Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 12: 752787, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35069190

RESUMO

Tight junctions (TJ) are formed by transmembrane and intracellular proteins that seal the intercellular space and control selective permeability of epithelia. Integrity of the epithelial barrier is central to tissue homeostasis and barrier dysfunction has been linked to many pathological conditions. TJ support the maintenance of cell polarity through interactions with the Par complex (Cdc42-Par-6-Par-3-aPKC) in which Par-6 is an adaptor and links the proteins of the complex together. Studies have shown that Par-6 overexpression delays the assembly of TJ proteins suggesting that Par-6 negatively regulates TJ assembly. Because restoring barrier integrity is of key therapeutic and prophylactic value, we focus on finding compounds that have epithelial barrier reinforcement properties; we developed a screening platform (theLiTE™) to identify compounds that modulate Par-6 expression in follicular epithelial cells from Par-6-GFP Drosophila melanogaster egg chambers. Hits identified were then tested whether they improve epithelial barrier function, using measurements of transepithelial electrical resistance (TEER) or dye efflux to evaluate paracellular permeability. We tested 2,400 compounds, found in total 10 hits. Here we present data on six of them: the first four hits allowed us to sequentially build confidence in theLiTE™ and two compounds that were shortlisted for further development (myricetin and quercetin). We selected quercetin due to its clinical and scientific validation as a compound that regulates TJ; food supplement formulated on the basis of this discovery is currently undergoing clinical evaluation in gastroesophageal reflux disease (GERD) sufferers.

2.
Cell Rep ; 8(1): 126-36, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-24981859

RESUMO

Nitric oxide (NO) and carbon monoxide (CO) are gasotransmitters that suppress the development of severe forms of malaria associated with Plasmodium infection. Here, we addressed the mechanism underlying their protective effect against experimental cerebral malaria (ECM), a severe form of malaria that develops in Plasmodium-infected mice, which resembles, in many aspects, human cerebral malaria (CM). NO suppresses the pathogenesis of ECM via a mechanism involving (1) the transcription factor nuclear factor erythroid 2-related factor 2 (NRF-2), (2) induction of heme oxygenase-1 (HO-1), and (3) CO production via heme catabolism by HO-1. The protection afforded by NO is associated with inhibition of CD4(+) T helper (TH) and CD8(+) cytotoxic (TC) T cell activation in response to Plasmodium infection via a mechanism involving HO-1 and CO. The protective effect of NO and CO is not associated with modulation of host pathogen load, suggesting that these gasotransmitters establish a crosstalk-conferring disease tolerance to Plasmodium infection.


Assuntos
Monóxido de Carbono/farmacologia , Tolerância Imunológica , Malária Cerebral/imunologia , Óxido Nítrico/farmacologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Monóxido de Carbono/metabolismo , Monóxido de Carbono/uso terapêutico , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Ativação Linfocitária , Malária Cerebral/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico/uso terapêutico , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia
3.
Redox Biol ; 1: 347-52, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24024170

RESUMO

Hydrogen peroxide (H2O2) at moderate steady-state concentrations synergizes with TNF-α, leading to increased nuclear levels of NF-κB p65 subunit and to a cell-type specific up-regulation of a limited number of NF-κB-dependent genes. Here, we address how H2O2 achieves this molecular specificity. HeLa and MCF-7 cells were exposed to steady-state H2O2 and/or TNF-α and levels of c-Rel, p65, IκB-α, IκB-ß and IκB-ε were determined. For an extracellular concentration of 25 µM H2O2, the intracellular H2O2 concentration is 3.7 µM and 12.5 µM for respectively HeLa and MCF-7 cells. The higher cytosolic H2O2 concentration present in MCF-7 cells may be a contributing factor for the higher activation of NF-κB caused by H2O2 in this cell line, when compared to HeLa cells. In both cells lines, H2O2 precludes the recovery of TNF-α-dependent IκB-α degradation, which may explain the observed synergism between H2O2 and TNF-α concerning p65 nuclear translocation. In MCF-7 cells, H2O2, in the presence of TNF-α, tripled the induction of c-Rel triggered either by TNF-α or H2O2. Conversely, in HeLa cells, H2O2 had a small antagonistic effect on TNF-α-induced c-Rel nuclear levels, concomitantly with a 50 % induction of IκB-ε, the preferential inhibitor protein of c-Rel dimers. The 6-fold higher c-Rel/IκB-ε ratio found in MCF-7 cells when compared with HeLa cells, may be a contributing factor for the cell-type dependent modulation of c-Rel by H2O2. Our results suggest that H2O2 might have an important cell-type specific role in the regulation of c-Rel-dependent processes, e.g. cancer or wound healing.


Assuntos
Peróxido de Hidrogênio/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Células HeLa , Humanos , Proteínas I-kappa B/metabolismo , Células MCF-7 , Inibidor de NF-kappaB alfa , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais
4.
Methods Enzymol ; 528: 173-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23849865

RESUMO

NF-κB is a transcription factor that plays key roles in health and disease. Learning how this transcription factor is regulated by hydrogen peroxide (H2O2) has been slowed down by the lack of methodologies suitable to obtain quantitative data. Literature is abundant with apparently contradictory information on whether H2O2 activates or inhibits NF-κB. There is increasing evidence that H2O2 is not just a generic modulator of transcription factors and signaling molecules but becomes a specific regulator of individual genes. Here, we describe a detailed protocol to obtain rigorous quantitative data on the effect of H2O2 on members of the NF-κB/Rel and IκB families, in which H2O2 is delivered as a steady-state addition instead of the usual bolus addition. Solutions, pilot experiments, and experimental set-ups are fully described. In addition, we outline a protocol to measure the impact of alterations in the promoter κB regions on the H2O2 regulation of the expression of individual genes. As important as evaluating the effects of H2O2 alone is the evaluation of the modulation elicited by this oxidant on cytokine regulation of NF-κB. We illustrate this for the cytokine tumor necrosis factor alpha.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Glucose/metabolismo , Glucose Oxidase/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , NF-kappa B/agonistas , NF-kappa B/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-rel/genética , Proteínas Proto-Oncogênicas c-rel/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo
5.
Antioxid Redox Signal ; 11(9): 2223-43, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19496701

RESUMO

Hydrogen peroxide (H2O2) has been implicated in the regulation of the transcription factor NF-kappaB, a key regulator of the inflammatory process and adaptive immunity. However, no consensus exists regarding the regulatory role played by H2O2. We discuss how the experimental methodologies used to expose cells to H2O2 produce inconsistent results that are difficult to compare, and how the steady-state titration with H2O2 emerges as an adequate tool to overcome these problems. The redox targets of H2O2 in the NF-kappaB pathway--from the membrane to the post-translational modifications in both NF-kappaB and histones in the nucleus--are described. We also review how H2O2 acts as a specific regulator at the level of the single gene, and briefly discuss the implications of this regulation for human health in the context of kappaB polymorphisms. In conclusion, after near 30 years of research, H2O2 emerges not as an inducer of NF-kappaB, but as an agent able to modulate the activation of the NF-kappaB pathway by other agents. This modulation is generic at the level of the whole pathway but specific at the level of the single gene. Therefore, H2O2 is a fine-tuning regulator of NF-kappaB-dependent processes, as exemplified by its dual regulation of inflammation.


Assuntos
Peróxido de Hidrogênio/metabolismo , NF-kappa B/metabolismo , Núcleo Celular/metabolismo , Humanos , Interleucina-1/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Ativação Transcricional , Fator de Necrose Tumoral alfa/metabolismo
6.
Antioxid Redox Signal ; 11(9): 2043-53, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19014314

RESUMO

We recently observed that H2O2 regulates inflammation via upexpression of a few NF-kappaB-dependent genes, while leaving expression of most NF-kappaB-dependent genes unaltered. Here we test the hypothesis that this differential gene expression depends on the apparent affinity of kappaB sites in the gene-promoter regions toward NF-kappaB. Accordingly, cells were transfected with three reporter plasmids containing kappaB sequences with different affinities for NF-kappaB. It was observed that the lower the affinity, the higher the range of TNF-alpha concentrations where H2O2 upregulated gene expression. Mathematical models reproduced the key experimental observations indicating that H2O2 upregulation ceased when NF-kappaB fully occupied the kappaB sites. In vivo, it is predicted that genes with high-affinity sites remain insensitive to H2O2, whereas genes with lower-affinity sites are upregulated by H2O2. In conclusion, a simple chemical mechanism is at the root of a complex biologic process such as differential gene expression caused by H2O2.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , NF-kappa B/fisiologia , Sequência de Bases , Primers do DNA , Genes Reporter , Células HeLa , Humanos , Transcrição Gênica
7.
J Immunol ; 178(6): 3893-902, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17339489

RESUMO

Although the germicide role of H(2)O(2) released during inflammation is well established, a hypothetical regulatory function, either promoting or inhibiting inflammation, is still controversial. In particular, after 15 years of highly contradictory results it remains uncertain whether H(2)O(2) by itself activates NF-kappaB or if it stimulates or inhibits the activation of NF-kappaB by proinflammatory mediators. We investigated the role of H(2)O(2) in NF-kappaB activation using, for the first time, a calibrated and controlled method of H(2)O(2) delivery--the steady-state titration--in which cells are exposed to constant, low, and known concentrations of H(2)O(2). This technique contrasts with previously applied techniques, which disrupt cellular redox homeostasis and/or introduce uncertainties in the actual H(2)O(2) concentration to which cells are exposed. In both MCF-7 and HeLa cells, H(2)O(2) at extracellular concentrations up to 25 microM did not induce significantly per se NF-kappaB translocation to the nucleus, but it stimulated the translocation induced by TNF-alpha. For higher H(2)O(2) doses this stimulatory role shifts to an inhibition, which may explain published contradictory results. The stimulatory role was confirmed by the observation that 12.5 microM H(2)O(2), a concentration found during inflammation, increased the expression of several proinflammatory NF-kappaB-dependent genes induced by TNF-alpha (e.g., IL-8, MCP-1, TLR2, and TNF-alpha). The same low H(2)O(2) concentration also induced the anti-inflammatory gene coding for heme oxygenase-1 (HO-1) and IL-6. We propose that H(2)O(2) has a fine-tuning regulatory role, comprising both a proinflammatory control loop that increases pathogen removal and an anti-inflammatory control loop, which avoids an exacerbated harmful inflammatory response.


Assuntos
Núcleo Celular/metabolismo , Peróxido de Hidrogênio/farmacologia , NF-kappa B/metabolismo , Oxidantes/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/imunologia , Núcleo Celular/imunologia , Quimiocina CCL2/biossíntese , Quimiocina CCL2/imunologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Células HeLa , Heme Oxigenase-1 , Homeostase/efeitos dos fármacos , Homeostase/imunologia , Humanos , Peróxido de Hidrogênio/agonistas , Peróxido de Hidrogênio/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-6/biossíntese , Interleucina-6/imunologia , Interleucina-8/biossíntese , Interleucina-8/imunologia , NF-kappa B/imunologia , Oxidantes/agonistas , Oxirredução/efeitos dos fármacos , Receptor 2 Toll-Like/biossíntese , Receptor 2 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/agonistas , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA