Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 204(4): 933-942, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31900337

RESUMO

Otitis media (OM) is the most common bacterial infection in children. It remains a major health problem and a substantial socioeconomic burden. Streptococcus pneumoniae (S. pneumoniae) is one of the most common bacterial pathogens causing OM. Innate inflammatory response plays a critical role in host defense against bacterial pathogens. However, if excessive, it has a detrimental impact on the middle ear, leading to middle ear inflammation, a hallmark of OM. Currently, there has been limited success in developing effective therapeutic agents to suppress inflammation without serious side effects. In this study, we show that vinpocetine, an antistroke drug, suppressed S. pneumoniae-induced inflammatory response in cultured middle ear epithelial cells as well as in the middle ear of mice. Interestingly, vinpocetine inhibited S. pneumoniae-induced inflammation via upregulating a key negative regulator cylindromatosis (CYLD). Moreover, CYLD suppressed S. pneumoniae-induced inflammation via inhibiting the activation of ERK. Importantly, the postinfection administration of vinpocetine markedly inhibited middle ear inflammation induced by S. pneumoniae in a well-established mouse OM model. These studies provide insights into the molecular mechanisms underlying the tight regulation of inflammation via inhibition of ERK by CYLD and identified vinpocetine as a potential therapeutic agent for suppressing the inflammatory response in the pathogenesis of OM via upregulating negative regulator CYLD expression.


Assuntos
Enzima Desubiquitinante CYLD/metabolismo , Otite Média/tratamento farmacológico , Infecções Pneumocócicas/tratamento farmacológico , Alcaloides de Vinca/farmacologia , Animais , Linhagem Celular , Enzima Desubiquitinante CYLD/genética , Modelos Animais de Doenças , Orelha Média/citologia , Orelha Média/efeitos dos fármacos , Orelha Média/imunologia , Células Epiteliais , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Camundongos Knockout , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Otite Média/imunologia , Otite Média/microbiologia , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/microbiologia , RNA Interferente Pequeno/metabolismo , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/isolamento & purificação , Regulação para Cima/efeitos dos fármacos , Alcaloides de Vinca/uso terapêutico
2.
Bioorg Med Chem ; 25(3): 1202-1218, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28057407

RESUMO

We herein disclose a series of compounds with potent inhibitory activities towards histone deacetylases (HDAC) and cyclooxygenases (COX). These compounds potently inhibited the growth of cancer cell lines consistent with their anti-COX and anti-HDAC activities. While compound 2b showed comparable level of COX-2 selectivity as celecoxib, compound 11b outperformed indomethacin in terms of selectivity towards COX-2 relative to COX-1. An important observation with our lead compounds (2b, 8, 11b, and 17b) is their enhanced cytotoxicity towards androgen dependent prostate cancer cell line (LNCaP) relative to androgen independent prostate cancer cell line (DU-145). Interestingly, compounds 2b and 17b arrested the cell cycle progression of LNCaP in the S-phase, while compound 8 showed a G0/G1 arrest, similar to SAHA. Relative to SAHA, these compounds displayed tumor-selective cytotoxicity as they have low anti-proliferative activity towards healthy cells (VERO); an attribute that makes them attractive candidates for drug development.


Assuntos
Antineoplásicos/farmacologia , Celecoxib/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Indometacina/farmacologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Antineoplásicos/síntese química , Antineoplásicos/química , Celecoxib/síntese química , Celecoxib/química , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/síntese química , Inibidores de Ciclo-Oxigenase/química , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/química , Humanos , Indometacina/síntese química , Indometacina/química , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
3.
PLoS One ; 10(12): e0144840, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26669856

RESUMO

Airway diseases such as asthma and chronic obstructive pulmonary disease (COPD) are characterized by excessive inflammation and are exacerbated by nontypeable Haemophilus influenzae (NTHi). Airway epithelial cells mount the initial innate immune responses to invading pathogens and thus modulate inflammation. While inflammation is necessary to eliminate a pathogen, excessive inflammation can cause damage to the host tissue. Therefore, the inflammatory response must be tightly regulated and deciphering the signaling pathways involved in this response will enhance our understanding of the regulation of the host inflammatory response. NTHi binds to TLR2 and signal propagation requires the adaptor molecule myeloid differentiation factor 88 (MyD88). An alternative spliced form of MyD88 is called MyD88 short (MyD88s) and has been identified in macrophages and embryonic cell lines as a negative regulator of inflammation. However, the role of MyD88s in NTHi-induced inflammation in airway epithelial cells remains unknown. Here we show that NTHi induces MyD88s expression and MyD88s is a negative regulator of inflammation in airway epithelial cells. We further demonstrate that MyD88s is positively regulated by IKKß and CREB and negatively regulated by ERK1/2 signaling pathways. Taken together these data indicate that airway inflammation is controlled in a negative feedback manner involving MyD88s and suggest that airway epithelial cells are essential to maintain immune homeostasis.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Haemophilus influenzae/fisiologia , Quinase I-kappa B/metabolismo , Sistema de Sinalização das MAP Quinases , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Infecções por Haemophilus/complicações , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/patologia , Humanos , Inflamação/complicações , Inflamação/patologia , Pulmão/patologia , Camundongos Endogâmicos C57BL , Modelos Biológicos
5.
PLoS One ; 9(11): e112516, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25389768

RESUMO

Nontypeable Haemophilus influenzae (NTHi), a Gram-negative bacterium, is the primary cause of otitis media in children and the exacerbation of chronic obstructive pulmonary disease in adults. A hallmark of both diseases is an overactive inflammatory response, including the upregulation of chemokines, such as interleukin-8 (IL-8). An appropriate inflammatory response is essential for eradicating pathogens. However, excessive inflammation can cause host tissue damage. Therefore, expression of IL-8 must be tightly regulated. We previously reported that NTHi induces IL-8 expression in an ERK-dependent manner. We also have shown that the deubiquitinase cylindromatosis (CYLD) suppresses NTHi-induced inflammation. However, the underlying molecular mechanism of how CYLD negatively regulates ERK-mediated IL-8 production is largely unknown. Here, we examine both human lung epithelial A549 cells and lung of Cyld-/- mice to show that CYLD specifically targets the activation of ERK. Interestingly, CYLD enhances NTHi-induced upregulation of another negative regulator, MAP Kinase Phosphatase-1 (MKP-1), which, in turn, leads to reduced ERK activation and subsequent suppression of IL-8. Taken together, the CYLD suppression of ERK-dependent IL-8 via MKP-1 may bring novel insights into the tight regulation of inflammatory responses and also lead to innovative therapeutic strategies for controlling these responses by targeting key negative regulators of inflammation.


Assuntos
Cisteína Endopeptidases/genética , Fosfatase 1 de Especificidade Dupla/genética , Infecções por Haemophilus/genética , Interleucina-8/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteínas Supressoras de Tumor/genética , Animais , Linhagem Celular Tumoral , Cisteína Endopeptidases/deficiência , Enzima Desubiquitinante CYLD , Fosfatase 1 de Especificidade Dupla/metabolismo , Regulação da Expressão Gênica , Infecções por Haemophilus/metabolismo , Infecções por Haemophilus/patologia , Haemophilus influenzae/genética , Haemophilus influenzae/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Interleucina-8/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo
6.
Nat Commun ; 4: 1684, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23575688

RESUMO

The deubiquitinase CYLD acts as a key negative regulator to tightly control overactive inflammation. Most anti-inflammatory strategies have focused on directly targeting the positive regulator, which often results in significant side effects such as suppression of the host defence response. Here, we show that inhibition of phosphodiesterase 4B (PDE4B) markedly enhances upregulation of CYLD expression in response to bacteria, thereby suggesting that PDE4B acts as a negative regulator for CYLD. Interestingly, in Cyld-deficient mice, inhibition of PDE4B no longer suppresses inflammation. Moreover, PDE4B negatively regulates CYLD via specific activation of JNK2 but not JNK1. Importantly, ototopical post-inoculation administration of a PDE4 inhibitor suppresses inflammation in this animal model, thus demonstrating the therapeutic potential of targeting PDE4. These studies provide insights into how inflammation is tightly regulated via the inhibition of its negative regulator and may also lead to the development of new anti-inflammatory therapeutics that upregulate CYLD expression.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/efeitos dos fármacos , Inflamação/prevenção & controle , Inibidores de Fosfodiesterase/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Células Cultivadas , Enzima Desubiquitinante CYLD , Regulação para Baixo , Ativação Enzimática , Humanos , Inflamação/enzimologia , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Regulação para Cima
7.
J Biol Chem ; 287(27): 22799-811, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22610099

RESUMO

Otitis media (OM) is the most common childhood bacterial infection and the major cause of conductive hearing loss in children. Mucus overproduction is a hallmark of OM. Streptococcus pneumoniae is the most common gram-positive bacterial pathogen causing OM. Among many mucin genes, MUC5AC has been found to be greatly up-regulated in the middle ear mucosa of human patients with OM. We previously reported that S. pneumoniae up-regulates MUC5AC expression in a MAPK ERK-dependent manner. We also found that MAPK phosphatase-1 (MKP-1) negatively regulates S. pneumoniae-induced ERK-dependent MUC5AC up-regulation. Therapeutic strategies for up-regulating the expression of negative regulators such as MKP-1 may have significant therapeutic potential for treating mucus overproduction in OM. However, the underlying molecular mechanism by which MKP-1 expression is negatively regulated during S. pneumoniae infection is unknown. In this study we show that phosphodiesterase 4B (PDE4B) mediates S. pneumoniae-induced MUC5AC up-regulation by inhibiting the expression of a negative regulator MKP-1, which in turn leads to enhanced MAPK ERK activation and subsequent up-regulation of MUC5AC. PDE4B inhibits MKP-1 expression in a cAMP-PKA-dependent manner. PDE4-specific inhibitor rolipram inhibits S. pneumoniae-induced MUC5AC up-regulation both in vitro and in vivo. Moreover, we show that PDE4B plays a critical role in MUC5AC induction. Finally, topical and post-infection administration of rolipram into the middle ear potently inhibited S. pneumoniae-induced MUC5AC up-regulation. Collectively, these data demonstrate that PDE4B mediates ERK-dependent up-regulation of mucin MUC5AC by S. pneumoniae by inhibiting cAMP-PKA-dependent MKP-1 pathway. This study may lead to novel therapeutic strategy for inhibiting mucus overproduction.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Fosfatase 1 de Especificidade Dupla/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Mucina-5AC/metabolismo , Infecções Pneumocócicas/metabolismo , Streptococcus pneumoniae/metabolismo , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Orelha Média/citologia , Orelha Média/imunologia , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Células Epiteliais/microbiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Muco/metabolismo , Otite Média/imunologia , Otite Média/metabolismo , Otite Média/microbiologia , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/microbiologia , RNA Interferente Pequeno/genética , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Regulação para Cima/fisiologia
8.
Nat Commun ; 3: 771, 2012 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-22491319

RESUMO

Lung injury, whether induced by infection or caustic chemicals, initiates a series of complex wound-healing responses. If uncontrolled, these responses may lead to fibrotic lung diseases and loss of function. Thus, resolution of lung injury must be tightly regulated. The key regulatory proteins required for tightly controlling the resolution of lung injury have yet to be identified. Here we show that loss of deubiquitinase CYLD led to the development of lung fibrosis in mice after infection with Streptococcus pneumoniae. CYLD inhibited transforming growth factor-ß-signalling and prevented lung fibrosis by decreasing the stability of Smad3 in an E3 ligase carboxy terminus of Hsc70-interacting protein-dependent manner. Moreover, CYLD decreases Smad3 stability by deubiquitinating K63-polyubiquitinated Akt. Together, our results unveil a role for CYLD in tightly regulating the resolution of lung injury and preventing fibrosis by deubiquitinating Akt. These studies may help develop new therapeutic strategies for preventing lung fibrosis.


Assuntos
Cisteína Endopeptidases/metabolismo , Regulação para Baixo , Infecções Pneumocócicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular , Cisteína Endopeptidases/genética , Enzima Desubiquitinante CYLD , Humanos , Pulmão/enzimologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções Pneumocócicas/enzimologia , Infecções Pneumocócicas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Streptococcus pneumoniae/fisiologia , Fator de Crescimento Transformador beta1/genética , Proteínas Supressoras de Tumor/genética , Ubiquitinação
9.
Mol Cell Biol ; 32(8): 1581-90, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22310660

RESUMO

Tumor suppressor genes regulate the antiviral host defense through molecular mechanisms that are not yet well explored. Here, we show that the tumor suppressor retinoblastoma (Rb) protein positively regulates Toll-like receptor 3 (TLR3) expression, the sensing receptor for viral double-stranded RNA and poly(I · C). TLR3 expression was lower in Rb knockout (Rb(-/-)) mouse embryonic fibroblasts (MEF) and in mammalian epithelial cells transfected with Rb small-interfering RNA (siRNA) than in control cells. Consequently, induction of cytokines interleukin-8 and beta interferon after poly(I · C) stimulation was impaired in Rb(-/-) MEF and Rb siRNA-transfected cells compared to controls. TLR3 promoter analysis showed that Rb modulates the transcription factor E2F1, which directly binds to the proximal promoter of TLR3. Exogenous addition of E2F1 decreased TLR3 promoter activity, while Rb dose dependently curbed the effect of E2F1. Interestingly, poly(I · C) increased the Rb expression, and the poly(I · C)-induced TLR3 expression was impaired in Rb-depleted cells, suggesting the importance of Rb in TLR3 induction by poly(I · C). Together, these data indicated that E2F1 suppresses TLR3 transcription, but during immune stimulation, Rb is upregulated to block the inhibitory effect of E2F1 on TLR3, highlighting a role of Rb-E2F1 axis in the innate immune response in epithelial cells.


Assuntos
Fator de Transcrição E2F1/metabolismo , Imunidade Inata/genética , Proteína do Retinoblastoma/metabolismo , Receptor 3 Toll-Like/metabolismo , Animais , Linhagem Celular , Fator de Transcrição E2F1/genética , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Fibroblastos/imunologia , Fibroblastos/metabolismo , Humanos , Camundongos , Proteína do Retinoblastoma/genética , Receptor 3 Toll-Like/genética , Transcrição Gênica
10.
Biochem J ; 417(2): 583-91, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18823280

RESUMO

The TGF-beta (transforming growth factor-beta) pathway represents an important signalling pathway involved in regulating diverse biological processes, including cell proliferation, differentiation and inflammation. Despite the critical role for TGF-beta in inflammatory responses, its role in regulating NF-kappaB (nuclear factor-kappaB)-dependent inflammatory responses still remains unknown. In the present study we show that TGF-beta1 synergizes with proinflammatory cytokine TNF-alpha (tumour necrosis factor-alpha) to induce NF-kappaB activation and the resultant inflammatory response in vitro and in vivo. TGF-beta1 synergistically enhances TNF-alpha-induced NF-kappaB DNA binding activity via induction of RelA acetylation. Moreover, synergistic enhancement of TNF-alpha-induced RelA acetylation and DNA-binding activity by TGF-beta1 is mediated by PKA (protein kinase A). Thus the present study reveals a novel role for TGF-beta in inflammatory responses and provides new insight into the regulation of NF-kappaB by TGF-beta signalling.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , NF-kappa B/metabolismo , Fator de Transcrição RelA/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Acetilação/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/genética , DNA/metabolismo , Humanos , Ligação Proteica , Fator de Transcrição RelA/genética
11.
Biochem Biophys Res Commun ; 377(3): 763-8, 2008 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-18957283

RESUMO

Glucocorticoids are highly effective in the control of many inflammatory and immune diseases. Despite the importance of glucocorticoids in suppressing immune and inflammatory responses, the molecular basis for the inhibitory effect of glucocorticoids on mucin overproduction, a hallmark of chronic respiratory diseases, still remains unclear. Here we show that glucocorticoids markedly inhibit up-regulation of MUC5AC induced by NTHi, a major human bacterial pathogen causing chronic obstructive pulmonary disease and otitis media. Inhibition of NTHi-induced MUC5AC expression by dexamethasone occurs at the level of p38 MAPK via glucocorticoid receptor. Moreover, glucocorticoids up-regulate MKP-1 expression, which in turn leads to p38 dephosphorylation and the subsequent inhibition of NTHi-induced MUC5AC expression. These studies provide new insight into the molecular mechanism underlying glucocorticoid therapy and may lead to novel therapeutic intervention for inhibiting mucin overproduction in patients with NTHi infections.


Assuntos
Dexametasona/uso terapêutico , Glucocorticoides/uso terapêutico , Infecções por Haemophilus/tratamento farmacológico , Haemophilus influenzae , Mucina-5AC/antagonistas & inibidores , Otite Média/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Linhagem Celular Tumoral , Fosfatase 1 de Especificidade Dupla/metabolismo , Infecções por Haemophilus/microbiologia , Humanos , Mucina-5AC/biossíntese , Otite Média/microbiologia , Fosforilação/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/microbiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Mol Cell Biol ; 28(21): 6557-67, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18779317

RESUMO

Toll-like receptors (TLRs) are important sensors of microbial pathogens and mediators of innate immune responses. Although the signal transduction of TLRs is well elucidated, their basal regulation is largely unexplored. Here we show that the tumor suppressor p53 positively regulates the transcription of TLR3, a receptor for viral double-stranded RNA and poly(I-C), by binding to the p53 site in the TLR3 promoter. TLR3 expression was lower in HCT116 p53(-/-) cells than in HCT116 p53(+/+) cells. Activation of p53 by 5-fluorouracil increased the TLR3 mRNA in epithelial cell lines with wild-type p53 but not in cell lines harboring mutant p53. Knockdown of p53 by small interfering RNA decreased the TLR3 expression. TLR3 mRNA was also lower in liver and intestine of p53(-/-) mice than in p53(+/+) mice. Furthermore, the poly(I-C)-induced phosphorylation of IkappaB-alpha, nuclear translocation of NF-kappaB, and phosphorylation of interferon regulatory transcription factor 3, were drastically reduced in HCT116 p53(-/-) cells, indicating a dysregulation of the two signaling pathways governed by TLR3. Consequently, induction of interleukin-8 and beta interferon after poly(I-C) stimulation was impaired in HCT116 p53(-/-) cells. These results suggest that p53 influences TLR3 expression and function and highlight a role of p53 in innate immune response in epithelial cells.


Assuntos
Células Epiteliais/metabolismo , Receptor 3 Toll-Like/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Camundongos , Poli I-C/farmacologia , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 3 Toll-Like/metabolismo , Ativação Transcricional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA