & dosagem , Sulfonamidas/efeitos adversos
15.
PLoS One ; 8(6): e67171, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840617

RESUMO

BACKGROUND: In this study we examined the in vivo and in vitro effects and mechanisms of action of curcumin on the development of acute graft-versus-host disease (GVHD) using a murine model. METHODOLOGY/PRINCIPAL FINDINGS: Mixed lymphocyte reactions were used to determine the in vitro effects of curcumin. Treatment with curcumin attenuated alloreactive T cell proliferation and inhibited the production of interferon (IFN)-γ and interleukin (IL)-17. In a murine acute GVHD model, transplantation of curcumin-treated allogeneic splenocytes into irradiated recipient mice significantly reduced the clinical severity scores of acute GVHD manifested in the liver, skin, colon and lung as compared with animals receiving vehicle-treated splenocytes. c-Fos and c-Jun expression levels in the skin and intestine, which are major target organs, were analyzed using immunohistochemical staining. Expression of both proteins was reduced in epithelial tissues of skin and intestine from curcumin-treated GVHD animals. The IFN-γ-expressing CD4(+) splenocytes and IFN-γ-expressing lymph node cells were dramatically decreased in curcumin-treated mice. In contrast, CD4(+)Foxp3(+) splenocytes were increased in the curcumin-treated acute GVHD animals. Flow cytometric analysis revealed that animals transplanted with curcumin-treated allogeneic splenocytes showed increased populations of CD4(+) regulatory T cells (Tregs) as well as CD8(+) Treg cells, compared to animals administered vehicle-treated splenocytes. Curcumin-treated acute GVHD animals could have a change in B cell subpopulations. CONCLUSION/SIGNIFICANCE: In the present study, we investigated the efficacy and mechanism of action of curcumin treatment against acute GVHD. The acute GVHD mice administered with curcumin-treated splenocytes showed significantly reduced severity of acute GVHD. Curcumin exerted in vivo preventive effects on acute GVHD by reciprocal regulation of T helper 1 (Th1) and Treg (both CD4(+) and CD8(+) Treg) cell lineages as well as B cell homeostasis.


Assuntos
Curcumina/farmacologia , Doença Enxerto-Hospedeiro/tratamento farmacológico , Fatores Imunológicos/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Transplante de Medula Óssea , Proliferação de Células , Células Cultivadas , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Curcumina/uso terapêutico , Doença Enxerto-Hospedeiro/imunologia , Fatores Imunológicos/uso terapêutico , Interferon gama/metabolismo , Interleucina-17/metabolismo , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Baço/imunologia , Baço/patologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/metabolismo
16.
PLoS One ; 8(4): e60900, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23613752

RESUMO

C57BL/6 mice are known to be resistant to the development of collagen-induced arthritis (CIA). However, they show a severe arthritic phenotype when the Ifng gene is deleted. Although it has been proposed that IFN-γ suppresses inflammation in CIA via suppressing Th17 which is involved in the pathogenesis of CIA, the exact molecular mechanism of the Th17 regulation by IFN-γ is poorly understood. This study was conducted to 1) clarify that arthritogenic condition of IFN-γ knockout (KO) mice is dependent on the disinhibition of Th17 and 2) demonstrate that IFN-γ-induced indoleamine-2,3-dioxgenase (IDO) is engaged in the regulation of Th17. The results showed that the IFN-γ KO mice displayed increased levels of IL-17 producing T cells and the exacerbation of arthritis. Also, production of IL-17 by the splenocytes of the IFN-γ KO mice was increased when cultured with type II collagen. When Il17 was deleted from the IFN-γ KO mice, only mild arthritis developed without any progression of the arthritis score. The proportion of CD44(high)CD62L(low) memory-like T cells were elevated in the spleen, draining lymph node and mesenteric lymph node of IFN-γ KO CIA mice. Meanwhile, CD44(low)CD62L(high) naïve T cells were increased in IFN-γ and IL-17 double KO CIA mice. When Th17 polarized CD4+ T cells of IFN-γ KO mice were co-cultured with their own antigen presenting cells (APCs), a greater increase in IL-17 production was observed than in co-culture of the cells from wild type mice. In contrast, when APCs from IFN-γ KO mice were pretreated with IFN-γ, there was a significant reduction in IL-17 in the co-culture system. Of note, pretreatment of 1-methyl-DL- tryptophan, a specific inhibitor of IDO, abolished the inhibitory effects of IFN-γ. Given that IFN-γ is a potent inducer of IDO in APCs, these results suggest that IDO is involved in the regulation of IL-17 by IFN-γ.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon gama/metabolismo , Interleucina-17/metabolismo , Animais , Artrite Experimental , Linfócitos T CD4-Positivos/metabolismo , Ensaio de Imunoadsorção Enzimática , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Interferon gama/genética , Interleucina-17/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
PLoS One ; 8(7): e69362, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894457

RESUMO

OBJECTIVE: To investigate the effect of CoenzymeQ10 (CoQ10) on pain severity and cartilage degeneration in an experimental model of rat osteoarthritis (OA). MATERIALS AND METHODS: OA was induced in rats by intra-articular injection of monosodium iodoacetate (MIA) to the knee. Oral administration of CoQ10 was initiated on day 4 after MIA injection. Pain severity was assessed by measuring secondary tactile allodynia using the von Frey assessment test. The degree of cartilage degradation was determined by measuring cartilage thickness and the amount of proteoglycan. The mankin scoring system was also used. Expressions of matrix metalloproteinase-13 (MMP-13), interleukin-1ß (IL-1ß), IL-6, IL-15, inducible nitric oxide synthase (iNOS), nitrotyrosine and receptor for advanced glycation end products (RAGE) were analyzed using immunohistochemistry. RESULTS: Treatment with CoQ10 demonstrated an antinociceptive effect in the OA animal model. The reduction in secondary tactile allodynia was shown by an increased pain withdrawal latency and pain withdrawal threshold. CoQ10 also attenuated cartilage degeneration in the osteoarthritic joints. MMP-13, IL-1ß, IL-6, IL-15, iNOS, nitrotyrosine and RAGE expressions were upregulated in OA joints and significantly reduced with CoQ10 treatment. CONCLUSION: CoQ10 exerts a therapeutic effect on OA via pain suppression and cartilage degeneration by inhibiting inflammatory mediators, which play a vital role in OA pathogenesis.


Assuntos
Cartilagem/efeitos dos fármacos , Citocinas/metabolismo , Óxido Nítrico/metabolismo , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Dor/tratamento farmacológico , Ubiquinona/análogos & derivados , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Cartilagem/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/metabolismo , Ácido Iodoacético/farmacologia , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Osteoartrite/induzido quimicamente , Osteoartrite/patologia , Dor/complicações , Ratos , Ratos Wistar , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Ubiquinona/farmacologia , Ubiquinona/uso terapêutico
18.
PLoS One ; 8(11): e78843, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24223854

RESUMO

Grape seed proanthocyanidin extract (GSPE) is a natural flavonoid that exerts anti-inflammatory properties. Obesity is an inflammatory condition and inflammatory cells and their secretion of pro-inflammatory molecules contribute to the pathogenesis of obesity. Rheumatoid arthritis (RA) is a chronic autoimmune disease that is characterized by inflammation of joints lined by synovium. Previously, we demonstrated that obesity augmented arthritis severity in collagen induced arthritis (CIA), a murine model of human RA. Here, we investigated whether oral administration of GSPE showed antiobesity and anti-arthritic effects in high-fat diet-induced obese (DIO) mice and in obese CIA mice, respectively. The pathophysiologic mechanisms by which GSPE attenuates weight gain and arthritis severity in vivo were also investigated. In DIO mice, GSPE administration significantly inhibited weight gain, reduced fat infiltration in liver and improved serum lipid profiles. The antiobesity effect of GSPE was associated with increased populations of regulatory T (Treg) cells and those of decreased Th17 cells. Decrease of Th17 cells was associated with significant inhibition of their key transcriptional factors, pSTAT3(Tyr705) and pSTAT3(Ser727). On the contrary, GSPE-induced Treg induction was associated with enhanced pSTAT5 expression. To identify the anti-arthritis effects of GSPE, GSPE was given orally for 7 weeks after type II collagen immunization. GSPE treatment significantly attenuated the development of autoimmune arthritis in obese CIA model. In line with DIO mice, GSPE administration decreased Th17 cells and reciprocally increased Treg cells by regulating STAT proteins in autoimmune arthritis model. The expressions of pro-inflammatory cytokines and nitrotyrosine in synovium were significantly inhibited by GSPE treatment. Taken together, GSPE functions as a reciprocal regulator of T cell differentiation - suppression of Th17 cells and induction of Tregs in both DIO and obese CIA mice. GSPE may act as a therapeutic agent to treat immunologic diseases related with enhanced STAT3 activity such as metabolic disorders and autoimmune diseases.


Assuntos
Artrite/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Extrato de Sementes de Uva/farmacologia , Obesidade/prevenção & controle , Proantocianidinas/farmacologia , Fator de Transcrição STAT3/metabolismo , Administração Oral , Animais , Artrite/complicações , Artrite Experimental/complicações , Artrite Experimental/prevenção & controle , Citocinas/genética , Citocinas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Extrato de Sementes de Uva/administração & dosagem , Humanos , Inflamação/complicações , Inflamação/prevenção & controle , Mediadores da Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Microscopia Confocal , Obesidade/complicações , Obesidade/etiologia , Proantocianidinas/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Células Th17/efeitos dos fármacos , Células Th17/metabolismo , Células Th17/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa