Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
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.
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
3.
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
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.
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
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.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Eur J Pharmacol ; 643(1): 84-92, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20599914

RESUMO

C-reactive protein (CRP) has emerged as a new marker for cardiovascular diseases. Activation of peroxisome proliferator-activated receptor delta (PPARdelta) plays beneficial roles in cardiac disorders. However, the relationship between CRP and PPARdelta in cardiac cells remains unclear. This study focused on the underlying molecular mechanisms of CRP and PPARdeltaagonists. Cardiomyocytes and cardiomyoblast cell line (H9c2) were used in different groups: Untreated; 15 microg/ml CRP with or without 1 microM PPARdelta agonists (L-165041). CRP increased PPARdelta and interleukin-6 expression in cardiomyocytes and H9c2 cardiomyoblasts. NF-kappaB inducing kinase (NIK) and NF-kappaB pathway also activated by CRP stimulation. These changes could be inhibited by L-165041 through p38MAPK and c-JNK pathways. However, transfection with siRNA of CD32 CRP receptor did not decrease CRP signaling or reverse the effects of L-165041 in CRP-treated cardiomyocytes and H9c2. Pretreatment with L-165041 attenuated apoptosis induced by hypoxia with or without CRP in H9c2 cardiomyoblasts. CRP up-regulated PPARdelta expression in cardiomyocytes and H9c2. L-165041 attenuated CRP-induced pro-inflammatory signaling through p38MAPK and c-JNK in H9c2 cardiomyoblasts. However, PPARdelta activation attenuated CRP-induced NF-kappaB pathway may be independent of CD32. These results may provide new evidence of PPARdelta beneficial effects for inflammatory cardiomyopathy.


Assuntos
Proteína C-Reativa/fisiologia , Mioblastos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , PPAR delta/agonistas , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Proteína C-Reativa/farmacologia , Cardiomiopatias/imunologia , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Técnicas de Cultura de Células , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Genes Reporter , Interleucina-6/biossíntese , Interleucina-6/imunologia , Luciferases/genética , Mioblastos Cardíacos/imunologia , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/patologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , NF-kappa B/genética , PPAR delta/biossíntese , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Regulação para Cima
15.
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
16.
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
17.
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
18.
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
19.
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
20.
J Theor Biol ; 250(1): 92-102, 2008 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-17980390

RESUMO

Peroxisome proliferator activated receptor alpha (PPAR alpha) regulates fatty acid beta-oxidation (FAO) and plays a central role in the metabolic and energetic homeostasis of striated muscles. The thermodynamic consequences of the absence of PPAR alpha were investigated in diaphragm muscle of PPAR alpha knockout mice (KO). Statistical mechanics provides a powerful tool for determining entropy production, which quantifies irreversible chemical processes generated by myosin molecular motors and which is the product of thermodynamic force A/T (chemical affinity A and temperature T) and thermodynamic flow (myosin crossbridge (CB) cycle velocity upsilon). The behavior of both wild type (WT) and KO diaphragm was shown to be near-equilibrium and in a stationary state, but KO was farther from equilibrium than WT. In KO diaphragm, a substantial decrease in contractile function was associated with an increase in both A/T and upsilon and with profound histological injuries such as contraction band necrosis. There were no changes in PPAR delta and gamma expression levels or myosin heavy chain (MHC) patterns. In KO diaphragm, a marked increase in entropy production (A/T x upsilon) accounted for major thermodynamic dysfunction and a dramatic increase in irreversible chemical processes during the myosin CB cycle.


Assuntos
Diafragma/fisiopatologia , PPAR alfa/deficiência , Animais , Peso Corporal , Diafragma/metabolismo , Diafragma/patologia , Entropia , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cadeias Pesadas de Miosina/análise , Miosinas/metabolismo , PPAR alfa/biossíntese , PPAR alfa/genética , PPAR delta/biossíntese , PPAR delta/genética , PPAR gama/biossíntese , PPAR gama/genética , Isoformas de Proteínas/análise , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA