Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
Int J Mol Sci ; 23(9)2022 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-35563088

RESUMO

Urolithin A is an active compound of gut-microbiota-derived metabolites of polyphenol ellagic acid that has anti-aging, antioxidative, and anti-inflammatory effects. However, the effects of urolithin A on polyinosinic acid-polycytidylic acid (poly(I:C))-induced inflammation remain unclear. Poly(I:C) is a double-stranded RNA (dsRNA) similar to a virus and is recognized by Toll-like receptor-3 (TLR3), inducing an inflammatory response in immune cells, such as macrophages. Inflammation is a natural defense process of the innate immune system. Therefore, we used poly(I:C)-induced RAW264.7 cells and attenuated the inflammation induced by urolithin A. First, our data suggested that 1-30 µM urolithin A does not reduce RAW264.7 cell viability, whereas 1 µM urolithin A is sufficient for antioxidation and the decreased production of tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and C-C chemokine ligand 5. The inflammation-related proteins cyclooxygenase-2 and inducible nitric oxide synthase were also downregulated by urolithin A. Next, 1 µM urolithin A inhibited the levels of interferon (INF)-α and INF-ß. Urolithin A was applied to investigate the blockade of the TLR3 signaling pathway in poly(I:C)-induced RAW264.7 cells. Moreover, the TLR3 signaling pathway, subsequent inflammatory-related pathways, and antioxidation pathways showed changes in nuclear factor-κB (NF-κB) signaling and blocked ERK/mitogen-activated protein kinase (MAPK) signaling. Urolithin A enhanced catalase (CAT) and superoxide dismutase (SOD) activities, but decreased malondialdehyde (MDA) levels in poly(I:C)-induced RAW264.7 cells. Thus, our results suggest that urolithin A inhibits TLR3-activated inflammatory and oxidative-associated pathways in macrophages, and that this inhibition is induced by poly(I:C). Therefore, urolithin A may have antiviral effects and could be used to treat viral-infection-related diseases.


Assuntos
Cumarínicos , NF-kappa B , Receptor 3 Toll-Like , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antioxidantes/farmacologia , Cumarínicos/farmacologia , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Poli I-C/farmacologia , Células RAW 264.7 , RNA de Cadeia Dupla/farmacologia , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/antagonistas & inibidores , Receptor 3 Toll-Like/metabolismo
2.
Carbohydr Polym ; 277: 118869, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-34893274

RESUMO

After regular chemotherapy, the expression of programmed cell death ligand 1 (PD-L1) in almost all kinds of cancers is significantly increased, leading to reduced efficacy of T cell mediated immune killing in tumors. To solve this, a lot of PD-L1 antibodies were produced and used, but their high cost and serious toxic side effects still limit its usage. Recently, small molecule compounds that could effectively regulate PD-L1 expression possess the edges to solve the problems of PD-L1 antibodies. Chitosan oligosaccharide (COS), a biomaterial derived from the N-deacetylation product of chitin, has a broad spectrum of biological activities in treating tumors. However, the mechanism of its anti-cancer effect is still not well understood. Here, for the first time, we clearly identified that COS could inhibit the upregulated PD-L1 expression induced by interferon γ (IFN-γ) in various tumors via the AMPK activation and STAT1 inhibition. Besides, COS itself significantly restricted the growth of CT26 tumors by enhancing the T cell infiltration in tumors. Furthermore, we observed that combining COS with Gemcitabine (GEM), one of the typical chemotherapeutic drugs, leaded to a more remarkable tumor remission. Therefore, it was demonstrated that COS could be used as a useful way to improve the efficacy of existing chemotherapies by effective PD-L1 downregulation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Quitosana/farmacologia , Neoplasias do Colo/terapia , Imunoterapia , Oligossacarídeos/farmacologia , Fator de Transcrição STAT1/antagonistas & inibidores , Animais , Antineoplásicos/química , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quitosana/química , Neoplasias do Colo/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Oligossacarídeos/química , Fator de Transcrição STAT1/metabolismo
3.
Inflamm Res ; 70(4): 389-405, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33608746

RESUMO

OBJECTIVE: Coronavirus disease 2019 (COVID-19) is a world-wide pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To date, treatment of severe COVID-19 is far from clear. Therefore, it is urgent to develop an effective option for the treatment of patients with COVID-19. Most patients with severe COVID-19 exhibit markedly increased serum levels of pro-inflammatory cytokines, including interferon (IFN)-α, IFN-γ, and interleukin (IL)-1ß. Immunotherapeutic strategies have an important role in the suppression of cytokine storm and respiratory failure in patients with COVID-19. METHODS: A systematic search in the literature was performed in PubMed, Scopus, Embase, Cochrane Library, Web of Science, as well as Google Scholar preprint database using all available MeSH terms for Coronavirus, SARS-CoV-2, anti-rheumatoid agents, COVID-19, cytokine storm, immunotherapeutic drugs, IFN, interleukin, JAK/STAT inhibitors, MCP, MIP, TNF. RESULTS: Here, we first review common complications of COVID-19 patients, particularly neurological symptoms. We next explain host immune responses against COVID-19 particles. Finally, we summarize the existing experimental and clinical immunotherapeutic strategies, particularly anti-rheumatoid agents and also plasma (with a high level of gamma globulin) therapy for severe COVID-19 patients. We discuss both their therapeutic effects and side effects that should be taken into consideration for their clinical application. CONCLUSION: It is suggested that immunosuppressants, such as anti-rheumatoid drugs, could be considered as a potential approach for the treatment of cytokine storm in severe cases of COVID-19. One possible limitation of immunosuppressant therapy is their inhibitory effects on host anti-viral immune response. So, the appropriate timing of administration should be carefully considered.


Assuntos
COVID-19/epidemiologia , COVID-19/terapia , Síndrome da Liberação de Citocina/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Animais , Antirreumáticos/uso terapêutico , COVID-19/imunologia , Citocinas/metabolismo , Inibidores Enzimáticos/uso terapêutico , Humanos , Imunização Passiva , Imunossupressores/uso terapêutico , Imunoterapia , Inflamação/tratamento farmacológico , Interferons/uso terapêutico , Interleucina-1/antagonistas & inibidores , Interleucina-6/antagonistas & inibidores , Janus Quinase 1/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Insuficiência Respiratória/terapia , Fator de Transcrição STAT1/antagonistas & inibidores , Transdução de Sinais , Soroterapia para COVID-19
4.
Vet Microbiol ; 254: 108980, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33445054

RESUMO

The Caprine parainfluenza virus 3 (CPIV3) is a novel Paramyxovirus that is isolated from goats suffering from respiratory diseases. Presently, the pathogenesis of CPIV3 infection has not yet been fully characterized. The Type I interferon (IFN) is a key mediator of innate antiviral responses, as many viruses have developed strategies to circumvent IFN response, whether or how CPIV3 antagonizes type I IFN antiviral effects have not yet been characterized. This study observed that CPIV3 was resistant to IFN-α treatment and antagonized IFN-α antiviral responses on MDBK and goat tracheal epithelial (GTE) cell models. Western blot analysis showed that CPIV3 infection reduced STAT1 expression and phosphorylation, which inhibited IFN-α signal transduction on GTE cells. By screening and utilizing specific monoclonal antibodies (mAbs), three CPIV3 accessory proteins C, V and D were identified during the virus infection process on the GTE cell models. Accessory proteins C and V, but not protein D, was identified to antagonize IFN-α antiviral signaling. Furthermore, accessory protein C, but not protein V, reduced the level of IFN-α driven phosphorylated STAT1 (pSTAT1), and then inhibit STAT1 signaling. Genetic variation analysis to the PIV3 accessory protein C has found two highly variable regions (VR), with VR2 (31-70th aa) being involved in for the CPIV3 accessory protein C to hijack the STAT1 signaling activation. The above data indicated that CPIV3 is capable of inhibiting IFN-α signal transduction by reducing STAT1 expression and activation, and that the accessory protein C, plays vital roles in the immune escape process.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Antivirais/farmacologia , Evasão da Resposta Imune , Interferon Tipo I/antagonistas & inibidores , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/genética , Infecções por Paramyxoviridae/veterinária , Animais , Anticorpos Monoclonais/farmacologia , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Feminino , Doenças das Cabras/virologia , Cabras/virologia , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos , Interferon-alfa/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Paramyxoviridae/tratamento farmacológico , Fosforilação , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Transdução de Sinais/efeitos dos fármacos
5.
Invest New Drugs ; 39(3): 644-657, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33300108

RESUMO

Objectives Malignant pleural mesothelioma (MPM) is an occupational disease mainly due to asbestos exposure. Effective therapies for MPM are lacking, making this tumour type a fatal disease. Materials and Methods In order to meet this need and in view of a future "drug repositioning" approach, here we screened five MPM (Mero-14, Mero-25, IST-Mes2, NCI-H28 and MSTO-211H) and one SV40-immortalized mesothelial cell line (MeT-5A) as a non-malignant model, with a library of 1170 FDA-approved drugs. Results Among several potential compounds, we found that fludarabine (F-araA) and, to a lesser extent, risedronic acid (RIS) were cytotoxic in MPM cells, in comparison to the non-malignant Met-5A cells. In particular, F-araA reduced the proliferation and the colony formation ability of the MPM malignant cells, in comparison to the non-malignant control cells, as demonstrated by proliferation and colony formation assays, in addition to measurement of the phospho-ERK/total-ERK ratio. We have shown that the response to F-araA was not dependent upon the expression of DCK and NT5E enzymes, nor upon their functional polymorphisms (rs11544786 and rs2295890, respectively). Conclusion This drug repositioning screening approach has identified that F-araA could be therapeutically active against MPM cells, in addition to other tumour types, by inhibiting STAT1 expression and nucleic acids synthesis. Further experiments are required to fully investigate this.


Assuntos
Antineoplásicos/farmacologia , Mesotelioma/tratamento farmacológico , Neoplasias Pleurais/tratamento farmacológico , Ácido Risedrônico/farmacologia , Fator de Transcrição STAT1/antagonistas & inibidores , Vidarabina/análogos & derivados , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Reposicionamento de Medicamentos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Mesotelioma/genética , Mesotelioma/metabolismo , Neoplasias Pleurais/genética , Neoplasias Pleurais/metabolismo , Fator de Transcrição STAT1/metabolismo , Vidarabina/farmacologia
6.
Bioorg Med Chem Lett ; 30(18): 127415, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32717616

RESUMO

Furanocoumarins, particularly furo[3,2-c]coumarins, are found in many natural products. However, coumarins annulated to a thiophene ring have received scarce attention to date in the literature. Therefore, we synthesized 4-oxo-4H-thieno[3,2-c]chromene derivatives and tested in vitro their anti-inflammatory activity. Anti-inflammatory potential of the synthesized compounds (1, 2, 6-8, 9a-e and 10a-c) has been evaluated by measuring various pSTAT (signal transducer and activator of transcription) inhibition within the JAK (Janus-activated family kinase)/STAT signaling pathway. Ethyl 7-hydroxy-4-oxo-4H-thieno[3,2-c]chromene-2-carboxylate (7) showed best inhibition properties on pSTAT5 in GM-CSF (Granulocyte-macrophage colony-stimulating factor)-triggered PBMC assay, with IC50 value of 5.0 µM.


Assuntos
Anti-Inflamatórios/síntese química , Benzopiranos/síntese química , Citocinas/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT5/antagonistas & inibidores , Sequência de Aminoácidos , Anti-Inflamatórios/farmacologia , Benzopiranos/farmacologia , Proteínas de Ligação a DNA , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Janus Quinases/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
7.
J Infect Dis ; 222(5): 734-745, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32563187

RESUMO

Clinical manifestations of coronavirus disease 2019 (COVID-19) vary from asymptomatic virus shedding, nonspecific pharyngitis, to pneumonia with silent hypoxia and respiratory failure. Dendritic cells and macrophages are sentinel cells for innate and adaptive immunity that affect the pathogenesis of severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS). The interplay between SARS-CoV-2 and these cell types remains unknown. We investigated infection and host responses of monocyte-derived dendritic cells (moDCs) and macrophages (MDMs) infected by SARS-CoV-2. MoDCs and MDMs were permissive to SARS-CoV-2 infection and protein expression but did not support productive virus replication. Importantly, SARS-CoV-2 launched an attenuated interferon response in both cell types and triggered significant proinflammatory cytokine/chemokine expression in MDMs but not moDCs. Investigations suggested that this attenuated immune response to SARS-CoV-2 in moDCs was associated with viral antagonism of STAT1 phosphorylation. These findings may explain the mild and insidious course of COVID-19 until late deterioration.


Assuntos
Betacoronavirus/fisiologia , Infecções por Coronavirus/imunologia , Células Dendríticas/imunologia , Interferons/imunologia , Monócitos/imunologia , Pneumonia Viral/imunologia , Fator de Transcrição STAT1/antagonistas & inibidores , Imunidade Adaptativa , Animais , Betacoronavirus/imunologia , Betacoronavirus/isolamento & purificação , Betacoronavirus/metabolismo , COVID-19 , Quimiocinas/metabolismo , Chlorocebus aethiops , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Monócitos/virologia , Pandemias , Fosforilação , Pneumonia Viral/metabolismo , Pneumonia Viral/virologia , SARS-CoV-2 , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Células Vero , Replicação Viral/fisiologia , Eliminação de Partículas Virais
8.
Inflammation ; 43(2): 752-764, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31873836

RESUMO

Cordycepin, a natural derivative of adenosine, has been shown to exert pharmacological properties including anti-oxidation, antitumor, and immune regulation. It is reported that cordycepin is involved in the regulation of macrophage function. However, the effect of cordycepin on inflammatory cell infiltration in inflammation remains ambiguous. In this study, we investigated the potential role of cordycepin playing in macrophage function in CFA-induced inflammation mice model. In this model, we found that cordycepin prevented against macrophage infiltration in paw tissue and reduced interferon-γ (IFN-γ) production in both serum and paw tissue. Using luciferase reporter assay, we found that cordycepin suppressed IFN-γ-induced activators of transcription-1 (STAT1) transcriptional activity in a dose-dependent manner. Moreover, western blotting data demonstrated that cordycepin inhibited IFN-γ-induced STAT1 activation through attenuating STAT1 phosphorylation. Further investigations revealed that cordycepin inhibited the expressions of IFN-γ-inducible protein 10 (IP-10) and monokine induced by IFN-γ (Mig), which were the effector genes in IFN-γ-induced STAT1 signaling. Meanwhile, the excessive inflammatory cell infiltration in paw tissue was reduced by cordycepin. These findings demonstrate that cordycepin alleviates excessive inflammatory cell infiltration through down-regulation of macrophage IP-10 and Mig expressions via suppressing STAT1 phosphorylation. Thus, cordycepin may be a potential therapeutic approach to prevent and treat inflammation-associated diseases.


Assuntos
Quimiocina CXCL10/antagonistas & inibidores , Quimiocina CXCL9/antagonistas & inibidores , Desoxiadenosinas/uso terapêutico , Interferon gama/toxicidade , Macrófagos/efeitos dos fármacos , Fator de Transcrição STAT1/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Quimiocina CXCL10/biossíntese , Quimiocina CXCL9/biossíntese , Desoxiadenosinas/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Adjuvante de Freund/toxicidade , Expressão Gênica , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , Distribuição Aleatória , Fator de Transcrição STAT1/metabolismo
9.
Sci Rep ; 9(1): 16710, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31723221

RESUMO

Nipah virus (NiV) is a pathogenic paramyxovirus and zoononis with very high human fatality rates. Previous protein over-expression studies have shown that various mutations to the common N-terminal STAT1-binding motif of the NiV P, V, and W proteins affected the STAT1-binding ability of these proteins thus interfering with he JAK/STAT pathway and reducing their ability to inhibit type-I IFN signaling, but due to differing techniques it was unclear which amino acids were most important in this interaction or what impact this had on pathogenesis in vivo. We compared all previously described mutations in parallel and found the amino acid mutation Y116E demonstrated the greatest reduction in binding to STAT1 and the greatest reduction in interferon antagonism. A similar reduction in binding and activity was seen for a deletion of twenty amino acids constituting the described STAT1-binding domain. To investigate the contribution of this STAT1-binding motif in NiV-mediated disease, we produced rNiVs with complete deletion of the STAT1-binding motif or the Y116E mutation for ferret challenge studies (rNiVM-STAT1blind). Despite the reduced IFN inhibitory function, ferrets challenged with these rNiVM-STAT1blind mutants had a lethal, albeit altered, NiV-mediated disease course. These data, together with our previously published data, suggest that the major role of NiV P, V, and W in NiV-mediated disease in the ferret model are likely to be in the inhibition of viral recognition/innate immune signaling induction with a minor role for inhibition of IFN signaling.


Assuntos
Infecções por Henipavirus/patologia , Infecções por Henipavirus/virologia , Vírus Nipah/fisiologia , Fosfoproteínas/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Sítios de Ligação , Modelos Animais de Doenças , Progressão da Doença , Feminino , Furões , Infecções por Henipavirus/metabolismo , Fosfoproteínas/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Proteínas Virais/genética , Proteínas Estruturais Virais/genética
10.
Int J Biochem Cell Biol ; 117: 105626, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31605752

RESUMO

Although Signal transducer and activator of transcription 1 (STAT1)-mediated transactivation potential is inhibited in cancer cells, the mechanism is poorly understood. In the present study, we implicated long non-coding RNA lncRNA625 in the inhibition of STAT1 activity. LncRNA625 knockdown up-regulated STAT1-mediated transcription and resulted in an increase of STAT1-mediated expression of IFITM2. Conversely, lncRNA625 upregulation inhibited STAT1 reporter activity. Mechanistically, lncRNA625 inhibited STAT1 binding to the promoter of IFITM2 in both untreated cells and following interferon-gamma (IFN-γ) stimulation. LncRNA625 interacted with the DNA-binding (DB) domain of STAT1 and promoted STAT1 interaction with T-cell protein tyrosine phosphatase TC45 to dephosphorylate pSTAT1. Taken together, the results show that lncRNA625 inhibits STAT1-mediated transactivation potential by causing formation of STAT1-TC45 complexes, resulting in STAT1 dephosphorylation.


Assuntos
Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , RNA Longo não Codificante/genética , Fator de Transcrição STAT1/antagonistas & inibidores , Linhagem Celular Tumoral , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , Células HEK293 , Células HeLa , Humanos , Interferon gama/farmacologia , Células K562 , Fosforilação , Regiões Promotoras Genéticas , Proteína Tirosina Fosfatase não Receptora Tipo 2/genética , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , RNA Longo não Codificante/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Transcrição Gênica , Ativação Transcricional , Transfecção
11.
Front Immunol ; 10: 1182, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31191546

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is a life-threatening infectious disease caused by a novel phlebovirus, SFTS virus (SFTSV). Currently, there is no vaccine or antiviral available and the viral pathogenesis remains largely unknown. In this study, we demonstrated that SFTSV infection results in substantial production of serum interferon-γ (IFN-γ) in patients and then that IFN-γ in turn exhibits a robust anti-SFTSV activity in cultured cells, indicating the potential role of IFN-γ in anti-SFTSV immune responses. However, the IFN-γ anti-SFTSV efficacy was compromised once viral infection had been established. Consistently, we found that viral nonstructural protein (NSs) expression counteracts IFN-γ signaling. By protein interaction analyses combined with mass spectrometry, we identified the transcription factor of IFN-γ signaling pathway, STAT1, as the cellular target of SFTSV for IFN-γ antagonism. Mechanistically, SFTSV blocks IFN-γ-triggered STAT1 action through (1) NSs-STAT1 interaction-mediated sequestration of STAT1 into viral inclusion bodies and (2) viral infection-induced downregulation of STAT1 protein level. Finally, the efficacy of IFN-γ as an anti-SFTSV drug in vivo was evaluated in a mouse infection model: IFN-γ pretreatment but not posttreatment conferred significant protection to mice against lethal SFTSV infection, confirming IFN-γ's anti-SFTSV effect and viral antagonism against IFN-γ after the infection establishment. These findings present a picture of virus-host arm race and may promote not only the understanding of virus-host interactions and viral pathogenesis but also the development of antiviral therapeutics.


Assuntos
Infecções por Bunyaviridae/imunologia , Interferon gama/imunologia , Phlebovirus/imunologia , Fator de Transcrição STAT1/imunologia , Animais , Antivirais/administração & dosagem , Antivirais/sangue , Antivirais/imunologia , Infecções por Bunyaviridae/tratamento farmacológico , Infecções por Bunyaviridae/virologia , Chlorocebus aethiops , Células HEK293 , Células Hep G2 , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferon gama/administração & dosagem , Interferon gama/sangue , Camundongos Endogâmicos ICR , Phlebovirus/efeitos dos fármacos , Phlebovirus/fisiologia , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Células Vero , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/metabolismo
12.
Inflammation ; 42(4): 1311-1316, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30820808

RESUMO

Carnosic acid, which is a bioactive compound isolated from rosemary, has various pharmacological effects. However, the anti-inflammatory effect of carnosic acid on periodontitis is still unknown. The aim of this study was to investigate the effect of carnosic acid on CXC chemokine receptor 3 (CXCR3) ligands, which are involved in Th1 cells migration and accumulation, production in interleukin (IL)-27-stimulated human oral epithelial cells (TR146 cells). Carnosic acid decreased CXC chemokine ligand (CXCL)9, CXCL10, and CXCL11 production in IL-27-stimulated TR146 cells in a dose-dependent fashion. Moreover, we disclosed that carnosic acid could suppress signal transducer and activator of transcription (STAT)1, STAT3, and protein kinase B (Akt) phosphorylation in IL-27-stimulated TR146 cells. Furthermore, STAT1, STAT3, and Akt inhibitors could suppress CXCR3 ligands production in IL-27-treated TR146 cells. In summary, carnosic acid could reduce CXCR3 ligands production in human oral epithelial cell by inhibiting STAT1, STAT3, and Akt activation.


Assuntos
Abietanos/farmacologia , Células Epiteliais/metabolismo , Interleucina-27/farmacologia , Receptores CXCR3/biossíntese , Células Cultivadas , Quimiocina CXCL10/antagonistas & inibidores , Quimiocina CXCL11/antagonistas & inibidores , Quimiocina CXCL9/antagonistas & inibidores , Humanos , Ligantes , Periodontite/tratamento farmacológico , Periodontite/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT3/antagonistas & inibidores
13.
Inflammopharmacology ; 27(6): 1255-1263, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30783895

RESUMO

Influenza viruses can bring about acute respiratory diseases and are a potential hazard to human health. Antiviral drugs are the main ways to control the influenza virus infection except the vaccine. In this study, the immune regulation activity of pterodontic acid isolated from Laggera pterodonta induced by influenza A virus in vitro was evaluated. In studies on anti-influenza activity, our results showed that it maybe target the influenza protein of polymerase basic 1 (PB1), polymerase basic 2 (PB2), polymerase acid (PA), nuclear protein (NP), non-structural protein (NS), and matrix protein (M) but not hemagglutinin (HA) and neuraminidase (NA). In studies on immune regulation, our results demonstrated that pterodontic acid can inhibit the Retinoic acid inducible gene-I (RIG-I) expression in mRNA and protein level at 100 µg/ml, then further to clarify its action on the signalling pathway, The results indicated that pterodontic acid can inhibit the Tumor Necrosis Factor-related Apoptosis-inducing Ligand/Fas Ligand (TRAIL/Fasl) expression in mRNA level at 100 µg/ml; the cleaved caspase 3/7, p-NF-KB, and p-ERK were all suppressed in protein level by pterodontic acid at 100 µg/ml. This confirmed its mechanism that restrained the nuclear export of viral RNPs. The interferon system was also affected, the STAT1, IFN-α, IFN-ß expression were also inhibited by pterodontic acid at 25-100 µg/ml and also, the important programmed death-ligand of PD-L1 and PD-L2 was inhibited at 50-100 µg/ml. The mechanisms of pterodontic acid against influenza virus infection may be a cascade inhibition and it has the anti-inflammatory activity, which has no side effect, and can be as a supplement drug in clinical influenza virus infection.


Assuntos
Antivirais/farmacologia , Asteraceae/química , Antígeno B7-H1/fisiologia , Proteína DEAD-box 58/antagonistas & inibidores , Vírus da Influenza A/efeitos dos fármacos , Interferon Tipo I/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Proteína 2 Ligante de Morte Celular Programada 1/antagonistas & inibidores , Sesquiterpenos/farmacologia , Células A549 , Antígeno B7-H1/antagonistas & inibidores , Humanos , Vírus da Influenza A/fisiologia , Proteína 2 Ligante de Morte Celular Programada 1/fisiologia , Receptores Imunológicos , Ribonucleoproteínas/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores
14.
Curr Cancer Drug Targets ; 19(10): 796-806, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30605061

RESUMO

BACKGROUND: Gefitinib is well-known as a tyrosine kinase inhibitor targeting non-smalllung- cancer (NSCLC) containing EGFR mutations. However, its effectiveness in treating mucoepidermoid carcinoma (MEC) without such EGFR mutations suggests additional targets. OBJECTIVE: The CRTC1-MAML2 (C1-M2) fusion typical for MEC has been proposed to be a gefitinib target. METHODS: To test this hypothesis, we developed a set of siRNAs to down-regulate C1-M2 expression. RNA-seq and Western blot techniques were applied to analyze the effects of gefitinib and siC1-M2 on the transcriptome of and the phosphorylation of tyrosine kinases in a MEC cell line H292. RESULTS: Deep-sequencing transcriptome analysis revealed that gefitinib extensively inhibited transcription of genes in JAK-STAT and MAPK/ERK pathways. Both siC1-M2 and gefitinib inhibited the phosphorylation of multiple signaling kinases in these signaling pathways, indicating that gefitinib inhibited JAK-STAT and MAPK/ERK pathways activated by C1-M2 fusion. Moreover, gefitinib inhibition of EGFR and MAPK/ERK was more effective than that of AKT, JAK2 and STATs, and their dependence on C1-M2 could be uncoupled. Taken together, our results suggest that gefitinib simultaneously represses phosphorylation of multiple key signaling proteins which are activated in MEC, in part by C1-M2 fusion. Gefitinib-repressed kinase phosphorylation explains the transcriptional repression of genes in JAK-STAT and MAPK/ERK pathways. CONCLUSION: These findings provide new insights into the efficacy of gefitinib in treating mucoepidermoid carcinoma, and suggest that a combination of gefitinib and other inhibitors specifically against C1-M2 fusion could be more effective.


Assuntos
Carcinoma Mucoepidermoide/tratamento farmacológico , Gefitinibe/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Janus Quinases/antagonistas & inibidores , Proteínas de Fusão Oncogênica/genética , Fator de Transcrição STAT1/antagonistas & inibidores , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Mucoepidermoide/metabolismo , Carcinoma Mucoepidermoide/patologia , Linhagem Celular Tumoral , Humanos , Proteínas de Fusão Oncogênica/metabolismo , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
J Pharm Pharmacol ; 71(1): 93-103, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28990659

RESUMO

OBJECTIVES: St John's wort extract (SJW) and its component hyperforin (HPF) were shown to potently inhibit cytokine-induced STAT-1 and NF-κB activation in pancreatic ß cells and protect them against injury. This study aimed at exploring the time course of STAT-1 inhibition afforded by these natural compounds in the ß-cell line INS-1E. METHODS: INS-1E cells were pre-incubated with SJW extract (2-5 µg/ml) or HPF (0.5-2 µm) and then exposed to a cytokine mixture. In some experiments, these compounds were added after or removed before cytokine exposure. STAT-1 activation was assessed by electrophoretic mobility shift assay, apoptosis by caspase-3 activity assay, mRNA gene expression by RT-qPCR. KEY FINDINGS: Pre-incubation with SJW/HPF for 1-2 h exerted a remarkable STAT-1 downregulation, which was maintained upon removal of the compounds before early or delayed cytokine addition. When the protective compounds were added after cell exposure to cytokines, between 15 and 90 min, STAT-1 inhibition also occurred at a progressively decreasing extent. Upon 24-h incubation, SJW and HPF counteracted cytokine-induced ß-cell dysfunction, apoptosis and target gene expression. CONCLUSIONS: SJW and HPF confer to ß cells a state of 'cytokine resistance', which can be elicited both before and after cytokine exposure and safeguards these cells from deleterious cytokine effects.


Assuntos
Hypericum/química , Células Secretoras de Insulina/efeitos dos fármacos , Floroglucinol/análogos & derivados , Extratos Vegetais/farmacologia , Terpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Floroglucinol/administração & dosagem , Floroglucinol/isolamento & purificação , Floroglucinol/farmacologia , Extratos Vegetais/administração & dosagem , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/metabolismo , Terpenos/administração & dosagem , Terpenos/isolamento & purificação , Fatores de Tempo
16.
Br J Pharmacol ; 176(2): 317-333, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30338847

RESUMO

BACKGROUND AND PURPOSE: Sepsis is a serious clinical condition with a high mortality rate. Anti inflammatory agents have been found to be beneficial for the treatment of sepsis. Here, we have evaluated the anti-inflammatory activity of seselin in models of sepsis and investigated the underlying molecular mechanism(s). EXPERIMENTAL APPROACH: In vivo therapeutic effects of seselin was evaluated in two models of sepsis, caecal ligation and puncture or injection of LPS, in C57BL/6 mice. In vitro, anti-inflammatory activity of seselin was assessed with macrophages stimulated with LPS and IFN-γ. Anti inflammatory actions were analysed with immunohistochemical methods, ELISA and Western blotting. Flow cytometry was used to assess markers of macrophage phenotype (pro- or anti-inflammatory). Other methods used included co-immunoprecipitation, cellular thermal shift assay and molecular docking. KEY RESULTS: In vivo, seselin clearly ameliorated sepsis induced by caecal ligation and puncture. In lung tissue from septic mice and in cultured macrophages, seselin down-regulated levels of proinflammatory factors and activity of STAT1 and p65, the master signal pathway molecules for polarization of macrophages into the proinflammatory phenotype. Importantly, adoptive transfer of bone marrow-derived macrophages, pretreated with seselin, lowered systemic proinflammatory factors in mice challenged with LPS. The underlying mechanism was that seselin targeted Jak2 to block interaction with IFNγ receptors and downstream STAT1. CONCLUSIONS AND IMPLICATIONS: Seselin exhibited anti-inflammatory activity through its action on Jak2. These results indicated a possible application of seselin to the treatment of inflammatory disease via blocking the development of the proinflammatory phenotype of macrophages.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Cumarínicos/farmacologia , Inflamação/tratamento farmacológico , Janus Quinase 2/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Sepse/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Cumarínicos/administração & dosagem , Modelos Animais de Doenças , Feminino , Inflamação/metabolismo , Janus Quinase 2/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Fenótipo , Receptores de Interferon/antagonistas & inibidores , Receptores de Interferon/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/metabolismo , Sepse/metabolismo
17.
Carbohydr Polym ; 197: 540-547, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30007645

RESUMO

The present study examined the role of a polysaccharide (LSP, 25 and 100 µg/ml) from the fruiting bodies of Lepista sordid on the immunosuppressive enzyme indoleamine 2, 3-dioxygenase (IDO) in HepG2 cells, and the possible mechanism of action. IDO expression and kynurenine production from LSP-treated HepG2 cells following IFN-γ stimulation were dramatically inhibited by LSP treatment. In line with this, the medium of HepG2 cells pretreated with LSP improved the survival rate of primary CD4+ and CD8+ T cells as compared with IFN-γ-treated control cells. Moreover, tyrosine 701 and serine 727 phosphorylation of STAT1 were dramatically reduced by LSP pretreatment in IFN-γ-stimulated HepG2 cells. Furthermore phosphorylation of JAK-1 and JAK-2 was also inhibited by LSP. Additionally, two IDO promoters (GAS and ISRE) were inhibited in cells pretreated with LSP prior to IFN-γ exposure. These findings suggest that LSP exerts antitumor effects on HepG2 cells by inhibiting IDO via JAK-PKC-δ-STAT1 signaling pathway.


Assuntos
Inibidores Enzimáticos/farmacologia , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Janus Quinase 1/antagonistas & inibidores , Polissacarídeos/farmacologia , Proteína Quinase C-delta/antagonistas & inibidores , Fator de Transcrição STAT1/antagonistas & inibidores , Animais , Relação Dose-Resposta a Droga , Descoberta de Drogas , Inibidores Enzimáticos/química , Inibidores Enzimáticos/isolamento & purificação , Carpóforos/química , Células Hep G2 , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Janus Quinase 1/metabolismo , Lepisma/química , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Proteína Quinase C-delta/metabolismo , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
18.
Int J Mol Sci ; 19(5)2018 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-29786649

RESUMO

Kudzu (Pueraria montana var. lobata (Willd.) Sanjappa & Pradeep) is a perennial leguminous vine, and its root and flower have been used for herbal medicine in Asia for a long time. Most dietary flavonoids are reported to be concentrated in its root, not in its aerial parts including leaves. In this study, we investigated whether kudzu leaf and its major constituent, robinin (kaempferol-3-O-robinoside-7-O-rhanmoside) possessed anti-inflammatory activity. To test this hypothesis, we used peritoneal macrophages isolated from BALB/c mice and stimulated the cells with lipopolysaccharide (LPS) or LPS plus interferon (IFN)-γ. Compared with kudzu root extract, its leaf extract was more potent in inhibiting the production of inducible nitric oxide synthase (iNOS), cyclooxygenase-2, tumor necrosis factor-α, and interleukin-6. Kudzu leaf extract decreased LPS-induced activation of c-Jun N-terminal kinase (JNK) and TANK-binding kinase 1(TBK1) with no effects on nuclear factor-κB and activator protein 1 transcriptional activity. Also, kudzu leaf extract inhibited LPS/IFN-γ-induced signal transducer and activator of transcription 1 (STAT1) activation partly via an altered level of STAT1 expression. Robinin, being present in 0.46% of dry weight of leaf extract, but almost undetected in the root, decreased iNOS protein involving modulation of JNK and STAT1 activation. However, robinin showed no impact on other inflammatory markers. Our data provide evidence that kudzu leaf is an excellent food source of as yet unknown anti-inflammatory constituents.


Assuntos
Inibidores Enzimáticos/farmacologia , Macrófagos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Pueraria/química , Animais , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Interleucina-6/metabolismo , MAP Quinase Quinase 4/antagonistas & inibidores , MAP Quinase Quinase 4/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico Sintase Tipo II/metabolismo , Folhas de Planta/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
19.
Mol Med Rep ; 17(2): 2515-2522, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29207093

RESUMO

Inflammatory skin disease are caused by multiple factors, including susceptibility genes, and immunologic and environmental factors, and are characterized by an increase in epidermal thickness and the infiltration of macrophages, keratinocytes, mast cells, eosinophils and other inflammatory cells. Keratinocytes may serve an important role in the pathogenesis of inflammatory skin diseases. The traditional herbal decoction Hyangso­san (HSS) has been used to treat symptoms of the common cold, including headache, pantalgia, fever and chills. However, to the best of our knowledge, there is no evidence regarding whether HSS has an effect on inflammatory skin diseases. The present study investigated the anti­skin inflammation activity of HSS using the HaCaT human keratinocyte cell line. The mRNA expression and production of inflammatory chemokines, including C­C motif chemokine ligand 22 (CCL22), CCL5, CCL17, and interleukin (IL)­8, was measured using reverse transcription polymerase chain reaction and ELISA analyses. Moreover, we evaluated the effect of HSS on signal transducer and activator of transcription 1 (STAT1) pathway in HaCaT cells. The cells were stimulated with tumor necrosis factor­α (TNF­α) and interferon­Î³ (IFN­Î³) to induce an inflammatory reaction. In the TNF­α­ and IFN­Î³­stimulated cells, the production and expression of inflammatory chemokines were observed, including CCL22, CCL5, CCL17 and IL­8. In addition, stimulation with TNF­α and IFN­Î³ increased the phosphorylation and nuclear translocation of STAT1 in HaCaT cells. By contrast, HSS extract treatment inhibited TNF­α­ and IFN­Î³­induced STAT1 activation. Results from the present study indicated that HSS exhibited inhibitory effects on TNF­α­ and IFN­Î³­mediated chemokine production and expression by targeting STAT1 in keratinocytes. Overall, the results indicated that HSS may be a potential candidate therapeutic drug for inflammatory skin diseases such as atopic dermatitis.


Assuntos
Anti-Inflamatórios/farmacologia , Quimiocinas/biossíntese , Dermatite/metabolismo , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Extratos Vegetais/farmacologia , Fator de Transcrição STAT1/antagonistas & inibidores , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Quimiocinas/genética , Cromatografia Líquida de Alta Pressão , Citocinas/biossíntese , Citocinas/genética , Dermatite/tratamento farmacológico , Dermatite/etiologia , Dermatite/patologia , Relação Dose-Resposta a Droga , Expressão Gênica , Humanos , Extratos Vegetais/química
20.
Endocrinology ; 159(1): 297-309, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29029022

RESUMO

The goal of this study was to characterize the function of microRNA-27a-3p (miR-27a-3p) in polycystic ovary syndrome (PCOS). miR-27a-3p expression was analyzed in excised granulosa cells (GCs) from 21 patients with PCOS and 12 normal patients undergoing in vitro fertilization cycle treatments and in 17 nontreated cuneiform ovarian resection PCOS samples and 13 control ovarian samples from patients without PCOS. We found that the expression of miR-27a-3p was significantly increased in both excised GCs and the ovaries of patients with PCOS compared with the controls. Insulin treatment of the human granulosa-like tumor cell line (KGN) resulted in decreased downregulated expression of miR-27a-3p, and this effect appeared to be mediated by signal transducer and activator of transcription STAT1 and STAT3. The overexpression of miR-27a-3p in KGN cells inhibited SMAD5, which in turn decreased cell proliferation and promoted cell apoptosis. After KGN cells were stimulated with insulin for 48 hours, there was increased expression of SMAD5 protein and decreased apoptosis. Additionally, knockdown/overexpression of SMAD5 in KGN cells reduced/increased cell number and promoted/inhibited cell apoptosis. Insulin-stimulated primary GCs isolated from patients with PCOS, in contrast to normal GCs or KGN cells, did not exhibit decreased miR-27a-3p expression. The differences in the expression levels in KGN cells and human PCOS GCs are likely explained by increased miR-27a-3p expression in the GCs caused by insulin resistance in PCOS. Taken together, our data provided evidence for a functional role of miR-27a-3p in the GCs' dysfunction that occurs in patients with PCOS.


Assuntos
Apoptose , Regulação da Expressão Gênica , Células da Granulosa/metabolismo , MicroRNAs/metabolismo , Síndrome do Ovário Policístico/metabolismo , Proteína Smad5/antagonistas & inibidores , Adulto , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Tumor de Células da Granulosa/tratamento farmacológico , Tumor de Células da Granulosa/metabolismo , Tumor de Células da Granulosa/patologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/patologia , Humanos , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Resistência à Insulina , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/patologia , RNA/metabolismo , Interferência de RNA , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Proteína Smad5/agonistas , Proteína Smad5/genética , Proteína Smad5/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA