Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Endocrinology ; 150(3): 1192-201, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18948393

RESUMEN

Seasonal obesity and fasting-associated hibernation are the two major metabolic events governing hepatic lipid metabolism in hibernating mammals. In this process, however, the role of the nuclear receptor known as peroxisome proliferator-activated receptor (PPAR)-alpha has not been elucidated yet. Here we show, as in human, that jerboa (Jaculus orientalis) liver expresses both active wild-type PPARalpha (PPARalpha1wt) and truncated PPARalpha forms and that the PPARalpha1wt to truncated PPARalpha2 ratio, which indicates the availability of active PPARalpha1wt, is differentially regulated during fasting-associated hibernation. Functional activation of hepatic jerboa PPARalpha, during prehibernating and hibernating states, was demonstrated by the induction of its target genes, which encode peroxisomal proteins such as acyl-CoA oxidase 1, peroxisomal membrane protein 70, and catalase, accompanied by a concomitant induction of PPARalpha thermogenic coactivator PPARgamma coactivator-1alpha. Interestingly, sustained activation of PPARalpha by its hypolipidemic ligand, ciprofibrate, abrogates the adaptive fasting response of PPARalpha during prehibernation and overinduces its target genes, disrupting the prehibernation fattening process. In striking contrast, during fasting-associated hibernation, jerboas exhibit preferential up-regulation of hepatic peroxisomal fatty acid oxidation instead of the mitochondrial pathway, which is down-regulated. Taken together, our results strongly suggest that PPARalpha is subject to a hibernation-dependent splicing regulation in response to feeding-fasting conditions, which defines the activity of PPARalpha and the activation of its target genes during hibernation bouts of jerboas.


Asunto(s)
Ayuno/fisiología , Ácidos Grasos/metabolismo , Hibernación/genética , Hígado/metabolismo , PPAR alfa/genética , Roedores/genética , Roedores/metabolismo , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/genética , Adaptación Fisiológica/fisiología , Animales , Ácido Clofíbrico/análogos & derivados , Ácido Clofíbrico/farmacología , Ayuno/metabolismo , Ácidos Fíbricos , Regulación de la Expresión Génica/efectos de los fármacos , Hibernación/fisiología , Hipolipemiantes/farmacología , Metabolismo de los Lípidos/genética , Mamíferos , Oxidación-Reducción , PPAR alfa/metabolismo , Peroxisomas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Int J Mol Med ; 22(6): 825-32, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19020782

RESUMEN

Peroxisome proliferator-activated receptor-gamma (PPARgamma) belongs to the nuclear hormone receptor family. This receptor is implicated in colon cell differentiation and in colon cancer. Receptor activation by specific agonists has been shown to protect against colon cancer progression. PPARgamma protein content within cells is modulated by several mechanisms, including proteasome degradation, activation of Wnt signalling pathways and presence of fermentation products such as butyrate. Herein, we investigated the impact of L-glutamine on PPARgamma expression during the differentiation of Caco-2 cells grown in medium containing dialyzed fetal calf serum supplemented or not with L-glutamine. Using RT-PCR and Western blotting, we demonstrated that PPARgamma expression was decreased when L-glutamine was added to the medium. Using immunohistochemistry, we demonstrated that PPARgamma immunostaining was mainly found in cytoplasm when cells were cultured with L-glutamine while it was found in nuclei and cytoplasm when cells were grown without the addition of L-glutamine. Supershift retardation assays demonstrated a decrease of PPARgamma binding onto consensus peroxisome proliferator response element. We concluded that L-glutamine modulated PPARgamma expression in Caco-2 cells.


Asunto(s)
Diferenciación Celular , Colon/citología , Glutamina/farmacología , PPAR gamma/metabolismo , Western Blotting , Células CACO-2 , Colon/metabolismo , Medios de Cultivo , Citoplasma/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Glutaminasa/genética , Glutaminasa/metabolismo , Glutamina/metabolismo , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
PLoS One ; 3(4): e2002, 2008 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-18431487

RESUMEN

The Damaged DNA binding protein 2 (DDB2), is involved in nucleotide excision repair as well as in other biological processes in normal cells, including transcription and cell cycle regulation. Loss of DDB2 function may be related to tumor susceptibility. However, hypothesis of this study was that DDB2 could play a role in breast cancer cell growth, resulting in its well known interaction with the proliferative marker E2F1 in breast neoplasia. DDB2 gene was overexpressed in estrogen receptor (ER)-positive (MCF-7 and T47D), but not in ER-negative breast cancer (MDA-MB231 and SKBR3) or normal mammary epithelial cell lines. In addition, DDB2 expression was significantly (3.0-fold) higher in ER-positive than in ER-negative tumor samples (P = 0.0208) from 16 patients with breast carcinoma. Knockdown of DDB2 by small interfering RNA in MCF-7 cells caused a decrease in cancer cell growth and colony formation. Inversely, introduction of the DDB2 gene into MDA-MB231 cells stimulated growth and colony formation. Cell cycle distribution and 5 Bromodeoxyuridine incorporation by flow cytometry analysis showed that the growth-inhibiting effect of DDB2 knockdown was the consequence of a delayed G1/S transition and a slowed progression through the S phase of MCF-7 cells. These results were supported by a strong decrease in the expression of S phase markers (Proliferating Cell Nuclear Antigen, cyclin E and dihydrofolate reductase). These findings demonstrate for the first time that DDB2 can play a role as oncogene and may become a promising candidate as a predictive marker in breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de Unión al ADN/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Fase G1 , Regulación Neoplásica de la Expresión Génica , Humanos , Células Madre Neoplásicas/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Receptores de Estrógenos/metabolismo , Fase S
4.
Breast Cancer Res Treat ; 108(2): 203-15, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17473980

RESUMEN

Manganese superoxide dismutase (MnSOD) is known to play a role in cancer. MnSOD exerts a tumor suppressive effect in estrogen-dependent human breast cancer cells. In the present study we investigated the in vitro role of MnSOD in the growth of some aggressive and highly metastatic estrogen-independent breast cancer cells, i.e., MDA-MB231 and SKBR3 cells. We show that estrogen-independent cells expressed a significantly higher basal MnSOD level compared to estrogen-dependent human breast cancer cell lines (MCF-7 and T47D). For MDA-MB231 cells, the high-MnSOD level was accompanied by an overproduction of intracellular hydrogen peroxide (H2O2) and by a low expression of the major H2O2-detoxifying enzymes, catalase, and peroxiredoxin 3, compared to MCF-7 cells. Suppression of MnSOD expression by antisense RNA was associated with a decrease of H2O2 content and caused a stimulation of growth with a reduced cell doubling time but induced a decrease of colony formation. Furthermore, treatment of MDA-MB231 cells with H2O2 scavengers markedly reduced tumor cell growth and colony formation. In addition, MnSOD suppression or treatment with H2O2 scavengers reduced the invasive properties of MDA-MB231 cells up to 43%, with a concomitant decrease of metalloproteinase-9 activity. We conclude that MnSOD plays a role in regulating tumor cell growth and invasive properties of estrogen-independent metastatic breast cancer cells. These action are mediated by MnSOD-dependent H2O2 production. In addition, these results suggest that MnSOD up-regulation may be one mechanism that contributes to the development of metastatic breast cancers.


Asunto(s)
Neoplasias de la Mama/enzimología , Movimiento Celular , Proliferación Celular , Superóxido Dismutasa/metabolismo , Antioxidantes/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Catalasa/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Estrógenos/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Invasividad Neoplásica , Peroxiredoxina III , Peroxirredoxinas/metabolismo , ARN sin Sentido/metabolismo , Superóxido Dismutasa/genética , Factores de Tiempo , Transfección
5.
Med Sci (Paris) ; 23(5): 515-8, 2007 May.
Artículo en Francés | MEDLINE | ID: mdl-17502068

RESUMEN

The peroxisome proliferator-activated receptors (PPARs) are transcription factors and belong to the superfamily of nuclear receptors. They are encoded by three genes located on different chromosomes: PPARalpha, PPARbeta/delta and PPARgamma. PPARalpha plays a key role in the control of lipid metabolism and homeostasis. PPARbeta/delta is expressed ubiquitously and participates in skeletal muscle physiology. PPARbeta/delta and PPARgamma are important factors for placental development and function as well as for embryo implantation. In addition, PPARgamma is mainly involved in adipogenesis. PPARs also participate more or less to cell proliferation, differentiation and apoptosis. Surprisingly, the involvement of these transcription factors in cell-cell and/or cell-matrix interactions has not yet been reviewed except for their role as therapeutic agents in inflammation. Nevertheless, several pioneer reports have recently provided some new insights in this research field, by suggesting that PPARs are involved, directly or indirectly, in these interactions and that their activation by specific ligands may lead to potential therapeutic approaches.


Asunto(s)
Adhesión Celular/fisiología , Comunicación Celular/fisiología , Matriz Extracelular/fisiología , Receptores Activados del Proliferador del Peroxisoma/fisiología , Apoptosis , Diferenciación Celular , División Celular , Humanos , Integrinas/genética , Hígado/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
6.
Biochimie ; 89(3): 329-36, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17070643

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are ligand-inducible transcription factors and belong to the nuclear hormone receptor superfamily. They form heterodimers with retinoid X receptor (RXR) and bind to specific PPAR-response elements. To identify novel PPAR target genes, we developed an affinity method to isolate human genomic fragments containing binding sites for PPARs. For this, an antibody raised against all PPAR subtypes was used. Immunoselected fragments were amplified and sequenced. One of them, ISF1029, was mapped by BLAT and BLAST searches on different human chromosomes, downstream of several POTE genes. ISF1029 contained three hexamers strongly related to the AGGTCA motif organized according to a DR0/3 motif. The latter was found to bind to PPARalpha in gel mobility shift and supershift assays and to exhibit a downregulation potentiality in transfection experiments under clofibrate treatment. POTE genes were shown to be highly expressed in human Caco-2 colorectal adenocarcinoma cells and downregulated by fenofibrate and 9-cis-retinoic acid, as attested by RT-PCR assays. Microarray analysis confirmed and extended to the human T98-G glioblastoma cells, the downregulation of several POTE genes expression by Wy-14,643, a potent PPARalpha activator. Our data provide new insights about the pleiotropic action of PPARs.


Asunto(s)
ADN/metabolismo , Genoma Humano , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Elementos de Respuesta/genética , Secuencia de Bases , Sitios de Unión/genética , Southern Blotting , Células CACO-2 , Línea Celular Tumoral , ADN/inmunología , ADN/aislamiento & purificación , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica , Células HCT116 , Humanos , Inmunoprecipitación , Modelos Genéticos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/inmunología , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Int J Oncol ; 29(6): 1601-10, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17089002

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a member of the nuclear hormone receptor family. In colon, this transcription factor is involved in differentiation of absorptive cells. PPARgamma participates also in colon carcinogenesis and cancer progression. Two isoforms, namely PPARgamma1 and PPARgamma2, have been described. Recently, new PPARgamma1 transcripts whose translation raises PPARgamma1 protein have been characterised. They differ from each other by combination of untranslated exons localised in the 5' UTR of the PPARG gene. Here, we studied whether such a diversity of PPARgamma transcripts occurs in human colon cell models. Based on bioinformatic analysis, putative untranslated exons were identified in the human PPARG gene. By RT-PCR analysis, we have demonstrated that several of these untranslated exons are included in PPARgamma transcripts from colon-derived cell lines or in those derived from other tissue. Using HT-29 cells, changes in PPARgamma1 mRNA levels were observed after treatment with PPARgamma agonists such as pioglitazone and troglitazone. These modifications correlated with particular PPARgamma transcripts excluding the untranslated exon A2. HT-29 cells treatment with actinomycin D or cycloheximide showed that the presence of PPARgamma mRNA including exon A2 was dependent on de novo protein synthesis. We concluded that diverse PPARgamma1 mRNA exist in colorectal cells. Levels of PPARgamma1 transcript varied according to the phenotype of colon cell model used. We suggest that regulation of PPARgamma1 mRNA levels could be dependent in part on the composition of untranslated exon(s) in the 5' UTR of PPARgamma1 mRNA.


Asunto(s)
Neoplasias del Colon/genética , PPAR gamma/genética , Secuencia de Bases , Células CACO-2 , Cromanos/farmacología , Mapeo Cromosómico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Cicloheximida/farmacología , Dactinomicina/farmacología , Exones , Células HCT116 , Células HT29 , Humanos , PPAR gamma/agonistas , PPAR gamma/metabolismo , Pioglitazona , Isoformas de Proteínas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Tiazolidinedionas/farmacología , Troglitazona , Regiones no Traducidas
8.
Int J Oncol ; 28(4): 977-84, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16525649

RESUMEN

This study tests the hypothesis that the activators of peroxisome proliferator-activated receptors (PPARs) and 9-cis-retinoic acid receptor (RXR) regulate human semaphorin 6B (Sema6B) gene expression. The human MCF-7 breast adenocarcinoma cell line was chosen because it expresses Sema6B at a high level. The Sema6B mRNA level was analyzed by RT-PCR and the semaphorin 6B protein content was determined using a polyclonal antibody that we have produced and characterized. Treatments with fenofibrate (a PPARalpha activator) and troglitazone (a PPARgamma ligand) strongly decreased the Sema6B mRNA. The drop in Sema6B mRNA level and in protein content was more important when the treatment combined the action of fenofibrate or troglitazone and 9-cis-retinoic acid. On the other hand, no significant change was observed in the Sema6B mRNA and protein levels when the cells were exposed to the combined action of GW610742 (a PPARbeta activator) and 9-cis-retinoic acid. These data suggest that PPARalpha/RXR and PPARgamma/RXR heterodimers are involved in the regulation of Sema6B gene expression and open new perspectives concerning the participation of these nuclear receptors in cell recognition and migration.


Asunto(s)
Cromanos , Fenofibrato , Semaforinas , Tiazolidinedionas , Humanos , Alitretinoína , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Cromanos/farmacología , Dimerización , Fenofibrato/farmacología , Expresión Génica/efectos de los fármacos , Células HT29 , Células K562 , PPAR alfa/agonistas , PPAR alfa/química , PPAR gamma/agonistas , PPAR gamma/química , Receptores X Retinoide/agonistas , Receptores X Retinoide/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Tiazoles/farmacología , Tiazolidinedionas/farmacología , Tretinoina/farmacología , Troglitazona
9.
Int J Mol Med ; 16(3): 483-92, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16077959

RESUMEN

Hypolipidemic drugs (HP drugs) are xenobiotics belonging to the peroxisome proliferator family which are used as pharmaceuticals in the treatment of human hyperlipidemia and hypercholesterolemia. They cause hepatocarcinogenesis in rodents by increasing cell proliferation. One hypothesis is that this hepatocarcinogenic effect is caused by induced oxidative stress resulting from the overproduction of reactive oxygen species (ROS) and from a decreasing antioxidant defense. In addition, ROS play a role in hepatocellular proliferation by activation of NF-kappaB and AP-1, leading to an increase in mitogenic cytokines such as tumor necrosis factor-alpha. No liver cancer incidence has been noted in individuals treated with HP drugs for brief periods of time. However, the observation that old rats and mice are more susceptible than young individuals to the hepatocarcinogenic effect caused by long term exposure to HP drugs raises the question of a potential health risk for the human population. In vitro, HP drugs cause an apoptogenic effect in human hepatocytes. This effect is related to a moderate antioxidant response, dysfunction of mitochondria caused by an overproduction of ROS and release of apoptogenic factors. Finally, the apoptogenic effect of HP drugs is observed in human hepatomas, suggesting a clinical interest of these agents in antitumoral activity.


Asunto(s)
Hipolipemiantes/efectos adversos , Hígado/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factores de Edad , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Hiperlipidemias/tratamiento farmacológico , Hipolipemiantes/uso terapéutico , Hígado/metabolismo , Hígado/patología , Ratones , FN-kappa B/metabolismo , Ratas , Superóxido Dismutasa/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Hypertension ; 46(2): 372-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15967870

RESUMEN

Specific treatment of age-related aortic wall arteriosclerosis and stiffening is lacking. Because ligands for peroxisome proliferator-activated receptor gamma have beneficial effects on the arterial wall in atherosclerosis, via an antiinflammatory mechanism, we investigated whether long-term pioglitazone (Pio) treatment protects against another form of vascular wall disease, arteriosclerosis. We evaluated, in a rat model of elastocalcinotic arteriosclerosis (hypervitaminosis D and nicotine [VDN]), whether Pio (3 mg . kg(-1) per day for 1.5 month PO) attenuated arteriosclerosis and its consequences: aortic wall rigidity, increased aortic pulse pressure, and left ventricular hypertrophy. In VDN rats, medial calcification was associated with monocyte/macrophage infiltration and induction of tumor necrosis factor alpha and interleukin 1beta. Pio increased nuclear peroxisome proliferator-activated receptor gamma immunostaining in the aortic wall, decreased tumor necrosis factor alpha (P <0.05 versus VDN Pio-), tended to decrease interleukin 1beta mRNA expression (P =0.08 versus VDN Pio-), blunted aortic wall calcification (271+/-69, P <0.05 versus VDN Pio- 562+/-87 micromol . g(-1) dry weight) and prevented fragmentation of elastic fibers (segments per 10,000 microm2: 8.4+/-0.3; P <0.05 versus VDN Pio- 10.5+/-0.6). Pio reduced aortic wall stiffness (elastic modulus/wall stress: 4.8+/-0.6; P <0.05 versus VDN Pio- 10.0+/-1.6), aortic pulse pressure (30+/-2 mm Hg; P <0.05 versus VDN Pio- 39+/-4) and left ventricular hypertrophy (1.58+/-0.05 g . kg(-1); P <0.05 versus VDN Pio- 1.76+/-0.06). In conclusion, long-term Pio treatment attenuates aortic wall elastocalcinosis and, thus, lowers aortic wall stiffness, aortic pulse pressure, and left ventricular hypertrophy.


Asunto(s)
Aorta/fisiopatología , Enfermedades de la Aorta/fisiopatología , Arteriosclerosis/fisiopatología , Calcinosis/fisiopatología , Tejido Elástico/fisiopatología , Tiazolidinedionas/farmacología , Animales , Aorta/patología , Aorta Torácica/metabolismo , Aorta Torácica/patología , Enfermedades de la Aorta/patología , Arteriosclerosis/patología , Presión Sanguínea/efectos de los fármacos , Calcinosis/patología , Citocinas/metabolismo , Tejido Elástico/patología , Elasticidad , Hipertrofia Ventricular Izquierda/patología , Macrófagos/patología , Masculino , Monocitos/patología , Miocardio/patología , Tamaño de los Órganos/efectos de los fármacos , PPAR gamma/metabolismo , Pioglitazona , Ratas , Ratas Wistar
11.
Neuropharmacology ; 48(5): 673-84, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15814102

RESUMEN

In order to approach the astroglial implication of addictive and neurotoxic processes associated with psychostimulant drug abuse, the effects of amphetamine or cocaine (1-100 microM) on redox status, AP-1 transcription factor and pro-enkephalin, an AP-1 target gene, were investigated in the human astrocyte-like U373 MG cells. We demonstrated an early increase in the generation of radical oxygen species and in the formation of 4-hydroxynonenal-adducts reflecting the pro-oxidant action of both substances. After 1 h or 96 h of treatment, Fos and Jun protein levels were altered and the DNA-binding activity of AP-1 was increased in response to both substances. Using supershift experiments, we observed that the composition of AP-1 dimer differed according to the substance and the duration of treatment. FRA-2 protein represented the main component of the chronic amphetamine- or cocaine-activated complexes, which suggests its relevance in the long-term effects of psychostimulant drugs. Concomitantly, the pro-enkephalin gene was differently regulated by either 6 h or 96 h of treatment. Because astrocytes interact extensively with the neurons in the brain, our data led us to conclude that oxidation-regulated AP-1 target genes may represent one of the molecular mechanisms underlying neuronal adaptation associated with psychostimulant dependence.


Asunto(s)
Anfetamina/farmacología , Astrocitos/efectos de los fármacos , Fármacos del Sistema Nervioso Central/farmacología , Cocaína/farmacología , Encefalinas/metabolismo , Precursores de Proteínas/metabolismo , Factor de Transcripción AP-1/metabolismo , Sistema de Transporte de Aminoácidos X-AG/genética , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Análisis de Varianza , Western Blotting/métodos , Línea Celular , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Ensayo de Cambio de Movilidad Electroforética/métodos , Encefalinas/genética , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Fluoresceínas , Antígeno 2 Relacionado con Fos , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Oxidación-Reducción/efectos de los fármacos , Precursores de Proteínas/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sales de Tetrazolio , Tiazoles , Factores de Tiempo , Factores de Transcripción/metabolismo
12.
Biochimie ; 86(9-10): 633-42, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15556273

RESUMEN

We showed that the metabolism of arachidonic acid (AA) in HepG2 cells generates reactive oxygen species (ROS), which activate the p38 mitogen-activated protein kinase (MAPK) pathway and the redox-sensitive transcription factors AP-1 and NF-kappaB, leading to the induction of the antioxidant manganese superoxide dismutase gene. The present study reports that AA decreases the HepG2 cell growth by 40% and 55% after a treatment for 24 and 48 h, respectively. This effect was blocked by an inhibitor of lipoxygenase/cytochrome P450 monooxygenase pathways and by the antioxidants. In addition, AA induced an oxidative stress, as an accumulation of malondialdehyde (MDA)-modified proteins, resulting to a generation of MDA and H(2)O(2) was observed after 24 h. This AA-induced oxidative stress was associated with the lack of an increase in the H(2)O(2)-degrading enzyme level. In contrast, 5,8,11,14-eicosatetraynoic acid, a nonmetabolizable analog of AA, had not effect. The peroxisome proliferator-activated receptor gamma (PPARgamma) with AA metabolites as ligands was upregulated by the fatty acid but was not involved in the AA effect because its transcriptional activity estimated by reporter gene assays was negatively controlled by p38 MAPK pathway. These findings suggest that the effect of AA on human hepatoma cell growth by inducing an oxidative stress may present a clinical interest in the treatment of the liver cancer.


Asunto(s)
Ácido Araquidónico/farmacología , Carcinoma Hepatocelular/metabolismo , Proliferación Celular/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Neoplasias Hepáticas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ácido Araquidónico/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
13.
Genomics ; 83(6): 1141-50, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15177567

RESUMEN

The peroxisome proliferator-activated receptors (PPARs) are ligand-inducible transcription factors and belong to the nuclear hormone receptor superfamily. They form heterodimers with the retinoid X receptor and bind to specific peroxisome proliferator-response elements. The latter are direct repeat elements of two hexanucleotides with the consensus sequence TG(A/T)CCT separated by a single nucleotide spacer. Such a sequence, or a similar one, has been found in numerous PPAR-inducible genes. We developed an affinity method to isolate human genomic fragments containing binding sites for PPARs and to identify novel PPAR target genes. For this, an antibody raised against all PPAR subtypes was used. Immunoselected fragments were amplified and sequenced and one of them, ISF5148, was found to bind specifically to PPARs in gel mobility shift, supershift, and competition assays and to exhibit a down transregulation potentiality in transfection experiments under clofibrate (a PPARalpha agonist) treatment. ISF5148 was mapped by BLAST analysis 8.5 kb upstream of the human semaphorin 6B [(HSA)SEMA6B] gene. The latter encodes a member of the semaphorin family of axon guidance molecules. Expression of this gene in human glioblastoma T98G cells was strongly down regulated after treatment with clofibrate or Wy-14,643, two PPARalpha agonists. Our study establishes for the first time that PPAR activators diminish the expression of the human (HSA)SEMA6B gene. These data are relevant to the fact that PPARs are implicated in brain development, neuronal differentiation, and lipid metabolism in the central nervous system. In addition, cross talk between the peroxisome proliferator and retinoic acid pathways is suggested.


Asunto(s)
Regulación hacia Abajo , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Elementos de Respuesta/genética , Semaforinas/genética , Región de Flanqueo 5'/genética , Secuencia de Bases , Sitios de Unión , Unión Competitiva/genética , Línea Celular Tumoral , Clofibrato/farmacología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Componentes Genómicos/genética , Glioblastoma/metabolismo , Humanos , Inmunoprecipitación , Datos de Secuencia Molecular , Receptores Activados del Proliferador del Peroxisoma/genética , Unión Proteica , Pirimidinas/farmacología , ARN Mensajero/análisis , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo
14.
Free Radic Biol Med ; 35(6): 636-47, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12957656

RESUMEN

Exogenous arachidonic acid (AA) has been shown to induce the antioxidant manganese superoxide dismutase gene by reactive oxygen species (ROS) derived from AA metabolism and the participation of the p38 mitogen-activated protein kinase (MAPK) pathway in human HepG2 hepatoma cells. The goal of this study was to investigate the effect of AA on the activation of the two redox-sensitive transcription factors AP-1 and NF-kappaB in HepG2 cells. Using electrophoretic mobility shift assays, DNA-binding activities of AP-1 and NF-kappaB were markedly increased in AA-treated HepG2 cells. The c-Jun and c-Fos proteins were identified as components of the AA-induced AP-1 complex and their levels were increased. AA-activated NF-kappaB complex was constituted as a p50 homodimer resulting in a nuclear translocation for this protein only. Moreover, no degradation of IkappaBalpha was observed. These results were contrasted to the interleukin-1beta-activated p50/p65 complex used as a positive control. Using 5,8,11,14-eicosatetraynoic acid and inhibitors of AA metabolism, AP-1 and NF-kappaB activation required the lipoxygenase/cytochrome P450 monooxygenase pathways. In addition, antioxidants inhibited the AA-induced AP-1 and NF-kappaB activation, suggesting a role of ROS released from the AA metabolism. In reporter gene assays, AA induced the transcriptional activity of AP-1 but not that of NF-kappaB. Further investigations showed that the AA-induced transcriptional activity of AP-1 was regulated by protein kinase C and p38 MAPK pathways. These results suggest that the functional AP-1 activated by AA and coupled to that of p38 MAPK pathway may play an important role in response to ROS induced by AA metabolism in HepG2 cells without the involvement of the NF-kappaB pathway.


Asunto(s)
Ácido Araquidónico/farmacología , FN-kappa B/metabolismo , Factor de Transcripción AP-1/metabolismo , Ácido Araquidónico/metabolismo , Línea Celular Tumoral , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Unión Proteica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
15.
Microsc Res Tech ; 61(2): 185-90, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12740825

RESUMEN

Peroxisome proliferator-activated receptor alpha (PPARalpha) is a member of the nuclear hormone receptor superfamily that can be activated by natural fatty acids and various xenobiotics, including clofibrate. This transcription factor primarily regulates genes involved in lipid metabolism and homeostasis. We present the expression pattern of the PPARalpha subtype in the adult jerboa Jaculus orientalis, determined by RT-PCR and Western blotting using specific probes and a polyclonal antibody for PPARalpha, respectively. PPARalpha is highly expressed in liver and kidney, and to a lesser extent in duodenum and colon. PPARalpha expression is increased at the mRNA and protein levels in liver and duodenum of jerboa treated for 2 weeks with the peroxisome proliferator (PP) clofibrate. The induction is tissue-specific as no significant changes are observed in kidney and colon. The present data indicate that the PP-induced PPARalpha gene expression is not dependent on the PPARalpha content in target cells.


Asunto(s)
Clofibrato/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proliferadores de Peroxisomas/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Roedores/metabolismo , Factores de Transcripción/metabolismo , Animales , Mucosa Intestinal/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética
16.
Int J Oncol ; 22(4): 899-905, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12632085

RESUMEN

Regulation of growth arrest and apoptosis are, in part, controlled by the tumor suppressor p53 after its phosphorylation which causes a determinant role in its functional activation. Moreover, PPAR regulate many functions such as proliferation and apoptosis. We compared the biological activity of diosgenin with hecogenin and tigogenin, plant steroids structurally close to diosgenin, on proliferation rate, cell cycle distribution and apoptosis in human 1547 osteosarcoma cells. We found that all three molecules have an antiproliferative effect but gel shift analysis demonstrated that none of the plant steroids transactivated PPAR in human 1547 osteosarcoma cells whereas these molecules induced NF-kappaB binding to DNA. Although these plant steroids have a very close structure, only diosgenin caused a cell cycle arrest associated with strong apoptosis. This biological action seems correlated with a large increase of p53 protein expression. This fact was showed by immunofluorescence analysis which confirmed that diosgenin strongly enhanced the activation of p53 in contrast to hecogenin and tigogenin actions.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Diosgenina/farmacología , Medicamentos Herbarios Chinos/farmacología , Osteosarcoma/tratamiento farmacológico , Sapogeninas/farmacología , Espirostanos/farmacología , Western Blotting , Ciclo Celular , División Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Electroforesis en Gel de Poliacrilamida , Humanos , Luciferasas/metabolismo , Microscopía Fluorescente , Modelos Químicos , Receptores Citoplasmáticos y Nucleares/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factores de Transcripción/metabolismo , Activación Transcripcional , Transfección , Proteína p53 Supresora de Tumor/metabolismo
17.
Biol Cell ; 94(1): 15-27, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12000143

RESUMEN

The expression of peroxisome proliferator-activated receptors alpha (PPARalpha) and gamma (PPARgamma) was studied in the human adenocarcinoma Caco-2 cells induced to differentiate by long term culture (15 days). The differentiation of Caco-2 cells was attested by increases in the activities of sucrase-isomaltase and alkaline phosphatase (two brush border enzymes), fatty acyl-CoA oxidase (AOX) and catalase (two peroxisomal enzymes), by an elevation in the protein levels of villin (a brush border molecular marker), AOX, peroxisomal bifunctional enzyme (PBE), catalase and peroxisomal membrane protein of 70 kDa (PMP70). and by the appearance of peroxisomes. The expression of PPARalpha and PPARgamma was investigated by Western blotting, immunocytochemistry, Northern blotting and S1 nuclease protection assay during the differentiation of Caco-2 cells. The protein levels of PPARalpha, PPARgamma, and PPARgamma2 increased gradually during the time-course of Caco-2 cell differentiation. Immunocytochemistry revealed that PPARalpha and gamma were localized in cell nuclei. The PPARgamma1 protein was encoded by PPARgamma3 mRNA because no signal was obtained for PPARgamma1 mRNA using a specific probe in S1 nuclease protection assay. The amount of PPARgamma3 mRNA increased concomitantly to the resulting PPARgamma1 protein. On the other hand, the mRNA of PPARalpha and PPARgamma2 were not significantly changed, suggesting that the increase in their respective protein was due to an elevation of the translational rate. The role played by the PPAR subtypes in Caco-2 cell differentiation is discussed.


Asunto(s)
Neoplasias del Colon/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Factores de Transcripción/biosíntesis , Northern Blotting , Western Blotting , Células CACO-2 , Diferenciación Celular , Neoplasias del Colon/patología , Humanos , Inmunohistoquímica , Ensayos de Protección de Nucleasas , Peroxisomas/metabolismo , Peroxisomas/ultraestructura , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética
18.
Free Radic Biol Med ; 32(11): 1132-42, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12031898

RESUMEN

Metabolism of arachidonic acid (AA) is known to induce in different cell types an oxidative stress via the production of reactive oxygen species. As these latter may be scavenged by antioxidant enzymes as manganese and copper/zinc-dependent superoxide dismutase (MnSOD and Cu/ZnSOD, respectively), we investigated the effects of AA on their expression in human HepG2 hepatoma cells. RT-PCR and Western blot data revealed that AA induced an increase in the MnSOD, but not Cu/ZnSOD, expression at the mRNA and protein levels, respectively. This induction was also marked by an increase in MnSOD activity. The AA-induced MnSOD expression required de novo transcription as demonstrated by cotreatment of HepG2 cells with AA and actinomycin D. The fact that MnSOD expression was not induced when HepG2 cells were cultured with 5,8,11,14-eicosatetraynoic acid (ETYA), a nonmetabolizable analog of AA, or with different inhibitors of the AA metabolism pathways suggested that the metabolism of AA was required. Further investigations into the mechanisms by which AA induced MnSOD expression showed that superoxide anions released from AA metabolism act as second messengers via a signal-controlling pathway involving protein kinase C and p38 mitogen activated protein kinase (MAPK). These results define a novel role of p38 MAPK dependent-pathway in the regulation of MnSOD gene.


Asunto(s)
Ácido Araquidónico/farmacología , Carcinoma Hepatocelular/enzimología , Neoplasias Hepáticas/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/genética , Superóxidos/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Ácido 5,8,11,14-Eicosatetrainoico/farmacología , Western Blotting , Carcinoma Hepatocelular/patología , División Celular/efectos de los fármacos , Cartilla de ADN/química , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Humanos , Neoplasias Hepáticas/patología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Naftalenos/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/biosíntesis , Sales de Tetrazolio , Tiazoles , Células Tumorales Cultivadas/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos
19.
Biochem Pharmacol ; 63(2): 237-45, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11841798

RESUMEN

Among the different hypotheses advanced to explain the peroxisome proliferator (PP)-induced hepatocarcinogenicity in rodents, one is based on the development of an oxidative stress due to an imbalance in the production of reactive oxygen species that leads to DNA damages and lipid peroxidation. On the other hand, human cells appear to be nonresponsive to PPs. As metallothionein proteins play an important antioxidant role, the aim of the present study was to investigate the expression of metallothionein IA (MTIA) and IIA (MTIIA) in HepG2 human hepatoma cells exposed to clofibric acid. When HepG2 cells were treated for 24 hr with 0.50 or 0.75 mM CA, a significant decrease was observed in MT protein-level determined by Western blotting and in the MTIIA mRNA content analyzed by RT-PCR and Northern blotting. No significant change was observed in the MTIA mRNA amount whatever the CA concentration and the duration of treatment. The decrease in MTIIA mRNA-level was not mediated via peroxisome proliferator-activated receptor alpha as attested by our data from gel mobility shift DNA binding assays, Dot blotting and cotransfection experiments with MTIIA promoter-driven luciferase reporter gene and PPARalpha expression vector. These results provide new insights about the pleiotropic effects of PPs on human cells.


Asunto(s)
Ácido Clofíbrico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Metalotioneína/metabolismo , Carcinoma Hepatocelular , Regulación hacia Abajo/efectos de los fármacos , Humanos , Hipolipemiantes/farmacología , Neoplasias Hepáticas , Metalotioneína/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
20.
Am J Physiol Cell Physiol ; 282(1): C125-33, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11742805

RESUMEN

This work demonstrated the constitutive expression of peroxisome proliferator-activated receptor (PPAR)-gamma and PPAR-alpha in rat synovial fibroblasts at both mRNA and protein levels. A decrease in PPAR-gamma expression induced by 10 microg/ml lipopolysaccharide (LPS) was observed, whereas PPAR-alpha mRNA expression was not modified. 15-Deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) dose-dependently decreased LPS-induced cyclooxygenase (COX)-2 (-80%) and inducible nitric oxide synthase (iNOS) mRNA expression (-80%), whereas troglitazone (10 microM) only inhibited iNOS mRNA expression (-50%). 15d-PGJ(2) decreased LPS-induced interleukin (IL)-1 beta (-25%) and tumor necrosis factor (TNF)-alpha (-40%) expression. Interestingly, troglitazone strongly decreased TNF-alpha expression (-50%) but had no significant effect on IL-1 beta expression. 15d-PGJ(2) was able to inhibit DNA-binding activity of both nuclear factor (NF)-kappa B and AP-1. Troglitazone had no effect on NF-kappa B activation and was shown to increase LPS-induced AP-1 activation. 15d-PGJ(2) and troglitazone modulated the expression of LPS-induced iNOS, COX-2, and proinflammatory cytokines differently. Indeed, troglitazone seems to specifically target TNF-alpha and iNOS pathways. These results offer new insights in regard to the anti-inflammatory potential of the PPAR-gamma ligands and underline different mechanisms of action of 15d-PGJ(2) and troglitazone in synovial fibroblasts.


Asunto(s)
Fibroblastos/metabolismo , Lipopolisacáridos/farmacología , Prostaglandina D2/análogos & derivados , Receptores Citoplasmáticos y Nucleares/metabolismo , Membrana Sinovial/citología , Tiazolidinedionas , Factores de Transcripción/metabolismo , Animales , Antineoplásicos/farmacología , Células Cultivadas , Cromanos/farmacología , Ciclooxigenasa 2 , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Interleucina-1/genética , Isoenzimas/genética , Ligandos , Masculino , FN-kappa B/antagonistas & inhibidores , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Prostaglandina D2/farmacología , Prostaglandina-Endoperóxido Sintasas/genética , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/genética , Membrana Sinovial/inmunología , Tiazoles/farmacología , Factor de Transcripción AP-1/antagonistas & inhibidores , Factores de Transcripción/genética , Troglitazona , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA