Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
1.
ScientificWorldJournal ; 2021: 1583154, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34531707

RESUMO

Ayu-narezushi, a traditional Japanese fermented food, comprises abundant levels of lactic acid bacteria (LAB) and free amino acids. This study aimed to examine the potential beneficial effects of ayu-narezushi and investigated whether ayu-narezushi led to improvements in the Tsumura Suzuki obese diabetes (TSOD) mice model of spontaneous metabolic syndrome because useful LAB are known as probiotics that regulate intestinal function. In the present study, the increased body weight of the TSOD mice was attenuated in those fed the ayu-narezushi-comprised chow (ayu-narezushi group) compared with those fed the normal rodent chow (control group). Serum triglyceride and cholesterol levels were significantly lower in the Ayu-narezushi group than in the control group at 24 weeks of age. Furthermore, hepatic mRNA levels of carnitine-palmitoyl transferase 1 and acyl-CoA oxidase, which related to fatty acid oxidation, were significantly increased in the ayu-narezushi group than in the control group at 24 weeks of age. In conclusion, these results suggested that continuous feeding with ayu-narezushi improved obesity and dyslipidemia in the TSOD mice and that the activation of fatty acid oxidation in the liver might contribute to these improvements.


Assuntos
Modelos Animais de Doenças , Alimentos Fermentados , Metabolismo dos Lipídeos , Síndrome Metabólica/dietoterapia , Osmeriformes , Acil-CoA Oxidase/biossíntese , Acil-CoA Oxidase/genética , Animais , Peso Corporal , Carnitina O-Palmitoiltransferase/biossíntese , Carnitina O-Palmitoiltransferase/genética , Colesterol/sangue , Dislipidemias/dietoterapia , Dislipidemias/genética , Indução Enzimática , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Gordura Intra-Abdominal/química , Gordura Intra-Abdominal/patologia , Japão , Fígado/metabolismo , Síndrome Metabólica/sangue , Síndrome Metabólica/genética , Camundongos , Camundongos Obesos , Obesidade/dietoterapia , Obesidade/genética , Oryza , Oxirredução , PPAR alfa/biossíntese , PPAR alfa/genética , Reação em Cadeia da Polimerase em Tempo Real , Cloreto de Sódio , Triglicerídeos/sangue
2.
J Ethnopharmacol ; 268: 113654, 2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33271248

RESUMO

HEADINGS ETHNOPHARMACOLOGICAL RELEVANCE: Sophora alopecuroides L. is a traditional ethnopharmacological plant, which is widely used in traditional Chinese medicine and Mongolian and Uighur medicine to ameliorate "thirst disease". AIM OF THE STUDY: This study aimed to investigate the antidiabetic activities and mechanisms of a flavonoid-rich extract from Sophora alopecuroides L. (SA-FRE) both in vivo and vitro. MATERIALS AND METHODS: The main six chemical constituents of SA-FRE were elucidated based on an off-line semi-preparative liquid chromatography nuclear magnetic resonance (LC-NMR) protocol. Myc-GLUT4-mOrange-L6 cell models and mouse model with diabetes induced by high-fat diet combined with STZ injection were respectively adopted to investigate the antidiabetic effects of SA-FRE both in vitro and vivo. RESULTS: In vivo, 4-week treatment of SA-FRE ameliorated hyperglycemia, dyslipidemia, and insulin resistance in diabetic mice. Mechanically, SA-FRE regulated PPARα and PPARγ expression in white adipose tissue (WAT) and liver, thereby ameliorating dyslipidemia. Moreover, SA-FRE increased the phosphorylation of PKC and further stimulated the GLUT4 expression in WAT and skeletal muscle, thus increasing the glucose utilization in vivo. In vitro, 50 µg/mL SA-FRE increased GLUT4 translocation to about 1.91-fold and glucose uptake to 1.82-fold in L6-myotubes. SA-FRE treatment increased the GLUT4 expression at both gene and protein levels. Furthermore, only Gö6983, a PKC inhibitor, reversed the SA-FRE-induced GLUT4 translocation and expression at the gene and protein levels. CONCLUSIONS: Generally, SA-FRE ameliorated hyperglycemia, dyslipidemia, and insulin resistance partly through activating PKC/GLUT4 pathway and regulating PPARα and PPARγ expression.


Assuntos
Transportador de Glucose Tipo 4/biossíntese , Hipoglicemiantes/uso terapêutico , PPAR alfa/biossíntese , PPAR gama/biossíntese , Proteína Quinase C/biossíntese , Sophora , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Dieta Hiperlipídica/efeitos adversos , Flavonoides/isolamento & purificação , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Expressão Gênica , Transportador de Glucose Tipo 4/genética , Hipoglicemiantes/isolamento & purificação , Hipoglicemiantes/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR alfa/genética , PPAR gama/genética , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Proteína Quinase C/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Estreptozocina
3.
Int J Mol Sci ; 21(18)2020 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-32967195

RESUMO

Dim light at night (dLAN) is associated with metabolic risk but the specific effects on lipid metabolism have only been evaluated to a limited extent. Therefore, to explore whether dLAN can compromise lipid metabolic homeostasis in healthy individuals, we exposed Wistar rats to dLAN (~2 lx) for 2 and 5 weeks and analyzed the main lipogenic pathways in the liver and epididymal fat pad, including the control mechanisms at the hormonal and molecular level. We found that dLAN promoted hepatic triacylglycerol accumulation, upregulated hepatic genes involved in de novo synthesis of fatty acids, and elevated glucose and fatty acid uptake. These observations were paralleled with suppressed fatty acid synthesis in the adipose tissue and altered plasma adipokine levels, indicating disturbed adipocyte metabolic function with a potential negative impact on liver metabolism. Moreover, dLAN-exposed rats displayed an elevated expression of two peroxisome proliferator-activated receptor family members (Pparα and Pparγ) in the liver and adipose tissue, suggesting the deregulation of important metabolic transcription factors. Together, our results demonstrate that an impaired balance of lipid biosynthetic pathways caused by dLAN can increase lipid storage in the liver, thereby accounting for a potential linking mechanism between dLAN and metabolic diseases.


Assuntos
Tecido Adiposo/metabolismo , Ritmo Circadiano , Regulação da Expressão Gênica , Iluminação/efeitos adversos , Metabolismo dos Lipídeos , Fígado/metabolismo , Animais , Ácidos Graxos/biossíntese , Masculino , PPAR alfa/biossíntese , PPAR gama/biossíntese , Ratos , Ratos Wistar , Triglicerídeos/biossíntese
4.
Biochem Pharmacol ; 180: 114194, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32800851

RESUMO

MiR-23a-5p is involved in the occurrence and development of some serious diseases, but its effects on intestinal ischemia-reperfusion (II/R) injury is unclear. In this research, the hypoxia/reoxygenation (H/R) model on IEC-6 cells and II/R model in mice were used. The data showed that the ROS level in model group was significantly increased compared with control group. The level of intestinal MPO was increased and serum SOD was decreased in mice compared with sham group. Moreover, the expression levels of miR-23a-5p in model groups were obviously increased in vitro and in vivo, while the expression levels of PPARα, FOXO3α, PGC-1α, Nrf2, CAT, NQO1, HO-1 and SOD2 were significantly decreased. The double luciferase reporter gene assay showed that there was binding site between miR-23a-5p and PPARα. When miR-23a-5p was inhibited or PPARα gene was overexpressed, H/R-caused cell damage was alleviated and ROS level was decreased compared with NC group. PPARα expression level was increased, accompanied by the increased levels of FOXO3α, PGC-1α, Nrf2, CAT, NQO1, HO-1 and SOD2. After enhancing miR-23a-5p expression or silencing PPARα gene, H/R-caused cell damage was further aggravated compared with NC group, and ROS level was increased associated with the decreased levels of FOXO3α, PGC-1α, Nrf2, CAT, NQO1, HO-1 and SOD2. Our study demonstrated that miR-23a-5p exacerbated II/R injury by promoting oxidative stress via targeting PPARα, which should be considered as one new drug target to treat II/R injury.


Assuntos
Sistemas de Liberação de Medicamentos , Mucosa Intestinal/metabolismo , MicroRNAs/administração & dosagem , Estresse Oxidativo/fisiologia , PPAR alfa/biossíntese , Traumatismo por Reperfusão/metabolismo , Animais , Linhagem Celular , Sistemas de Liberação de Medicamentos/métodos , Mucosa Intestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , Estresse Oxidativo/efeitos dos fármacos , PPAR alfa/antagonistas & inibidores , Ratos , Traumatismo por Reperfusão/patologia
5.
Biomed Res Int ; 2020: 6708061, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32420361

RESUMO

The increased prevalence of nonalcoholic fatty liver disease (NAFLD), which develops from hepatic steatosis, represents a public health challenge. Catalpol, a natural component extracted from the roots of Radix Rehmanniae, has several pharmacological activities. The present study is aimed at examining whether catalpol prevents hepatic steatosis in cell and animal experiments and elucidating the possible mechanisms. HepG2 cells were treated with 300 µM palmitate (PA) and/or catalpol for 24 h in vitro, and male C57BL/6J mice fed a high-fat diet (HFD) were administered catalpol for 18 weeks in vivo. The results revealed that catalpol significantly decreased lipid accumulation in PA-treated HepG2 cells. Moreover, catalpol drastically reduced body weight and lipid accumulation in the liver, whereas it ameliorated hepatocyte steatosis in HFD-fed mice. Notably, catalpol remarkably promoted the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase. Subsequently, catalpol repressed the expressions of lipogenesis-associated genes such as sterol regulatory element-binding protein 1c and fatty acid synthase but promoted the expressions of genes associated with fatty acid ß-oxidation such as peroxisome proliferator-activated receptor α together with its target genes carnitine palmitoyltransferase 1 and acyl-CoA oxidase 1 (ACOX1). However, the preincubation of the HepG2 cells with compound C (10 µM), an AMPK inhibitor, prevented catalpol-mediated beneficial effects. These findings suggest that catalpol ameliorates hepatic steatosis by suppressing lipogenesis and enhancing fatty acid ß-oxidation in an AMPK-dependent manner. Therefore, catalpol has potential as a novel agent in the treatment of NAFLD.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Fígado Gorduroso/tratamento farmacológico , Glucosídeos Iridoides/farmacologia , Lipogênese/efeitos dos fármacos , Acil-CoA Oxidase/biossíntese , Animais , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Células Hep G2 , Humanos , Masculino , Camundongos , PPAR alfa/biossíntese
6.
Metab Eng ; 60: 77-86, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32247827

RESUMO

Maintaining a metabolic steady state is essential for an organism's fitness and survival when confronted with environmental stress, and metabolic imbalance can be reversed by exposing the organism to fasting. Here, we attempted to apply this physiological principle to mammalian cell cultures to improve cellular fitness and consequently their ability to express recombinant proteins. We showed that transient vitamin B5 deprivation, an essential cofactor of central cellular metabolism, can quickly and irreversibly affect mammalian cell growth and division. A selection method was designed that relies on mammalian cell dependence on vitamin B5 for energy production, using the co-expression of the B5 transporter SLC5A6 and a gene of interest. We demonstrated that vitamin B5 selection persistently activates peroxisome proliferator-activated receptors (PPAR), a family of transcription factors involved in energy homeostasis, thereby altering lipid metabolism, improving cell fitness and therapeutic protein production. Thus, stable PPAR activation may constitute a cellular memory of past deprivation state, providing increased resistance to further potential fasting events. In other words, our results imply that cultured cells, once exposed to metabolic starvation, may display an improved metabolic fitness as compared to non-exposed cells, allowing increased resistance to cellular stress.


Assuntos
Homeostase , Ácido Pantotênico/deficiência , Ácido Pantotênico/metabolismo , Proteínas Recombinantes/biossíntese , Animais , Células CHO , Divisão Celular , Células Cultivadas , Cricetinae , Cricetulus , Metabolismo Energético , Vetores Genéticos , Metabolismo dos Lipídeos/fisiologia , PPAR alfa/biossíntese , PPAR alfa/genética , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Estresse Fisiológico , Simportadores/metabolismo
7.
Nutr Cancer ; 72(5): 892-902, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31403341

RESUMO

PPAR receptors are ligand-dependent transcription factors activated in response to various small lipophilic ligands controlling the expression of different genes involved in cellular differentiation, development, metabolism, and tumorigenesis. Unexpectedly, our previous studies have shown that single plasmid-based expression of PPARs under the control of CMV promoter/enhancer was significantly elevated in the presence of PPAR agonists. Here we show that the PPAR reporters controlled by the CMV promoter/enhancer, that was shown to contain three internal non-canonical PPRE elements, can be used as a fast screening system for more effective PPAR ligands. This model allowed us to confirm our previous results indicating that fatty acids of CLA-enriched egg yolks (EFA-CLAs) are efficient PPAR ligands that can specifically upregulate the expression of PPARα and PPARγ leading to downregulation of MCF-7 cancer cell proliferation. We also show that synthetic cis9,trans11CLA is more effective in transactivation of PPARγ, while trans10,cis12CLA of PPARα receptor indicating the selectivity of the CLA isomers. This report presents a novel, fast, and reliable strategy for simple testing of PPAR ligands using PPAR expressing plasmids containing the CMV promoter/enhancer that can trigger the positive feedback loop of PPAR self-transcription in the presence of PPAR ligands.


Assuntos
Citomegalovirus/genética , Ácidos Linoleicos Conjugados/metabolismo , PPAR alfa/agonistas , PPAR alfa/biossíntese , PPAR gama/agonistas , PPAR gama/biossíntese , Animais , Linhagem Celular Tumoral , Proliferação de Células , Gema de Ovo/química , Humanos , Ligantes , Células MCF-7 , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ativação Transcricional
8.
Toxicol Lett ; 319: 85-94, 2020 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-31730885

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is a chronic hepatic disease associated with the excessive accumulation of lipids in the liver. Premenopausal women are protected from the liver metabolic complications of obesity compared with body mass index (BMI)-matched men. This protection may be related to estrogen's ability to limit liver fat accumulation. Aryl hydrocarbon receptor (AhR), a novel regulator of NAFLD, may be an important target for regulating estrogen homeostasis. In present study, we used benzo[a]pyrene (BaP), a classic and potent ligand of AhR, to activate AhR pathway causes overexpression of the estrogen-metabolizing enzyme cytochrome P450 1A1 (CYP1A1) and affects the expression of important genes involved in hepatic lipid regulation. BaP induces CYP1A1 expression through AhR signaling and inhibits the protective effect of 17ß-estradiol (E2) on hepatic steatosis, characterized by triglyceride accumulation, and markers of liver damage are significantly elevated. The expression of adipogenic genes involved in the hepatic lipid metabolism of sterol regulatory element-binding protein-1c (SREBP-1c) was increased compared with that in the control group. Furthermore, the mRNA and protein levels of peroxisome proliferator-activated receptor alpha (PPARα), which is involved in fatty acid oxidation, were significantly reduced. Taken together, our results revealed that the steatotic effect of AhR is likely due to overexpression of the E2 metabolic enzyme CYP1A1, which affects the estrogen signaling pathway, leading to the suppression of fatty acid oxidation, inhibition of the hepatic export of triglycerides, and an increase in peripheral fat mobilization. The results from this study may help establish AhR as a novel therapeutic and preventive target for fatty liver disease.


Assuntos
Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Benzo(a)pireno/farmacologia , Citocromo P-450 CYP1A1/biossíntese , Citocromo P-450 CYP1A1/genética , Estradiol/farmacologia , Estrogênios/metabolismo , Feminino , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , PPAR alfa/biossíntese , PPAR alfa/genética , Receptores de Hidrocarboneto Arílico/agonistas , Transdução de Sinais/efeitos dos fármacos , Proteína de Ligação a Elemento Regulador de Esterol 1/biossíntese , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Triglicerídeos/metabolismo
9.
Am J Physiol Endocrinol Metab ; 317(3): E436-E445, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31211621

RESUMO

Hepatic trefoil factor 3 (Tff3) was identified as a potential protein for the treatment of diabetes, yet the effect of Tff3 on nonalcoholic fatty liver disease (NAFLD) has never been explored. Here, we found that the expression of hepatic Tff3 was significantly decreased in NAFLD mice models, suggesting that Tff3 was a potential marker gene for NAFLD. Restoring the expression of Tff3 in the liver of NAFLD mice, including diabetic (db), obese (ob/ob), and diet-induced obese mice, with adenovirus-mediated Tff3 (Ad-Tff3) apparently attenuates the fatty liver phenotype. In contrast, adenovirus-mediated knockdown of Tff3 (Ad-shTff3) in C57BL/6J mice results in an obvious fatty liver phenotype. Furthermore, our molecular experiments indicated that hepatic Tff3 could alleviate hepatic steatosis via upregulating the expression of peroxisome proliferator-activated receptor-α (PPARα) directly, thereby enhancing the fatty acid oxidation process in the liver. Notably, we found that Tff3 attenuates the fatty liver phenotype independent of modulation of lipogenesis and improves the capacity of anti-inflammation. Overall, our results suggested that hepatic Tff3 could be effectively used as a potential therapy target for the treatment of NAFLD.


Assuntos
Ácidos Graxos/metabolismo , Hepatopatia Gordurosa não Alcoólica/terapia , PPAR alfa/biossíntese , Fator Trefoil-3/genética , Animais , Biomarcadores , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Dieta Hiperlipídica , Técnicas de Silenciamento de Genes , Terapia Genética , Hepatócitos/metabolismo , Lipogênese/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/metabolismo , Oxirredução , PPAR alfa/genética
10.
Exp Cell Res ; 382(1): 111455, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31163124

RESUMO

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


Assuntos
Nefropatias Diabéticas/metabolismo , Ácidos Hidroxieicosatetraenoicos/fisiologia , PPAR alfa/fisiologia , PPAR gama/fisiologia , PPAR beta/fisiologia , Amidinas/farmacologia , Anilidas/farmacologia , Animais , Linhagem Celular , Citocromo P-450 CYP4A/metabolismo , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/patologia , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/toxicidade , Ácidos Hidroxieicosatetraenoicos/biossíntese , Indóis/farmacologia , Túbulos Renais/citologia , Masculino , Camundongos , PPAR alfa/biossíntese , PPAR alfa/genética , PPAR gama/biossíntese , PPAR gama/genética , PPAR beta/biossíntese , PPAR beta/genética , Ratos , Sulfonas/farmacologia , Tiofenos/farmacologia
11.
Antioxid Redox Signal ; 30(5): 713-732, 2019 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-29631413

RESUMO

AIMS: Metabolic remodeling of cardiac muscles during pathological hypertrophy is characterized by downregulation of fatty acid oxidation (FAO) regulator, peroxisome proliferator-activated receptor alpha (PPARα). Thereby, we hypothesized that a cardiac-specific induction of PPARα might restore the FAO-related protein expression and resultant energy deficit. In the present study, consequences of PPARα augmentation were evaluated for amelioration of chronic oxidative stress, myocyte apoptosis, and cardiac function during pathological cardiac hypertrophy. RESULTS: Nanotized PPARα overexpression targeted to myocardium was done by a stearic acid-modified carboxymethyl-chitosan (CMC) conjugated to a 20-mer myocyte-targeted peptide (CMCP). Overexpression of PPARα ameliorated pathological hypertrophy and improved cardiac function. Augmented PPARα in hypertrophied myocytes revealed downregulated p53 acetylation (lys 382), leading to reduced apoptosis. Such cells showed increased binding of PPARα with p53 that in turn reduced interaction of p53 with glycogen synthase kinase-3ß (GSK3ß), which upregulated inactive phospho-GSK3ß (serine [Ser]9) expression within mitochondrial protein fraction. Altogether, the altered molecular milieu in PPARα-overexpressed hypertrophy groups restored mitochondrial structure and function both in vitro and in vivo. INNOVATION: Cardiomyocyte-targeted overexpression of a protein of interest (PPARα) by nanotized plasmid has been described for the first time in this study. Our data provide a novel insight towards regression of pathological hypertrophy by ameliorating mitochondrial oxidative stress in targeted PPARα-overexpressed myocardium. CONCLUSION: PPARα-overexpression during pathological hypertrophy showed substantial betterment of mitochondrial structure and function, along with downregulated apoptosis. Myocardium-targeted overexpression of PPARα during pathological cardiac hypertrophy led to an overall improvement of cardiac energy deficit and subsequent cardiac function, thereby, opening up a potential avenue for cardiac tissue engineering during hypertrophic cardiac pathophysiology.


Assuntos
Cardiomegalia/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Mitocôndrias/patologia , Miocárdio/metabolismo , Nanopartículas/metabolismo , PPAR alfa/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Animais , Humanos , Mitocôndrias/metabolismo , Nanopartículas/química , Estresse Oxidativo , PPAR alfa/química , PPAR alfa/genética
12.
Arch Biochem Biophys ; 662: 219-225, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30553767

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are a family of ligand-activated nuclear transcription factors, with three characterized subtypes: PPARα, PPARß/δ, and PPARγ. The biological correlation between the two PPAR subtypes PPARα and γ and carcinogenesis is well-characterized; however, substantially less is known about the biological functions of PPARß/δ. PPARß/δ has been reported to repress transcription when PPARß/δ and PPARα or PPARγ are simultaneously expressed in some cells, and MDA-MB-231 cells express functional levels of PPARß/δ. We have previously reported that Δ9-tetrahydrocannabinol (Δ9-THC), a major cannabinoid component of the drug-type cannabis plant, can stimulate the expression of fatty acid 2-hydroxylase (FA2H) via upregulation of PPARα expression in human breast cancer MDA-MB-231 cells. Although the possibility of an inhibitory interaction between PPARα and PPARß/δ has not been demonstrated in MDA-MB-231 cells, we reasoned if this interaction were to exist, Δ9-THC should make PPARα free to achieve FA2H induction. Here, we show that a PPARß/δ-mediated suppression of PPARα function, but not of PPARγ, exists in MDA-MB-231 cells and Δ9-THC causes FA2H induction via mechanisms underlying the cancellation of PPARß/δ-mediated inhibition of PPARα, in addition to the upregulation of PPARα.


Assuntos
Dronabinol/farmacologia , Oxigenases de Função Mista/genética , PPAR alfa/biossíntese , PPAR delta/metabolismo , PPAR beta/metabolismo , Regulação para Cima/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , PPAR delta/genética , PPAR beta/genética , Sulfonas/farmacologia , Tiofenos/farmacologia , Transcrição Gênica/efeitos dos fármacos
13.
Artigo em Inglês | MEDLINE | ID: mdl-30528668

RESUMO

The crosstalk between peroxisome proliferator-activated receptor α (PPARα) and estrogenic pathways are shared from fish to humans. Salmonid fish had an additional genome duplication, and two PPARα isoforms (PPARαBa and PPARαBb) were previously identified. Since a negative regulation between estrogen signaling and PPARα was described, a post-transcriptional gene silencing for PPARαBb was designed in primary brown trout hepatocytes. The aims of the study were to: (i) decipher the effects of PPARαBb knock-down on peroxisome morphology and on mRNA expression of potential target genes, and (ii) to assess the cross-interferences caused by an estrogenic compound (17α-ethinylestradiol - EE2) and a PPARα agonist (Wy-14,643 - Wy) using the established knock-down model. A knock-down efficiency of 70% was achieved for PPARαBb and its silencing significantly reduced the volume density of peroxisomes, but did not alter mRNA levels of the studied genes. Exposure to Wy did not change peroxisome morphology or mRNA expression, but under silencing conditions Wy rescued the volume density of peroxisomes to control levels, and increased acyl-coenzyme A oxidase 1-3l (Acox1-3l) mRNA. Exposure to EE2 caused a reduction of peroxisome volume density, but under silencing conditions this effect was abolished and ApoA1 mRNA level was diminished. The morphological alterations of peroxisomes by WY and EE2 demonstrated that obtained results are PPARαBb dependent, and suggest the regulation of unknown downstream targets of PPARαBb. In summary, PPARαBb is involved in the control of peroxisome size and/or number, which opens future opportunities to explore its regulation and molecular targets.


Assuntos
Estrogênios/farmacologia , Proteínas de Peixes , Inativação Gênica/efeitos dos fármacos , Hepatócitos/metabolismo , Subunidade 1 do Complexo Mediador/biossíntese , PPAR alfa , Pirimidinas/farmacologia , Animais , Proteínas de Peixes/agonistas , Proteínas de Peixes/biossíntese , Hepatócitos/citologia , Humanos , PPAR alfa/agonistas , PPAR alfa/biossíntese , Cultura Primária de Células , Truta
14.
Biomed Pharmacother ; 111: 99-108, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30579258

RESUMO

BACKGROUND AND AIMS: The lack of valid therapeutic approach that can ameliorate the manifestations of NASH is a barrier to therapeutic development. Therefore, we investigate the novel role of Methyl Palmitate (MP) in preventing NASH and the possible mechanism involved. METHODS: 50 Male C57BL/6 J mice were randomly divided into 5 groups (n = 10). The control group was fed control diet; model group was fed MCD diet; MP 1 group was fed MCD diet supplemented with MP (75 mg/kg/day); MP 2 group was fed MCD plus MP diet (150 mg/kg/day); and MP 3 group was fed MCD plus MP diet (300 mg/kg/day). Histological staining's, and commercially available kits for serum ALT and AST and hepatic contents of TG, TC, MDA, SOD, and GSH were used to assess NASH. Furthermore, relative liver protein and gene expression levels were determined by Western Blot and qPCR, respectively. RESULTS: Mice fed MCD diet developed NASH, which was markedly improved by MP in a dose-dependent manner. MP treatment improved hepatic content of TG, TC, MDA, SOD and GSH and serum levels of ALT and AST. In vivo studies showed that MP treatment activated PPARα expression, that in turns, promoted ß-oxidation protein and gene expressions, suppressed TNFα, MCP1, TGFß1 and Colla1 protein and gene expression levels, contributing to the prevention of NASH. CONCLUSIONS: Our results indicated that MP could successfully prevent NASH. This effect of MP was mediated through induction of PPARα pathway. This study provides a novel therapeutic target that plays pivotal role in the prevention of NASH.


Assuntos
Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , PPAR alfa/biossíntese , Palmitatos/uso terapêutico , Animais , Deficiência de Colina/complicações , Deficiência de Colina/metabolismo , Células Hep G2 , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/fisiologia , Masculino , Metionina/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/etiologia , Palmitatos/farmacologia
15.
PLoS One ; 13(9): e0203466, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30212482

RESUMO

Despite the important functions of PPARγ in various cell types of the lung, PPARγ-deficiency in club cells induces only mild emphysema. Peroxisomes are distributed in a similar way as PPARγ in the lung and are mainly enriched in club and AECII cells. To date, the effects of PPARγ-deficiency on the overall peroxisomal compartment and its metabolic alterations in pulmonary club cells are unknown. Therefore, we characterized wild-type and club cell-specific PPARγ knockout-mice lungs and used C22 cells to investigate the peroxisomal compartment and its metabolic roles in the distal airway epithelium by means of 1) double-immunofluorescence labelling for peroxisomal proteins, 2) laser-assisted microdissection of the bronchiolar epithelium and subsequent qRT-PCR, 3) siRNA-transfection of PPARγand PPRE dual-luciferase reporter activity in C22 cells, 4) PPARg inhibition by GW9662, 5) GC-MS based lipid analysis. Our results reveal elevated levels of fatty acids, increased expression of PPARα and PPRE activity, a strong overall upregulation of the peroxisomal compartment and its associated gene expression (biogenesis, α-oxidation, ß-oxidation, and plasmalogens) in PPARγ-deficient club cells. Interestingly, catalase was significantly increased and mistargeted into the cytoplasm, suggestive for oxidative stress by the PPARγ-deficiency in club cells. Taken together, PPARα-mediated metabolic induction and proliferation of peroxisomes via a PPRE-dependent mechanism could compensate PPARγ-deficiency in club cells.


Assuntos
Brônquios/metabolismo , Regulação da Expressão Gênica , PPAR alfa/biossíntese , PPAR gama/deficiência , Peroxissomos/metabolismo , Enfisema Pulmonar/metabolismo , Anilidas/farmacologia , Animais , Brônquios/patologia , Camundongos , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , PPAR alfa/antagonistas & inibidores , PPAR alfa/genética , Peroxissomos/genética , Enfisema Pulmonar/genética , Enfisema Pulmonar/patologia
16.
Sci Rep ; 8(1): 13766, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30214009

RESUMO

The rapid decline in fertility that has been occurring to high-producing dairy cows in the past 50 years seems to be associated with metabolic disturbances such as ketosis, supporting the need for research to improve our understanding of the relations among the diet, metabolism and embryonic development. Recently, the ketone body ß-hydroxybutyrate (BOHB) was demonstrated to be a potent inhibitor of histone deacetylases (HDACs). Herein, we performed a series of experiments aiming to investigate the epigenetic effects of BOHB on histone acetylation in somatic cells, cumulus-oocyte complexes (COCs) and somatic cell nuclear transfer (SCNT) embryos. Treatment with BOHB does not increase histone acetylation in cells but stimulates genes associated with ketolysis and master regulators of metabolism. We further demonstrated that maturing COCs with high levels of BOHB does not affect their maturation rate or histone acetylation but increases the expression of PPARA in cumulus cells. Treatment of somatic cell nuclear transfer zygotes with BOHB causes hyperacetylation, which is maintained until the blastocyst stage, causing enhanced FOXO3A expression and blastocyst production. Our data shed light on the epigenetic mechanisms caused by BOHB in bovine cells and embryos and provide a better understanding of the connection between nutrition and reproduction.


Assuntos
Ácido 3-Hidroxibutírico/farmacologia , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/efeitos dos fármacos , Fertilidade/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Oócitos/metabolismo , Ácido 3-Hidroxibutírico/biossíntese , Ácido 3-Hidroxibutírico/genética , Acetilação , Animais , Blastocisto/citologia , Bovinos , Linhagem Celular , Células do Cúmulo/metabolismo , Feminino , Proteína Forkhead Box O3/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Inibidores de Histona Desacetilases/metabolismo , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Técnicas de Transferência Nuclear , Estresse Oxidativo/efeitos dos fármacos , PPAR alfa/biossíntese , Gravidez
17.
Proc Natl Acad Sci U S A ; 115(33): E7871-E7880, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30061404

RESUMO

Smyd1, a muscle-specific histone methyltransferase, has established roles in skeletal and cardiac muscle development, but its role in the adult heart remains poorly understood. Our prior work demonstrated that cardiac-specific deletion of Smyd1 in adult mice (Smyd1-KO) leads to hypertrophy and heart failure. Here we show that down-regulation of mitochondrial energetics is an early event in these Smyd1-KO mice preceding the onset of structural abnormalities. This early impairment of mitochondrial energetics in Smyd1-KO mice is associated with a significant reduction in gene and protein expression of PGC-1α, PPARα, and RXRα, the master regulators of cardiac energetics. The effect of Smyd1 on PGC-1α was recapitulated in primary cultured rat ventricular myocytes, in which acute siRNA-mediated silencing of Smyd1 resulted in a greater than twofold decrease in PGC-1α expression without affecting that of PPARα or RXRα. In addition, enrichment of histone H3 lysine 4 trimethylation (a mark of gene activation) at the PGC-1α locus was markedly reduced in Smyd1-KO mice, and Smyd1-induced transcriptional activation of PGC-1α was confirmed by luciferase reporter assays. Functional confirmation of Smyd1's involvement showed an increase in mitochondrial respiration capacity induced by overexpression of Smyd1, which was abolished by siRNA-mediated PGC-1α knockdown. Conversely, overexpression of PGC-1α rescued transcript expression and mitochondrial respiration caused by silencing Smyd1 in cardiomyocytes. These findings provide functional evidence for a role of Smyd1, or any member of the Smyd family, in regulating cardiac energetics in the adult heart, which is mediated, at least in part, via modulating PGC-1α.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Metabolismo Energético/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Mitocôndrias Cardíacas/metabolismo , Proteínas Musculares/metabolismo , Miocárdio/enzimologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/biossíntese , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/genética , Proteínas Musculares/genética , PPAR alfa/biossíntese , PPAR alfa/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Receptor X Retinoide alfa/biossíntese , Receptor X Retinoide alfa/genética , Fatores de Transcrição/genética
18.
Sci Rep ; 8(1): 12964, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30154499

RESUMO

Chronic intoxication of mice with the porphyrinogenic compound 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) leads to morphological and metabolic changes closely resembling steatohepatitis, a severe form of metabolic liver disease in humans. Since human steatohepatitis (both the alcoholic and non-alcoholic type) is characterized by reduced expression of PPARα and disturbed lipid metabolism we investigated the role of this ligand-activated receptor in the development of DDC-induced liver injury. Acute DDC-intoxication was accompanied by early significant downregulation of Pparα mRNA expression along with PPARα-controlled stress-response and lipid metabolism genes that persisted in the chronic stage. Administration of the specific PPARα agonist fenofibrate together with DDC prevented the downregulation of PPARα-associated genes and also improved the stress response of Nrf2-dependent redox-regulating genes. Moreover, oxidative stress and inflammation were strongly reduced by DDC/fenofibrate co-treatment. In addition, fenofibrate prevented the disruption of hepatocyte intermediate filament cytoskeleton and the formation of Mallory-Denk bodies at late stages of DDC intoxication. Our findings show that, like in human steatohepatitis, PPARα is downregulated in the DDC model of steatohepatitis-like hepatocellular damage. Its downregulation and the pathomorphologic features of steatohepatitis are prevented by co-administration of fenofibrate.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Fígado Gorduroso , Fenofibrato/farmacologia , Corpos de Mallory/metabolismo , PPAR alfa , Agregados Proteicos/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/prevenção & controle , Humanos , Masculino , Corpos de Mallory/patologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , PPAR alfa/agonistas , PPAR alfa/biossíntese , Piridinas/toxicidade
19.
Diabetes Metab Syndr ; 12(5): 711-714, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29699951

RESUMO

BACKGROUND: Metabolic syndrome (MS) is considered one of the major worldwide epidemics. It accounts for billions of cardiovascular disease events and deaths. Till now, major basics of MS are not fully clarified. Peroxisome Proliferator-Activated Receptor-α (PPARα) displays a ligand-activated transcription factor. It is involved in the regulation of many metabolic processes including inflammation, lipid, and glucose metabolism. Therefore, this study investigated the leucocytic expression of PPARα in a metabolic patient in comparison to healthy controls. METHODS: 100 subjects with MS were recruited, in addition to 100 subjects without any obvious metabolic disorders as healthy controls. Expression of PPARα and CD 36 were analyzed on different leucocytic populations using optimized flow-cytometric analysis. Correlations of the expression of both indexes with different clinical and laboratory parameters were analyzed. RESULTS: The eosinophilic expression of PPARα was found to be lower in subjects with MS in comparison to the healthy controls (p value 0.001). Also, PPARα expression, on most of the leucocytic populations, was inversely correlated with waist circumferences among the study populations. CONCLUSION: Circulated eosinophilic expression of PPARα protein is reduced in MS subjects. This conclusion may explain the endothelial dysfunction and obesity associated with MS, as well as it may help in the management of this worldwide health problem.


Assuntos
Eosinófilos/metabolismo , Leucócitos Mononucleares/metabolismo , Síndrome Metabólica/sangue , Síndrome Metabólica/diagnóstico , PPAR alfa/biossíntese , PPAR alfa/sangue , Adulto , Estudos de Casos e Controles , Egito/epidemiologia , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Síndrome Metabólica/epidemiologia , Pessoa de Meia-Idade , PPAR alfa/genética
20.
Obes Res Clin Pract ; 12(Suppl 2): 71-79, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28320596

RESUMO

OBJECTIVE: Previous investigations have shown that the peroxisome proliferator activated receptor beta/delta (PPAR/)-angiopoietin-like protein 4 (ANGPTL4) pathways may be a new pharmacologic target for treatment of obesity. The present study was conducted to test the effect of phillyrin, a glucoside, on obesity in mice. METHOD: Fifty mice were randomly divided into 5 groups (n=10): control group (C57BL/6J mice), obese mice group, two groups of obese mice treated with phillyrin (15 or 45mg/kg/day), one group of obese mice treated with PPAR/ agonist GW0742 (3mg/kg/day). Twelve weeks after treatment, body weight, liver weight, fat weight, lipid levels in the liver, serum levels of tumour necrosis factor-(TNF-), leptin, and insulin, expression of PPAR/, ANGPTL4, and AMP-activated protein kinase (AMPK) were determined. RESULTS: Treatment with phillyrin (15 or 45mg/kg) significantly decreased body weight, liver weight, fat weight, hepatic total cholesterol, free fatty acid, and triglyceride concentrations, serum levels of TNF-, leptin, and insulin concomitantly with up-regulated expression of PPAR/, ANGPTL4, and p-AMPK-. In addition, GW0742 has similar effect of phillyrin. CONCLUSIONS: The present results suggest that phillyrin could regulate the PPAR/-ANGPTL 4 pathway to lower body weight in obese C57BL/6J mice.


Assuntos
Angiopoietinas/biossíntese , Glucosídeos/farmacologia , Obesidade/tratamento farmacológico , PPAR alfa/biossíntese , Transdução de Sinais/efeitos dos fármacos , Angiopoietinas/genética , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/metabolismo , PPAR alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA