Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Arq. neuropsiquiatr ; 79(3): 216-221, Mar. 2021. graf
Article in English | LILACS | ID: biblio-1285354

ABSTRACT

ABSTRACT Background: Sleep disorders induce anxiety and forgetfulness and change habits. The chemical hypnotic drugs currently used have serious side effects and, therefore, people are drawn towards using natural compounds such as plant-based healing agents. Abscisic acid (ABA) is produced in a variety of mammalian tissues and it is involved in many neurophysiological functions. Objective: To investigate the possible effect of ABA on pentobarbital-induced sleep and its possible signaling through GABA-A and PPAR (γ and β) receptors, in male Wistar rats. Methods: The possible effect of ABA (5 and 10 µg/rat, intracerebroventricularly) on sleep onset latency time and duration was evaluated in a V-maze model of sleep. Pentobarbital sodium (40 mg/kg, intraperitoneally) was injected to induce sleep 30 min after administration of ABA. PPARβ (GSK0660, 80 nM/rat), PPARγ (GW9662, 3 nM/rat) or GABA-A receptor (bicuculline, 6 µg/rat) antagonists were given 15 min before ABA injection. Diazepam (2 mg/kg, intraperitoneally) was used as a positive control group. Results: ABA at 5 µg significantly boosted the pentobarbital-induced subhypnotic effects and promoted induction of sleep onset in a manner comparable to diazepam treatment. Furthermore, pretreatment with bicuculline significantly abolished the ABA effects on sleep parameters, while the amplifying effects of ABA on the induction of sleep onset was not significantly affected by PPARβ or PPARγ antagonists. The sleep prolonging effect of ABA was significantly prevented by both PPAR antagonists. Conclusions: The data showed that ABA boosts pentobarbital-induced sleep and that GABA-A, PPARβ and PPARγ receptors are, at least in part, involved in ABA signaling.


RESUMO Introdução: Os distúrbios do sono induzem a ansiedade e esquecimento e mudam hábitos. Os medicamentos hipnóticos químicos utilizados atualmente têm efeitos colaterais graves e, portanto, as pessoas são atraídas para o uso de compostos naturais, como agentes de cura à base de plantas. O ácido abscísico (ABA) é produzido em uma variedade de tecidos de mamíferos e está envolvido em muitas funções neurofisiológicas. Objetivo: Investigar o possível efeito do ABA no sono induzido por pentobarbital e sua possível sinalização por meio dos receptores GABA-A e PPAR (γ e β), em ratos Wistar machos. Métodos: O possível efeito do ABA (5 e 10 µg/rato, intracerebroventricularmente) no tempo de latência e duração do início do sono foi avaliado em um modelo de labirinto em V de sono. Pentobarbital sódico (40 mg/kg, intraperitonealmente) foi injetado para induzir o sono 30 minutos após a administração de ABA. PPARβ (GSK0660, 80 nM/rato), PPARγ (GW9662, 3 nM/rato) ou antagonistas do receptor GABA-A (bicuculina, 6 µg/rato) foram administrados 15 minutos antes da injeção de ABA. Diazepam (2 mg/kg, intraperitonealmente) foi utilizado como grupo de controle positivo. Resultados: ABA a 5 µg aumentou significativamente os efeitos sub-hipnóticos induzidos por pentobarbital e promoveu a indução do início do sono de forma comparável ao tratamento com diazepam. Além disso, o pré-tratamento com bicuculina aboliu significativamente os efeitos do ABA nos parâmetros do sono, ao passo que os efeitos amplificadores do ABA na indução do início do sono não foram significativamente afetados pelos antagonistas do PPARβ ou PPARγ. O efeito de prolongamento do sono do ABA foi significativamente prevenido por ambos os antagonistas do PPAR. Conclusões: Os dados mostraram que o ABA estimula o sono induzido por pentobarbital e que os receptores GABA-A, PPARβ e PPARγ estão, pelo menos em parte, envolvidos na sinalização ABA.


Subject(s)
Animals , Male , Rats , Sleep , Abscisic Acid/pharmacology , Receptors, GABA-A/metabolism , PPAR-beta/metabolism , PPAR gamma/metabolism , Pentobarbital/pharmacology , Plant Growth Regulators/pharmacology , Signal Transduction , Rats, Wistar
3.
Arq. bras. cardiol ; 113(6): 1121-1127, Dec. 2019. tab
Article in English | LILACS | ID: biblio-1055071

ABSTRACT

Abstract Background: Oxidative stress and inflammation are present in coronary artery disease (CAD) and are linked to the activation of the transcription nuclear factor kappa B (NF-κB). To attenuate these complications, transcription factors like nuclear factor erythroid 2-related factor 2 (Nrf2) and peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) can be activated to inhibit NF-κB. However, the available data on expression of NF-κB, Nrf2 and PPARβ/δ in CAD patients are limited. Objective: To evaluate the expression of the transcription factors NF-κB and Nrf2 and PPAR��/�� in CAD patients. Methods: Thirty-five patients (17 men, mean age 62.4 ? 7.55 years) with CAD and twelve patients (5 men, mean age 63.50 ? 11.46 years) without CAD were enrolled. Peripheral blood mononuclear cells (PBMCs) were isolated and processed for mRNA expression of Nrf2, NF-κB, NADPH: quinone oxidoreductase 1 (NQO1) and PPARβ/δ mRNAs using quantitative real-time polymerase chain reaction (qPCR). p < 0.05 was considered statistically significant. Results: There was no difference in the mRNA expressions of Nrf2 (1.35 ? 0.57), NF-κB (1.08 ? 0.50) or in the antioxidant enzyme NQO1 (1.05 ? 0.88) in the CAD group compared to the group without CAD (1.16 ? 0.76, 0.95 ? 0.33, 0.81 ? 0.55, respectively). However, PPARβ/δ was highest expressed in the CAD group (1.17 ? 0.86 vs. 0.56 ? 0.34, p = 0.008). Conclusion: The main finding of this study was the PPARβ/δ being more expressed in the PBMC of patients with CAD compared to the control group, whereas no differences were observed in Nrf2 or NF-κB mRNA expressions.


Resumo Fundamentos: O estresse oxidativo e a inflamação estão presentes na doença arterial coronariana (DAC) e estão ligados à ativação do fator de transcrição nuclear kappa B (NF-κB). Para atenuar essas complicações, fatores de transcrição como o fator nuclear eritroide 2-relacionado ao fator 2 (Nrf2) e o receptor ativado por proliferador de peroxissoma β/δ (PPARβ/δ) podem ser ativados para inibir o NF-κB. No entanto, os dados disponíveis sobre a expressão de NF-κB, Nrf2 e PPARβ/δ em pacientes com DAC são limitados. Objetivo: Avaliar a expressão dos fatores transcricionais NF-κB e Nrf2 e o PPARβ/δ em pacientes com DAC. Métodos: Trinta e cinco pacientes (17 homens, idade média de 62,4 ± 7,55 anos) com DAC e doze pacientes (5 homens, com idade média de 63,50 ± 11,46 anos) sem DAC foram incluídos. Células mononucleares do sangue periférico (PBMCs) foram isoladas e processadas para a expressão de mRNA do Nrf2, NF-κB, NADPH: quinona oxidoredutase 1 (NQO1) e mRNAs do PPARβ/δ por meio de reação em cadeia da polimerase quantitativa em tempo real (qPCR). Valores de p < 0,05 foram considerados como estatisticamente significativos. Resultados: Não houve diferença nas expressões de mRNA do Nrf2 (1,35 ± 0,57), NF-κB (1,08 ± 0,50) ou na enzima antioxidante NQO1 (1,05 ± 0,88) no grupo DAC em comparação com o grupo sem DAC (1,16 ± 0,76, 0,95 ± 0,33, 0,81 ± 0,55, respectivamente). Entretanto, o PPARβ/δ apresentou maior expressão no grupo com DAC (1,17 ± 0,86 vs. 0,56 ± 0,34, p = 0,008). Conclusão: O principal achado do presente estudo foi o PPARβ/δ apresentar maior expressão nas PBMCs de pacientes com DAC comparados ao grupo controle, ao passo que não foram observadas diferenças nas expressões de mRNA do Nrf2 ou NF-κB.


Subject(s)
Humans , Male , Female , Middle Aged , Aged , Coronary Artery Disease/metabolism , RNA, Messenger/metabolism , NF-kappa B/metabolism , PPAR-beta/metabolism , PPAR delta/metabolism , NF-E2-Related Factor 2/metabolism , Biomarkers/metabolism , Body Mass Index , Gene Expression Regulation , Polymerase Chain Reaction , Oxidative Stress , Inflammation/metabolism
4.
Chinese Medical Sciences Journal ; (4): 245-251, 2015.
Article in English | WPRIM | ID: wpr-242814

ABSTRACT

<p><b>OBJECTIVE</b>To explore the effect of atorvastatin on cardiac hypertrophy and to determine the potential mechanism involved.</p><p><b>METHODS</b>An in vitro cardiomyocyte hypertrophy from neonatal rats was induced with angiotensin II (Ang II) stimulation. Before Ang II stimulation, the cultured rat cardiac myocytes were pretreated with atorvastatin at different concentrations (0.1, 1, and 10 μmol/L). The following parameters were evaluated: the myocyte surface area, 3H-leucine incorporation into myocytes, mRNA expressions of atrial natriuretic peptide, brain natriuretic peptide, matrix metalloproteinase 9, matrix metalloproteinase 2, and interleukin-1β, mRNA and protein expressions of the δ/β peroxisome proliferator-activated receptor (PPAR) subtypes.</p><p><b>RESULTS</b>It was shown that atorvastatin could ameliorate Ang II-induced neonatal cardiomyocyte hypertrophy in the area of cardiomyocytes, 3H-leucine incorporation, and the expression of atrial natriuretic peptide and brain natriuretic peptide markedly. Meanwhile, atorvastatin also inhibited the augmented mRNA level of several cytokines in hypertrophic myocytes. Furthermore, the down-regulated expression of PPAR- δ/β at both the mRNA and protein levels in hypertrophic myocytes could be significantly reversed by atorvastatin treatment.</p><p><b>CONCLUSIONS</b>Atorvastatin could improve Ang II-induced cardiac hypertrophy and inhibit the expression of cytokines. Such effect might be partly achieved through activation of the PPAR-δ/β pathway.</p>


Subject(s)
Animals , Rats , Angiotensin II , Pharmacology , Atorvastatin , Pharmacology , Therapeutic Uses , Cardiomegaly , Metabolism , Pathology , Cells, Cultured , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Pharmacology , PPAR delta , Genetics , PPAR-beta , Genetics , Rats, Wistar
5.
Chinese Medical Journal ; (24): 2129-2137, 2014.
Article in English | WPRIM | ID: wpr-241712

ABSTRACT

<p><b>BACKGROUND</b>Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are the first steps in the development of multiple organ failure induced by sepsis. A systemic excessive inflammatory reaction is currently the accepted mechanism of the pathogenesis of sepsis. Several studies have suggested a protective role of the peroxisome proliferator activated receptor-β/δ (PPAR-β/δ) in related inflammatory diseases. But the role of PPARβ/δ in ALI remains uncertain. The aim of this study was to investigate the role and possible mechanism of PPARβ/δ in ALI induced by sepsis.</p><p><b>METHODS</b>Cecal ligation and puncture (CLP) was used as a sepsis model. Rats were randomly divided into four groups, the control group (CON, n = 6), sham-operation group (SHAM, n = 12), cecal ligation and puncture group (CLP, n = 30), GW501516 group (CLP+GW, n = 25), which underwent CLP and were subcutaneously injected with the PPAR-β/δ agonist GW501516 (0.05 mg/100 g body weight). Survival was monitored to 24 hours after operation. Blood pressure, serum creatinine, blood urea nitrogen, aspartate aminotrasferase and alanine aminotrasferase were measured after CLP. Concentrations of tumor necrosis factor α (TNF-α) and interleukin (IL)-1β in serum were detected by enzyme linked immunosorbent assay (ELISA) kits. Lung tissue samples were stained with H&E and scored according to the degree of inflammation. Bacterial colonies were counted in the peritoneal fluid. Alveolar macrophages were cultured and incubated with GW501516 (0.15 µmol/L) and PPARβ/δ adenovirus and then treated with Lipopolysaccharide (2 µg/ml) for 2 hours. The TNF-α, IL-1β and IL-6 RNA in lung and alveolar macrophages were determined by real-time PCR. Phosphorylation of signal transducer and activator of transcription 3 (STAT3) in lung and alveolar macrophages was detected by Western blotting.</p><p><b>RESULTS</b>GW501516 significantly increased the survival of septic rats, decreased histological damage of the lungs, reduced inflammatory cytokines in serum and lung tissues of septic rats and did not increase counts of peritoneal bacteria. In vitro, GW501516 and over-expression of PPARβ/δ attenuated gene expression of TNF-α, IL-1β and IL-6 in alveolar macrophages. Both in vivo and in vitro, PPARβ/δ inhibited the phosphorylation of STAT3.</p><p><b>CONCLUSION</b>PPARβ/δ plays a protective role in sepsis induced ALI via suppressing excessive inflammation.</p>


Subject(s)
Animals , Male , Rats , Acute Lung Injury , Drug Therapy , Cells, Cultured , PPAR delta , Metabolism , PPAR-beta , Metabolism , Rats, Sprague-Dawley , Sepsis , Drug Therapy , Thiazoles , Therapeutic Uses
6.
Korean Journal of Hepato-Biliary-Pancreatic Surgery ; : 89-108, 2013.
Article in English | WPRIM | ID: wpr-63501

ABSTRACT

BACKGROUNDS/AIMS: During the acute phase response, cytokines induce marked alterations in lipid metabolism including an increase in serum triglyceride levels and a decrease in hepatic fatty acid oxidation, in bile acid synthesis, and in high-density lipoprotein levels. METHODS: Peroxisome proliferator-activated receptors (PPARs: PPARalpha, beta/delta, and gamma) regulate fatty acid metabolism, glucose homeostasis, cell proliferation, differentiation and inflammation. Proinflammatory profiles including tumor necrosis factor alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6) are the important pathological factors in inflammatory responses during the pathological progression of the acute phase response. Lipopolysaccarides (LPS) induced the expression of TNF-alpha, IL-1beta, and IL-6. LPS-induced inflammation decrease the expression of peroxisome proliferator-activated receptor alpha (PPARalpha), PPARbeta/delta, PPARgamma, and coactivators PPARgamma co-activator 1 alpha (PGC-1alpha), PGC-1beta messenger RNA (mRNA) in the liver of Balb/c mouse. In addition, LPS-induced inflammation diminishes the protein level of PPARalpha, PPARbeta/delta, and PPARgamma. Proinflammatory cytokines including TNFalpha, IL-1beta, and IL-6 are the principal reducer of PPARs. However, the knockout mouse model against TNFalpha and IL-6 does not block decrease of PPARs in serum and liver. The mice were pretreated with fenofibrate at 100 mg/kg for 2 days. RESULTS: These treatment protocols increased the amount of PPARs mRNA in the liver. Fenofibrate inhibited LPS-induced TNF-alpha, IL-1beta, and IL-6 production in the serum and liver. Similar results were obtained when human hepatoma HepG2 cells exposed to LPS were co-incubated with fenofibrate. LPS-treated HepG2 cells decreased expression of IkappaB. Moreover, activation of PPARs abrogated LPS-induced degradation of IkappaB, thus suppressing LPS-induced NF-kappaB activities. CONCLUSIONS: Therefore, fenofibrate decreases the expression and secretion of TNF-alpha, IL-1beta, and IL-6 via the NF-kappaB signaling pathway, thus serving as therapeutic targets to attenuate inflammation that is involved in hepatic pathological progression.


Subject(s)
Animals , Humans , Mice , Bile , Carcinoma, Hepatocellular , Cell Proliferation , Clinical Protocols , Cytokines , Fenofibrate , Glucose , Hep G2 Cells , Homeostasis , Inflammation , Interleukin-1beta , Interleukin-6 , Lipid Metabolism , Lipoproteins , Liver , Mice, Knockout , NF-kappa B , Peroxisome Proliferator-Activated Receptors , Peroxisomes , PPAR alpha , PPAR-beta , PPAR delta , PPAR gamma , RNA, Messenger , Tumor Necrosis Factor-alpha
7.
Indian J Biochem Biophys ; 2012 Aug; 49(4): 219-227
Article in English | IMSEAR | ID: sea-140239

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone receptor family of ligand-inducible transcription factors. Our previous study has shown that in human umbilical vein endothelial cells PPARβ initiates a protective mechanism that limits the extent of damage due to H2O2-induced injury. Although fibroblasts are one of the main cell types involved in wound repair, the role of PPARβ in the fibroblast response to heat injury has not been investigated. Thus, in this study, we examined possible protective role of PPARβ in fibroblasts from heat injury. We developed a novel dermal fibroblast heat injury model to characterize the mechanisms of the heat injury healing response that involved PPARβ. The specific PPARβ ligand GW0742, a PPARβ activator and a short hairpin RNA (shRNA) plasmid against PPARβ were used to reveal the action mechanism of PPARβ in heat injury-induced fibroblast changes in morphology and increased proliferation. In response to heat injury (52˚C for 30 s), fibroblast activation of PPARß, increased 1.56-fold. Administration of GW0742 significantly induced a protective effect on heat injury-induced fibroblasts by minimizing the structural damage and increasing the cell proliferation response. Likewise, inhinition of PPARß, usingh shRNA exacerbated the damage by inhibiting the de novo synthesis of PPARß. These results indicated that heat injury enhanced PPARß expression and PPARß protected fibroblast structure and proliferation.


Subject(s)
Fibroblasts/physiology , Heat Exhaustion/therapy , Heat Stress Disorders/therapy , Heat Stroke/therapy , Humans , PPAR-beta
8.
Chinese Journal of Burns ; (6): 446-450, 2011.
Article in Chinese | WPRIM | ID: wpr-257829

ABSTRACT

<p><b>OBJECTIVE</b>To study the effect of freeze-dried mouse epidermal growth factor (mEGF) on the expression of peroxisome proliferator-activated receptor β (PPAR-β) in mice during wound healing.</p><p><b>METHODS</b>Full-thickness skin defect with area of 1.5 cm × 1.5 cm was reproduced on both sides of the back of 70 BALB/c mice (2 wounds in each mouse). The wound on the left side in each mouse was treated with 5 µg/mL mEGF solution (experiment group), and that on the right side in each mouse was treated with saline (control group). On post injury day (PID) 7, 11, and 16, 20 mice were used for determination of wound healing rate at each time point. On PID 1, 3, 7, 11, 14, and 18, specimens of wound edge were harvested for determination of protein and gene expression of PPAR-β with immunohistochemical staining and in situ hybridization, with 10 specimens at each time point (denoted as integral absorbance value). Data were processed with t test.</p><p><b>RESULTS</b>(1) Wound healing rate. The wound healing rate in experiment group on PID 7, 11, and 16 was respectively higher than that in control group (with t value respectively 3.03, 6.05, 11.9, P values all below 0.01). (2) Immunohistochemical observation. In both groups, the PPAR-β proteins highly expressed in fibroblasts of wound granulation tissues and nuclei of keratinocytes located in wound edge at early stage after injury, and they highly expressed in newly formed epidermis and their fibroblasts in the lower layer after wound epithelization. The expression of PPAR-β protein was gradually decreased after wound healing. The expression of PPAR-β protein at each time point in experiment group was respectively higher than that in control group (with t values from 2.15 to 7.37, P < 0.05 or P < 0.01). The expression of PPAR-β protein peaked on PID 3 in experiment group [(3.46 ± 1.33) × 10(3)], which was (2.35 ± 1.09) × 10(3) in control group. (3) In situ hybridization. The expression levels of PPAR-β mRNA in both groups were up-regulated after injury, which were mainly observed in fibroblasts of wound and cytoplasm of KC in wound edge, but they were down-regulated after wound epithelization. The expression of PPAR-β mRNA at each time point in experiment group was respectively higher than that in control group (with t values from 2.35 to 6.64, P < 0.05 or P < 0.01). The expression of PPAR-β mRNA in both groups peaked on PID 3 [(7.3 ± 2.6) × 10(6), (4.5 ± 3.0) × 10(6), respectively].</p><p><b>CONCLUSIONS</b>mEGF can up-regulate the expression of PPAR-β in wound tissue of mice and promote wound healing.</p>


Subject(s)
Animals , Female , Male , Mice , Epidermal Growth Factor , Pharmacology , Granulation Tissue , Metabolism , Mice, Inbred BALB C , PPAR-beta , Metabolism , Skin , Wounds and Injuries , Metabolism , Wound Healing
9.
The Korean Journal of Internal Medicine ; : 19-24, 2011.
Article in English | WPRIM | ID: wpr-75333

ABSTRACT

The nuclear hormone receptor PPARgamma is activated by several agonists, including members of the thiazolidinedione group of insulin sensitizers. Pleiotropic beneficial effects of these agonists, independent of their blood glucose-lowering effects, have recently been demonstrated in the vasculature. PPARgamma agonists have been shown to lower blood pressure in animals and humans, perhaps by suppressing the renin-angiotensin (Ang)-aldosterone system (RAAS), including the inhibition of Ang II type 1 receptor expression, Ang-II-mediated signaling pathways, and Ang-II-induced adrenal aldosterone synthesis/secretion. PPARgamma agonists also inhibit the progression of atherosclerosis in animals and humans, possibly through a pathway involving the suppression of RAAS and the thromboxane A2 system, as well as the protection of endothelial function. Moreover, PPARgamma-agonist-mediated renal protection, especially the reduction of albuminuria, has been observed in diabetic nephropathy, including animal models of the disease, and in non-diabetic renal dysfunction. The renal protective activities may reflect, at least in part, the ability of PPARgamma agonists to lower blood pressure, protect endothelial function, and cause vasodilation of the glomerular efferent arterioles. Additionally, anti-neoplastic effects of PPARgamma agonists have recently been described. Based on the multiple therapeutic actions of PPARgamma agonists, they will no doubt lead to novel approaches in the treatment of lifestyle-related and other diseases.


Subject(s)
Animals , Humans , Atherosclerosis/prevention & control , Hypertension/drug therapy , Hypoglycemic Agents/pharmacology , Kidney Diseases/etiology , PPAR gamma/agonists , PPAR-beta/agonists
10.
Rio de Janeiro; s.n; 2010. 138 p. ilus, tab.
Thesis in Portuguese | LILACS | ID: lil-601469

ABSTRACT

Este trabalho teve o objetivo de estudar o efeito de medicamentos com diferentes ações agonista PPAR (rosiglitazona, fenofibrato e bezafibrato) sobre o perfil lipídico, glicídico e alterações na massa corporal e morfologia do tecido adiposo e pancreático em modelo de diabetes e sobrepeso induzido por dieta. Camundongos C57BL/6 (2 meses de idade) foram alimentados com dieta padrão (SC, n=10) ou dieta hiperlipídica rica em sacarose (HFHS, n=40) por 6 semanas. Logo após, os animais HFHS foram subdividos em: HFHS não tratado e HFHS tratado com rosiglitazona (HFHS-Ro), fenofibrato (HFHS-Fe) ou bezafibrato (HFHS-BZ) (5 semanas). Os camundongos alimentados com dieta HFHS apresentaram maior glicemia e insulina de jejum (+33% e +138%, respectivamente), intolerância à glicose, resistência à insulina, aumento da massa corporal (MC) (+20%) e adiposidade, hipertrofia de adipócitos e redução da imunocoloração para adiponectina no tecido adiposo. No pâncreas houve aumento da massa (+28%), acúmulo de gordura (+700%), hipertrofia da ilhota (+38%) e redução da imunocoloração para GLUT-2 (-60%). A rosiglitazona diminuiu a glicemia e insulina de jejum, porém induziu o ganho de MC e hipertrofia cardíaca. O fenofibrato estabilizou a MC, enquanto o bezafibrato levou a perda de MC. Apenas o bezafibrato impediu a hipertrofia da ilhota. A imunocoloração para GLUT-2 foi aumentada por todos os medicamentos, e não houve alterações na imunocoloração para o PPARalfa. Sinais morfológicos de pancreatite foram vistos no grupo HFHS-Fe, apesar dos níveis normais de amilase e lipase séricos. A rosiglitazona exacerbou a infiltração intrapancreática de gordura (+75% vs. HFHS), e o bezafibrato aumentou a imunocoloração para o PPARbeta/delta nas ilhotas pancreáticas. Em conclusão, o bezafibrato apresentou um efeito mais amplo sobre as alterações metabólicas, morfológicas e biométricas decorrentes da dieta HFHS, sugerindo que a inibição das três isoformas do PPAR seria melhor do que a inibição...


This work aimed to evaluate the effect of peroxisome proliferator-activated receptor (PPAR) agonists (rosiglitazone, fenofibrate and bezafibrate) on lipid and glucose metabolism, body mass, and adipose and pancreatic tissue morphology in a model of diet-induced type 2 diabetes and overweight in mice. Two-month-old male C57BL/6 mice were fed a standard chow (SC, n=10) or a high-fat high-sucrose chow (HFHS, n=40) for 6 weeks, and then HFHS-fed mice were subdivided by treatment: untreated HFHS and HFHS treated with rosiglitazone (HFHS-Ro), fenofibrate (HFHS-Fe), or bezafibrate (HFHS-Bz) (5 weeks on medication). HFHS-fed mice have altered fasting glucose (+33%) and insulin (+138%), GI, IR, increased body mass (+20%) and fat pad weight, adipocyte hypertrophy, and decreased adiponectin immunostain. They also presented increased pancreatic (+28%) mass, intrapancreatic fat (+700%), islet hypertrophy (+38%), and decreased GLUT-2 immunostain (-60%). Rosiglitazone reduced fasting glucose and insulin but induced weight gain and heart hypertrophy. Fenofibrate impaired body mass gain, while bezafibrate induced weight loss. Only bezafibrate impaired islet hypertrophy. GLUT-2 immunostain was improved by all treatments, and there were no alterations in PPAR-alfa stain. There were morphological signs of pancreatitis in fenofibrate-treated mice, although there was no alteration in serum amylase and lipase. Rosiglitazone exacerbated pancreatic fat infiltration (+75% vs. HFHS group), and bezafibrate increased PPAR-beta expression in pancreatic islets. In conclusion, bezafibrate showed a wider range of action on metabolic, morphologic, and biometric alterations due to HFHS intake, suggesting that inhibiting the three PPAR isoforms is better than inhititing each isoform alone. Rosiglitazone exacerbated body mass gain, pancreatic fat infiltration and induced heart hyperthophy as well, thus, precaution has to be taken in prescribing rosiglitazone to obese patients.


Subject(s)
Animals , Mice , Adiponectin , Bezafibrate/agonists , Dietary Fats , Fenofibrate/agonists , Lipid Metabolism , PPAR alpha/metabolism , PPAR-beta/metabolism , PPAR gamma/metabolism , Sucrose , Thiazolidinediones/agonists , /chemically induced , Cardiovascular Diseases/metabolism , Models, Animal , Pancreas/metabolism
11.
Rio de Janeiro; s.n; 2009. 113 p. ilus.
Thesis in Portuguese | LILACS | ID: lil-564731

ABSTRACT

A insuficiência cardíaca (IC) é a evolução final das várias formas de doenças cardiovascular, sendo resultado de modificações estruturais, metabólicas e de contratilidade miocárdica. A fim de compreender o papel na dinâmica do metabolismo cardíaco no estado basal e na sobrecarga de pressão, utilizamos os modelos de cre-lox com deleção específica no coração para substrato do receptor de insulina (IRS) e co-ativador do PPAR (PGC-1b) e analisamos a estrutura cardíaca (histologia e estereologia), função cardíaca (ecocardiograma e técnica de Working heart), o metabolismo (isolamento de cardiomiócito e captação de glicose), ação hormonal (Western Blotting), expressão gênica (PCR-RT) de enzimas do metabolismo (lipídico, glicídico, da cadeia respiratória fatores transcricionais e hipertróficos) e a função mitocondrial. Verificamos, nos CIRS12KO, disfunção cardíaca grave, disfunção mitocondrial e prejuízo na expressão gênica das enzimas do metabolismo energético. Nos PGC-1BKO observamos disfunção mitocondrial e alteração de expressão gênica das enzimas do metabolismo energético quando submetidos à sobrecarga de pressão. Através do estudo do metabolismo cardíaco e da expressão gênica nestes diferentes modelos conseguimos explorar as vias metabólicas que levam a hipertrofia compensada à IC. Sugerimos que o mecanismo responsável pela descompensação seja a disfunção mitocondrial em conseqüência à alteração da expressão gênica. E que IRS e o PGC-1B são fatores chaves da dinâmica cardíaca, e que são indispensáveis para a estrutura e funcionamento cardíaco. Além de representar alvo promissor para limitar a transição de hipertrofia cardíaca compensada a insuficiência cardíaca...


Heart failure (HF) is the end stage of different types of cardiovascular diseases and it is characterized by changes in the metabolic and myocardial contractility. We use the models cre-lox with specific knockout for insulin receptor substrate (IRS) and co-activator of PPAR (PGC-1b) (basal and pressure overload). The objective was understood the role in the dynamics of cardiac metabolism. We analyzed cardiac structure (histology and stereology), cardiac function (echocardiography and the working heart technique), metabolism (glucose uptake), hormonal action (Western Blotting), gene expression (RT-PCR) from enzyme metabolism (lipid, carbohydrates, respiratory chain, transcriptional and hypertrophic factors) and mitochondrial function. We found in CIRS12KO, severe cardiac dysfunction, mitochondrial dysfunction and reduction of gene expression. And in the PGC-1bKO when subjected to pressure overload, the progression to heart failure, with mitochondrial dysfunction, and alteration of gene expression from enzyme metabolism. The data show that changes on cardiac metabolism and gene expression in both models explain the metabolic pathways that lead to compensated hypertrophy to HF. We suggest that the mitochondrial dysfunction and the gene expression was possible mechanisms for HF. We conclude that IRS and PGC-1b are key factors of cardiac dynamics, which are essential to the structure and heart function. IRS and PGC-1b represent a promising target for limiting the transition from compensated cardiac hypertrophy to heart failure...


Subject(s)
Animals , Rats , Cardiomegaly/complications , Gene Expression/genetics , Heart Failure/etiology , Heart Failure/pathology , Mice , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , PPAR-beta/therapeutic use , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mice, Knockout
12.
Chinese Journal of Burns ; (6): 294-297, 2009.
Article in Chinese | WPRIM | ID: wpr-257398

ABSTRACT

<p><b>OBJECTIVE</b>To explore the role of EGF in regulating HaCaT apoptosis through peroxisome proliferator-activated receptor beta (PPARbeta).</p><p><b>METHODS</b>Cultured HaCaT cells were divided into different groups with different additives in culture medium as control (normal culture), TNF-alpha (with addition of 10 ng/mL TNF-alpha), EGF (with addition of 20 ng/mL EGF), EGF + TNF-alpha (cells were treated with 10 ng/mL TNF-alpha for 60 mins after the exposure to 20 ng/mL EGF for 4 hs) groups. Conjugation activity and transcription activity of PPARbeta of HaCaT cells in each group were detected by electrophoretic mobility shift assay (EMSA) and luciferase gene analysis (LGA). Protein expression of PPARbeta of HaCaT cells after transfected by missense oligonucleotide (scrODN) and antisense oligonucleotide (asODN) was determined by Western blot. Caspase-3 activity and apoptosis rate were detected by flow cytometry.</p><p><b>RESULTS</b>Conjugation and transcription activity of PPARbeta DNA were enhanced as shown in EMSA and LGA. Compared with that of cells in groups transfected by scrODN, protein expression of PPARbeta in cells of groups transfected by asODN was obviously inhibited as shown in Western blot. Caspase-3 activity of cells in TNF-alpha and EGF + TNF-alpha groups transfected by asODN was stronger than that of cells in TNF-alpha and EGF + TNF-alpha groups transfected by scrODN (P < 0.01). Apoptosis rate of cells in control, EGF, TNF-alpha, and EGF + TNF-alpha groups which were transfected by scrODN was (7.31 +/- 0.45)%, (7.43 +/- 0.21)%, (39.78 +/- 0.65)%, (28.34 +/- 0.54)% respectively, and that in those groups transfected by asODN was (8.22 +/- 0.51)%, (7.83 +/- 0.67)%, (46.78 +/- 0.48)%, (44.69 +/- 0.83)%. Apoptosis rate of cells in TNF-alpha and EGF + TNF-alpha groups transfected by asODN was respectively higher than that in TNF-alpha and EGF + TNF-alpha groups transfected by scrODN (P < 0.01).</p><p><b>CONCLUSIONS</b>EGF inhibits HaCaT KC apoptosis caused by TNF-alpha in a PPARbeta-dependent manner.</p>


Subject(s)
Humans , Apoptosis , Cell Culture Techniques , Cell Line , Epidermal Growth Factor , Pharmacology , PPAR-beta , Genetics , Metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha
13.
Arq. bras. endocrinol. metab ; 51(4): 526-533, jun. 2007. ilus, tab
Article in Portuguese | LILACS | ID: lil-457088

ABSTRACT

Os receptores ativados por proliferadores de peroxissoma (PPARs) são fatores de transcrição pertencentes à família de receptores nucleares que regulam a homeostase da glicose, metabolismo de lipídeos e inflamação. Três proteínas, codificadas por genes distintos, têm sido identificadas: PPARalfa, PPARbeta e PPARgama, que controlam a expressão gênica pela ligação a elementos responsivos específicos (PPREs) localizados na região promotora. Estudos recentes sugerem que a ativação do PPARgama pode diminuir a progressão da aterosclerose e aumentar a sensibilidade à insulina, podendo ser um potencial alvo terapêutico para o tratamento de diversas enfermidades, incluindo o diabetes melito do tipo 2 e dislipidemia. Esta revisão destaca os estudos recentes e os avanços das principais funções que esse receptor desempenha no metabolismo, com ênfase nos mecanismos moleculares e eficácia terapêutica.


The peroxisome proliferators-activated receptors (PPARs) are transcription factors belonging to the family of nuclear receptors that regulate glucose homeostasis, lipid metabolism and inflammation. Three proteins, encoded by distinct genes, have been identified: PPARalpha, PPARbeta and PPARgamma, which control gene expression by binding to specific response elements (PPREs) in the promoters. Recent studies suggest that activation of PPARgamma might decrease atherosclerosis progression and increase the insulin sensitivity, might be a potential therapeutic target for the treatment of a diverse array of disorders, including type 2 diabetes and dyslipidaemia. This review highlights recent studies, which have advanced our understanding of the pivotal role that this receptor plays in metabolism, with particular reference to the molecular mechanisms and therapeutic efficacy.


Subject(s)
Humans , Atherosclerosis/prevention & control , Glucose/metabolism , Homeostasis/physiology , Lipid Metabolism/physiology , PPAR gamma/genetics , Transcription Factors/genetics , /drug therapy , Dyslipidemias/drug therapy , Inflammation Mediators/metabolism , Inflammation/metabolism , Polymorphism, Genetic , PPAR alpha/genetics , PPAR alpha/therapeutic use , PPAR gamma/therapeutic use , PPAR-beta/genetics , PPAR-beta/therapeutic use
14.
Chinese Journal of Burns ; (6): 284-287, 2007.
Article in Chinese | WPRIM | ID: wpr-347686

ABSTRACT

<p><b>OBJECTIVE</b>To explore the effect of epidermal growth factor (EGF) on apoptosis induced by TNF-alpha and the expression of PPARbeta in HaCaT keratinocytes.</p><p><b>METHODS</b>HaCaT keratinocytes were cultured and randomly divided into A (normal control), B (with treatment of 10 ng/ml TNF-alpha for 24 hours), C (with treatment of 20 ng/ml TNF-alpha for 24 hours), D (with treatment of 10 ng/ml TNF-alpha after 20 ng/ml EGF treatment for 4 hours), E (with treatment of 20 ng/ml TNF-alpha after 20 ng/ml EGF treatment for 4 hours) groups. The apoptosis of HaCaT keratinocytes was observed by flow cytometry. The proliferative activity of HaCaT keratinocytes was evaluated by MTT method. The activity of caspase-3 was analyzed with caspase colorimetric assay Kit. The changes in the mRNA and protein expression of PPARbeta in HaCaT keratinocytes were observed by RT-PCR and western-blotting after treatment with different concentrations (5, 10, 20, 40 ng/ml) of EGF for 4 or 24 hrs.</p><p><b>RESULTS</b>Compared with A and B groups [(32 +/- 6)%, (57 +/- 6)%], the apoptosis of HaCaT keratinocytes in D and E groups were significantly increased [(20 +/- 3)%, (28 +/- 4)%, respectively, P < 0.01], while the survival rate of HaCaT keratinocytes in D and E groups increased, and the caspase-3 activity were decreased (P < 0.01). The expression of PPARbeta mRNA and protein in HaCaT keratinocytes reached the peak with the treatment of 20 ng/ml EGF.</p><p><b>CONCLUSION</b>EGF can inhibit the apoptosis of HaCaT keratinocytes induced by TNF-alpha, and it can also increase the expression of PPARbeta.</p>


Subject(s)
Humans , Apoptosis , Caspase 3 , Metabolism , Cell Line , Epidermal Growth Factor , Pharmacology , Keratinocytes , Cell Biology , Metabolism , PPAR-beta , Metabolism , RNA, Messenger , Metabolism , Tumor Necrosis Factor-alpha , Pharmacology
15.
Chinese Journal of Burns ; (6): 369-373, 2006.
Article in Chinese | WPRIM | ID: wpr-331561

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the influence of antisense phosphorothioate oligonucleotides on peroxisome proliferator-activated receptors (PPARbeta) in the TNF-alpha mediated apoptosis of HaCat cells.</p><p><b>METHODS</b>HaCat cells were resuscitated and randomly divided into normal control (without transfection), sham (merely with liposome transfection), scrODN (with transfection of 4 micromol/L PPARbeta scrODN), asODN (with transfection of 4 micromol/L PPARbeta asODN), TNF-alpha with transfection of 10 micromol/L TNF-alpha), scrODN + TNF-alpha with 10 micromol/L TNF-alpha stimulation after transfection of 4 micromol/L PPARbeta scrODN), asODN + TNF-alpha with 10 micromol/L TNF-alpha stimulation after transfection of 4 micromol/L PPARbeta asODN) groups. The mRNA and protein levels of PPARbeta were determined with RT-PCR and Western blotting, respectively. The changes in cell morphology were observed with Hoechst 33258 fluorescent staining to quantitate apoptotic rate of nuclei. The effect of PPARbeta asODN on HaCat cell viability was assayed with MTT method. Activation of caspase-3 was evaluated with caspase colorimetric analysis kit.</p><p><b>RESULTS</b>The mRNA and protein expression of PPARbeta in normal control, sham, scrODN groups were similar, but it decreased obviously in asODN group. The nuclear apoptotic rate in normal control, scrODN and asODN groups were rather low, and the caspase-3 activity in these groups was also low. After 24 hours of culture, the nuclear apoptotic rate in TNF-alpha and scrODN + TNF-alpha groups were (33.1 +/- 2.7)% and (32.9 +/- 3.0)%, respectively, while that in asODN + TNF-alpha group was obviously increased (58.8 +/- 4.6)%, with the caspase-3 activity significantly higher, but the number of live cells markedly lower than that in the former 2 groups (P < 0.05).</p><p><b>CONCLUSION</b>PPARbeta expression can promote the apoptosis of HaCat cells mediated by TNF-alpha.</p>


Subject(s)
Humans , Apoptosis , Caspase 3 , Metabolism , Cell Cycle , Cell Line , Cell Proliferation , Oligonucleotides, Antisense , Genetics , Pharmacology , PPAR-beta , Genetics , Pharmacology , RNA, Messenger , Metabolism , Transfection , Tumor Necrosis Factor-alpha , Pharmacology
16.
Chinese Journal of Cardiology ; (12): 1080-1084, 2005.
Article in Chinese | WPRIM | ID: wpr-253008

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effects of atorvastatin on angiotensin II (Ang II)-induced hypertrophy of cardiac myocytes (MC) and the changes of mRNA expression of peroxisome proliferators-activated receptor alpha, gamma (PPAR alpha, gamma) subtypes in vitro.</p><p><b>METHODS</b>Hypertrophy in neonatal rat MC was established with Ang II and treated with atorvastatin. The surface area of MC was analyzed by the aid of NIH Image J software, and the synthetic rate of protein in MC was detected by (3)H-leucine incorporation. mRNA expression of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), matrix metalloproteinase (MMP) 9, MMP2, interleukin1beta (IL-1beta) and PPARalpha, gamma was measured by reverse transcription-polymerase chain reaction (RT-PCR).</p><p><b>RESULTS</b>Changes of MC were detected induced by Ang II, including increases in surface area, mRNA expression of ANP, BNP, MMP9, MMP2 and IL-1beta, and (3)H-leucine incorporation, as well as a decrease in mRNA expression of PPARalpha, gamma. Treatment with atorvastatin inhibited the changes above in a dose-dependent manner, but no change was found in treated with DMSO.</p><p><b>CONCLUSION</b>Atorvastatin inhibits cardiac hypertrophy in vitro. It is suggested that atorvastatin has a potential role in the prevention and treatment of cardiac diseases such as cardiac hypertrophy, and PPAR alpha and gamma maybe involved in this process.</p>


Subject(s)
Animals , Rats , Angiotensin II , Atorvastatin , Cardiomegaly , Metabolism , Cells, Cultured , Gene Expression Regulation , Heptanoic Acids , Pharmacology , Myocytes, Cardiac , Metabolism , PPAR alpha , Metabolism , PPAR-beta , Metabolism , Pyrroles , Pharmacology , Rats, Wistar , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL