Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Clin Sci (Lond) ; 120(10): 451-62, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21143191

RESUMO

rIPC [remote IPC (ischaemic preconditioning)] has been shown to invoke potent myocardial protection in animal studies and recent clinical trials. Although the important role of PI3K (phosphoinositide 3-kinase)/Akt activation in the cardioprotection afforded by local IPC is well described, our understanding of the intracellular signalling of rIPC remains incomplete. We therefore examined the hypothesis that the myocardial protection afforded by rIPC is mediated via the PI3K/Akt/GSK3ß (glycogen synthase kinase 3ß) signalling pathway, activation of which is associated with nuclear accumulation of ß-catenin. rIPC was induced in mice using four cycles of 5 min of ischaemia and 5 min of reperfusion of the hindlimb using a torniquet. This led to reduced infarct size (19 ± 4% in rIPC compared with 39 ± 7% in sham; P<0.05), improved functional recovery and reduced apoptosis after global I/R (ischaemia/reperfusion) injury using a Langendorff-perfused mouse heart model. These effects were reversed by pre-treatment with an inhibitor of PI3K activity. Furthermore, Western blot analysis demonstrated that, compared with control, rIPC was associated with activation of the PI3K/Akt signalling pathway, resulting in phosphorylation and inactivation of GSK3ß, accumulation of ß-catenin in the cytosol and its translocation to the nucleus. Finally, rIPC increased the expression of ß-catenin target genes involved in cell-survival signalling, including E-cadherin and PPARδ (peroxisome-proliferator-activated receptor δ). In conclusion, we show for the first time that the myocardial protection afforded by rIPC is mediated via the PI3K/Akt/GSK3ß signalling pathway, activation of which is associated with nuclear accumulation of ß-catenin and the up-regulation of its downstream targets E-cadherin and PPARδ involved in cell survival.


Assuntos
Precondicionamento Isquêmico Miocárdico/métodos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , beta Catenina/metabolismo , Animais , Apoptose , Caderinas/biossíntese , Caderinas/genética , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , PPAR delta/biossíntese , PPAR delta/genética , RNA Mensageiro/genética , Transdução de Sinais/fisiologia , Regulação para Cima
10.
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
11.
Am J Physiol Endocrinol Metab ; 298(6): E1140-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20215574

RESUMO

Accumulating evidence suggests that alcohol, hepatitis C virus infection, steatosis with obesity, and insulin resistance are accompanied by iron overload states. Phlebotomy and oral iron chelators are effective treatments for these conditions and for hemochromatosis. However, the mechanisms by which iron depletion improves clinical factors remain unclear. We examined the effect of iron depletion in a model of type 2 diabetes, Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Age-matched Long-Evans Tokushima Otsuka (LETO) rats were used as controls for all experiments. Iron restriction was performed by eliminating iron in the diet from 15 wk of age or by phlebotomy. Phlebotomy was commenced at 29 wk of age by removing 4 and 3 ml of blood from the tail vein every week in OLETF and LETO rats, respectively. Rats were euthanized at 43 wk of age, and detailed analyses were performed. The plasma ferritin concentration was markedly higher in OLETF rats and decreased in iron-deficient (ID) diet and phlebotomy rats. Hemoglobin A(1c) (Hb A(1c)) was decreased significantly in OLETF rats fed the ID diet and in the phlebotomy group. Increased levels of triglycerides, glucose, free fatty acids, and total cholesterol were found in ID OLETF rats. Plasma, liver, and pancreas lipid peroxidation and hepatic superoxide production decreased in both groups. Pancreatic fibrosis and insulin levels improved in both groups of OLETF rats. Pancreatic levels of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) ligands and hypoxia-inducible factor (HIF)-1alpha were decreased significantly in OLETF rats. These factors were normalized in both rats fed ID and phlebotomy groups of OLETF rats. In conclusion, iron depletion improved diabetic complications by inhibition of oxidative stress and TGFbeta signal pathways and the maintenance of pancreatic PPARbeta/delta and HIF-1alpha pathways.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Deficiências de Ferro , PPAR delta/metabolismo , Animais , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/genética , Ferritinas/sangue , Expressão Gênica , Hemoglobinas Glicadas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Insulina/sangue , Insulina/metabolismo , Ferro/administração & dosagem , Ferro/metabolismo , Fígado/metabolismo , Masculino , Malondialdeído/sangue , Estresse Oxidativo/fisiologia , PPAR delta/biossíntese , PPAR delta/genética , Pâncreas/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos OLETF , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxido Dismutase/sangue , Fator de Crescimento Transformador beta/metabolismo
12.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 27(2): 400-6, 2010 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-20481327

RESUMO

Peroxisome proliferator-activated receptordelta (PPARdelta), as a downstream target of adenomatous polyposis coli (APC) signaling pathway, has been presumed to play some roles in colorectal carcinogenesis. However, the exact role of PPARdelta in colorectal cancer remains unclear. An HIV-1-based lentivirus packaging system was used for the construction of a lentiviral vector (lentivector) mediating RNA interference against PPARB. The direct sequencing demonstrated that the resulting lentivector containing the short-hairpin RNA expression cassette specifically targeting PPARdelta (sh-PPARdelta) was successfully constructed, and designated as pLVshPPARdelta. The control vector was designated as pLVControl. After the transduction, we observed highly efficient transduction (> 90%) of lentivirus in KM12C cells by fluorescent microscopy and fluorescence-activated cell sorting. Quantitative RT-PCR showed that pLVshPPARdelta lentivirus reduced PPARdelta mRNA expression by about 70.0% in KM12C cells as compared with that of the untreated cells (P < 0.05), while pLVControl had no significant effect on the PPARdelta mRNA level (P > 0.05). Western blot revealed an obvious reduction of PPARdelta protein expression in pLVshPPARdelta treated cells and showed no obvious difference between the control group and the untreated group. The results demonstrated that the lentivector mediating RNAi against PPARdelta was successfully constructed, which could stably knock down the PPARdelta expression in KM12C cells. This study finally provided a new cell model for the study of PPARdelta's function in colorectal cancer.


Assuntos
Neoplasias do Colo/genética , Vetores Genéticos/genética , Lentivirus/genética , PPAR delta/genética , RNA Interferente Pequeno/genética , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Técnicas de Silenciamento de Genes , Humanos , Lentivirus/metabolismo , PPAR delta/biossíntese , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
13.
Carcinogenesis ; 30(2): 230-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19056929

RESUMO

A number of reports indicate that peroxisome proliferator-activated receptor (PPAR) delta is involved in the molecular control of monocyte-macrophage differentiation. In this regard, the recent demonstration that PPARdelta is a primary response gene of 1alpha,25-dihydroxyvitamin D3 (VD), i.e. a powerful inducer of such process, allowed us to hypothesize the existence of a cross talk between PPARdelta and VD receptor pathways. To address this issue, we analyzed the effects promoted by stimulation with PPARdelta ligands and by overexpression of this nuclear receptor in monoblastic cell lines undergoing exposure to VD. The results obtained evidenced that, although promoting a weak differentiation effect by themselves, PPARdelta ligands efficiently co-operated with VD treatment. In spite of this, PPARdelta overexpression exerted a remarkable inhibitory effect on monocyte-macrophage differentiation induced by VD that was, at least partly, reverted by stimulation with a highly specific PPARdelta ligand. These data indicate that, although acting through a ligand-dependent modality, PPARdelta is a negative regulator of VD-mediated monocyte differentiation, allowing us to hypothesize a role of the investigated nuclear receptor in the differentiation block of M5 type (monoblastic) acute myeloid leukemias (AMLs). Bioinformatic analysis of a microarray database, containing the expression profiles of 285 AML cases, further supported this hypothesis demonstrating the existence of a subset of M5 type (monoblastic) AMLs that overexpress PPARdelta gene.


Assuntos
Diferenciação Celular/fisiologia , Colecalciferol/farmacologia , Monócitos/citologia , PPAR delta/fisiologia , Antígenos CD34/metabolismo , Antígenos de Diferenciação/metabolismo , Ácido Araquidônico/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Biologia Computacional , Bases de Dados Factuais , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Perfilação da Expressão Gênica , Hematopoese , Humanos , Leucemia Mieloide Aguda/metabolismo , Ligantes , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , PPAR delta/biossíntese , Tiazóis/farmacologia , Regulação para Cima
14.
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
15.
Cancer Res ; 67(11): 5285-92, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545608

RESUMO

The underlying causes of epithelial ovarian cancer (EOC) are unclear, and treatment options for patients with advanced disease are limited. There is evidence that the use of nonsteroidal anti-inflammatory drugs is associated with decreased risk of developing EOC. Nonsteroidal anti-inflammatory drugs inhibit cyclooxygenase (COX)-1 and COX-2, which catalyze prostaglandin biosynthesis. We previously showed that mouse and human EOCs have increased levels of COX-1, but not COX-2, and a COX-1-selective inhibitor, SC-560, attenuates prostaglandin production and tumor growth. However, the downstream targets of COX-1 signaling in EOC are not yet known. To address this question, we evaluated peroxisome proliferator-activated receptor delta (PPARdelta) expression and function in EOC. We found that EOC cells express high levels of PPARdelta, and neutralizing PPARdelta function reduces tumor growth in vivo. More interestingly, aspirin, a nonsteroidal anti-inflammatory drug that preferentially inhibits COX-1, compromises PPARdelta function and cell growth by inhibiting extracellular signal-regulated kinases 1/2, members of the mitogen-activated protein kinase family. Our study, for the first time, shows that whereas PPARdelta can be a target of COX-1, extracellular signal-regulated kinase is a potential target of PPARdelta. The ability of aspirin to inhibit EOC growth in vivo is an exciting finding because of its low cost, lack of cardiovascular side effects, and availability.


Assuntos
Ciclo-Oxigenase 1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias Ovarianas/metabolismo , PPAR delta/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Inibidores de Ciclo-Oxigenase/farmacologia , Ativação Enzimática , Células Epiteliais/patologia , Epoprostenol/metabolismo , Feminino , Humanos , Masculino , Camundongos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , PPAR delta/antagonistas & inibidores , PPAR delta/biossíntese , Pirazóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
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
17.
Arterioscler Thromb Vasc Biol ; 27(5): 1115-22, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17322100

RESUMO

OBJECTIVE: Reduced plasma concentrations of high-density lipoprotein-cholesterol (HDL-C) are a significant risk factor for cardiovascular disease. Mechanisms that regulate HDL-C concentrations represent an important area of investigation. METHODS AND RESULTS: Comparative transcriptome analyses of monocyte-derived macrophages (MDM) from a large population of low HDL-C subjects and age- and sex-matched controls revealed a cluster of inflammatory genes highly expressed in low HDL-C subjects. The expression levels of peroxisome proliferator activated receptor (PPAR) gamma and several antioxidant metallothionein genes were decreased in MDM from all low HDL-C groups compared with controls, as was the expression of other genes regulated by PPARgamma, including CD36, adipocyte fatty acid binding protein (FABP4), and adipophilin (ADFP). In contrast, PPARdelta expression was increased in MDM from low HDL-C groups. Quantitative RT-PCR corroborated all major findings from the microarray analysis in two separate patient cohorts. Expression of several inflammatory cytokine genes including interleukin 1beta, interleukin 8, and tumor necrosis factor alpha were highly increased in low HDL-C subjects. CONCLUSIONS: The activated proinflammatory state of monocytes and MDM in low HDL-C subjects constitutes a novel parameter of risk associated with HDL deficiency, related to altered expression of metallothionein genes and the reciprocal regulation of PPARgamma and PPARdelta.


Assuntos
HDL-Colesterol/deficiência , Expressão Gênica , Hipolipoproteinemias/sangue , Macrófagos/metabolismo , PPAR delta/genética , PPAR gama/genética , RNA/genética , Aterosclerose/sangue , Aterosclerose/etiologia , Biomarcadores/sangue , HDL-Colesterol/sangue , Proteínas de Ligação a Ácido Graxo/biossíntese , Proteínas de Ligação a Ácido Graxo/genética , Genótipo , Humanos , Hipolipoproteinemias/complicações , Hipolipoproteinemias/genética , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Interleucina-8/biossíntese , Interleucina-8/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Análise em Microsséries , Mutação , PPAR delta/biossíntese , PPAR gama/biossíntese , Perilipina-2 , Fenótipo , Reação em Cadeia da Polimerase , Fatores de Risco , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
18.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 28(3): 229-33, 2008 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-18476423

RESUMO

OBJECTIVE: To observe the effect of Bushen Antai Recipe (BAR) on expression of prostaglandin I2 (PGI2) and its nuclear receptor peroxisome proliferators-activated receptor delta (PPARdelta) at implantation site in mice with blastocyst implantation dysfunction. METHODS: Pregnant mice were divided into three groups randomly, the normal group, the model group and the BAR group. The pregnant uterus of all mice was cut off on the 4th (D4), 5th (D5), 6th (D6) and 8th (D8) day of pregnancy for determining the PGI2 expression with radio immunoassay; and the mRNA and protein expression of PPARdelta with RT-PCR and immunohistochemistry at implantation site. RESULTS: PGI2 expression in the model group was obviously lower than that in the normal group (P < 0.01), and also lower than that in the BAR group (P < 0.01), while the index was insignificantly different between the normal and the BAR group. Compared with the normal group, the expression of PPARdelta in the model group was delayed temporally and spatially (P < 0.05), while that in the BAR group was not significantly different. CONCLUSION: BAR can improve the implantation in mice with blastocyst implantation dysfunction through promoting the PGI2 expression and its nuclear receptor PPARdelta at implantation site.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Implantação Tardia do Embrião/efeitos dos fármacos , Epoprostenol/biossíntese , PPAR delta/biossíntese , Animais , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Feminino , Imuno-Histoquímica , Masculino , Camundongos , PPAR delta/genética , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Radioimunoensaio , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA