Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Korean Med Sci ; 35(6): e40, 2020 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-32056400

RESUMEN

BACKGROUND: Immune cells express the vitamin (vit) D receptor, and vit D is a potent immune-modulator. A negative correlation between serum vit D levels and rheumatoid arthritis (RA) disease activity has been reported. Therefore, we aimed to investigate if the sufficient serum vit D level is helpful to control disease activity in RA patients treated with interleukin (IL)-6 receptor antibody tocilizumab. METHODS: RA patients taking tocilizumab were enrolled, and data were collected retrospectively. Disease activity scores (DAS) 28, serum vit D levels, modified Sharp scores of hand X-ray at the time of tocilizumab initiation, and follow-up data were analysed. Peripheral blood mononuclear cells were differentiated into T-helper (Th) 17 or osteoclasts in the presence of various concentrations of tocilizumab and/or 1,25(OH)2D. Th17 proportions were analysed by fluorescence-activated cell sorting. Supernatant cytokine levels were determined by enzyme-linked immunosorbent assay. RESULTS: Among 98 RA patients taking tocilizumab, 34 (34.7%) had sufficient serum 25(OH)D levels (≥ 30 ng/mL) when tocilizumab was initiated. At 24 weeks, vit D sufficient patients had greater DAS28 reduction (64.6% ± 15.5% vs. 52.7% ± 20.7%, P = 0.004), and lower disease activity (91.2% vs. 70.3%, P = 0.018) or remission (82.4% vs. 57.8%, P = 0.014). These differences in DAS28 reduction and the proportion of patients with remission persisted at 48 weeks. However, there was no significant difference in hand and wrist erosion progression. In vitro, tocilizumab and 1,25(OH)2D treatment synergistically suppressed IL-17 production and osteoclastogenesis. CONCLUSION: RA patients treated with IL-6 antibody show a better response when they have sufficient serum vit D. Tocilizumab and 1,25(OH)2D synergistically suppress IL-17 production and osteoclast differentiation in RA patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Antirreumáticos , Artritis Reumatoide , Vitamina D/análogos & derivados , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Humanos , Leucocitos Mononucleares , Osteogénesis/efectos de los fármacos , Receptores de Interleucina-6/inmunología , Estudios Retrospectivos , Vitamina D/sangre , Vitamina D/uso terapéutico
2.
J Autoimmun ; 100: 114-119, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30922727

RESUMEN

Despite advances in the understanding of the pathogenesis, disease-specific biomarkers have not been included in the classification criteria for Primary Sjogren's syndrome (pSS). Based on a microarray of peripheral blood mononuclear cells (PBMCs) from patients with primary Sjogren's syndrome (pSS), we aimed to investigate whether soluble sialic acid-binding immunoglobulin-like lectin (siglec)-5 in saliva might be a biomarker for pSS. The concentration of siglec-5 in saliva and sera was determined by ELISA. Clinical parameters related with pSS were obtained from pSS registry and correlation with salivary siglec-5 level was evaluated. Receiver operating curve (ROC) analysis was performed to determine cut off value. A separate validation cohort consisted of subjects with suspicious pSS was evaluated to determine the performance. The level of salivary siglec-5 was significantly higher in pSS patients (n = 170) compared with HCs (n = 25), non SS sicca patients (n = 78) or patients with systemic lupus erythematosus (SLE) (n = 43) (1346.8 [202.8-4280.0] pg/mL, 6.08 [0-134.0] pg/mL, 195 [0-947.5] pg/mL, and 0 [0-238.7] pg/mL, median [interquartile range], P < 0.001). Salivary siglec-5 level negatively correlated with salivary flow rate (spearman's rho: -0.420, P < 0.001), and positively correlated with ocular surface score (rho: 0.331, P < 0.001) and serum immunoglobulin G level (rho = 0.202, P = 0.008). In ROC analysis, area under the curve was 0.774[0.724-0.826]. With a cut off value of 400 pg/mL, sensitivity and specificity was 0.69 and 0.70 respectively. In validation cohort (45 pSS patients and 45 non SS sicca patients), sensitivity and specificity of siglec-5 was 64.4% and 77.8%, respectively. In conclusion, the level of soluble siglec-5 is significantly higher in the saliva from pSS patients, which reflects the severity of hyposalivation and ocular surface damage. This novel salivary biomarker may provide benefits for pSS diagnosis.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Lectinas/inmunología , Saliva/inmunología , Adulto , Biomarcadores , Femenino , Humanos , Masculino , Persona de Mediana Edad , Síndrome de Sjögren/diagnóstico , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/patología
3.
Rheumatology (Oxford) ; 55(1): 156-61, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26320130

RESUMEN

OBJECTIVE: Triggering receptor expressed on myeloid cells 1 (TREM-1), which amplifies the inflammation elicited by the Toll-like receptor pathway, was originally implicated in sepsis and bacterial infection. However, it has been suggested that TREM-1 may also play an important role in non-infectious inflammation. The present study was conducted to investigate whether TREM-1 is involved in human acute gouty inflammation. METHODS: A total of 37 gout patients were recruited between March 2011 and January 2014 from Seoul St Mary's Hospital. The expression of TREM-1 on mononuclear cells was assessed using FACS analysis, immunostaining and real-time RT-PCR. To block the TREM-1 signal, soluble TREM-1 (sTREM-1) or the synthetic blocking peptide LP17 was used. The concentration of sTREM-1 was assessed by ELISA. RESULTS: FACS analysis and real-time RT-PCR demonstrated that TREM-1 expression was higher in the SF mononuclear cells of acute gouty arthritis patients than in peripheral blood mononuclear cells (PBMCs). Immunohistochemical staining of tophi tissues revealed TREM-1 expression, with confocal microscopy demonstrating TREM-1 expression on tophi tissue macrophages. We also demonstrated that MSU treatment induced TREM-1 expression on the PBMCs of acute gout patients in vitro. Although blockade of TREM-1 did not directly suppress MSU-induced IL-1ß production of PBMCs in vitro, the concentration of soluble TREM-1 was higher in the SF of gout vs OA patients and was positively correlated with serum CRP. CONCLUSION: TREM-1 is induced by MSU and is associated with the inflammation of human acute gouty arthritis.


Asunto(s)
Regulación de la Expresión Génica , Gota/genética , Glicoproteínas de Membrana/genética , Células Mieloides/metabolismo , ARN/genética , Receptores Inmunológicos/genética , Ácido Úrico/metabolismo , Enfermedad Aguda , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Gota/metabolismo , Gota/patología , Humanos , Inmunohistoquímica , Glicoproteínas de Membrana/biosíntesis , Células Mieloides/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Inmunológicos/biosíntesis , Estudios Retrospectivos , Receptor Activador Expresado en Células Mieloides 1
4.
Acta Pharmacol Sin ; 35(9): 1177-87, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25087995

RESUMEN

AIM: Ursolic acid (UA) is a pentacyclic triterpenoid found in most plant species, which has been shown anti-inflammatory and anti-oxidative activities. In this study, we examined the effects of UA on collagen-induced arthritis (CIA) in mice, and to identify the mechanisms underlying the effects. METHODS: CIA was induced in mice. Two weeks later, the mice were treated with UA (150 mg/kg, ip, 3 times per week) for 4 weeks. The expression of cytokines and oxidative stress markers in joint tissues was measured with immunohistochemistry. The numbers of CD4+IL-17+, CD4+CD25+Foxp3+ and pSTAT3 cells in spleens were determined using confocal immunostaining or flowcytometric analyses. Serum antibody levels and B cell-associated marker mRNAs were analyzed with ELISAs and qRT-PCR, respectively. CD4+ T cells and CD19+ B cells were purified from mice spleens for in vitro studies. RESULTS: UA treatment significantly reduced the incidence and severity of CIA-induced arthritis, accompanied by decreased expression of proinflammatory cytokines (TNF-α, IL-1ß, IL-6, IL-21 and IL-17) and oxidative stress markers (nitrotyrosine and iNOS) in arthritic joints. In CIA mice, UA treatment significantly decreased the number of Th17 cells, while increased the number of Treg cells in the spleens, which was consistent with decreased expression of pSTAT3, along with IL-17 and RORγt in the splenocytes. In addition, UA treatment significantly reduced the serum CII-specific IgG levels in CIA mice. The inhibitory effects of UA on Th17 cells were confirmed in an in vitro model of Th17 differentiation. Furthermore, UA dose-dependently suppressed the expression of B cell-associated markers Bcl-6, Blimp1 and AID mRNAs in purified CD19+ B cells pretreated with IL-21 or LPS in vitro. CONCLUSION: UA treatment significantly ameliorates CIA in mice via suppression of Th17 and differentiation. By targeting pathogenic Th17 cells and autoantibody production, UA may be useful for the treatment of autoimmune arthritis and other Th17-related diseases.


Asunto(s)
Artritis Experimental/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Células Th17/inmunología , Triterpenos/inmunología , Animales , Masculino , Ratones , Ratones Endogámicos DBA , Ácido Ursólico
5.
J Korean Med Sci ; 28(8): 1145-53, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23960440

RESUMEN

Interleukin (IL)-33 is an important mediator of innate immunity. Behcet's disease (BD) is an autoinflammatory disorder characterized by hyperactivity of the innate immune response. We measured serum levels of IL-33 and its receptor soluble ST2 (sST2) in patients with BD to investigate their association with disease activity. Serum levels of both IL-33 and sST2 were higher in patients with BD compared with those in normal controls (IL-33: 594.48±175.04 pg/mL in BD and 224.23±56.64 pg/mL in normal controls [P=0.048], sST2: 99.01±15.92 pg/mL in BD and 23.56±3.25 pg/mL in normal controls [P<0.001]). IL-33 and sST2 expression in skin tissue, as shown by immunohistochemistry, was higher in patients with BD compared with that in the normal controls. Serum sST2 level correlated significantly with the BD currently active form (BDCAF), Iranian BD dynamic activity measure (IBDDAM), erythrocyte sedimentation rate and C-reactive protein. Multiple linear regression showed that serum sST2 was an independent factor associated with IBBDAM (regression coefficient, 0.374; P=0.004), and BDCAF (regression coefficient, 0.236; P=0.047). These results demonstrate that IL-33 and sST2 are highly expressed in patients with BD and that serum sST2 is an independent factor associated with IBDDAM and BDCAF, suggesting a potential role for sST2 as a surrogate marker of disease activity in patients with BD.


Asunto(s)
Síndrome de Behçet/patología , Interleucinas/sangre , Receptores de Superficie Celular/sangre , Adulto , Síndrome de Behçet/sangre , Sedimentación Sanguínea , Proteína C-Reactiva/análisis , Femenino , Humanos , Inmunohistoquímica , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Interleucinas/metabolismo , Masculino , Persona de Mediana Edad , Receptores de Superficie Celular/metabolismo , Índice de Severidad de la Enfermedad , Piel/metabolismo , Piel/patología
6.
Front Immunol ; 12: 704526, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34497607

RESUMEN

Objective: Baricitinib, a selective inhibitor for janus kinase (JAK) 1 and JAK2, is approved for use in rheumatoid arthritis. Systemic lupus erythematosus (SLE) is recently regarded as a potential candidate targeted by JAK inhibitors because of the relationship between its pathogenesis and JAK/signal transducer and activator of transcription (STAT) pathway-mediated cytokines such as type I interferons. The objective of this study was to determine whether baricitinib could effectively ameliorate SLE using a murine model. Methods: To investigate effects of baricitinib on various autoimmune features, especially renal involvements in SLE, eight-week-old MRL/Mp-Faslpr (MRL/lpr) mice were used as a lupus-prone animal model and treated with baricitinib for eight weeks. Immortalized podocytes and primary podocytes and B cells isolated from C57BL/6 mice were used to determine the in vitro efficacy of baricitinib. Results: Baricitinib remarkably suppressed lupus-like phenotypes of MRL/lpr mice, such as splenomegaly, lymphadenopathy, proteinuria, and systemic autoimmunity including circulating autoantibodies and pro-inflammatory cytokines. It also modulated immune cell populations and effectively ameliorated renal inflammation, leading to the recovery of the expression of structural proteins in podocytes. According to in vitro experiments, baricitinib treatment could mitigate B cell differentiation and restore disrupted cytoskeletal structures of podocytes under inflammatory stimulation by blocking the JAK/STAT pathway. Conclusions: The present study demonstrated that baricitinib could effectively attenuate autoimmune features including renal inflammation of lupus-prone mice by suppressing aberrant B cell activation and podocyte abnormalities. Thus, baricitinib as a selective JAK inhibitor could be a promising therapeutic candidate in the treatment of SLE.


Asunto(s)
Azetidinas/farmacología , Lupus Eritematoso Sistémico , Podocitos , Purinas/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/inmunología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/inmunología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos MRL lpr , Podocitos/inmunología , Podocitos/patología , Transducción de Señal/efectos de los fármacos
7.
Arthritis Rheumatol ; 70(12): 2057-2066, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29927095

RESUMEN

OBJECTIVE: To examine whether a JAK inhibitor regulates functional responses of human salivary gland epithelial cells (SGECs) and disease parameters in an animal model of Sjögren's syndrome (SS). METHODS: Common differentially expressed genes (DEGs) were analyzed among peripheral blood mononuclear cells from patients with primary SS and other data sets, using blood and SG tissue. Validation of expression in SGs was analyzed by focus score. Inhibition of messenger RNA expression of DEGs and BAFF by filgotinib was analyzed using reverse transcription-polymerase chain reaction in primary SGECs. SG organoid cultures were used to determine the association between DEGs and BAFF via knockdown using small interfering RNAs or to determine regulation of BAFF by JAK inhibitor. Filgotinib (1.5 mg/kg) was intraperitoneally injected into 8-week-old NOD/ShiLtJ mice 3 times per week to analyze manifestations of disease. Finally, STAT signaling was assessed in human and mouse SGECs. RESULTS: Expression of the DEGs IFNG and BAFF increased in SGs from patients with primary SS, as assessed by focus score. There was a significant correlation between IFIT2 and BAFF expression. JAK inhibitor suppressed interferon (IFN)-induced transcription of DEGs and BAFF in human primary SGECs. Knockdown of DEGs or inhibition of JAK caused reduced secretion of BAFF in human SG organoid cultures. In addition, filgotinib-treated mice exhibited increased salivary flow rates and marked reductions in lymphocytic infiltration of SGs. JAK inhibitor regulated IFNα- and IFNγ-induced pSTAT-1Y701 , pSTAT-3Y705 , and protein inhibitor of activated STAT-3 (PIAS-3) in human SGECs as well as IFNγ-induced pSTAT-1Y701 , pSTAT-3S727 , and PIAS-1 in mouse SGECs. CONCLUSION: JAK inhibition controls aberrant activation of SGECs and may be a novel therapeutic approach for primary SS.


Asunto(s)
Janus Quinasa 1/antagonistas & inhibidores , Piridinas/farmacología , Glándulas Salivales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Síndrome de Sjögren/tratamiento farmacológico , Triazoles/farmacología , Animales , Factor Activador de Células B/biosíntesis , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Humanos , Interferones/fisiología , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos NOD , ARN Mensajero/efectos de los fármacos
8.
Int Immunopharmacol ; 26(1): 103-11, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25819229

RESUMEN

Digoxin is a cardiac glycoside that is commonly used to treat heart failure. Based on its known anti-inflammatory effect, this study was undertaken to investigate the effect of digoxin on collagen-induced arthritis (CIA) and to delineate the underlying mechanism. Digoxin or vehicle was injected intraperitoneally thrice weekly in mice with CIA, from day 7 or day 35 after immunization to investigate preventive or therapeutic effect, respectively. The incidence and severity of arthritis was evaluated. Digoxin treatment suppressed the incidence of arthritis and joint inflammation in mice with CIA. The expression of IL-17 and other proinflammatory cytokines, including IL-1ß, IL-6, TNF-α and IL-21, were markedly reduced in the arthritic joints of digoxin-treated CIA mice. Th17 cells and CD4(+) pSTAT3(+) cells were less frequently observed in the spleen of digoxin-treated CIA mice than controls. The mRNA expression of IL-17 and ROR γt was consistently lower in total splenocytes or draining lymph node cells obtained from digoxin-treated CIA mice. Digoxin also reduced in vitro Th17 differentiation and LPS-stimulated IgG production. The number of osteoclasts in the arthritic joint was lower in digoxin-treated mice, whereas digoxin treatment did not directly suppress in vitro osteoclastogenesis. Our findings suggest that digoxin can regulate Th17 and reciprocally promote Treg cells and suppress joint inflammation and bone erosion in CIA. Digoxin may be a therapeutic option by targeting pathogenic Th17 and immunoglobulin production, for treatment of autoimmune arthritis and other Th17-related diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Enfermedades Autoinmunes/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Digoxina/uso terapéutico , Células Th17/efectos de los fármacos , Animales , Antiinflamatorios/administración & dosificación , Artritis Experimental/inmunología , Artritis Experimental/patología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Digoxina/administración & dosificación , Modelos Animales de Enfermedad , Articulaciones/efectos de los fármacos , Articulaciones/inmunología , Articulaciones/patología , Masculino , Ratones Endogámicos DBA
9.
PLoS One ; 10(11): e0138201, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26544846

RESUMEN

INTRODUCTION: Oxidative stress plays a role in the pathogenesis of rheumatoid arthritis (RA). Anthocyanin is a plant antioxidant. We investigated the therapeutic effects of anthocyanin extracted from black soybean seed coats (AEBS) in a murine model of collagen-induced arthritis (CIA) and human peripheral blood mononuclear cells (PBMCs) and explored possible mechanisms by which AEBS might exert anti-arthritic effects. MATERIAL AND METHODS: CIA was induced in DBA/1J mice. Cytokine levels were measured via enzyme-linked immunosorbent assays. Joints were assessed in terms of arthritis incidence, clinical arthritis scores, and histological features. The extent of oxidative stress in affected joints was determined by measuring the levels of nitrotyrosine and inducible nitric oxide synthase. NF-κB activity was assayed by measuring the ratio of phosphorylated IκB to total IκB via Western blotting. Th17 cells were stained with antibodies against CD4, IL-17, and STAT3. Osteoclast formation was assessed via TRAP staining and measurement of osteoclast-specific mRNA levels. RESULTS: In the CIA model, AEBS decreased the incidence of arthritis, histological inflammation, cartilage scores, and oxidative stress. AEBS reduced the levels of proinflammatory cytokines in affected joints of CIA mice and suppressed NF-κB signaling. AEBS decreased Th17 cell numbers in spleen of CIA mice. Additionally, AEBS repressed differentiation of Th17 cells and expression of Th17-associated genes in vitro, in both splenocytes of naïve DBA/1J mice and human PBMCs. In vitro, the numbers of both human and mouse tartrate-resistant acid phosphatase+ (TRAP) multinucleated cells fell, in a dose-dependent manner, upon addition of AEBS. CONCLUSIONS: The anti-arthritic effects of AEBS were associated with decreases in Th17 cell numbers, and the levels of proinflammatory cytokines synthesized by such cells, mediated via suppression of NF-κB signaling. Additionally, AEBS suppressed osteoclastogenesis and reduced oxidative stress levels.


Asunto(s)
Antocianinas/farmacología , Artritis Experimental/tratamiento farmacológico , Enfermedades Autoinmunes/tratamiento farmacológico , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , FN-kappa B/antagonistas & inhibidores , Extractos Vegetales/farmacología , Células Th17/inmunología , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/metabolismo , Artritis Experimental/patología , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Western Blotting , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/toxicidad , Citocinas/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos DBA , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Semillas/química , Transducción de Señal/efectos de los fármacos , Glycine max/química , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/efectos de los fármacos , Células Th17/metabolismo
10.
Arthritis Rheumatol ; 66(5): 1195-207, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24782183

RESUMEN

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.


Asunto(s)
Artritis/prevención & control , Enfermedades Autoinmunes/prevención & control , Diferenciación Celular/fisiología , Osteoclastos/patología , Piperidinas/uso terapéutico , Inhibidores de la Síntesis de la Proteína/uso terapéutico , Quinazolinonas/uso terapéutico , Linfocitos T Reguladores/patología , Células Th17/patología , Animales , Artritis/patología , Artritis/fisiopatología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Factores de Transcripción Forkhead/metabolismo , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Osteoclastos/efectos de los fármacos , Osteoprotegerina/farmacología , Osteoprotegerina/uso terapéutico , Piperidinas/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Quinazolinonas/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA