Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Mol Model ; 27(1): 14, 2021 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33403456

RESUMO

Rheumatoid arthritis (RA) is a systemic autoimmune disorder that commonly affects multiple joints of the body. Currently, there is no permanent cure to the disease, but it can be managed with several potent drugs that cause serious side effects on prolonged use. Traditional remedies are considered promising for the treatment of several diseases, particularly chronic conditions, because they have lower side effects compared to synthetic drugs. In folklore, the rhizome of Alpinia calcarata Roscoe (Zingiberaceae) is used as a major ingredient of herbal formulations to treat RA. Phytoconstituents reported in A. calcarata rhizomes are diterpenoids, sesquiterpenoid, flavonoids, phytosterol, and volatile oils. The present study is intended to understand the molecular-level interaction of phytoconstituents present in A. calcarata rhizomes with RA molecular targets using computational approaches. A total of 30 phytoconstituents reported from the plant were used to carry out docking with 36 known targets of RA. Based on the docking results, 4 flavonoids were found to be strongly interacting with the RA targets. Further, molecular dynamics simulation confirmed stable interaction of quercetin with 6 targets (JAK3, SYK, MMP2, TLR8, IRAK1, and JAK1), galangin with 2 targets (IRAK1 and JAK1), and kaempferol (IRAK1) with one target of RA. Moreover, the presence of these three flavonoids was confirmed in the A. calcarata rhizome extract using LC-MS analysis. The computational study suggests that flavonoids present in A. calcarata rhizome may be responsible for RA modulatory activity. Particularly, quercetin and galangin could be potential development candidates for the treatment of RA. Investigation of Alpinia calcarata constituent interactions with molecular targets of rheumatoid arthritis: docking, molecular dynamics, and network approach.


Assuntos
Alpinia/química , Artrite Reumatoide/tratamento farmacológico , Biologia Computacional , Flavonoides/farmacologia , Compostos Fitoquímicos/farmacologia , Artrite Reumatoide/metabolismo , Cromatografia Líquida , Flavonoides/análise , Flavonoides/química , Humanos , Quinases Associadas a Receptores de Interleucina-1/química , Quinases Associadas a Receptores de Interleucina-1/efeitos dos fármacos , Janus Quinase 1/química , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 3/química , Janus Quinase 3/efeitos dos fármacos , Quempferóis/química , Quempferóis/farmacologia , Espectrometria de Massas , Metaloproteinase 2 da Matriz/química , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Compostos Fitoquímicos/análise , Compostos Fitoquímicos/química , Extratos Vegetais/química , Quercetina/química , Quercetina/farmacologia , Rizoma/química , Quinase Syk/química , Quinase Syk/efeitos dos fármacos , Receptor 8 Toll-Like/química , Receptor 8 Toll-Like/efeitos dos fármacos
2.
Aging (Albany NY) ; 13(1): 411-423, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33290262

RESUMO

As the first clinical proteasome inhibitor, Bortezomib (BTZ) has been reported to improve the outcome of lymphoma. However, due to the unstable property, low bioavailability, and hydrophobic properties of BTZ, it is needed to develop effective drug delivery systems to deliver BTZ into targeted cells or organs. Here we developed a bortezomib (BTZ)-loaded HMSNs (BTZ@HMSNs) system, which can sustain the release of BTZ in targeted tissues. In vitro assays showed that BTZ@HMSNs limited cell proliferation and augmented apoptosis of lymphoma SNK-1 cells. Moreover, BTZ@HMSNs significantly diminished migration and invasion of SNK-1 cells as compared with BTZ. In contrast to the upregulation of SHP-1, BTZ@HMSNs decreased the mRNA levels of c-Kit, NF-κB, and JAK1, which elicit oncogenic role in lymphoma development. Importantly, lymphoma mice model showed that BTZ@HMSNs significantly activated p53 signaling and reduced tumor volume and weight compared with free BTZ. Our data thus demonstrate that BTZ@HMSNs manifests improved tumor-suppressing effect in vitro and in vivo compared to free BTZ. We believe that HMSNs is a promising strategy for delivering therapeutic agents for cancer treatment.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Bortezomib/administração & dosagem , Bortezomib/farmacologia , Proliferação de Células/efeitos dos fármacos , Linfoma/tratamento farmacológico , Nanosferas , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Portadores de Fármacos , Humanos , Técnicas In Vitro , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 1/genética , Linfoma/genética , Linfoma/metabolismo , Camundongos , Camundongos Nus , NF-kappa B/efeitos dos fármacos , NF-kappa B/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Proteínas Proto-Oncogênicas c-kit/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Dióxido de Silício , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Dermatol Ther ; 32(5): e13053, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31381252

RESUMO

Alopecia areata (AA) is a non-scarring alopecia, which often carries a major impact on patients' quality of life. Currently there is no single approved treatment that effectively induces permanent remission. Recently, the JAK-STAT signaling pathway has emerged as a possible therapeutic target leading to increased interest in the use of Janus kinase (JAK) inhibitors (JAKis) in the treatment of this pathology. This review of the literature summarizes information on patients with AA who underwent treatment with JAKis and discusses the current evidence on the efficacy and safety of its use. A literature search was conducted in different databases to identify clinical trials and case reports published in January 2019. Several clinical studies have shown very promising results in the treatment of AA with oral formulas of JAKis. These agents, however, need chronic administration to maintain response. Topical formulations did not show satisfactory responses. The safety profile of these agents appears to be favorable. Current evidence is promising regarding the efficacy and safety of oral JAKis. However, the data obtained are of low quality, originating predominantly from reports of clinical cases. Further studies are needed to confirm these data and to optimize its long-term efficacy and safety.


Assuntos
Alopecia em Áreas/tratamento farmacológico , Janus Quinase 1/efeitos dos fármacos , Inibidores de Janus Quinases/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Alopecia em Áreas/genética , Ensaios Clínicos como Assunto , Feminino , Humanos , Janus Quinase 1/genética , Masculino , Terapia de Alvo Molecular , Prognóstico , Resultado do Tratamento
5.
Environ Toxicol ; 34(4): 486-494, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30609183

RESUMO

Macrophages not only play an important role in the innate immune response but also participate in many inflammatory and infectious diseases including asthma, diabetes, obesity, cardiovascular diseases, and cancers. Bisphenol A (BPA) is the most commonly used component for plastic products. However, BPA is an endocrine disruptor for mammals and participates in several inflammatory and infectious diseases. Up until now, there are no researches demonstrated the potential role of BPA in macrophage activation and its relative mechanism. BPA promoted the generation of proinflammatory cytokines IL-1ß, IL-6, and TNFα in a concentration-dependent manner (P < 0.05). BPA was identified to increase the expression of proinflammatory mediators NO and PGE2, and its upstream factors iNOS, COX2, and cPLA2 in a concentration-dependent manner (P < 0.05). Phosphorylation and nuclear translocation of NF-κB p65 were significantly induced by BPA via IκB degradation (P < 0.05). In addition, phosphorylation of ERK significantly induced by BPA at a concentration which was less than that for phosphorylation of p38 MAPK and JNK (P < 0.05). Furthermore, phosphorylation of STAT3 significantly induced by BPA at a concentration lower than that for phosphorylation of STAT1 (P < 0.05). Phosphorylation of JAK1 and JAK2 was also significantly induced by BPA in a concentration-dependent manner (P < 0.05).


Assuntos
Compostos Benzidrílicos/toxicidade , Citocinas/genética , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 2/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Fenóis/toxicidade , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Relação Dose-Resposta a Droga , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Fosforilação , Células RAW 264.7
6.
J Eur Acad Dermatol Venereol ; 33(2): 367-375, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30357932

RESUMO

BACKGROUND: Janus kinase (JAK) inhibition may be a promising new treatment modality for inflammatory (skin) diseases. However, little is known about direct effects of kinase inhibitors on keratinocyte differentiation and function as well as skin barrier formation. OBJECTIVE: Our aim was to address the direct impact of kinase inhibition of the JAK1/3 pathways by tofacitinib on keratinocyte immune function and barrier formation in atopic dermatitis (AD) and psoriasis. METHODS: 3D skin equivalents of both diseases were developed and concurrently pretreated with tofacitinib. To induce AD, 3D skin equivalents were stimulated with recombinant human IL-4 and IL-13. Psoriasis-like conditions were induced by incubation with IL-17A, IL-22 and tumour necrosis factor α (TNFα). The activation of signal transducer and activator of transcription (STAT)1, STAT3 and STAT6 was assessed by Western blot analysis. Microarray analysis and quantitative real-time PCR were used for gene expression analysis. RESULTS: Tofacitinib pretreatment preserved epidermal morphology and reduced STAT3 and STAT6 phosphorylation of AD-like and STAT3 phosphorylation of psoriasis-like culture conditions in 3D skin models compared to sham-controls. Filaggrin expression was fully maintained in the AD-like models, but only partially in psoriasis-like conditions after pretreatment with tofacitinib. In addition, tofacitinib upregulated DSC1, FLG and KRT1. Using gene expression analysis, downregulation of POSTN and IL24 was observed in AD-like conditions, whereas downregulation of IL20 and IL1B was observed in psoriasis-like conditions. CONCLUSION: JAK1/3 inhibition counteracted cytokine-induced AD- and psoriasis-like epidermal morphology and enhanced keratinocyte differentiation in 3D skin models. This effect was more pronounced in the AD-like models compared to the psoriasis-like 3D skin models.


Assuntos
Dermatite Atópica/patologia , Imageamento Tridimensional , Proteínas de Filamentos Intermediários/farmacologia , Janus Quinase 1/efeitos dos fármacos , Piperidinas/farmacologia , Psoríase/patologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Proliferação de Células/efeitos dos fármacos , Simulação por Computador , Dermatite Atópica/tratamento farmacológico , Proteínas Filagrinas , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Psoríase/tratamento farmacológico , Fator de Transcrição STAT6/efeitos dos fármacos , Sensibilidade e Especificidade
7.
Ann Rheum Dis ; 76(8): 1467-1475, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28478401

RESUMO

OBJECTIVES: Janus kinase 2 (JAK2) has recently been described as a novel downstream mediator of the pro-fibrotic effects of transforming growth factor-ß. Although JAK2 inhibitors are in clinical use for myelodysplastic syndromes, patients often rapidly develop resistance. Tumour cells can escape the therapeutic effects of selective JAK2 inhibitors by mutation-independent transactivation of JAK2 by JAK1. Here, we used selective JAK2 inhibition as a model to test the hypothesis that chronic treatment may provoke resistance by facilitating non-physiological signalling pathways in fibroblasts. METHODS: The antifibrotic effects of long-term treatment with selective JAK2 inhibitors and reactivation of JAK2 signalling by JAK1-dependent transphosphorylation was analysed in cultured fibroblasts and experimental dermal and pulmonary fibrosis. Combined JAK1/JAK2 inhibition and co-treatment with an HSP90 inhibitor were evaluated as strategies to overcome resistance. RESULTS: The antifibrotic effects of selective JAK2 inhibitors on fibroblasts decreased with prolonged treatment as JAK2 signalling was reactivated by JAK1-dependent transphosphorylation of JAK2. This reactivation could be prevented by HSP90 inhibition, which destabilised JAK2 protein, or with combined JAK1/JAK2 inhibitors. Treatment with combined JAK1/JAK2 inhibitors or with JAK2 inhibitors in combination with HSP90 inhibitors was more effective than monotherapy with JAK2 inhibitors in bleomycin-induced pulmonary fibrosis and in adTBR-induced dermal fibrosis. CONCLUSION: Fibroblasts can develop resistance to chronic treatment with JAK2 inhibitors by induction of non-physiological JAK1-dependent transactivation of JAK2 and that inhibition of this compensatory signalling pathway, for example, by co-inhibition of JAK1 or HSP90 is important to maintain the antifibrotic effects of JAK2 inhibition with long-term treatment.


Assuntos
Fibroblastos/efeitos dos fármacos , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 2/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fibrose Pulmonar/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia , Escleroderma Sistêmico , Sulfonamidas/farmacologia , Adulto , Animais , Antibióticos Antineoplásicos/toxicidade , Benzoquinonas/farmacologia , Bleomicina/toxicidade , Western Blotting , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibrose , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Imuno-Histoquímica , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 1/metabolismo , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Lactamas Macrocíclicas/farmacologia , Pulmão/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Nitrilas , Fosforilação/efeitos dos fármacos , Fibrose Pulmonar/induzido quimicamente , Reação em Cadeia da Polimerase em Tempo Real , Fator de Crescimento Transformador beta/farmacologia
8.
J Trauma Acute Care Surg ; 78(5): 980-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25909419

RESUMO

BACKGROUND: JAK/STAT signal pathway plays an important role in the inflammation process of acute lung injury (ALI). This study aimed to investigate the correlation between recombinant human brain natriuretic peptide (rhBNP) and the JAK/STAT signaling pathway and to explore the protective mechanism of rhBNP against trauma-induced ALI. METHODS: The arterial partial pressure in oxygen, lung wet-dry weight ratios, protein content in bronchoalveolar lavage fluid, the histopathologic of the lung, as well as the protein expressions of STAT1, JAK2, and STAT3 were detected. RESULTS: Sprague-Dawley rats were randomly divided into five groups: a control group, a sham-operated group, an ALI group, an ALI + rhBNP group, and an ALI + AG490 group. At 4 hours, 12 hours, 1 day, 3 days, and 7 days after injury, injured lung specimens were harvested. rhBNP pretreatment significantly ameliorated hypoxemia and histopathologic changes and alleviated pulmonary edema in trauma-induced ALI rats. rhBNP pretreatment reduced the phosphorylated protein and total protein level of STAT1. Similarly to JAK-specific inhibitor AG490, rhBNP was shown to significantly inhibit the phosphorylation of JAK2 and STAT3 in rats with trauma-induced ALI. CONCLUSION: Our experimental findings indicated that rhBNP can protect rats against trauma-induced ALI and that its underlying mechanism may be related to the inhibition of JAK/STAT signaling pathway activation.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Janus Quinase 1/efeitos dos fármacos , Peptídeo Natriurético Encefálico/uso terapêutico , Fatores de Transcrição STAT/efeitos dos fármacos , Ferimentos e Lesões/complicações , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Imuno-Histoquímica , Janus Quinase 1/metabolismo , Masculino , Natriuréticos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Ann Rheum Dis ; 74(6): 1311-6, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25398374

RESUMO

OBJECTIVE: Tofacitinib is an oral Janus kinase (JAK) inhibitor for the treatment of rheumatoid arthritis (RA). The pathways affected by tofacitinib and the effects on gene expression in situ are unknown. Therefore, tofacitinib effects on synovial pathobiology were investigated. METHODS: A randomised, double-blind, phase II serial synovial biopsy study (A3921073; NCT00976599) in patients with RA with an inadequate methotrexate response. Patients on background methotrexate received tofacitinib 10 mg twice daily or placebo for 28 days. Synovial biopsies were performed on Days -7 and 28 and analysed by immunoassay or quantitative PCR. Clinical response was determined by disease activity score and European League Against Rheumatism (EULAR) response on Day 28 in A3921073, and at Month 3 in a long-term extension study (A3921024; NCT00413699). RESULTS: Tofacitinib exposure led to EULAR moderate to good responses (11/14 patients), while placebo was ineffective (1/14 patients) on Day 28. Tofacitinib treatment significantly reduced synovial mRNA expression of matrix metalloproteinase (MMP)-1 and MMP-3 (p<0.05) and chemokines CCL2, CXCL10 and CXCL13 (p<0.05). No overall changes were observed in synovial inflammation score or the presence of T cells, B cells or macrophages. Changes in synovial phosphorylation of signal transducer and activator of transcription 1 (STAT1) and STAT3 strongly correlated with 4-month clinical responses (p<0.002). Tofacitinib significantly decreased plasma CXCL10 (p<0.005) at Day 28 compared with placebo. CONCLUSIONS: Tofacitinib reduces metalloproteinase and interferon-regulated gene expression in rheumatoid synovium, and clinical improvement correlates with reductions in STAT1 and STAT3 phosphorylation. JAK1-mediated interferon and interleukin-6 signalling likely play a key role in the synovial response. TRIAL REGISTRATION NUMBER: NCT00976599.


Assuntos
Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Janus Quinase 1/efeitos dos fármacos , Piperidinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Pirróis/uso terapêutico , RNA Mensageiro/efeitos dos fármacos , Fatores de Transcrição STAT/efeitos dos fármacos , Membrana Sinovial/efeitos dos fármacos , Adulto , Idoso , Antirreumáticos/farmacologia , Artrite Reumatoide/metabolismo , Quimiocinas/efeitos dos fármacos , Quimiocinas/genética , Quimiocinas/metabolismo , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Humanos , Janus Quinase 1/metabolismo , Masculino , Metaloproteinase 1 da Matriz/efeitos dos fármacos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/efeitos dos fármacos , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 3 da Matriz/metabolismo , Metotrexato/uso terapêutico , Pessoa de Meia-Idade , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , RNA Mensageiro/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/efeitos dos fármacos , Membrana Sinovial/metabolismo , Resultado do Tratamento
10.
Nat Rev Rheumatol ; 9(3): 173-82, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23419429

RESUMO

The molecular biology revolution coupled with the development of monoclonal antibody technology enabled remarkable progress in rheumatology therapy, comprising an array of highly effective biologic agents. With advances in understanding of the molecular nature of immune cell receptors came elucidation of intracellular signalling pathways downstream of these receptors. These discoveries raise the question of whether selective targeting of key intracellular factors with small molecules would add to the rheumatologic armamentarium. In this Review, we discuss several examples of this therapeutic strategy that seem to be successful, and consider their implications for the future of immune-targeted treatments. We focus on kinase inhibitors, primarily those targeting Janus kinase family members and spleen tyrosine kinase, given their advanced status in clinical development and application. We also summarize other targets involved in signalling pathways that might offer promise for therapeutic intervention in the future.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Antirreumáticos/administração & dosagem , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Janus Quinase 1/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Receptores de Citocinas/efeitos dos fármacos , Transdução de Sinais/fisiologia , Quinase Syk , Tiazóis
11.
Indian J Exp Biol ; 49(11): 840-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22126015

RESUMO

For understanding of signaling molecules important in lung cancer growth and progression, IL-1beta effect was analyzed on iNOS expression and key signaling molecules in human lung carcinoma A549 cells and established the role of specific signaling molecules by using specific chemical inhibitors. IL-1beta exposure (10 ng/ml) induced strong iNOS expression in serum starved A549 cells. Detailed molecular analyses showed that IL-1beta increased expression of phosphorylated STAT1 (Tyr701 and Ser727) and STAT3 (Tyr705 and Ser727) both in total cell lysates and nuclear lysates. Further, IL-1beta exposure strongly activated MAPKs (ERK1/2, JNK1/2 and p38) and Akt as well as increased nuclear levels of NF-kappaB and HIF-1alpha in A549 cells. Use of specific chemical inhibitors for JAK1 kinase (piceatannol), JAK2 kinase (AG-490), MEK1/2 (PD98059) and JNK1/2 (SP600125) revealed that IL-1beta-induced iNOS expression involved signaling pathways in addition to JAK-STAT and ERK1/2-JNK1/2 activation. Overall, these results suggested that instead of specific pharmacological inhibitors, use of chemopreventive agents with broad spectrum efficacy to inhibit IL-1beta-induced signaling cascades and iNOS expression would be a better strategy towards lung cancer prevention and/or treatment.


Assuntos
Carcinoma/metabolismo , Neoplasias Pulmonares/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Carcinoma/prevenção & controle , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Janus Quinase 1/efeitos dos fármacos , Neoplasias Pulmonares/prevenção & controle , Sistema de Sinalização das MAP Quinases/genética , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Transdução de Sinais , Estilbenos/farmacologia
12.
ACS Chem Biol ; 6(5): 432-43, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21226522

RESUMO

Stat3 is a latent transcription factor that promotes cell survival and proliferation and is often constitutively active in multiple cancers. Inhibition of Stat3 signaling pathways suppresses cell survival signals and leads to apoptosis in cancer cells, suggesting direct inhibition of Stat3 function is a viable therapeutic approach. Herein, we identify a small molecule, C48, as a selective Stat3-family member inhibitor. To determine its mechanism of action, we used site-directed mutagenesis and multiple biochemical techniques to show that C48 alkylates Cys468 in Stat3, a residue at the DNA-binding interface. We further demonstrate that C48 blocks accumulation of activated Stat3 in the nucleus in tumor cell lines that overexpress active Stat3, leading to impressive inhibition of tumor growth in mouse models. Collectively, these findings suggest Cys468 in Stat3 represents a novel site for therapeutic intervention and demonstrates the promise of alkylation as a potentially effective chemical approach for Stat3-dependent cancers.


Assuntos
Aminopiridinas/química , Benzodioxóis/química , Cisteína/metabolismo , Fator de Transcrição STAT3/metabolismo , Alquilação , Sequência de Aminoácidos , Aminopiridinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Benzodioxóis/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Ensaio de Desvio de Mobilidade Eletroforética , Células HeLa , Humanos , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 2/efeitos dos fármacos , Espectrometria de Massas , Camundongos , Transplante de Neoplasias , Fosforilação
13.
Neuroscience ; 167(3): 872-9, 2010 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-20206237

RESUMO

Chondroitin sulfate (CS) is a glucosaminoglycan (GAG) currently used for the treatment of osteoarthritis because of its antiinflammatory and antiapoptotic actions. Recent evidence has revealed that those peripheral effects of CS may also have therapeutic interest in diseases of the CNS. Since neuroinflammation has been implicated in different neuronal pathologies, this study was planned to investigate how CS could modulate the inflammatory response in the CNS by using rat astrocyte cultures stimulated with lipopolysaccharide (LPS). We have evaluated different proteins implicated in the nuclear factor kappa B (NFkappaB) and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways employing RT-PCR, western blot and immunofluorescence techniques. At 10 microM, CS prevented translocation of p65 to the nucleus, reduced tumour necrosis factor alpha (TNF-alpha) mRNA and mitigated cyclooxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS) induction by LPS. However, it did not modify LPS-induced IP-10 and SOCS-1 mRNA, proteins that participate in the JAK/STAT pathway. The results of this study indicate that CS can potentially reduce neuroinflammation by inhibition of NFkappaB. Therefore endogenous GAGs could afford neuroimmunomodulatory actions under neurotoxic conditions.


Assuntos
Anti-Inflamatórios/farmacologia , Astrócitos/efeitos dos fármacos , Sulfatos de Condroitina/farmacologia , Encefalite/tratamento farmacológico , Gliose/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Anti-Inflamatórios/uso terapêutico , Astrócitos/metabolismo , Células Cultivadas , Sulfatos de Condroitina/uso terapêutico , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Encefalite/metabolismo , Encefalite/fisiopatologia , Gliose/metabolismo , Gliose/fisiopatologia , Mediadores da Inflamação/farmacologia , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 1/metabolismo , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição STAT/efeitos dos fármacos , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/genética
14.
Neuroimmunomodulation ; 16(3): 177-84, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19246940

RESUMO

Gonadotropin-releasing hormone (GnRH), which was originally found to be involved in the reproductive process, has also been implicated in the modulation of immune system function. However, the underlying mechanisms of this involvement remain largely unclear. In this study, we found that GnRH increased the intracellular calcium levels in murine Raw264.7 macrophages. Furthermore, the production of nitric oxide, costimulated with lipopolysaccharide and interferon-gamma, was suppressed by exposure to GnRH. Moreover, the modulatory effects of GnRH on calcium and nitric oxide were observed in freshly isolated primary peritoneal macrophages. In addition, the activity of nuclear factor-kappaB was suppressed by GnRH exposure. On the other hand, the phosphorylation of the Janus kinase-signal transducer and activator of transcription pathway was not affected by cotreatment with GnRH. Taken together, these results demonstrate that GnRH participates in the macrophage function and indicate that the nuclear factor-kappaB signaling pathway may be responsible for GnRH-mediated immune system modulation.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Fatores Imunológicos/metabolismo , Macrófagos/imunologia , NF-kappa B/metabolismo , Neuroimunomodulação/imunologia , Transdução de Sinais/imunologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/imunologia , Linhagem Celular , Hormônio Liberador de Gonadotropina/farmacologia , Fatores Imunológicos/farmacologia , Mediadores da Inflamação/farmacologia , Interferon gama/efeitos dos fármacos , Interferon gama/imunologia , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 1/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/efeitos dos fármacos , Neuroimunomodulação/efeitos dos fármacos , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Biochem Biophys Res Commun ; 370(4): 557-60, 2008 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-18381062

RESUMO

Two members of Jak kinases, Jak1 and Jak3, are associated with the cytoplasmic domains of the interleukin-2 (IL-2) receptor (IL-2R) beta chain (IL-2Rbeta) and the common cytokine receptor gamma chain (gammac), respectively, and accumulating evidence indicates their functional importance in IL-2 signaling. Here, I showed that coumermycin-induced chemical heterodimerization of Jak1 and Jak3 but not homodimerization of Jak1 or Jak3 induces cell proliferation of an IL-2R-reconstituted cell line. In this regard, expression of IL-2Rbeta was essential for cell proliferation by chemical heterodimerization of Jak1 and Jak3, indicating that dimerized Jak1 and Jak3 induce heterodimerization of IL-2Rbeta and gammac, which may activate receptor-bound signaling molecules. Previous reports using chemical dimerization suggest that dimerization of Jak kinases is sufficient to induce cell proliferation. The present study indicates that re-evaluation of this conclusion is necessary and that interpretation of functional analysis of signaling molecules using chemical dimerizers needs more careful assessment.


Assuntos
Proliferação de Células , Subunidade beta de Receptor de Interleucina-2/metabolismo , Janus Quinase 1/metabolismo , Janus Quinase 3/metabolismo , Aminocumarinas/farmacologia , Animais , Dimerização , Humanos , Subunidade beta de Receptor de Interleucina-2/genética , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 3/efeitos dos fármacos , Camundongos
16.
J Cardiovasc Pharmacol ; 50(2): 126-41, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17703129

RESUMO

Activation of the transcription factor signal transducers and activators of transcription (STAT) 3 is a defining feature of the interleukin (IL)-6 family of cytokines, which include IL-6, leukemia inhibitory factor, and cardiotrophin-1. These cytokines, as well as STAT3 activation, have been shown to be protective for cardiac myocytes and necessary for ischemia preconditioning. However, the mechanisms that regulate IL-6-type cytokine signaling in cardiac myocytes are largely unexplored. We propose that the protective character of IL-6-type cytokine signaling in cardiac myocytes is determined principally by three mechanisms: redox status of the nonreceptor tyrosine kinase Janus kinase 1 (JAK) 1 that activates STAT3, phosphorylation of STAT3 within the transcriptional activation domain on serine 727, and STAT3-mediated induction of suppressor of cytokine signaling (SOCS) 3 that terminates IL-6-type cytokine signaling. Moreover, we hypothesize that hyperactivation of the JAK kinases, particularly JAK2, mismatched STAT3 serine-tyrosine phosphorylation or heightened STAT3 transcriptional activity, and SOCS3 induction may ultimately prove detrimental. Here we summarize recent evidence that supports this hypothesis, as well as additional possible mechanisms of JAK-STAT regulation. Understanding how IL-6-type cytokine signaling is regulated in cardiac myocytes has great significance for exploiting the therapeutic potential of these cytokines and the phenomenon of preconditioning.


Assuntos
Fármacos Cardiovasculares/farmacologia , Cardiopatias/tratamento farmacológico , Interleucina-6/metabolismo , Fator de Transcrição STAT3/efeitos dos fármacos , Animais , Fármacos Cardiovasculares/administração & dosagem , Citocinas/metabolismo , Sistemas de Liberação de Medicamentos , Cardiopatias/fisiopatologia , Humanos , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 1/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/metabolismo
17.
J Neurochem ; 100(3): 628-38, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17263792

RESUMO

Multipotent neural stem cells (NSCs) are competent for commitment to the oligodendrocyte (OL) lineage both in vitro and in vivo. We exploited this property to develop a rat neurospheres (NS)/oligospheres (OS)-based culture system to generate large numbers of highly enriched late OL progenitors (preOLs) and mature OLs (MatOLs). CNS neuroblastoma cell line B104-derived conditioned medium promoted the generation of nearly pure populations of preOLs from dissociated OS. The subsequent culture of preOLs with ciliary neurotrophic factor (CNTF) and 3,3',5'-triiodo-L-thyronine (T(3)) generated nearly pure populations of MatOLs. OL lineage specificity was confirmed by immunocytochemistry, quantitative RT-PCR and gene expression profiling, which demonstrated large differences between preOLs and MatOLs. The insulin-like growth factors (IGFs) are potent neuro-protective agents required for OL survival. We used this system to systematically define maturation-dependent changes in IGF signaling during the course of OL differentiation. The IGF-I and insulin receptors, insulin receptor substrate-1 (IRS-1) and IRS-2, protein kinase B (PKB)/Akt and Janus kinase (JNK) were expressed at higher levels in NS and preOLs compared with OS and MatOLs. Erk expression increased markedly from NS to OS, decreased only partially upon commitment to preOLs, and, in MatOLs, returned to a low level similar to NS. IGF activation of the generally proliferative Erk pathway was gradually acquired during NSC differentiation, whereas IGF activation of the generally pro-survival, anti-apoptotic PI3K/PKB pathway was consistently robust at each developmental stage.


Assuntos
Transplante de Tecido Encefálico/métodos , Diferenciação Celular/fisiologia , Células-Tronco Multipotentes/metabolismo , Oligodendroglia/metabolismo , Somatomedinas/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/fisiologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Fator Neurotrófico Ciliar/metabolismo , Fator Neurotrófico Ciliar/farmacologia , Meios de Cultivo Condicionados/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Substratos do Receptor de Insulina , Fator de Crescimento Insulin-Like I/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/metabolismo , Janus Quinase 1/efeitos dos fármacos , Janus Quinase 1/metabolismo , Células-Tronco Multipotentes/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Somatomedinas/farmacologia , Células-Tronco/efeitos dos fármacos , Tri-Iodotironina/metabolismo , Tri-Iodotironina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA