Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 79(5): 954-969, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30679176

RESUMO

APC mutations activate aberrant ß-catenin signaling to drive initiation of colorectal cancer; however, colorectal cancer progression requires additional molecular mechanisms. PPAR-delta (PPARD), a downstream target of ß-catenin, is upregulated in colorectal cancer. However, promotion of intestinal tumorigenesis following deletion of PPARD in Apcmin mice has raised questions about the effects of PPARD on aberrant ß-catenin activation and colorectal cancer. In this study, we used mouse models of PPARD overexpression or deletion combined with APC mutation (ApcΔ580 ) in intestinal epithelial cells (IEC) to elucidate the contributions of PPARD in colorectal cancer. Overexpression or deletion of PPARD in IEC augmented or suppressed ß-catenin activation via up- or downregulation of BMP7/TAK1 signaling and strongly promoted or suppressed colorectal cancer, respectively. Depletion of PPARD in human colorectal cancer organoid cells inhibited BMP7/ß-catenin signaling and suppressed organoid self-renewal. Treatment with PPARD agonist GW501516 enhanced colorectal cancer tumorigenesis in ApcΔ580 mice, whereas treatment with PPARD antagonist GSK3787 suppressed tumorigenesis. PPARD expression was significantly higher in human colorectal cancer-invasive fronts versus their paired tumor centers and adenomas. Reverse-phase protein microarray and validation studies identified PPARD-mediated upregulation of other proinvasive pathways: connexin 43, PDGFRß, AKT1, EIF4G1, and CDK1. Our data demonstrate that PPARD strongly potentiates multiple tumorigenic pathways to promote colorectal cancer progression and invasiveness. SIGNIFICANCE: These findings address long-standing, important, and unresolved questions related to the potential role of PPARD in APC mutation-dependent colorectal tumorigenesis by showing PPARD activation enhances APC mutation-dependent tumorigenesis.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , PPAR delta/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Benzamidas/farmacologia , Carcinogênese , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Progressão da Doença , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica , PPAR delta/biossíntese , PPAR delta/genética , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/genética , Sulfonas/farmacologia , Tiazóis/farmacologia
2.
Neuropharmacology ; 140: 150-161, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30086290

RESUMO

Activation of peroxisome proliferator-activated receptor beta/delta (PPAR-ß/δ), a nuclear receptor acting as a transcription factor, was shown to be protective in various models of neurological diseases. However, there is no information about the role of PPAR-ß/δ as well as its molecular mechanisms in neonatal hypoxia-ischemia (HI). In the present study, we hypothesized that PPAR-ß/δ agonist GW0742 can activate miR-17-5p, consequently inhibiting TXNIP and ASK1/p38 pathway leading to attenuation of apoptosis. Ten-day-old rat pups were subjected to right common carotid artery ligation followed by 2.5 h hypoxia. GW0742 was administered intranasally 1 and 24 h post HI. PPAR-ß/δ receptor antagonist GSK3787 was administered intranasally 1 h before and 24 h after HI, antimir-17-5p and TXNIP CRISPR activation plasmid were administered intracerebroventricularly 24 and 48 h before HI, respectively. Brain infarct area measurement, neurological function tests, western blot, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), Fluoro-Jade C and immunofluorescence staining were conducted. GW0742 reduced brain infarct area, brain atrophy, apoptosis, and improved neurological function at 72 h and 4 weeks post HI. Furthermore, GW0742 treatment increased PPAR-ß/δ nuclear expression and miR-17-5p level and reduced TXNIP in ipsilateral hemisphere after HI, resulting in inhibition of ASK1/p38 pathway and attenuation of apoptosis. Inhibition of PPAR-ß/δ receptor and miR-17-5p and activation of TXNIP reversed the protective effects. For the first time, we provide evidence that intranasal administration of PPAR-ß/δ agonist GW0742 attenuated neuronal apoptosis at least in part via PPAR-ß/δ/miR-17/TXNIP pathway. GW0742 could represent a therapeutic target for treatment of neonatal hypoxic ischemic encephalopathy (HIE).


Assuntos
Apoptose/fisiologia , Proteínas de Transporte/fisiologia , Hipóxia-Isquemia Encefálica/fisiopatologia , MicroRNAs/fisiologia , PPAR delta/fisiologia , PPAR beta/fisiologia , Animais , Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/patologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , MAP Quinase Quinase Quinase 5/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Neurônios/patologia , PPAR delta/agonistas , PPAR delta/antagonistas & inibidores , PPAR delta/biossíntese , PPAR beta/agonistas , PPAR beta/antagonistas & inibidores , PPAR beta/biossíntese , Ratos , Transdução de Sinais/fisiologia , Sulfonas/farmacologia , Tiazóis/farmacologia , Tiazóis/uso terapêutico
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.
J Lipid Res ; 58(2): 317-324, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28007964

RESUMO

α-Chlorofatty aldehydes are generated from myeloperoxidase-derived HOCl targeting plasmalogens, and are subsequently oxidized to α-chlorofatty acids (α-ClFAs). The catabolic pathway for α-ClFA is initiated by ω-oxidation. Here, we examine PPAR-α activation as a mechanism to increase α-ClFA catabolism. Pretreating both HepG2 cells and primary mouse hepatocytes with the PPAR-α agonist, pirinixic acid (Wy 14643), increased the production of α-chlorodicarboxylic acids (α-ClDCAs) in cells treated with exogenous α-ClFA. Additionally, α-ClDCA production in Wy 14643-pretreated wild-type mouse hepatocytes was accompanied by a reduction in cellular free α-ClFA. The dependence of PPAR-α-accelerated α-ClFA catabolism was further demonstrated by both impaired metabolism in mouse PPAR-α-/- hepatocytes and decreased clearance of plasma α-ClFA in PPAR-α-/- mice. Furthermore, Wy 14643 treatments decreased plasma 2-chlorohexadecanoic acid levels in wild-type mice. Additional studies showed that α-ClFA increases PPAR-α, PPAR-δ, and PPAR-γ activities, as well as mRNA expression of the PPAR-α target genes, CD36, CPT1a, Cyp4a10, and CIDEC. Collectively, these results indicate that PPAR-α accelerates important pathways for the clearance of α-ClFA, and α-ClFA may, in part, accelerate its catabolism by serving as a ligand for PPAR-α.


Assuntos
Hepatócitos/metabolismo , PPAR alfa/genética , Ácidos Palmíticos/metabolismo , Animais , Ácidos Graxos/metabolismo , Hepatócitos/efeitos dos fármacos , Humanos , Metabolismo/genética , Camundongos , Camundongos Knockout , Oxirredução , PPAR alfa/metabolismo , PPAR delta/biossíntese , RNA Mensageiro/biossíntese
5.
Neurotox Res ; 31(2): 259-268, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27815817

RESUMO

Impaired hippocampal neurogenesis and neuroinflammation are involved in the pathogenesis of radiation-induced brain injury. Kukoamine A (KuA) was demonstrated to have neuroprotective effects through inhibiting oxidative stress and apoptosis after whole-brain irradiation (WBI) in rats. The aim of this study was to investigate whether administration of KuA would prevent radiation-induced neuroinflammation and the detrimental effect on hippocampal neurogenesis. For this study, male Wistar rats received either sham irradiation or WBI (30 Gy single dose of X-rays) followed by the immediate injection of either KuA or vehicle intravenously. The dose of KuA was 5, 10, and 20 mg/kg, respectively. The levels of pro-inflammatory cytokines were assayed by ELISA kits. The newborn neurons were detected by 5-bromo-2-deoxyuridine (BrdU)/neuronal nuclei (NeuN) double immunofluorescence. Microglial activation was measured by Iba-1 immunofluorescence. The expression of Cox-2 and the activation of nuclear factor κB (NF-κB), activating protein 1(AP-1), and PPARδ were evaluated by western blot. WBI led to a significant increase in the level of TNF-α, IL-1ß, and Cox-2, and it was alleviated by KuA administration. KuA attenuated microglial activation in rat hippocampus after WBI. Neurogenesis impairment induced by WBI was ameliorated by KuA. Additionally, KuA alleviated the increased translocation of NF-κB p65 subunit and phosphorylation of c-jun induced by WBI and elevated the expression of PPARδ. These data indicate that KuA could ameliorate the neuroinflammatory response and protect neurogenesis after WBI, partially through regulating the activation of NF-κB, AP-1, and PPARδ.


Assuntos
Hipocampo/efeitos da radiação , Inflamação/prevenção & controle , NF-kappa B/metabolismo , Neurogênese/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Espermina/análogos & derivados , Fator de Transcrição AP-1/metabolismo , Animais , Encéfalo/efeitos da radiação , Ciclo-Oxigenase 2/biossíntese , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Hipocampo/metabolismo , Hipocampo/fisiologia , Masculino , Microglia/metabolismo , PPAR delta/biossíntese , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-jun/metabolismo , Lesões Experimentais por Radiação/prevenção & controle , Ratos , Espermina/farmacologia
6.
Hum Mol Genet ; 25(11): 2269-2282, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27008868

RESUMO

Huntington's disease (HD) is a devastating illness and at present there is no disease modifying therapy or cure for it; and management of the disease is limited to a few treatment options for amelioration of symptoms. Recently, we showed that the administration of bezafibrate, a pan-PPAR agonist, increases the expression of PGC-1α and mitochondrial biogenesis, and improves phenotype and survival in R6/2 transgenic mouse model of HD. Since the R6/2 mice represent a 'truncated' huntingtin (Htt) mouse model of HD, we tested the efficacy of bezafibrate in a 'full-length' Htt mouse model, the BACHD mice. Bezafibrate treatment restored the impaired PPARγ, PPARδ, PGC-1α signaling pathway, enhanced mitochondrial biogenesis and improved antioxidant defense in the striatum of BACHD mice. Untreated BACHD mice show robust and progressive motor deficits, as well as late-onset and selective neuropathology in the striatum, which was markedly ameliorated in the BACHD mice treated with bezafibrate. Our data demonstrate the efficacy of bezafibrate in ameliorating both neuropathological features and disease phenotype in BACHD mice, and taken together with our previous studies with the R6/2 mice, highlight the strong therapeutic potential of bezafibrate for treatment of HD.


Assuntos
Proteína Huntingtina/genética , Doença de Huntington/tratamento farmacológico , PPAR delta/biossíntese , PPAR gama/biossíntese , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/biossíntese , Animais , Bezafibrato/administração & dosagem , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Modelos Animais de Doenças , Humanos , Doença de Huntington/genética , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Biogênese de Organelas , PPAR delta/genética , PPAR gama/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/antagonistas & inibidores , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Transdução de Sinais/efeitos dos fármacos
7.
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
8.
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
9.
J Vasc Res ; 52(1): 12-21, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25896311

RESUMO

The peroxisome proliferator-activated receptor (PPAR) is downregulated in the cortex of experimental subarachnoid hemorrhage (SAH) animals. This study is to examine the effect of glycyrrhizin on the alternation of PPARs and proinflammatory cytokines in a rodent SAH model. CSF cytokines were evaluated by RT-PCR. Basilar arteries (BAs) were harvested to examine PPARs (RT-PCR and Western blot), and a morphological examination was conducted. Deformed endothelium and tortuous elastic lamina were observed in the BAs of the SAH groups, but they were absent in the glycyrrhizin groups or the healthy controls. The PPAR-γ and -δ protein levels were reduced in the SAH groups (p < 0.01). Glycyrrhizin significantly increased the expressed PPAR-γ protein and mRNA (preconditioning) and PPAR-δ mRNA (both treatment and preconditioning), which corresponded to the reduced IL-1ß and TNF-α levels. The administration of a PPAR-γ inhibitor, BADGE, halted the reduction of IL-1ß and TNF-α in the glycyrrhizin groups. Conclusively, glycyrrhizin exerts anti-inflammatory effects on SAH-induced vasospasm and attenuates the expression of PPARs, especially PPAR-γ, which corresponds to the severity of SAH-related inflammation. These findings also offer credit to the antivasospastic effect of glycyrrhizin and its vasculoprotective effect in animals subjected to SAH.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Medicamentos de Ervas Chinesas/uso terapêutico , Ácido Glicirrízico/uso terapêutico , PPAR gama/fisiologia , Fitoterapia , Hemorragia Subaracnóidea/tratamento farmacológico , Vasoespasmo Intracraniano/prevenção & controle , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Artéria Basilar/metabolismo , Citocinas/biossíntese , Citocinas/líquido cefalorraquidiano , Citocinas/genética , Medicamentos de Ervas Chinesas/farmacologia , Endotélio Vascular/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glicirrízico/farmacologia , Inflamação , Bombas de Infusão , Masculino , PPAR delta/biossíntese , PPAR delta/genética , PPAR gama/antagonistas & inibidores , PPAR gama/biossíntese , PPAR gama/genética , Pré-Medicação , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Método Simples-Cego , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/genética , Vasoespasmo Intracraniano/etiologia , Vasoespasmo Intracraniano/fisiopatologia
10.
J Neurochem ; 130(4): 563-74, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24806616

RESUMO

Peroxisome proliferator-activated receptor ß/δ (PPARß/δ) is a potential regulator of neuroinflammation. Toll-like receptors (TLR) are innate immunity-related receptors of inflammatory stimuli. In the present report, we evaluate the molecular mechanisms of regulation of mRNA, protein, and transcriptional activity levels of PPARß/δ by agonists of TLR4, TLR1/2, and TLR5, using lipopolysaccharide (LPS), peptidoglycan, and flagellin, respectively. We found that these stimuli increase the PPARß/δ levels in astrocytes. Expression and activity of PPARß/δ are separately regulated by inhibitors of p38, MEK1/2, extracellular signal-regulated kinases 1/2, and c-Jun N-terminal Kinase mitogen-activated protein kinases. The LPS-induced kinetics of PPARß/δ expression is similar to that of the proinflammatory gene cyclooxygenase 2. Moreover, for both genes the expression depends on nuclear factor kappa-light-chain-enhancer of activated B cells and p38, and is induced after inhibition of protein synthesis. The up-regulation of the expression after inhibition of protein synthesis signifies the participation of a labile protein in regulation of PPARß/δ expression. In contrast to cyclooxygenase 2, the cycloheximide-sensitive PPARß/δ expression was not responsive to nuclear factor kappa-light-chain-enhancer of activated B cells inhibition. Measurements of PPARß/δ mRNA stability showed that the PPARß/δ mRNA levels are regulated post-transcriptionally. We found that in LPS-stimulated astrocytes, the half-life of PPARß/δ mRNA was 50 min. Thus, we demonstrate that PPARß/δ expression and activity are regulated in TLR agonist-stimulated astrocytes by mechanisms that are widely used for regulation of proinflammatory genes. Protein expression level of nuclear receptor PPARß/δ is important for functions of this transcription factor. We investigate the regulatory mechanisms of PPARß/δ in rat primary astrocytes stimulated by agonists of toll-like receptors (TLR): TLR4, TLR1/2, and TLR5. Expression, activity, mRNA stability, and superinduction of PPARß/δ were up-regulated after TLR stimulation. These processes are sensitive to MAPKs and NF-kB inhibitors. Superinduction is up-regulation of mRNA expression after inhibition of protein synthesis.


Assuntos
Astrócitos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , PPAR delta/biossíntese , Inibidores de Proteínas Quinases/farmacologia , Receptores Toll-Like/agonistas , Animais , Astrócitos/efeitos dos fármacos , Western Blotting , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Indução Enzimática/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Lipopolissacarídeos/farmacologia , PPAR delta/efeitos dos fármacos , PPAR delta/genética , Cultura Primária de Células , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA/biossíntese , RNA/genética , RNA/isolamento & purificação , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
11.
Exp Eye Res ; 118: 100-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24309288

RESUMO

Selective killing of RPE cells in vivo by sodium iodate develops cardinal phenotypes of atrophic age-related macular degeneration. However, the molecular mechanisms are elusive. We tried to search for small cyto-protective molecules against sodium iodate and explore their mechanisms of action. Sodium iodate-mediated RPE cell death was associated with increased levels of reactive oxygen species (ROS) and IL-8. Resveratrol, a natural occurring polyphenol compound, was found to strongly protect RPE cells from sodium iodate with inhibition of production of ROS and IL-8. Resveratrol activated all isoforms of PPARs. Treatment with PPARα and PPARδ agonists inhibited sodium iodate-induced ROS production and protected RPE cells from sodium iodate. A PPARα antagonist significantly reduced resveratrol's protection of RPE cells from sodium iodate. Paradoxically, knocking down PPARδ also rendered RPE cells resistant to sodium iodate. Moreover, PPAR agonists reversed sodium iodate-induced production of IL-8. However, neutralizing extracellular IL-8 failed to protect RPE cells from sodium iodate. Taken together, these observations show that resveratrol protects RPE cells from sodium iodate injury through the activation of PPARα and alteration of PPARδ conformation. PPARα and δ modulators might ameliorate stress-induced RPE degeneration in vivo.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Iodatos/toxicidade , Degeneração Macular/psicologia , PPAR alfa/genética , PPAR delta/genética , RNA/genética , Estilbenos/farmacologia , Inibidores da Angiogênese , Antioxidantes/farmacologia , Células Cultivadas , Citoproteção , Humanos , Degeneração Macular/induzido quimicamente , Degeneração Macular/genética , PPAR alfa/biossíntese , PPAR delta/biossíntese , Resveratrol
12.
J Physiol Pharmacol ; 64(3): 289-97, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23959725

RESUMO

Liver X receptors (LXR) have been characterized as key transcriptional regulators of hepatic lipid and carbohydrate metabolism. LXR are expressed also in skeletal muscle, however, their role in this tissue is poorly investigated and the vast majority of available data comes from studies on cultured myotubes. Therefore, we aimed to examine effects of in vivo LXR activation on muscle lipid metabolism. The experiments were performed on male Wistar rats fed on a standard rodent chow. The animals were divided into two groups (n=10) receiving either LXR activator (T0901317, 10 mg/kg/day) or vehicle for one week. Samples of the soleus as well as red and white sections of the gastrocnemius muscle were excised. T0901317 increased muscle expression of peroxisome proliferator-activated receptor-δ and its target genes involved in fatty acid uptake and oxidation. In addition, LXR agonist enhanced palmitate oxidation (by 55%) in isolated soleus muscle. However, palmitate incorporation into triacylglycerol was decreased (by 38%), which was associated with reduced diacylglycerol acyltransferase expression (by 66%). Despite markedly increased plasma lipid concentration upon T0901317 treatment, muscle triacylglycerol level was elevated only in the red section of the gastrocnemius muscle. We conclude that T0901317 enhances muscle fatty acid oxidation, which prevents overt accumulation of intramuscular lipids that could be expected considering T0901317-induced hyperlipidemia.


Assuntos
Anticolesterolemiantes/efeitos adversos , Ácidos Graxos não Esterificados/metabolismo , Hidrocarbonetos Fluorados/efeitos adversos , Hipertrigliceridemia/induzido quimicamente , Músculo Esquelético/efeitos dos fármacos , Receptores Nucleares Órfãos/agonistas , PPAR delta/biossíntese , Sulfonamidas/efeitos adversos , Animais , Diacilglicerol O-Aciltransferase/antagonistas & inibidores , Diacilglicerol O-Aciltransferase/genética , Diacilglicerol O-Aciltransferase/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ácidos Graxos não Esterificados/sangue , Hipertrigliceridemia/sangue , Hipertrigliceridemia/metabolismo , Receptores X do Fígado , Masculino , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Especificidade de Órgãos , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Oxirredução/efeitos dos fármacos , PPAR delta/genética , PPAR delta/metabolismo , Ácido Palmítico/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Triglicerídeos/sangue , Triglicerídeos/metabolismo , Regulação para Cima/efeitos dos fármacos
13.
Auton Neurosci ; 177(2): 211-6, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23701913

RESUMO

BACKGROUND: Role of peroxisome proliferator-activated receptor δ (PPARδ) in cardiac contraction has recently been established. Dopamine is one of the agents used to treat heart failure in clinics. But the mediation of PPARδ in cardiac action of dopamine is still unclear. METHODS: The present study is aimed to clarify this point using neonatal rat cardiomyocytes to investigate the changes of PPARδ expression and cardiac troponin I (cTnI) phosphorylation by Western blotting analysis. Antagonists of receptors, inhibitor of phospholipase C (PLC) (U73122), calcium chelator (BAPTA-AM), and inhibitor of protein kinase A (PKAI) were also applied. We silenced PPARδ by RNAi to identify the major role of PPARδ in dopamine-induced actions. RESULTS: Dopamine increases PPARδ expression and cardiac troponin I (cTnI) phosphorylation in a time- and dose-dependent manner in neonatal rat cardiomyocytes. Moreover, both actions of dopamine were blocked by DA1 receptor antagonist and PLC inhibitor but not by PKAI. The increase of cTnI phosphorylation by dopamine was also inhibited in cardiomyocytes silenced by RNAi of PPARδ. CONCLUSION: We suggest that dopamine can enhance cardiac contraction mainly through an activation of DA1 receptor-linked PLC pathway to increase cellular calcium ions for the increase of PPARδ expression.


Assuntos
Dopamina/fisiologia , Miócitos Cardíacos/metabolismo , PPAR delta/biossíntese , Receptores de Dopamina D1/metabolismo , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Inibidores Enzimáticos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Dopamina D1/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fosfolipases Tipo C/antagonistas & inibidores
14.
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
15.
Int J Oncol ; 41(3): 839-48, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22710759

RESUMO

Transducer of ErbB-2.1 (Tob1), a tumor suppressor protein, is inactivated in a variety of cancers including stomach cancer. However, the role of Tob1 in gastric carcinogenesis remains elusive. The present study aimed to investigate whether Tob1 could inhibit gastric cancer progression in vitro, and to elucidate its underlying molecular mechanisms. We found differential expression of Tob1 in human gastric cancer (MKN28, AGS and MKN1) cells. The overexpression of Tob1 induced apoptosis in MKN28 and AGS cells, which was associated with sub-G1 arrest, activation of caspase-3, induction of Bax, inhibition of Bcl-2 and cleavage of poly (ADP-ribose) polymerase (PARP). In addition, Tob1 inhibited proliferation, migration and invasion, which were reversed in MKN1 and AGS cells transfected with Tob1 siRNA. Overexpression of Tob1 in MKN28 and AGS cells induced the expression of Smad4, leading to the increased expression and the promoter activity of p15, which was diminished by silencing of Tob1 using specific siRNA. Tob1 decreased the phosphorylation of Akt and glycogen synthase kinase-3ß (GSK3ß) in MKN28 and AGS cells, resulting in the reduced protein expression and the transcriptional activity of ß­catenin, which in turn decreased the expression of cyclin D1, cyclin-dependent kinase-4 (CDK4), urokinase plasminogen activator receptor (uPAR) and peroxisome proliferator and activator receptor-δ (PPARδ). Conversely, silencing of Tob1 induced the phosphorylation of Akt and GSK-3ß, and increased the expression of ß­catenin and its target genes. Collectively, our study demonstrates that the overexpression of Tob1 inhibits gastric cancer progression by activating Smad4- and inhibiting ß­catenin-mediated signaling pathways.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transdução de Sinais , Proteína Smad4/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteínas Supressoras de Tumor/metabolismo , beta Catenina/metabolismo , Apoptose , Caspase 3/biossíntese , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Ciclina D1/biossíntese , Quinase 4 Dependente de Ciclina/biossíntese , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Invasividade Neoplásica , PPAR delta/biossíntese , Fosforilação , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores de Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Proteínas Supressoras de Tumor/genética
16.
Clin Cancer Res ; 17(11): 3760-70, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21531809

RESUMO

PURPOSE: To investigate the expression significance of PPAR ß/δ in relation to radiotherapy (RT), clinicopathologic, and prognostic variables of rectal cancer patients. EXPERIMENTAL DESIGN: We included 141 primary rectal cancer patients who participated in a Swedish clinical trial of preoperative RT. Tissue microarray samples from the excised rectal cancers and the adjacent or distant normal mucosa and lymph node metastases were stained with PPAR δ antibody. Survival probability was computed by the Kaplan-Meier method and Cox regression model. The proliferation of colon cancer cell lines KM12C, KM12SM, and KM12L4a was assayed after PPAR δ knockdown. RESULTS: PPAR δ was increased from adjacent or distant normal mucosa to primary cancers, whereas it decreased from primary cancers to lymph node metastases. After RT, PPAR δ was increased in normal mucosa, whereas it decreased in primary cancers and lymph node metastases. In primary cancers, the high expression of PPAR δ was related to higher frequency of stage I cases, lower lymph node metastasis rate, and low expression of Ki-67 in the unirradiated cases, and related to favorable survival in the cases either with or without RT. The proliferation of the KM12C, KM12SM, or KM12L4a cells was significantly accelerated after PPAR δ knockdown. CONCLUSIONS: RT decreases the PPAR δ expression in primary rectal cancers and lymph node metastases. PPAR δ is related to the early development of rectal cancer and inhibits the proliferation of colorectal cancer cells. Increase of PPAR δ predicts favorable survival in the rectal cancer patients either with or without preoperative RT.


Assuntos
PPAR delta/biossíntese , Neoplasias Retais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Ciclo-Oxigenase 2/biossíntese , Feminino , Humanos , Proteínas Inibidoras de Apoptose/biossíntese , Antígeno Ki-67/biossíntese , Antígeno Ki-67/genética , Metástase Linfática , Masculino , Mucosa/citologia , Proteínas de Neoplasias/biossíntese , PPAR delta/genética , PPAR delta/imunologia , Prognóstico , Proteínas Tirosina Fosfatases/biossíntese , Neoplasias Retais/diagnóstico , Neoplasias Retais/radioterapia , Survivina
17.
Biochem Biophys Res Commun ; 406(4): 564-9, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21352808

RESUMO

This study evaluated peroxisome proliferator-activated receptor (PPAR) δ as a potential target for therapeutic intervention in Ang II-induced senescence in human vascular smooth muscle cells (hVSMCs). Activation of PPARδ by GW501516, a specific agonist of PPARδ, significantly inhibited the Ang II-induced premature senescence of hVSMCs. Agonist-activated PPARδ suppressed the generation of Ang II-triggered reactive oxygen species (ROS) with a concomitant reduction in DNA damage. Notably, GW501516 up-regulated the expression of antioxidant genes, such as glutathione peroxidase 1, thioredoxin 1, manganese superoxide dismutase and heme oxygenase 1. siRNA-mediated down-regulation of these antioxidant genes almost completely abolished the effects of GW501516 on ROS production and premature senescence in hVSMCs treated with Ang II. Taken together, the enhanced transcription of antioxidant genes is responsible for the PPARδ-mediated inhibition of premature senescence through sequestration of ROS in hVSMCs treated with Ang II.


Assuntos
Senescência Celular/genética , Regulação da Expressão Gênica , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , PPAR delta/fisiologia , Angiotensina II/farmacologia , Angiotensina II/fisiologia , Senescência Celular/efeitos dos fármacos , Dano ao DNA/genética , Glutationa Peroxidase/genética , Heme Oxigenase-1/genética , Humanos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , PPAR delta/agonistas , PPAR delta/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/genética , Tiazóis/farmacologia , Tiorredoxinas/genética , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima , Glutationa Peroxidase GPX1
18.
Br J Pharmacol ; 164(1): 192-208, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21426320

RESUMO

BACKGROUND AND PURPOSE: Humanized mice for the nuclear receptor peroxisome proliferator-activated receptor δ (PPARδ), termed PPARδ knock-in (PPARδ KI) mice, were generated for the investigation of functional differences between mouse and human PPARδ and as tools for early drug efficacy assessment. EXPERIMENTAL APPROACH: Human PPARδ function in lipid metabolism was assessed at baseline, after fasting or when challenged with the GW0742 compound in mice fed a chow diet or high-fat diet (HFD). KEY RESULTS: Analysis of PPARδ mRNA levels revealed a hypomorph expression of human PPARδ in liver, macrophages, small intestine and heart, but not in soleus and quadriceps muscles, white adipose tissue and skin. PPARδ KI mice displayed a small decrease of high-density lipoprotein-cholesterol whereas other lipid parameters were unaltered. Plasma metabolic parameters were similar in wild-type and PPARδ KI mice when fed chow or HFD, and following physiological (fasting) and pharmacological (GW0742 compound) activation of PPARδ. Gene expression profiling in liver, soleus muscle and macrophages showed similar gene patterns regulated by mouse and human PPARδ. The anti-inflammatory potential of human PPARδ was also similar to mouse PPARδ in liver and isolated macrophages. CONCLUSIONS AND IMPLICATIONS: These data indicate that human PPARδ can compensate for mouse PPARδ in the regulation of lipid metabolism and inflammation. Overall, this novel PPARδ KI mouse model shows full responsiveness to pharmacological challenge and represents a useful tool for the preclinical assessment of PPARδ activators with species-specific activity.


Assuntos
Inflamação/tratamento farmacológico , Inflamação/genética , PPAR delta/genética , PPAR delta/metabolismo , Animais , DNA Complementar/genética , Dieta Hiperlipídica/métodos , Jejum/metabolismo , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Inflamação/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Fígado/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , PPAR delta/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tiazóis/farmacologia
19.
Br J Pharmacol ; 163(2): 390-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21232041

RESUMO

BACKGROUND AND PURPOSE: Digoxin has been used as an inotropic agent in heart failure for a long time. Troponin I (TnI) phosphorylation is related to cardiac contractility, and the genes are regulated by peroxisome proliferator-activated receptors (PPARs). Our previous studies indicated that cardiac abnormality related to the depressed expression of PPARδ in the hearts of STZ rats is reversed by digoxin. However, the cellular mechanisms for this effect of digoxin have not been elucidated. The aim of the present study was to investigate possible mechanisms for this effect of digoxin using the H9c2 cell line cultured in high glucose (HG) conditions. METHODS: The effects of digoxin on PPARδ expression, intracellular calcium and TnI phosphorylation were investigated in cultured H9c2 cells, maintained in a HG medium, by using Western blot analysis. RESULTS: Digoxin increased PPARδ expression in H9c2 cells subjected to HG conditions, and increase the intracellular calcium concentration. This effect of digoxin was blocked by BAPTA-AM at concentrations sufficient to chelate calcium ions. In addition, the calcineurin inhibitor cyclosporine A and KN93, an inhibitor of calcium/calmodulin-dependent protein kinase, inhibited this action. Digoxin also increased TnI phosphorylation and this was inhibited when PPARδ was silenced by the addition of RNAi to the cells. Similar changes were observed on the contraction of H9c2 cells. CONCLUSION: The results suggest that digoxin appears, through calcium-triggered signals, to reverse the reduced expression of PPARδ in H9c2 cells caused by HG treatment.


Assuntos
Cardiotônicos/farmacologia , Digoxina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , PPAR delta/biossíntese , Animais , Cálcio/metabolismo , Linhagem Celular , Tamanho Celular/efeitos dos fármacos , Quelantes/farmacologia , Cães , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Glucose/metabolismo , Espaço Intracelular/metabolismo , Miócitos Cardíacos/metabolismo , PPAR delta/genética , Fosforilação , RNA Interferente Pequeno/genética , Ratos , Troponina I/metabolismo
20.
Dig Dis Sci ; 56(4): 1194-200, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20824502

RESUMO

BACKGROUND: The role of peroxisome proliferator-activated receptor delta (PPAR δ) in the development and progression of colorectal cancer (CRC) remains controversial. AIMS: We investigated the impact of PPAR δ expression in tissues on liver metastasis of CRC. METHODS: We analyzed samples of primary CRC and matched normal adjacent tissues from 52 patients for the expression of PPAR δ, cyclooxygenase (COX)-2, vascular endothelial growth factor (VEGF)-A, and CXC chemokine receptor 4 (CXCR4). Correlations of the molecules expressions with clinical characteristics and prognosis of patients were studied. RESULTS: The number of patients positive for PPAR δ, COX-2, CXCR4, and VEGF-A was 25, 33, 18, and 19, respectively. Among the PPAR δ (+)/COX-2 (+), PPAR δ (-)/COX-2 (+), PPAR δ (+)/COX-2 (-), and PPAR δ (-)/COX-2 (-) patient groups, PPAR δ (+)/COX-2 (+) patients had the highest incidence of liver metastasis (p<0.01). PPAR δ (+)/COX-2 (+) expression was a significant independent prognostic factor (HR=7.108, 95% CI 1.231-41.029, p=0.0283) by Cox proportional analysis. PPAR δ (+)/COX-2 (+) patients had the highest positivity for CXCR4 or VEGF-A in tissues (p<0.01). Among the patients in the CXCR4 (+)/VEGF-A (+), CXCR4 (+)/VEGF-A (-), CXCR4 (-)/VEGF-A (+), and CXCR4 (-)/VEGF-A (-) groups, CXCR4 (+)/VEGF-A (+) patients had the highest incidence of liver metastasis (p<0.01). CONCLUSIONS: The expression of both PPAR δ and COX-2 in tissues may lead to liver metastasis and consequent poor prognosis in CRC patients.


Assuntos
Carcinoma/secundário , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Ciclo-Oxigenase 2/biossíntese , Neoplasias Hepáticas/secundário , PPAR delta/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma/metabolismo , Feminino , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Pessoa de Meia-Idade , Prognóstico , Receptores CXCR4/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...