يعرض 1 - 5 نتائج من 5 نتيجة بحث عن '"Manuel D. Díaz-Muñoz"', وقت الاستعلام: 0.85s تنقيح النتائج
  1. 1

    المساهمون: UAM. Departamento de Biología Molecular, Comunidad de Madrid, Red Temática de Investigación Cooperativa en Enfermedades Cardiovasculares (España), European Commission, Ministerio de Ciencia e Innovación (España), Federación Española de Enfermedades Raras, Fundación Ramón Areces

    المصدر: Biblos-e Archivo. Repositorio Institucional de la UAM
    instname
    Digital.CSIC. Repositorio Institucional del CSIC

    الوصف: PG (prostaglandin) E2 plays an important role in the modulation of the immune response and the inflammatory process. In the present study, we describe a PGE2 positive feedback for COX (cyclo oxygenase)-2 and mPGES-1 (microsomal PGES (PGE synthase)-1) expression in the macrophage cell line RAW 264.7. Our results show that PGE2 induces COX-2 and mPGES-1 expression, an effect mimicked by dbcAMP (dibutyryl-cAMP) or forskolin. Furthermore, the cAMP signalling pathway cooperates with LPS (lipopolysaccharide) in the induction of COX-2 and mPGES-1 transcriptional activation. Analysis of the involvement of PGE receptors (EPs (E-prostanoids)) showed that incubation with EP2 agonists up-regulated both COX2 and mPGES-1 mRNA levels. Moreover, EP2 receptor overexpression enhanced the transcriptional activation of COX2 and mPGES-1 promoters. This induction was repressed by the PKA (protein kinase A) inhibitor H89. Activation of the PGE2/EP2/PKA signalling pathway induced the phosphorylation of CREB (CRE (cAMP-response element)-binding protein) in macrophages and stimulated the specific binding of this transcription factor toCOX2 and mPGES-1 promoters. Deletion or mutation of potential CRE sites in both promoters diminished their transcriptional activity. In summary, the results of the present study demonstrate that activation of PKA/CREB signalling through the EP2 receptor by PGE2 plays a key role in the expression of COX-2 and mPGES-1 in activated macrophages
    Comunidad Autónoma de Madrid (CCG08-UAM/BIO-4299); the Cardiovascular Network (RECAVA) of the Instituto de Salud Carlos III (grant number RD06/0014/1013); the European EICOSANOX integrated project (grant number LSH-CT-2004-005033); the Ministerio deCiencia e Innovación; FEDER (grant numbers SAF2007-61716, SAF2010-18733 and BFU2007-62659/BMC, BFU2010-21055 (toM.A.I.); Fundación Ramón Areces

    وصف الملف: Application/pdf

  2. 2

    المصدر: International Immunopharmacology. 10:1295-1304

    الوصف: Anti-inflammatory efficacy of non-steroidal anti-inflammatory drugs (NSAIDs) has been related to their properties as inhibitors of cyclooxygenase (COX)-mediated prostaglandin (PG) synthesis. However, recent studies have suggested that variations of the in vivo anti-inflammatory actions among different NSAIDs could not be solely explained by COX inhibition. Here, we have analyzed the effects on T cell activation of novel 4,5-dihydro-3 trifluoromethyl pyrazole anti-inflammatory drugs with different potencies as COX-2 inhibitors, namely E-6087, E-6232, E-6231, E-6036 and E-6259 as well as the chemically related COX-2 inhibitor Celecoxib. These drugs inhibited mitogen-mediated T cell proliferation as well as Interleukin (IL)-2, tumor necrosis factor (TNF)-α and Interferon (IFN)-γ synthesis by activated T cells, independently of their ability to inhibit COX-2 enzymatic activity. Immunosuppressive effects of these drugs seem to be due to their interference on transcription factor activation as induced transcription from Nuclear Factor (NF)-κB and Nuclear Factor of Activated T cells (NFAT)-dependent enhancers was inhibited in a dose-dependent manner, being the latter effect the most sensitive to the action of those compounds. Both NFAT dephosphorylation, required for its nuclear translocation, as well as transcriptional activity of a GAL4-NFAT chimera were diminished in the presence of these compounds. These findings provide new insights into the molecular mechanisms involved in the immunomodulatory and anti-inflammatory actions of NSAIDs, which may have important implications in anti-inflammatory therapy, through inhibition of NFAT.

  3. 3

    المصدر: Cellular Signalling. 22:1427-1436

    الوصف: Prostaglandin (PG) E(2) is a potent lipid mediator that plays an essential role in inflammation, fever and pain. It is produced from arachidonic acid (AA) by a cascade of enzymatic reactions involving cyclooxygenases (COX-1 and -2) and prostaglandin E synthases (cPGES, mPGES-1 and -2). Functional coupling of the inducible enzymes COX-2 and mPGES-1 has been proposed for increased production of PGE(2) in different cell types. PGE(2) produced by macrophages plays an essential role in the pathogenesis of inflammatory diseases. Here, we have investigated the mechanisms involved in the regulation of COX-2 and mPGES-1 expressions in murine macrophages upon bacterial lipopolysaccharide (LPS) treatment. LPS stimulation induced the coordinated synthesis of COX-2 and mPGES-1 that resulted in an enhanced production of PGE(2) in RAW 264.7 macrophages. Furthermore, we show the involvement of NF-kappaB and Egr-1 transcription factors in the transcriptional induction of these enzymes. LPS treatment promoted specific binding of NF-kappaB to both COX-2 and mPGES-1 promoters. Site-directed mutagenesis, electrophoretic mobility shift assays and ChIP assays allowed the identification of a sequence acting as a NF-kappaB recognition site in the murine mPGES-1 promoter. Furthermore, LPS induced the expression of Egr-1 that cooperated with NF-kappaB in the up-regulation of COX-2 and mPGES-1. Inhibition of Egr-1 expression reduced substantially LPS-mediated induction of COX-2 and mPGES-1 expression, resulting in a decrease in PGE(2) production. Our findings point out to Egr-1 and NF-kappaB cooperation as determinant for PGE2 synthesis by macrophages in inflammatory processes through the coordinated regulation of COX-2 and mPGES-1.

  4. 4

    المصدر: Journal of leukocyte biology. 81(1)

    الوصف: IFN-gamma induces NO production, inducible NO synthase (iNOS) protein, and promoter expression in mouse macrophage cells. Mutation of IFN regulatory factor 1 responsive element, gamma-activated site, as well as NF-kappaB elements in the murine iNOS promoter strongly reduced IFN-gamma-induced iNOS transcriptional activity. The role of NF-kappaB activation in iNOS induction by IFN-gamma was corroborated by overexpression of the NF-kappaB inhibitory protein IkappaBalpha, which inhibited iNOS promoter activity induced by IFN-gamma. In addition, IFN-gamma treatment induced p65 binding to the iNOS promoter by chromatin immunoprecipitation assay and NF-kappaB binding to DNA by EMSA, although with a delayed kinetics, suggesting an indirect autocrine role for another cytokine produced in response to IFN-gamma. It is interesting that we found that IFN-gamma induced TNF-alpha secretion, and the induction of iNOS expression by IFN-gamma was abolished in primary peritoneal macrophages from TNF-alpha-deficient (TNF-alpha-/-) mice or in RAW 264.7 cells treated with anti-TNF-alpha neutralizing antibodies. Moreover, exogenous addition of recombinant mouse TNF-alpha restored iNOS expression induced by IFN-gamma in TNF-alpha-/- mice. It is intriguing that NF-kappaB binding to DNA in response to IFN-gamma treatment was absent in TNF-alpha-/- mice. Taken together, our data suggest that the TNF-alpha produced in response to IFN-gamma is required for iNOS induction by activating NF-kappaB transcription factor.

  5. 5

    المصدر: Cellular signalling. 18(8)

    الوصف: Growing evidence shows that Interleukin (IL)-1beta and Cyclooxygenase 2 (COX-2) play a crucial role in the pathogenesis of inflammatory diseases and tumor growth, particularly in the gastrointestinal tract. Here, we have analyzed the regulation of COX-2 by IL-1beta in the human colon carcinoma cell line Caco-2, showing that COX-2 induction by this cytokine is due to both nuclear factor (NF)-kappaB-dependent transcriptional and p38 mitogen-activated protein kinase (MAPK)-mediated post-transcriptional mechanisms. Treatment of these cells with IL-1beta increased the levels of COX-2 mRNA and protein and hence the production of PGE2. IL-1beta induced NF-kappaB activation in Caco-2 cells, promoting the binding of this transcription factor to DNA and increasing NF-kappaB-dependent transcription. Inhibition of NF-kappaB activation diminished IL-1beta-mediated transcriptional activation of COX-2. Furthermore, mutation or deletion of a putative NF-kappaB binding site in the human COX-2 promoter greatly diminished its induction by IL-1beta. In addition, this cytokine induced a rapid increase in p38 MAPK activation. Interestingly, inhibition of p38 MAPK by SB203580 severely decreased induction of COX-2 expression by IL-1beta. p38 MAPK signalling was required for IL-1beta-dependent stabilization of COX-2 transcript. Given the importance of COX-2 expression in intestinal inflammation and colon carcinogenesis, these findings contribute to determine the key signalling pathways involved in the regulation of COX-2 expression in colorectal cells by inflammatory stimuli, such as IL-1beta.