Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Exp Cell Res ; 382(1): 111455, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31163124

RESUMO

Diabetic nephropathy (DN) is one of the most severe complications of diabetes mellitus. The pathomolecular events behind DN remain uncertain. Peroxisome proliferator-activated receptors (PPARs) play essential functions in the development of DN. Meanwhile, 20-hydroxyeicosatetraenoic acid (20-HETE) also plays central roles in the regulation of renal function. However, the relationship between PPARs and 20-HETE is rarely studied in DN. It was revealed in our study that both PPARs expression and CYP4A-20-HETE level were decreased under DN conditions in vivo and in vitro. Supplementation with bezafibrate, a PPAR pan-agonist, improved the damage of kidney in DN mice and in high glucose-induced NRK-52E cells, following the up-regulation of PPARs and the increase of CYP4A-20-HETE. PPARα antagonist (MK886), PPARß antagonist (GSK0660), and PPARγ antagonist (GW9662) reversed the protection of bezafibrate in NRK-52E, and abrogated the up-regulation of CYP4A-20-HETE produced by bezafibrate. Noteworthily, 20-HETE synthetase inhibitor, HET0016, also blocked the bezafibrate-mediated improvement of NRK-52E, and abolished the up-regulation of PPARs expression. Collectively, our data suggest that the concurrent down-regulation and interaction of PPARs and 20-HETE play crucial roles in the pathogenesis process of DN, and we provide a novel evidence that PPARs/20-HETE signaling may be served as a therapeutic target for DN patients.


Assuntos
Nefropatias Diabéticas/metabolismo , Ácidos Hidroxieicosatetraenoicos/fisiologia , PPAR alfa/fisiologia , PPAR gama/fisiologia , PPAR beta/fisiologia , Amidinas/farmacologia , Anilidas/farmacologia , Animais , Linhagem Celular , Citocromo P-450 CYP4A/metabolismo , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/patologia , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/toxicidade , Ácidos Hidroxieicosatetraenoicos/biossíntese , Indóis/farmacologia , Túbulos Renais/citologia , Masculino , Camundongos , PPAR alfa/biossíntese , PPAR alfa/genética , PPAR gama/biossíntese , PPAR gama/genética , PPAR beta/biossíntese , PPAR beta/genética , Ratos , Sulfonas/farmacologia , Tiofenos/farmacologia
2.
Environ Pollut ; 244: 588-599, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30384064

RESUMO

Adenosine triphosphate-binding cassette (ABC) transporters, including P-glycoprotein (Pgp) and multi-resistance associated proteins (Mrps), have been considered important participants in the self-protection of zebrafish embryos against environmental pollutants, but their possible involvement in the efflux and detoxification of quantum dots (QDs), as well as their regulation mechanism are currently unclear. In this work, gene expression alterations of ABC transporters, nuclear receptors, and oxidative stress signaling in zebrafish embryos after the treatment of mercaptopropionic acid (MPA)CdTe QDs and MPA-CdSCdTe QDs were investigated. It was observed that both QDs caused concentration-dependent delayed hatching effects and the subsequent induction of transporters like mrp1&2 in zebrafish embryos, indicating the protective role of corresponding proteins against CdTe QDs. Accompanying these alterations, expressions of nuclear receptors including the pregnane X receptor (pxr), aryl hydrocarbon receptor (ahr) 1b, and peroxisome proliferator-activated receptor (ppar)-ß were induced by QDs in a concentration- and time-dependent manner. Moreover, elevated oxidative stress, reflected by the reduction of glutathione (GSH) level and superoxide dismutase (SOD) activities, as well as the dramatic induction of nuclear factor E2 related factor (nrf) 2, was also found. More importantly, alterations of pxr and nrf2 were more pronounced than that of mrps, and these receptors exhibited an excellent correlation with delayed hatching rate in the same embryos (R2 > 0.8). Results from this analysis demonstrated that the induction of mrp1 and mrp2 could be important components for the detoxification of QDs in zebrafish embryos. These transporters could be modulated by nuclear receptors and oxidative stress signaling. In addition, up-regulation of pxr and nrf2 could be developed as toxic biomarkers of CdTe QDs.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Biotransformação/fisiologia , Compostos de Cádmio/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Pontos Quânticos/toxicidade , Receptores Citoplasmáticos e Nucleares/metabolismo , Telúrio/toxicidade , Peixe-Zebra/metabolismo , Ácido 3-Mercaptopropiônico/química , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Embrião não Mamífero/efeitos dos fármacos , Glutationa/metabolismo , Inativação Metabólica , Proteínas Associadas à Resistência a Múltiplos Medicamentos/biossíntese , Fator 2 Relacionado a NF-E2/metabolismo , PPAR beta/biossíntese , Receptor de Pregnano X/biossíntese , Pontos Quânticos/química , Receptores de Hidrocarboneto Arílico/biossíntese , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Peixe-Zebra/embriologia
3.
Toxicology ; 404-405: 25-32, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29729928

RESUMO

To examine the functional role of peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) and PPARγ in skin cancer, stable cell lines were created in the A431 human squamous cell carcinoma cell line. Expression of PPAR target genes was greatly enhanced in response to ligand activation of PPARß/δ or PPARγ in A431 cells expressing these receptors. PPARß/δ expression blocked the cell cycle at the G2/M phase, and this effect was increased by ligand activation. Ligand activation of PPARß/δ markedly inhibited clonogenicity as compared to vehicle-treated controls. Similarly, ligand activation of PPARγ in A431 cells expressing PPARγ resulted in reduced clonogenicity. Expression of either PPARß/δ or PPARγ markedly reduced tumor volume in ectopic xenografts, while ligand activation of these receptors had little further influence on tumor volume. Collectively, these studies demonstrate that stable expression and activation of PPARß/δ or PPARγ in A431 cells led to reduced tumorigenicity. Importantly, PPAR expression or ligand activation had major impacts on clonogenicity and/or tumor volume. Thus, PPARß/δ or PPARγ could be therapeutically targeted for the treatment of squamous cell carcinomas.


Assuntos
Carcinogênese/metabolismo , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular/fisiologia , PPAR delta/biossíntese , PPAR beta/biossíntese , Neoplasias Cutâneas/metabolismo , Animais , Carcinoma de Células Escamosas/prevenção & controle , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos Nus , Neoplasias Cutâneas/prevenção & controle , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
4.
Stroke ; 47(1): 196-205, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26628385

RESUMO

BACKGROUND AND PURPOSE: Early brain injury is proposed to be the primary cause of the poor outcome after subarachnoid hemorrhage (SAH), which is closely related to the neural apoptosis. To date, the relationship between peroxisome proliferator-activated receptor ß/δ (PPARß/δ) and nuclear factor-κB/matrix metalloproteinase-9 (NF-κB/MMP-9) pathway, both of which are closely related to apoptotic effects, has been poorly studied in SAH. The present study was undertaken to evaluate the effects of PPARß/δ on early brain injury and NF-κB/MMP-9 pathway after SAH in rats. METHODS: SAH model was established by injecting nonheparinized autologous arterial blood into the prechiasmatic cistern in male Sprague-Dawley rats. Adenoviruses or small interfering RNAs were injected into the right lateral cerebral ventricle to, respectively, up- or downregulate PPARß/δ expression before SAH. All animals were assessed with a neurological score and then killed at 24 hours after SAH surgery. The indexes of brain water content, blood-brain barrier permeability, and apoptosis were used to detect brain injury. The expression of PPARß/δ, NF-κB, and MMP-9 were measured by immunohistochemistry, gelatin zymography, and Western Blot methods, respectively. In addition, GW0742, a specific agonist of PPARß/δ, was used to treat SAH in rats, the effects of which were evaluated by neurological scoring and Evans blue extravasation. RESULTS: Overexpression of PPARß/δ by adenoviruses treatment significantly ameliorated brain injury with improvement in neurological deficits, brain edema, blood-brain barrier impairment, and neural cell apoptosis at 24 hours after SAH in rats, whereas downregulation of PPARß/δ by small interfering RNAs administration resulted in the reverse effects of the above. The expression levels of NF-κB and MMP-9 were markedly downregulated when PPARß/δ increased after PPARß/δ adenovirus transfection and upregulated when PPARß/δ decreased by PPARß/δ small interfering RNAs treatment. Moreover, GW0742 improved neurological deficits and reduced Evans blue extravasation at 24 hours after SAH. CONCLUSIONS: PPARß/δ's overexpression may attenuate early brain injury after rats' SAH administration, which reduces neural apoptosis possibly through blocking NF-κB/MMP-9 pathway.


Assuntos
Lesões Encefálicas/metabolismo , Lesões Encefálicas/prevenção & controle , PPAR delta/biossíntese , PPAR beta/biossíntese , Hemorragia Subaracnóidea/metabolismo , Animais , Lesões Encefálicas/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/patologia
5.
Vopr Onkol ; 62(5): 626-631, 2016.
Artigo em Russo | MEDLINE | ID: mdl-30695588

RESUMO

The significance of quantitative changes of ALDH1A1 and RDH10 gene expression in 22 non-treated multiple myeloma patients were studied. We found a direct correlation between the expression of ALDH1A1 and RDH10 genes. We showed that ALDHA1 and RDH10 expression were inversely related with expression of a key gene for all-trans-retinoic acid catabolism, CYP26A1, and correlated with expression of RARα and PPARß/ genes. In addition for the first time it was re- vealed that increased expression of ALDH1A1-RDH10-RARα- PPARß/δ pattern could be considered as adverse prognostic factor associated with a higher concentration of paraprotein and worst overall survival of patients with newly diagnosed multiple myeloma.


Assuntos
Oxirredutases do Álcool/biossíntese , Aldeído Desidrogenase/biossíntese , Regulação Neoplásica da Expressão Gênica , Mieloma Múltiplo , Proteínas de Neoplasias/biossíntese , PPAR delta/biossíntese , PPAR beta/biossíntese , Receptor alfa de Ácido Retinoico/biossíntese , Tretinoína/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Oxirredutases do Álcool/genética , Aldeído Desidrogenase/genética , Família Aldeído Desidrogenase 1 , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/genética , PPAR delta/genética , PPAR beta/genética , Retinal Desidrogenase , Receptor alfa de Ácido Retinoico/genética
6.
PLoS One ; 7(9): e46009, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23049921

RESUMO

Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a nuclear receptor involved in regulation of lipid and glucose metabolism, wound healing and inflammation. PPARß/δ has been associated also with cancer. Here we investigated the expression of PPARß/δ and components of the prostaglandin biosynthetic pathway in non-small cell lung cancer (NSCLC). We found increased expression of PPARß/δ, Cox-2, cPLA(2), PGES and VEGF in human NSCLC compared to normal lung. In NSCLC cell lines PPARß/δ activation increased proliferation and survival, while PPARß/δ knock-down reduced viability and increased apoptosis. PPARß/δ agonists induced Cox-2 and VEGF transcription, suggesting the existence of feed-forward loops promoting cell survival, inflammation and angiogenesis. These effects were seen only in high PPARß/δ expressing cells, while low expressing cells were less or not affected. The effects were also abolished by PPARß/δ knock-down or incubation with a PPARß/δ antagonist. Induction of VEGF was due to both binding of PPARß/δ to the VEGF promoter and PI3K activation through a non-genomic mechanism. We found that PPARß/δ interacted with the PI3K regulatory subunit p85α leading to PI3K activation and Akt phosphorylation. Collectively, these data indicate that PPARß/δ might be a central element in lung carcinogenesis controlling multiple pathways and representing a potential target for NSCLC treatment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , PPAR delta/biossíntese , PPAR beta/biossíntese , Transcrição Gênica , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Inibidores Enzimáticos/farmacologia , Genômica , Humanos , Inflamação , PPAR delta/fisiologia , PPAR beta/fisiologia , Fosforilação , Regiões Promotoras Genéticas , Interferência de RNA
7.
J Biol Chem ; 287(30): 25038-48, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22685301

RESUMO

ß-Adrenergic receptors (ß-ARs) promote brown adipose tissue (BAT) thermogenesis by mobilizing fatty acids and inducing the expression of oxidative genes. ß-AR activation increases the expression of oxidative genes by elevating cAMP, but whether lipolytic products can modulate gene expression is not known. This study examined the role that adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) plays in the induction of gene expression. Activation of brown adipocytes by ß-AR agonism or 8-bromo-cyclic AMP increased the expression of PGC1α, PDK4, PPARα, uncoupling protein 1 (UCP1), and neuron-derived orphan receptor-1 (NOR-1), and concurrent inhibition of HSL reduced the induction of PGC1α, PDK4, PPARα, and UCP1 but not NOR-1. Similar results were observed in the BAT of mice following pharmacological or genetic inhibition of HSL and in brown adipocytes with stable knockdown of ATGL. Conversely, treatments that increase endogenous fatty acids elevated the expression of oxidative genes. Pharmacological antagonism and siRNA knockdown indicate that PPARα and PPARδ modulate the induction of oxidative genes by ß-AR agonism. Using a live cell fluorescent reporter assay of PPAR activation, we demonstrated that ligands for PPARα and -δ, but not PPARγ, were rapidly generated at the lipid droplet surface and could transcriptionally activate PPARα and -δ. Knockdown of ATGL reduced cAMP-mediated induction of genes involved in fatty acid oxidation and oxidative phosphorylation. Consequently, ATGL knockdown reduced maximal oxidation of fatty acids, but not pyruvate, in response to cAMP stimulation. Overall, the results indicate that lipolytic products can activate PPARα and PPARδ in brown adipocytes, thereby expanding the oxidative capacity to match enhanced fatty acid supply.


Assuntos
Adipócitos Marrons/metabolismo , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/fisiologia , Lipólise/fisiologia , PPAR alfa/biossíntese , PPAR beta/biossíntese , 8-Bromo Monofosfato de Adenosina Cíclica/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Células Cultivadas , Ácidos Graxos/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Canais Iônicos/genética , Canais Iônicos/metabolismo , Lipase/genética , Lipase/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Oxirredução , PPAR alfa/genética , PPAR beta/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição , Proteína Desacopladora 1
8.
Biochem Biophys Res Commun ; 386(3): 459-62, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19527689

RESUMO

Macrophages adapt their response to micro-environmental signals. While Th1 cytokines promote pro-inflammatory M1 macrophages, Th2 cytokines promote an "alternative" anti-inflammatory M2 macrophage phenotype. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors expressed in macrophages where they control the inflammatory response. It has been shown that PPARgamma promotes the differentiation of monocytes into anti-inflammatory M2 macrophages in humans and mice, while a role for PPARbeta/delta in this process has been reported only in mice and no data are available for PPARalpha. Here, we show that in contrast to PPARgamma, expression of PPARalpha and PPARbeta/delta overall does not correlate with the expression of M2 markers in human atherosclerotic lesions, whereas a positive correlation with genes of lipid metabolism exists. Moreover, unlike PPARgamma, PPARalpha or PPARbeta/delta activation does not influence human monocyte differentiation into M2 macrophages in vitro. Thus, PPARalpha and PPARbeta/delta do not appear to modulate the alternative differentiation of human macrophages.


Assuntos
Aterosclerose/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , PPAR alfa/biossíntese , PPAR delta/biossíntese , PPAR beta/biossíntese , Diferenciação Celular , Células Cultivadas , Humanos , Macrófagos/metabolismo , Monócitos/imunologia , PPAR alfa/agonistas , PPAR alfa/genética , PPAR delta/agonistas , PPAR delta/genética , PPAR gama/agonistas , PPAR gama/biossíntese , PPAR gama/genética , PPAR beta/agonistas , PPAR beta/genética
9.
Hum Pathol ; 40(5): 705-13, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19157507

RESUMO

Peroxisome proliferator-activated receptors regulate lipid metabolism, affecting inflammation and cancer. The present study analyzed the anatomical site-related expression and prognostic role of peroxisome proliferator-activated receptors in ovarian carcinoma. Fresh-frozen effusions (n = 79), primary carcinomas (n = 44), and solid metastases (n = 16) were studied for peroxisome proliferator-activated receptor-alpha, -beta, and -gamma messenger RNA expression using reverse transcriptase polymerase chain reaction. Peroxisome proliferator-activated receptor-gamma messenger RNA expression was further assessed in 60 tumors (30 effusions, 20 primary carcinomas, 10 metastases) using in situ hybridization. Peroxisome proliferator-activated receptor-gamma protein expression was immunohistochemically analyzed in 160 effusions. All peroxisome proliferator-activated receptors were expressed in most tumors at all anatomical sites using reverse transcriptase polymerase chain reaction, but peroxisome proliferator-activated receptor-alpha (P = .004) and peroxisome proliferator-activated receptor-beta (P = .002) messenger RNA levels were higher in effusions compared with primary carcinomas and solid metastases. In situ hybridization localized peroxisome proliferator-activated receptor-gamma messenger RNA to carcinoma cells in both effusions and solid lesions. Peroxisome proliferator-activated receptor-gamma protein was detected in carcinoma cells in 102 of 160 (64%) effusions. Higher effusion messenger RNA levels of all peroxisome proliferator-activated receptors were associated with less favorable response to chemotherapy at diagnosis (P = .009). In univariate survival analysis, higher messenger RNA expression of all peroxisome proliferator-activated receptors was associated with poor progression-free (P = .045) and overall (P = .014) survival. Higher peroxisome proliferator-activated receptor-gamma protein expression was similarly associated with poor overall survival for the entire cohort (P = .046) and for patients with disease recurrence effusions (P = .009). Peroxisome proliferator-activated receptors were not independent predictors of survival in Cox multivariate analysis. Peroxisome proliferator-activated receptor members are frequently expressed in ovarian carcinoma, with upregulated expression in effusions. Peroxisome proliferator-activated receptor expression in effusions is associated with poor response to chemotherapy at disease recurrence and poor survival, suggesting a role in tumor biology at this unique microenvironment.


Assuntos
Carcinoma/metabolismo , Neoplasias Ovarianas/metabolismo , PPAR alfa/biossíntese , PPAR gama/biossíntese , PPAR beta/biossíntese , Adulto , Idoso , Antígenos de Plaquetas Humanas/biossíntese , Antineoplásicos/uso terapêutico , Líquido Ascítico , Carcinoma/mortalidade , Carcinoma/patologia , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Derrame Pleural Maligno , Prognóstico , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Resultado do Tratamento
10.
Fertil Steril ; 87(3): 706-9, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17156782

RESUMO

To determine the levels of peroxisome proliferators-activated receptors (PPARs) in normal and adhesion fibroblasts, we utilized real-time reverse transcription-polymerase chain reaction to measure messenger RNA (mRNA) levels in fibroblasts from normal peritoneum and adhesions from five patients in both the presence or absence of dichloroacetic acid (DCA) and a cyclooxygenase-2 (COX-2) inhibitor, NS-398. Peroxisome proliferators-activated receptor alpha, PPARbeta, PPARgamma1, and PPARgamma2 mRNA are all present in normal peritoneal and adhesion fibroblasts, and selectively rose in response to hypoxia and either DCA or NS-398.


Assuntos
Ácido Dicloroacético/farmacologia , Fibroblastos/metabolismo , Nitrobenzenos/farmacologia , Doenças Peritoneais/metabolismo , Peritônio/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/biossíntese , Sulfonamidas/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Hipóxia/metabolismo , PPAR alfa/biossíntese , PPAR gama/biossíntese , PPAR beta/biossíntese , Peritônio/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Aderências Teciduais/metabolismo
11.
Toxicology ; 228(2-3): 140-50, 2006 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-17046140

RESUMO

The lipid regulator gemfibrozil (GEM) is one of many human pharmaceuticals found in the aquatic environment. We previously demonstrated that GEM bioconcentrates in blood and reduces plasma testosterone levels in goldfish (Carassius auratus). In this study, we address the potential of an environmentally relevant waterborne concentration of GEM (1.5 microg/l) to induce oxidative stress in goldfish liver and whether this may be linked to GEM acting as a peroxisome proliferator (PP). We also investigate the autoregulation of the peroxisome proliferator-activated receptors (PPARs) as a potential index of exposure. The three PPAR subtypes (alpha, beta, and gamma) were amplified from goldfish liver cDNA. Goldfish exposed to a concentration higher (1500 microg/l) than environmentally relevant for 14 and 28 days significantly reduce hepatic PPARbeta mRNA levels (p<0.001). Levels of CYP1A1 mRNA were unchanged. GEM exposure significantly induced the antioxidant defense enzymes catalase (p<0.001), glutathione peroxidase (p<0.001) and glutathione-S-transferase (p=0.006) but not acyl-CoA oxidase or glutathione reductase. As GEM exposure failed to increase levels of thiobarbituric reactive substances (TBARS), we conclude that a sub-chronic exposure to GEM upregulates the antioxidant defense status of the goldfish as an adaptive response to this human pharmaceutical.


Assuntos
Antioxidantes/metabolismo , Genfibrozila/toxicidade , Carpa Dourada/metabolismo , Hipolipemiantes/toxicidade , Fígado/metabolismo , PPAR beta/biossíntese , RNA Mensageiro/biossíntese , Poluentes Químicos da Água/toxicidade , Actinas/biossíntese , Actinas/metabolismo , Acil-CoA Oxidase/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Citocromo P-450 CYP1A1/biossíntese , Citocromo P-450 CYP1A1/metabolismo , Regulação para Baixo/efeitos dos fármacos , Glutationa/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Dados de Sequência Molecular , Estresse Oxidativo/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Distribuição Tecidual
12.
Biochem Biophys Res Commun ; 340(2): 656-60, 2006 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-16378595

RESUMO

To investigate the role of peroxisome proliferator-activated receptors (PPARs) alpha and beta in the differentiation of colon cancer cells, we differentiated HT-29 cells using sodium butyrate (NaB) and culturing post-confluence and assessed differentiation using the marker intestinal alkaline phosphatase. While PPARalpha levels only changed with culturing post confluence, PPARbeta levels increased independent of the method of differentiation. To explore further the differences induced by NaB, we assessed changes in both PPAR isoforms in MCF-7 breast cancer cells cultured in the presence of NaB over 48h. Again a very different expression pattern was observed with PPARalpha increasing after 4h and remaining elevated, while PPARbeta increased transiently. Our studies suggest that the expression of PPARs is dependent upon both the method of differentiation and on time. Moreover, these studies show that changes in PPARalpha levels are not required for the differentiation of colon cancer cell lines, whereas changes in PPARbeta are more closely associated with differentiation.


Assuntos
Neoplasias da Mama/metabolismo , Diferenciação Celular/fisiologia , Neoplasias do Colo/genética , PPAR alfa/biossíntese , PPAR beta/biossíntese , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Butiratos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Feminino , Células HT29 , Humanos , PPAR alfa/genética , PPAR beta/genética , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , RNA Mensageiro/biossíntese
13.
Anticancer Res ; 25(6B): 3781-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16309164

RESUMO

BACKGROUND: In Apc(Min/+) (Min; multiple intestinal neoplasia) mice two separate populations of aberrant crypt foci (ACF) develop in the colon after azoxymethane (AOM) exposure. ACF(Min), with a flat appearance, severe dysplasia and increased beta-catenin expression, are related to adenoma development, whereas classic ACF, with elevated structure, hyperplasia and normal beta-catenin level, are probably not. MATERIALS AND METHODS: The expressions of peroxisome proliferator-activated receptors (PPARs) beta/delta, cyclin D1 and beta-catenin in ACF, adenoma and normal tissue from AOM-treated Apc(Min/+) mice and a familial adenomatous polyposis (FAP) patient colon tumour were assessed by immunohistochemistry and immunoblotting. RESULTS: The flat ACF (ACF(Min)) displayed increased cytoplasmic levels of beta-catenin, and increased levels of cyclin D1 and PPARbeta/delta. In contrast, the expression in classic ACF resembled normal mucosa. Adenomas from Apc(Min/+) mice, as well as a FAP patient colon tumour, displayed increased nuclear and cytoplasmic levels of beta-catenin, and the same expression patterns of cyclin D1 and PPARbeta/delta as those found in flat ACF. CONCLUSION: In addition to activation of the Wnt signalling pathway in both flat ACF and in adenomas in Apc(Min/+) mice, the increased expression of PPARbeta/delta in these lesions could be a target for pro-inflammatory signals important for growth and reduced apoptosis.


Assuntos
Adenoma/metabolismo , Neoplasias do Colo/metabolismo , Ciclina D1/biossíntese , PPAR delta/biossíntese , PPAR beta/biossíntese , Lesões Pré-Cancerosas/metabolismo , Adenoma/induzido quimicamente , Adenoma/genética , Animais , Azoximetano , Carcinógenos , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Genes APC , Hiperplasia , Immunoblotting , Camundongos , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/genética , beta Catenina/biossíntese
14.
J Am Soc Nephrol ; 16(8): 2395-402, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15944338

RESUMO

Ischemic acute renal failure is characterized by damages to the proximal straight tubule in the outer medulla. Lesions include loss of polarity, shedding into the tubule lumen, and eventually necrotic or apoptotic death of epithelial cells. It was recently shown that peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) increases keratinocyte survival after an inflammatory reaction. Therefore, whether PPARbeta/delta could contribute also to the control of tubular epithelium death after renal ischemia/reperfusion was tested. It was found that PPARbeta/delta+/- and PPARbeta/delta-/- mutant mice exhibited much greater kidney dysfunction and injury than wild-type counterparts after a 30-min renal ischemia followed by a 36-h reperfusion. Conversely, wild-type mice that were given the specific PPARbeta/delta ligand L-165041 before renal ischemia were completely protected against renal dysfunction, as indicated by the lack of rise in serum creatinine and fractional excretion of Na+. This protective effect was accompanied by a significant reduction in medullary necrosis, apoptosis, and inflammation. On the basis of in vitro studies, PPARbeta/delta ligands seem to exert their role by activating the antiapoptotic Akt signaling pathway and, unexpectedly, by increasing the spreading of tubular epithelial cells, thus limiting potentially their shedding and anoikis. These results point to PPARbeta/delta as a remarkable new target for preconditioning strategies.


Assuntos
Isquemia , Rim/citologia , PPAR delta/fisiologia , PPAR beta/fisiologia , Insuficiência Renal/patologia , Acetatos/farmacologia , Animais , Apoptose , Western Blotting , Células Cultivadas , Creatinina/sangue , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Humanos , Marcação In Situ das Extremidades Cortadas , Inflamação , Queratinócitos/metabolismo , Rim/metabolismo , Rim/patologia , Túbulos Renais/metabolismo , Ligantes , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Necrose , Neutrófilos/patologia , PPAR delta/biossíntese , PPAR beta/biossíntese , Peroxidase/metabolismo , Fenóis/farmacologia , Fenótipo , Fenoxiacetatos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Sódio/química , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA