Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 10(7): e0132498, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26162096

RESUMEN

Secretory Phospholipase A2 of type IIA (sPLA2 IIA) plays a crucial role in the production of lipid mediators by amplifying the neointimal inflammatory context of the vascular smooth muscle cells (VSMCs), especially during atherogenesis. Phenformin, a biguanide family member, by its anti-inflammatory properties presents potential for promoting beneficial effects upon vascular cells, however its impact upon the IL-1ß-induced sPLA2 gene expression has not been deeply investigated so far. The present study was designed to determine the relationship between phenformin coupling AMP-activated protein kinase (AMPK) function and the molecular mechanism by which the sPLA2 IIA expression was modulated in VSMCs. Here we find that 5-aminoimidazole-4-carboxamide-1-ß-D-ribonucleotide (AICAR) treatment strongly repressed IL-1ß-induced sPLA2 expression at least at the transcriptional level. Our study reveals that phenformin elicited a dose-dependent inhibition of the sPLA2 IIA expression and transient overexpression experiments of constitutively active AMPK demonstrate clearly that AMPK signaling is involved in the transcriptional inhibition of sPLA2-IIA gene expression. Furthermore, although the expression of the transcriptional repressor B-cell lymphoma-6 protein (BCL-6) was markedly enhanced by phenformin and AICAR, the repression of sPLA2 gene occurs through a mechanism independent of BCL-6 DNA binding site. In addition we show that activation of AMPK limits IL-1ß-induced NF-κB pathway activation. Our results indicate that BCL-6, once activated by AMPK, functions as a competitor of the IL-1ß induced NF-κB transcription complex. Our findings provide insights on a new anti-inflammatory pathway linking phenformin, AMPK and molecular control of sPLA2 IIA gene expression in VSMCs.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Fosfolipasas A2 Grupo II/genética , Interleucina-1beta/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/enzimología , Transducción de Señal/efectos de los fármacos , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Sitios de Unión , Bovinos , Separación Celular , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Fosfolipasas A2 Grupo II/metabolismo , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/metabolismo , Fenformina/farmacología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Ribonucleótidos/farmacología , Activación Transcripcional/efectos de los fármacos
2.
Biochem Pharmacol ; 79(8): 1149-55, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19962969

RESUMEN

Bacillus anthracis, the etiological agent of anthrax, produces lethal toxin (LT) that displays a metallo-proteolytic activity toward the N-terminus of the MAPK-kinases. We have previously shown that secreted type-IIA phospholipase A(2) (sPLA(2)-IIA) exhibits potent anthracidal activity. In vitro expression of sPLA(2)-IIA in guinea pig alveolar macrophages (AMs), the major source of this enzyme in lung tissues, is inhibited by LT. Here, we examined the mechanisms involved in sPLA(2)-IIA inhibition by LT. We first showed that chemical inhibitors of p38 and ERK MAPKs reduced sPLA(2)-IIA expression in AMs indicating that these kinases play a role in sPLA(2)-IIA expression. LT inhibited IL-1beta-induced p38 phosphorylation as well as sPLA(2)-IIA promoter activity in CHO cells. Inhibition of sPLA(2)-IIA promoter activity was mimicked by co-transfection with dominant negative construct of p38 (DN-p38) and reversed by the active form of p38-MAPK (AC-p38). Both LT and DN-p38 decreased IL-1beta-induced NF-kappaB luciferase activity. This contrasted with the effect of AC-p38, which enhanced this activity. However, neither LT nor specific p-38 inhibitor interfered with LPS-induced IkappaBalpha degradation or NF-kappaB nuclear translocation in AMs. Subcutaneous administration of LT to guinea pig before LPS challenge reduced sPLA(2)-IIA levels in broncho-alveolar lavages and ears. We conclude that sPLA(2)-IIA expression is induced via a sequential MAPK-NF-kappaB activation and that LT inhibits this expression likely by interfering with the transactivation of NF-kappaB in the nucleus. This inhibition, which is operating both in vitro and in vivo, may represent a mechanism by which B. anthracis subvert host defense.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Fosfolipasas A2 Grupo II/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Células CHO , Cricetinae , Cricetulus , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Fosfolipasas A2 Grupo II/biosíntesis , Cobayas , Masculino , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
3.
J Biol Chem ; 285(1): 565-75, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19850938

RESUMEN

Atherosclerosis is a multifactorial vascular disease characterized by formation of inflammatory lesions. Elevated circulating acute phase proteins indicate disease risk. Serum amyloid A (SAA) is one such marker but its function remains unclear. To determine the role of SAA on aortic smooth muscle cell gene expression, a preliminary screen of a number of genes was performed and a strong up-regulation of expression of secretory phospholipase A(2), group IIA (sPLA(2)) was identified. The SAA-induced increase in sPLA(2) was validated by real time PCR, Western blot analysis, and enzyme activity assays. Demonstrating that SAA increased expression of sPLA(2) heteronuclear RNA and that inhibiting transcription eliminated the effect of SAA on sPLA(2) mRNA suggested that the increase was transcriptional. Transient transfections and electrophoretic mobility shift assays identified CAAT enhancer-binding protein (C/EBP) and nuclear factor kappaB (NFkappaB) as key regulatory sites mediating the induction of sPLA(2). Moreover, SAA activated the inhibitor of NF-kappaB kinase (IKK) in cultured smooth muscle cells. Previous reports showed that interleukin (IL)-1beta up-regulates Pla2g2a gene transcription via C/EBPbeta and NFkappaB. Interestingly, SAA activated smooth muscle cell IL-1beta mRNA expression, however, blocking IL-1 receptors had no effect on SAA-mediated activation of sPLA(2) expression. Thus, the observed changes in sPLA(2) expression were not secondary to SAA-induced IL-1 receptor activation. The association of SAA with high density lipoprotein abrogated the SAA-induced increase in sPLA(2) expression. These data suggest that during atherogenesis, SAA can amplify the involvement of smooth muscle cells in vascular inflammation and that this can lead to deposition of sPLA(2) and subsequent local changes in lipid homeostasis.


Asunto(s)
Fosfolipasas A2 Grupo II/genética , Miocitos del Músculo Liso/enzimología , Proteína Amiloide A Sérica/metabolismo , Animales , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Activación Enzimática/efectos de los fármacos , Fosfolipasas A2 Grupo II/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipoproteínas HDL/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Interleucina-1/metabolismo , Proteína Amiloide A Sérica/farmacología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
4.
J Neurochem ; 109(4): 945-58, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19250336

RESUMEN

In several neurodegenerative diseases of the CNS, oligodendrocytes are implicated in an inflammatory process associated with altered levels of oxysterols and inflammatory enzymes such as secreted phospholipase A2 (sPLA2). In view of the scarce literature related to this topic, we investigated oxysterol effects on these myelinating glial cells. Natural oxysterol 25-hydroxycholesterol (25-OH; 1 and 10 microM) altered oligodendrocyte cell line (158N) morphology and triggered apoptosis (75% of apoptosis after 72 h). These effects were mimicked by 22(S)-OH (1 and 10 microM) which does not activate liver X receptor (LXR) but not by a synthetic LXR ligand (T0901317). Therefore, oxysterol-induced apoptosis appears to be independent of LXR. Interestingly, sPLA2 type IIA (sPLA2-IIA) over-expression partially rescued 158N cells from oxysterol-induced apoptosis. In fact, 25-OH, 24(S)-OH, and T0901317 stimulated sPLA2-IIA promoter and sPLA2 activity in oligodendrocyte cell line. Accordingly, administration of T0901317 to mice enhanced sPLA2 activity in brain extracts by twofold. Short interfering RNA strategy allowed to establish that stimulation of sPLA2-IIA is mediated by pregnane X receptor (PXR) at high oxysterol concentration (10 microM) and by LXR beta at basal oxysterol concentration. Finally, GC coupled to mass spectrometry established that oligodendrocytes contain oxysterols and express their biosynthetic enzymes, suggesting that they may act through autocrine/paracrine mechanism. Our results show the diversity of oxysterol signalling in the CNS and highlight the positive effects of the LXR/PXR pathway which may open new perspectives in the treatment of demyelinating and neurodegenerative diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Unión al ADN/efectos de los fármacos , Fosfolipasas A2 Grupo II/metabolismo , Hidroxicolesteroles/farmacología , Oligodendroglía/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores de Esteroides/efectos de los fármacos , Animales , Activación Enzimática/efectos de los fármacos , Citometría de Flujo , Cromatografía de Gases y Espectrometría de Masas , Hidrocarburos Fluorados/farmacología , Hidroxicolesteroles/antagonistas & inhibidores , Hidroxicolesteroles/toxicidad , Receptores X del Hígado , Ratones , Microscopía de Fuerza Atómica , Oligodendroglía/ultraestructura , Receptores Nucleares Huérfanos , Receptor X de Pregnano , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología , Transfección
5.
J Mol Biol ; 384(3): 564-76, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-18848838

RESUMEN

Macrophage-derived reactive oxygen species contribute to the initiation and development of atherosclerosis. The cellular balance between oxidative and reductive states depends on the endogenous antioxidant capacity, with the thioredoxin-1 (Trx-1) system playing a major role. Peroxisome proliferator-activated receptor-alpha (PPARalpha) is expressed by human macrophages and exhibits anti-inflammatory properties. Here we show that the selective PPARalpha activator GW647 significantly increased the Trx-1 mRNA and protein expression in human macrophages as determined by quantitative polymerase chain reaction and Western immunoblotting. Consistently, the Trx-1 activity was significantly increased by PPARalpha activation. By contrast, PPARalpha activation led to the down-regulation of vitamin D(3) up-regulated protein 1 (VDUP-1), the physiological inhibitor of Trx-1. Analysis of the Trx-1 and VDUP-1 promoters with gene reporter assays, mutational analysis, gel shift assays and chromatin immunoprecipitation analyses revealed the presence of a functional response element specific for PPARalpha in the Trx-1 promoter and the presence of a functional activator protein 1 (AP-1) site in the VDUP-1 promoter. The interference of PPARalpha/retinoid X receptor alpha with the AP-1 transcription factor elements c-Jun/c-Fos resulted in the inhibition of AP-1 binding and down-regulation of the VDUP-1 gene expression. Finally, PPARalpha activation reduced the lidocaine-induced caspase-3 activity and apoptosis, which might be due to the VDUP-1-mediated regulation of the Bax/Bcl-2 ratio. Together these data indicate that stimulation of PPARalpha in human macrophages might reduce arterial inflammation through differential regulation of the Trx-1 and VDUP-1 gene expression.


Asunto(s)
Proteínas Portadoras/biosíntesis , Regulación de la Expresión Génica , Macrófagos/metabolismo , PPAR alfa/metabolismo , Tiorredoxinas/biosíntesis , Apoptosis , Secuencia de Bases , Sitios de Unión , Butiratos/farmacología , Cromatina/metabolismo , Humanos , Datos de Secuencia Molecular , Estrés Oxidativo , PPAR alfa/agonistas , Compuestos de Fenilurea/farmacología , Regiones Promotoras Genéticas , Unión Proteica , Factor de Transcripción AP-1/metabolismo
6.
PLoS Pathog ; 3(12): e187, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18069891

RESUMEN

Bacillus anthracis, the etiological agent of anthrax, is a spore-forming gram-positive bacterium. Infection with this pathogen results in multisystem dysfunction and death. The pathogenicity of B. anthracis is due to the production of virulence factors, including edema toxin (ET). Recently, we established the protective role of type-IIA secreted phospholipase A2 (sPLA2-IIA) against B. anthracis. A component of innate immunity produced by alveolar macrophages (AMs), sPLA2-IIA is found in human and animal bronchoalveolar lavages at sufficient levels to kill B. anthracis. However, pulmonary anthrax is almost always fatal, suggesting the potential impairment of sPLA2-IIA synthesis and/or action by B. anthracis factors. We investigated the effect of purified ET and ET-deficient B. anthracis strains on sPLA2-IIA expression in primary guinea pig AMs. We report that ET inhibits sPLA2-IIA expression in AMs at the transcriptional level via a cAMP/protein kinase A-dependent process. Moreover, we show that live B. anthracis strains expressing functional ET inhibit sPLA2-IIA expression, whereas ET-deficient strains induced this expression. This stimulatory effect, mediated partly by the cell wall peptidoglycan, can be counterbalanced by ET. We conclude that B. anthracis down-regulates sPLA2-IIA expression in AMs through a process involving ET. Our study, therefore, describes a new molecular mechanism implemented by B. anthracis to escape innate host defense. These pioneering data will provide new molecular targets for future intervention against this deadly pathogen.


Asunto(s)
Carbunco/prevención & control , Antígenos Bacterianos/toxicidad , Bacillus anthracis/enzimología , Toxinas Bacterianas/toxicidad , Fosfolipasas A2 Grupo II/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Animales , Carbunco/enzimología , Carbunco/inmunología , Lavado Broncoalveolar , Células Cultivadas , Regulación hacia Abajo , Regulación Bacteriana de la Expresión Génica , Silenciador del Gen , Fosfolipasas A2 Grupo II/inmunología , Cobayas , Interacciones Huésped-Patógeno , Inmunidad Innata , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/microbiología , Masculino , ARN Mensajero/metabolismo
7.
Mol Cell Biol ; 27(23): 8374-87, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17908795

RESUMEN

The inflammation that occurs during atherosclerosis is characterized by the release of large amounts of group IIA secretory phospholipase A2 (sPLA2-IIA). This study was designed to define the function of the three peroxisome proliferator-activated receptors (PPARs) on sPLA2 expression in vascular smooth muscle cells (VSMCs). We found that PPAR ligands decreased sPLA2-IIA activity and inhibited mRNA accumulation under inflammatory conditions. Furthermore, interleukin-1beta-induced sPLA2-IIA promoter activity was inhibited by the three PPAR ligands and in a similar way when cells were cotransfected with PPARalpha, PPARbeta, or PPARgamma, plus retinoid X receptor alpha (RXRalpha). Our study revealed that the regulation of sPLA2-IIA gene transcription by PPARalpha/RXR and PPARgamma/RXR heterodimers requires an interaction with a PPAR response element (PPRE) of the sPLA2-IIA promoter. In contrast, PPARbeta operates through a PPRE-independent mechanism. In addition, we demonstrated that VSMCs expressed the transcriptional repressor BCL-6. Overexpression of BCL-6 markedly reduced sPLA2-IIA promoter activity in VSMCs, while a dominant negative form of BCL-6 abrogated sPLA2 repression by PPARbeta. The PPARbeta agonist induced a BCL-6 binding to the sPLA2 promoter in VSMCs under inflammatory conditions. The knockdown of BCL-6 by short interfering RNA abolished the inhibitory effect of the PPARbeta ligand on sPLA2 activity and prostaglandin E2 release. Thus, the inhibition of sPLA2-IIA activity by PPARbeta agonists may provide a promising approach to impacting the initiation and progression of atherosclerosis.


Asunto(s)
Fosfolipasas A2 Grupo II/biosíntesis , Interleucina-1beta/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , PPAR-beta/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Animales , Bovinos , Inducción Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Fosfolipasas A2 Grupo II/genética , Ligandos , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Proteínas Represoras/metabolismo , Elementos de Respuesta , Receptores X Retinoide/metabolismo , Eliminación de Secuencia
8.
FASEB J ; 20(10): 1727-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16807371

RESUMEN

Lipid mediators such as prostaglandin E2 (PGE2) play a central role during atherogenesis as a consequence of inflammation. PGE2 is produced from phospholipids by a cascade of enzymatic reactions involving phospholipase A2 (PLA2), cyclooxygenase (COX), and prostaglandin E synthase (PGES). It is released by several cell types, including vascular smooth muscle cells (VSMCs). Recent work has shown that the secretory PLA2-IIA (sPLA2-IIA), the most abundant isoform of secreted PLA2 in VSMCs, acts as a potent cytokine and activates VSMCs through a positive feedback loop. High mobility group protein 1 (HMGB1), also known as amphoterin, is a ubiquitous protein that plays various roles in the nucleus. HMGB1 is released by necrotic cells and by immune cells in response to various inflammatory mediators and acts as a potent proinflammatory cytokine. The present study investigates the role of HMGB1 in the activation of sPLA2-IIA expression and PGE2 production in VSMCs. Recombinant HMGB1 slightly activated the sPLA2-IIA, COX-2, and mPGES-1 genes but dramatically stimulated these genes in VSMCs that had been incubated with the proinflammatory cytokine IL-1beta for 24 h. This effect was accompanied by significantly increased PGE2 release. Induction of the three known receptors of HMGB1, namely RAGE, TLR-2, and TLR-4, by IL-1beta suggests that proinflammatory cytokines sensitize VSMCs to HMGB1. This provides new insights into the role of HMGB1 in VSMCs, suggesting it may be essential for the progression of atherosclerosis.


Asunto(s)
Dinoprostona/biosíntesis , Proteína HMGB1/farmacología , Interleucina-1/farmacología , Músculo Liso Vascular/metabolismo , Fosfolipasas A/metabolismo , Animales , Aterosclerosis/etiología , Activación Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Fosfolipasas A2 Grupo II , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Fosfolipasas A2 , Ratas , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/genética , Proteínas Recombinantes , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
9.
J Cell Physiol ; 208(3): 495-505, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16741924

RESUMEN

Transition of vascular smooth muscle cells from a contractile/quiescent to a secretory/proliferative phenotype is one of the critical steps in atherosclerosis and is instigated by pro-inflammatory cytokines released from macrophages that have infiltrated into the vascular wall. In most inflammatory diseases, cell activation induced by these compounds leads to a massive production of type E2 prostaglandin (PGE2) which often takes over and even potentiates the pro-inflammatory cytokine-related effects. To evaluate PGE2 incidence on atheroma plaque development, we investigated whether and how this compound could enhance the dedifferentiation of smooth muscle cells initially induced by interleukin-1beta (IL-1beta). To address this issue, we took advantage of vascular smooth muscle cells in primary culture and tracked two markers: PLA2 secretion and alpha-actin filament disorganization. In such a context, we found that PGE2 synergizes with IL-1beta to further enhance the phenotype transition of smooth muscle cells, through cAMP-protein kinase A. As indicated by pharmacological studies, the full PGE2-dependent potentiation of IL-1beta induced PLA2 secretion is associated with a change of regulation exerted by the subtypes 3 G(i)-coupled PGE2 receptors toward adenylyl cyclase(s) activated by the subtype 4 G(s)-linked PGE2 receptor. Whereas on contractile cells, stimulated subtypes 3 inhibit type 4-dependent PLA2 secretion, this negative regulation is switched to positive on IL-1beta-treated cells. Using real time PCR, pharmacological tools and small interfering RNA (siRNA), we demonstrated that the different integration of PGE2 signals depends on the upregulation of calcium/calmodulin stimulable adenylyl cyclase 8.


Asunto(s)
Adenilil Ciclasas/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Dinoprostona/farmacología , Interleucina-1/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Fosfolipasas A/genética , Receptores de Prostaglandina E/genética , Adenilil Ciclasas/metabolismo , Animales , Aorta Torácica , Células Cultivadas , AMP Cíclico/metabolismo , Sinergismo Farmacológico , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Masculino , Músculo Liso Vascular/efectos de los fármacos , Fosfolipasas A/metabolismo , Fosfolipasas A2 , ARN Interferente Pequeño/genética , Ratas , Receptores de Prostaglandina E/clasificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/efectos de los fármacos , Vasoconstricción/efectos de los fármacos
10.
Arterioscler Thromb Vasc Biol ; 25(6): 1161-7, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15802623

RESUMEN

OBJECTIVE: The inflammation that occurs during the development of atherosclerosis is characterized by a massive release of sPLA2-IIA (group IIA secretory phospholipase A2) from vascular smooth muscle cells (VSMCs). We have investigated the autocrine function of sPLA2-IIA in rat aortic and human VSMCs. METHODS AND RESULTS: We found that the transcription of the endogenous sPLA2-IIA gene increased by adding a cell supernatant containing human sPLA2-IIA proteins. We show that this effect was independent of the sPLA2 activity using sPLA2-IIA proteins lacking enzyme activity. Transient transfections with various sPLA2-IIA rat promoter-luciferase constructs demonstrated that the C/EBP, NK-kappaB, and Ets transcription factors are involved in the increase in sPLA2-IIA gene transcription. We also found the M-type sPLA2 receptor mRNA in VSMCs, and we showed that the sPLA2-luciferase reporter gene was induced by the specific agonist of the sPLA2 receptor, aminophenylmannopyranoside (APMP), and that this induction was mediated by the same transcription factor-binding sites. Finally, we used a sPLA2-IIA mutant unable to bind heparan-sulfate proteoglycans to show that the binding of wild-type sPLA2-IIA to proteoglycans is essential for the induction of an autocrine loop. CONCLUSIONS: We have thus identified new autocrine and paracrine pathways activating sPLA2-IIA gene expression in rat and human VSMCs.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Músculo Liso Vascular/enzimología , Fosfolipasas A/genética , Fosfolipasas A/metabolismo , Animales , Aorta Torácica/citología , Comunicación Autocrina/fisiología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Fosfolipasas A2 Grupo II , Humanos , Masculino , Músculo Liso Vascular/citología , FN-kappa B/metabolismo , Comunicación Paracrina/fisiología , Fosfolipasas A2 , Unión Proteica , Proteoglicanos/metabolismo , Ratas , Ratas Wistar , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Fosfolipasa A2 , Activación Transcripcional/fisiología , Factores de Transcripción Winged-Helix/metabolismo
11.
Eur J Biochem ; 271(22): 4462-73, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15560787

RESUMEN

The proliferation of smooth muscle cells (SMC) is a key event in the development of atherosclerosis. In addition to growth factors or cytokines, we have shown previously that n-3 polyunsaturated fatty acids (PUFAs) act in opposition to n-6 PUFAs by modulating various steps of the inflammatory process. We have investigated the molecular mechanisms by which the incorporation of the n-6 PUFA, arachidonic acid, increases the proliferation of rat SMC treated with interleukin-1beta, while the n-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), elicit no mitogenic response. Incorporation of EPA or DHA into SMC, which are then activated by interleukin-1beta to mimic inflammation, decreases promoter activity of the cyclin D1 gene and phosphorylation of the retinoblastoma protein. Together, our data demonstrate that n-3 effects are dependent on the Ras/Raf-1/extracellular signal regulated kinase (ERK)/mitogen-activated protein kinase pathway, and that down-regulation of the cyclin D1 promoter activity is mediated by the specific binding of the early growth response factor-1. Finally, we have shown that the incorporation of EPA and DHA also increased the concentration of caveolin-1 and caveolin-3 in caveolae, which correlated with n-3 PUFA inhibition of SMC proliferation through the mitogen-activated protein kinase pathway. We provide evidence indicating that, in contrast to n-6 PUFAs, n-3 PUFAs exert antiproliferative effects on SMC through the mitogen-activated protein kinase/ERK pathway.


Asunto(s)
Ciclina D1/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Ácidos Grasos Insaturados/farmacología , Proteínas Inmediatas-Precoces/biosíntesis , Interleucina-1/farmacología , Músculo Liso Vascular/efectos de los fármacos , Factores de Transcripción/biosíntesis , Animales , Bovinos , Caveolina 1 , Caveolina 2 , Caveolinas/biosíntesis , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Proteínas de Unión al ADN/genética , Proteína 1 de la Respuesta de Crecimiento Precoz , Activación Enzimática , Ácidos Grasos Insaturados/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Proteínas Inmediatas-Precoces/genética , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Regiones Promotoras Genéticas/efectos de los fármacos , Ratas , Ratas Wistar , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción/genética , Quinasas raf/metabolismo , Proteínas ras/metabolismo
12.
J Biol Chem ; 279(27): 28411-8, 2004 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-15090544

RESUMEN

Interleukin-1beta (IL-1beta) induces degradation via hyperexpression of an array of genes, including metalloproteinases (MMP), in cartilage cells during articular degenerative diseases. In contrast, natural ligands for peroxisome proliferator-activated receptors (PPARs) display protective anti-cytokine effects in these cells. We used the PPAR agonist rosiglitazone (Rtz) to investigate PPAR-gamma isotype on IL-1beta-target genes. Immunocytochemistry, electrophoretic mobility shift, and transient transfection assays revealed a functional PPAR-gamma in chondrocytes in vitro. Rtz displayed significant inhibition of IL-1beta effects in chondrocytes. Low Rtz concentrations (close to K(d) values for PPAR-gamma, 0.1 to 1 microm) inhibited the effects of IL-1beta on (35)S-sulfated proteoglycan production and gelatinolytic activities and downregulated MMP1 expression at mRNA and protein levels. We have investigated the mechanism of action of Rtz against IL-1beta-mediated MMP1 gene hyperexpression. Rtz effect occurs at the transcriptional level of the MMP1 promoter, as observed in transiently transfected cells with pMMP1-luciferase vector. Transient expression of wild type PPAR-gamma enhanced Rtz inhibitory effect in chondrocytes, whereas a mutated dominant negative PPAR-gamma abolished it, supporting the role of PPAR-gamma in this effect. MMP1 gene promoter analysis revealed the involvement of a cis-acting element located at -83 to -77, shown to be a composite PPRE/AP1 site. Gel mobility and supershift assays demonstrated that PPAR-gamma and c-Fos/c-Jun proteins bind this cis-acting element in a mutually exclusive way. Our data highlight a new PPAR-gamma-dependent inhibitory mechanism on IL-1beta-mediated cartilage degradation occurring through DNA binding competition on the composite PPRE/AP1 site in the MMP1 promoter.


Asunto(s)
Condrocitos/metabolismo , Regulación hacia Abajo , Metaloproteinasa 1 de la Matriz/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Northern Blotting , Western Blotting , Cartílago/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Clonación Molecular , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Fibrinolíticos/farmacología , Genes Dominantes , Humanos , Inmunohistoquímica , Interleucina-1/metabolismo , Cinética , Ligandos , Luciferasas/metabolismo , Mutagénesis Sitio-Dirigida , Mutación , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteoglicanos/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rosiglitazona , Sulfatos/metabolismo , Tiazolidinedionas/farmacología , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo , Transcripción Genética , Transfección
13.
Biochem J ; 376(Pt 2): 351-60, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12882648

RESUMEN

The inflammation that occurs during rheumatoid arthritis or atherosclerosis is characterized by the release of large amounts of sPLA(2) (group IIA secretory phospholipase A(2)). We have shown previously that the sPLA(2) promoter in SMC (smooth-muscle cells) is activated by interleukin-1beta and cAMP-signalling pathways, through the interplay of multiple transcription factors [Antonio, Brouillet, Janvier, Monne, Bereziat, Andreani, and Raymondjean (2002) Biochem. J. 368, 415-424]. In the present study, we have investigated the regulation of sPLA(2) gene expression in rat aortic SMCs by oxysterols. We found that oxysterol ligands that bind to the LXR (liver X receptor), including 25-HC (25-hydroxycholesterol) and 22( R )-HC, cause the accumulation of sPLA(2) mRNA and an increased enzyme activity. Transient transfection experiments demonstrated that the sPLA(2) promoter is synergistically activated by 22( R )-HC in combination with 9- cis -retinoic acid, a ligand for the LXR heterodimeric partner RXR (retinoid X receptor). Promoter activity was also increased in a sterol-responsive fashion when cells were co-transfected with LXRalpha/RXRalpha or LXRbeta/RXRalpha. Mutagenesis studies and gel mobility-shift assays revealed that LXR/RXR heterodimers regulate sPLA(2) transcription directly, by interacting with a degenerated LXRE (LXR response element) at position [-421/-406] of the sPLA(2) promoter. Chromatin immunoprecipitation revealed the in vivo occupancy of LXR on the sPLA(2) promoter. In addition, the orphan nuclear receptor LRH-1 (liver receptor homologue-1) potentiated the sterol-dependent regulation of the sPLA(2) promoter by binding to an identified promoter element (TCAAGGCTG). Finally, we have demonstrated that oxysterols act independent of interleukin-1beta and cAMP pathways to activate the sPLA(2) promoter. In the present study, we have identified a new pathway activating sPLA(2) gene expression in SMCs.


Asunto(s)
Hidroxicolesteroles/farmacología , Músculo Liso Vascular/enzimología , Fosfolipasas A/genética , Activación Transcripcional , Tretinoina/farmacología , Alitretinoína , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas de Unión al ADN , Inducción Enzimática , Fosfolipasas A2 Grupo II , Interleucina-1/farmacología , Receptores X del Hígado , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Receptores Nucleares Huérfanos , Fosfolipasas A/biosíntesis , Fosfolipasas A/metabolismo , Fosfolipasas A2 , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Ratas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Ácido Retinoico/metabolismo , Elementos de Respuesta , Receptores X Retinoide , Factores de Transcripción/metabolismo
14.
J Lipid Res ; 44(3): 601-11, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12562859

RESUMEN

There is good evidence that the n-3 polyunsaturated fatty acids (PUFAs) in fish oil have antiinflammatory effects and reduce the pathogenesis of atherosclerosis. However, the mechanisms underlying these actions are largely unknown. This study was designed to investigate the effects of membrane incorporation of two major components of fish oil [eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)], on rat smooth muscle cells (SMCs) activation induced by interleukin-1 beta (IL1 beta). We compared their effects with those of n-6 arachidonic acid (AA). Expression of vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1 adhesion molecules involved in SMCs migration was enhanced by AA, whereas EPA and DHA had no similar effects. We established that AA potentiates IL1 beta-induced expression of the type IIA secreted phospholipase A2 (sPLA2) gene, whereas EPA and DHA reduce this stimulation. EPA and DHA also abolished proinflammatory prostaglandin PGE2 production by inhibiting the IL1 beta-induced production of cyclooxygenase-2 (COX-2) mRNA. Much interest was then focused on three transcriptional factors implicated in inflammation control and especially in modulating rat sPLA2 and COX-2 gene transcription: nuclear factor-kappa B, CCAAT/enhancer binding protein beta, and E26 transformation-specific-1. electrophoretic mobility shift assay revealed that the binding activity of all three factors was increased by AA and reduced (or not affected) by n-3 PUFA. These results indicate that EPA and DHA act in opposition to AA by modulating various steps of the inflammatory process induced by IL1 beta, probably by reducing mitogen-activated protein kinase p42/p44 activity.


Asunto(s)
Ácidos Grasos Insaturados/farmacología , Interleucina-1/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Animales , Aorta , Quimiocina CCL2/genética , Ciclooxigenasa 2 , Dinoprostona/biosíntesis , Dinoprostona/metabolismo , Activación Enzimática/efectos de los fármacos , Ácidos Grasos Omega-3 , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/genética , Inflamación/metabolismo , Isoenzimas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Fosfolipasas A/metabolismo , Fosfolipasas A2 , Fosfolípidos/análisis , Fosfolípidos/metabolismo , Prostaglandina-Endoperóxido Sintasas/genética , Ratas , Factores de Transcripción/metabolismo , Triglicéridos/farmacología , Molécula 1 de Adhesión Celular Vascular/genética
15.
Biochem J ; 368(Pt 2): 415-24, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12188923

RESUMEN

The abundant secretion of type IIA secreted phospholipase A(2) (sPLA(2)) is a major feature of the inflammatory process of atherosclerosis. sPLA(2) is crucial for the development of inflammation, as it catalyses the production of lipid mediators and induces the proliferation of smooth muscle cells. We have analysed the activation of sPLA(2) transcription by cAMP and interleukin-1beta (IL-1beta), and shown that the 500 bp region upstream of the transcription start site of the rat sPLA(2) gene is implicated in activation by synergistically acting cAMP and IL-1beta. We transiently transfected and stimulated rat smooth muscle cells in primary culture and measured the promoter activities of serial and site-directed deletion mutants of sPLA(2)-luciferase constructs. A distal region, between -488 and -157 bp, bearing a CAAT/enhancer binding protein (C/EBP)-responsive element (-242 to -223) was sufficient for cAMP/protein kinase A-mediated sPLA(2) promoter activation. We find evidence for the first time that activation of the sPLA(2) promoter by IL-1beta requires activation of an Ets-responsive element in the -184 to -180 region of the distal promoter via the Ras pathway and a nuclear factor-kappaB site at positions -141 to -131 of the proximal promoter. We also used electrophoretic mobility shift assays to identify five binding sites for the Sp1 factor; a specific inhibitor of Sp1, mithramycin A, showed that this factor is crucial for the basal activity of the sPLA(2) promoter.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , AMP Cíclico/metabolismo , Interleucina-1/metabolismo , Músculo Liso Vascular/fisiología , FN-kappa B/metabolismo , Fosfolipasas A/genética , Animales , Arteriosclerosis/genética , Secuencia de Bases , Sitios de Unión , Células CHO , Células Cultivadas , Cricetinae , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación Enzimológica de la Expresión Génica , Fosfolipasas A2 Grupo II , Humanos , Interleucina-1/farmacología , Masculino , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Fosfolipasas A/efectos de los fármacos , Fosfolipasas A/metabolismo , Fosfolipasas A2 , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets , Ratas , Ratas Wistar , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas ras/genética , Proteínas ras/metabolismo
16.
FEBS Lett ; 518(1-3): 67-71, 2002 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-11997019

RESUMEN

We have designed a chimeric promoter that can be stimulated by various pro-inflammatory mediators and so drive the expression of therapeutic genes under inflammatory conditions. The promoter has two parts, the [-247/+20] fragment of the human type IIA secreted phospholipase A2 gene promoter, which is stimulated by the pro-inflammatory cytokine interleukin-1beta (IL-1beta), and a double peroxisome proliferator-activated receptor response element that is activated by some eicosanoids and by non-steroidal anti-inflammatory drugs (NSAIDs). Transfection experiments using rabbit articular chondrocytes in primary culture showed that this chimeric promoter produced a low basal activity and was induced by NSAIDs, WY-14643, IL-1beta, and 15-deoxy Delta12,14 prostaglandin J2. The latter two compounds stimulated the promoter synergistically.


Asunto(s)
Cartílago Articular/citología , Condrocitos/metabolismo , Mediadores de Inflamación/farmacología , Regiones Promotoras Genéticas , Activación Transcripcional , Animales , Antiinflamatorios no Esteroideos/farmacología , Secuencia de Bases , Sitios de Unión , Células COS , Condrocitos/efectos de los fármacos , ADN Recombinante/metabolismo , Dexametasona/farmacología , Eicosanoides/farmacología , Femenino , Glucocorticoides/farmacología , Fosfolipasas A2 Grupo II , Humanos , Interleucina-1/farmacología , Fosfolipasas A/genética , Fosfolipasas A2 , Conejos , Receptores Citoplasmáticos y Nucleares/metabolismo , Elementos de Respuesta , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA