Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Phytomedicine ; 126: 155445, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38412666

RESUMO

BACKGROUND: Diabetic nephropathy (DN) is the primary cause of end-stage renal disease (ESRD), and the therapeutic strategies for DN are limited. Notoginsenoside Fc (Fc), a novel saponin isolated from Panax Notoginseng (PNG), has been reported to alleviate vascular injury in diabetic rats. However, the protective effects of Fc on DN remain unclear. PURPOSE: To investigate the beneficial effects and mechanisms of Fc on DN. METHODS: Db/db mice were treated with 2.5, 5 and 10 mg·kg-1·d-1 of Fc for 8 weeks. High glucose (HG) induced mouse glomerular endothelial cells (GECs) were treated with 2.5, 5 and 10 µM of Fc for 24 h. RESULTS: Our data found that Fc ameliorated urinary microalbumin level, kidney dysfunction and histopathological damage in diabetic mice. Moreover, Fc alleviated the accumulation of oxidative stress, the collapse of mitochondrial membrane potential and the expression of mitochondrial fission proteins, such as Drp-1 and Fis1, while increased the expression of mitochondrial fusion protein Mfn2. Fc also decreased pyroptosis-related proteins levels, such as TXNIP, NLRP3, cleaved caspase-1, and GSDMD-NT, indicating that Fc ameliorated GECs pyroptosis. In addition, 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2) expression was increased in diabetic group, which was partially abrogated by Fc. Our data further proved that knockdown of HMGCS2 could restrain HG-induced GECs mitochondrial dysfunction and pyroptosis. These results indicated that the inhibitory effects of Fc on mitochondrial damage and pyroptosis were associated with the suppression of HMGCS2. CONCLUSION: Taken together, this study clearly demonstrated that Fc ameliorated GECs pyroptosis and mitochondrial dysfunction partly through regulating HMGCS2 pathway, which might provide a novel drug candidate for DN.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Ginsenosídeos , Doenças Mitocondriais , Ratos , Camundongos , Animais , Nefropatias Diabéticas/metabolismo , Células Endoteliais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Piroptose , Doenças Mitocondriais/metabolismo , Hidroximetilglutaril-CoA Sintase/metabolismo , Proteínas de Ciclo Celular/metabolismo
2.
Front Med ; 17(2): 339-351, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36602721

RESUMO

Ketone bodies have beneficial metabolic activities, and the induction of plasma ketone bodies is a health promotion strategy. Dietary supplementation of sodium butyrate (SB) is an effective approach in the induction of plasma ketone bodies. However, the cellular and molecular mechanisms are unknown. In this study, SB was found to enhance the catalytic activity of 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), a rate-limiting enzyme in ketogenesis, to promote ketone body production in hepatocytes. SB administrated by gavage or intraperitoneal injection significantly induced blood ß-hydroxybutyrate (BHB) in mice. BHB production was induced in the primary hepatocytes by SB. Protein succinylation was altered by SB in the liver tissues with down-regulation in 58 proteins and up-regulation in 26 proteins in the proteomics analysis. However, the alteration was mostly observed in mitochondrial proteins with 41% down- and 65% up-regulation, respectively. Succinylation status of HMGCS2 protein was altered by a reduction at two sites (K221 and K358) without a change in the protein level. The SB effect was significantly reduced by a SIRT5 inhibitor and in Sirt5-KO mice. The data suggests that SB activated HMGCS2 through SIRT5-mediated desuccinylation for ketone body production by the liver. The effect was not associated with an elevation in NAD+/NADH ratio according to our metabolomics analysis. The data provide a novel molecular mechanism for SB activity in the induction of ketone body production.


Assuntos
Corpos Cetônicos , Sirtuínas , Camundongos , Animais , Ácido Butírico/farmacologia , Ácido Butírico/metabolismo , Corpos Cetônicos/metabolismo , Fígado/metabolismo , Hidroxibutiratos/metabolismo , Regulação para Baixo , Sirtuínas/genética , Sirtuínas/metabolismo , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo
3.
Frontiers of Medicine ; (4): 339-351, 2023.
Artigo em Inglês | WPRIM | ID: wpr-982565

RESUMO

Ketone bodies have beneficial metabolic activities, and the induction of plasma ketone bodies is a health promotion strategy. Dietary supplementation of sodium butyrate (SB) is an effective approach in the induction of plasma ketone bodies. However, the cellular and molecular mechanisms are unknown. In this study, SB was found to enhance the catalytic activity of 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), a rate-limiting enzyme in ketogenesis, to promote ketone body production in hepatocytes. SB administrated by gavage or intraperitoneal injection significantly induced blood ß-hydroxybutyrate (BHB) in mice. BHB production was induced in the primary hepatocytes by SB. Protein succinylation was altered by SB in the liver tissues with down-regulation in 58 proteins and up-regulation in 26 proteins in the proteomics analysis. However, the alteration was mostly observed in mitochondrial proteins with 41% down- and 65% up-regulation, respectively. Succinylation status of HMGCS2 protein was altered by a reduction at two sites (K221 and K358) without a change in the protein level. The SB effect was significantly reduced by a SIRT5 inhibitor and in Sirt5-KO mice. The data suggests that SB activated HMGCS2 through SIRT5-mediated desuccinylation for ketone body production by the liver. The effect was not associated with an elevation in NAD+/NADH ratio according to our metabolomics analysis. The data provide a novel molecular mechanism for SB activity in the induction of ketone body production.


Assuntos
Camundongos , Animais , Ácido Butírico/metabolismo , Corpos Cetônicos/metabolismo , Fígado/metabolismo , Hidroxibutiratos/metabolismo , Regulação para Baixo , Sirtuínas/metabolismo , Hidroximetilglutaril-CoA Sintase/metabolismo
4.
Molecules ; 27(22)2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36432116

RESUMO

Primary liver cancer is the fifth leading death of cancers in men, and hepatocellular carcinoma (HCC) accounts for approximately 90% of all primary liver cancer cases. Sorafenib is a first-line drug for advanced-stage HCC patients. Sorafenib is a multi-target kinase inhibitor that blocks tumor cell proliferation and angiogenesis. Despite sorafenib treatment extending survival, some patients experience side effects, and sorafenib resistance does occur. 3-Hydroxymethyl glutaryl-CoA synthase 2 (HMGCS2) is the rate-limiting enzyme for ketogenesis, which synthesizes the ketone bodies, ß-hydroxybutyrate (ß-HB) and acetoacetate (AcAc). ß-HB is the most abundant ketone body which is present in a 4:1 ratio compared to AcAc. Recently, ketone body treatment was found to have therapeutic effects against many cancers by causing metabolic alternations and cancer cell apoptosis. Our previous publication showed that HMGCS2 downregulation-mediated ketone body reduction promoted HCC clinicopathological progression through regulating c-Myc/cyclin D1 and caspase-dependent signaling. However, whether HMGCS2-regulated ketone body production alters the sensitivity of human HCC to sorafenib treatment remains unclear. In this study, we showed that HMGCS2 downregulation enhanced the proliferative ability and attenuated the cytotoxic effects of sorafenib by activating expressions of phosphorylated (p)-extracellular signal-regulated kinase (ERK), p-P38, and p-AKT. In contrast, HMGCS2 overexpression decreased cell proliferation and enhanced the cytotoxic effects of sorafenib in HCC cells by inhibiting ERK activation. Furthermore, we showed that knockdown HMGCS2 exhibited the potential migratory ability, as well as decreasing zonula occludens protein (ZO)-1 and increasing c-Myc expression in both sorafenib-treated Huh7 and HepG2 cells. Although HMGCS2 overexpression did not alter the migratory effect, expressions of ZO-1, c-Myc, and N-cadherin decreased in sorafenib-treated HMGCS2-overexpressing HCC cells. Finally, we investigated whether ketone treatment influences sorafenib sensitivity. We showed that ß-HB pretreatment decreased cell proliferation and enhanced antiproliferative effect of sorafenib in both Huh7 and HepG2 cells. In conclusion, this study defined the impacts of HMGCS2 expression and ketone body treatment on influencing the sorafenib sensitivity of liver cancer cells.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Masculino , Humanos , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Carcinoma Hepatocelular/metabolismo , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Cetonas/uso terapêutico , Neoplasias Hepáticas/patologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Corpos Cetônicos/metabolismo , Corpos Cetônicos/uso terapêutico , MAP Quinases Reguladas por Sinal Extracelular , Resultado do Tratamento
5.
Gut ; 69(8): 1423-1431, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31753852

RESUMO

OBJECTIVE: Food intake normally stimulates release of satiety and insulin-stimulating intestinal hormones, such as glucagon-like peptide (GLP)-1. This response is blunted in obese insulin resistant subjects, but is rapidly restored following Roux-en-Y gastric bypass (RYGB) surgery. We hypothesised this to be a result of the metabolic changes taking place in the small intestinal mucosa following the anatomical rearrangement after RYGB surgery, and aimed at identifying such mechanisms. DESIGN: Jejunal mucosa biopsies from patients undergoing RYGB surgery were retrieved before and after very-low calorie diet, at time of surgery and 6 months postoperatively. Samples were analysed by global protein expression analysis and Western blotting. Biological functionality of these findings was explored in mice and enteroendocrine cells (EECs) primary mouse jejunal cell cultures. RESULTS: The most prominent change found after RYGB was decreased jejunal expression of the rate-limiting ketogenic enzyme mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (mHMGCS), corroborated by decreased ketone body levels. In mice, prolonged high-fat feeding induced the expression of mHMGCS and functional ketogenesis in jejunum. The effect of ketone bodies on gut peptide secretion in EECs showed a ∼40% inhibition of GLP-1 release compared with baseline. CONCLUSION: Intestinal ketogenesis is induced by high-fat diet and inhibited by RYGB surgery. In cell culture, ketone bodies inhibited GLP-1 release from EECs. Thus, we suggest that this may be a mechanism by which RYGB can remove the inhibitory effect of ketone bodies on EECs, thereby restituting the responsiveness of EECs resulting in increased meal-stimulated levels of GLP-1 after surgery.


Assuntos
Restrição Calórica , Células Enteroendócrinas/metabolismo , Derivação Gástrica , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Corpos Cetônicos/biossíntese , Ácido 3-Hidroxibutírico/sangue , Ácido 3-Hidroxibutírico/farmacologia , Anastomose em-Y de Roux , Animais , Células Cultivadas , Gorduras na Dieta/administração & dosagem , Emulsões/farmacologia , Emulsões Gordurosas Intravenosas/farmacologia , Feminino , Peptídeo 1 Semelhante ao Glucagon/antagonistas & inibidores , Humanos , Hidroximetilglutaril-CoA Sintase/metabolismo , Corpos Cetônicos/metabolismo , Cetonas/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fosfolipídeos/farmacologia , Período Pós-Operatório , Período Pré-Operatório , Cultura Primária de Células , Óleo de Soja/farmacologia
6.
Chin J Nat Med ; 17(8): 575-584, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31472894

RESUMO

3-Hydroxy-3-methylglutaryl-CoA synthase (HMGS) is the first committed enzyme in the MVA pathway and involved in the biosynthesis of terpenes in Tripterygium wilfordii. The full-length cDNA and a 515 bp RNAi target fragment of TwHMGS were ligated into the pH7WG2D and pK7GWIWG2D vectors to respectively overexpress and silence, TwHMGS was overexpressed and silenced in T. wilfordii suspension cells using biolistic-gun mediated transformation, which resulted in 2-fold increase and a drop to 70% in the expression level compared to cells with empty vector controls. During TwHMGS overexpression, the expression of TwHMGR, TwDXR and TwTPS7v2 was significantly upregulated to the control. In the RNAi group, the expression of TwHMGR, TwDXS, TwDXR and TwMCT visibly displayed downregulation to the control. The cells with TwHMGS overexpressed produced twice higher than the control value. These results proved that differential expression of TwHMGS determined the production of triptolide in T. wilfordii and laterally caused different trends of relative gene expression in the terpene biosynthetic pathway. Finally, the substrate acetyl-CoA was docked into the active site of TwHMGS, suggesting the key residues including His247, Lys256 and Arg296 undergo electrostatic or H-bond interactions with acetyl-CoA.


Assuntos
Diterpenos/metabolismo , Regulação da Expressão Gênica de Plantas , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Fenantrenos/metabolismo , Proteínas de Plantas/metabolismo , Tripterygium/metabolismo , Acetilcoenzima A/metabolismo , Sequência de Aminoácidos , Vias Biossintéticas , Domínio Catalítico , Compostos de Epóxi/metabolismo , Hidroximetilglutaril-CoA Sintase/química , Modelos Moleculares , Triterpenos Pentacíclicos , Proteínas de Plantas/química , Proteínas de Plantas/genética , Interferência de RNA , Terpenos/metabolismo , Tripterygium/enzimologia , Tripterygium/genética , Triterpenos/metabolismo
7.
Oxid Med Cell Longev ; 2017: 9032792, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28814987

RESUMO

SCOPE: Several reports in the literature have suggested the renoprotective effects of ketone bodies and green tea polyphenols (GTPs). Our previous study found that GTP consumption could elevate the renal expression of the ketogenic rate-limiting enzyme, which was decreased by a high-fat diet (HFD) in rats. Here, we investigated whether ketogenesis can mediate renoprotection by GTPs against an HFD. METHODS AND RESULTS: Wistar rats were fed a standard or HFD with or without GTPs for 18 weeks. The renal oxidative stress level, kidney function, renal expression, and activity levels of mitochondrial 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) synthase 2 (HMGCS2) and sirtuin 3(SIRT3) were detected. The increased renal oxidative stress and the loss of renal function induced by the HFD were ameliorated by GTPs. Renal ketogenesis and SIRT3 expression and activity levels, which were reduced by the HFD, were restored by GTPs. In vitro, HEK293 cells were transfected with the eukaryotic expression plasmid pcDNA HMGCS2. GTP treatment could upregulate HMGCS2 and SIRT3 expression. Although SIRT3 expression was not affected by HMGCS2 transfection, the 4-hydroxy-2-nonenal (4-HNE) level and the acetyl-MnSOD (K122)/MnSOD ratio were reduced in HMGCS2-transfected cells in the context of H2O2. CONCLUSION: The ketogenesis/SIRT3 pathway mediates the renoprotection of GTPs against the oxidative stress induced by an HFD.


Assuntos
Dieta Hiperlipídica , Rim/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Polifenóis/farmacologia , Sirtuína 3/metabolismo , Chá/química , Aldeídos/química , Aldeídos/metabolismo , Animais , Glicemia/análise , Peso Corporal/efeitos dos fármacos , Colesterol/sangue , Proteína Forkhead Box O3/metabolismo , Células HEK293 , Humanos , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Insulina/sangue , Rim/metabolismo , Rim/patologia , Masculino , Polifenóis/química , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Chá/metabolismo
8.
J Nutr Sci Vitaminol (Tokyo) ; 62(4): 249-256, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27725410

RESUMO

Western diets induce obesity associated with an increased risk of hypercholesterolaemia. Indeed, obesity-induced hypercholesterolaemia is correlated with increased coronary cardiovascular disease (CVD) risk. Male C57BL/6J mice were fed a normal diet, high-fat and high-sucrose diet (HF/HS), HF/HS with green tea extract powder diet (HF/HS+GT), HF/HS with eriodictyol diet (HF/HS+Eri), or HF/HS with green tea extract powder and eriodictyol diet (HF/HS+GT+Eri) for 8 wk. Body weight was lower in the HF/HS+GT+Eri group than in the HF/HS group (-8.3%, p<0.01). The HF/HS diet elicited an upregulation of total cholesterol levels (-63%, p<0.001), and low-density lipoprotein (LDL) levels (-89%, p<0.001) were significantly suppressed by the GT+Eri diet. Conversely, no change (p>0.05) was observed in the HF/HS+GT and HF/HS+Eri groups. The HF/HS diet-induced hepatic mRNA increase in 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) was ameliorated (-73%) by the oral administration of green tea extract and eriodictyol. Moreover, the GT+Eri diet suppressed HF/HS diet-induced upregulation of 3-hydroxy-3-methylglutaryl-coenzyme A synthase (HMGCS) (-75%, p<0.05). Furthermore, the LDL receptor (LDLR) levels were higher in the HF/HS+GT+Eri group (+50%, p<0.05) than in the HF/HS group. These results suggest that a combination of green tea and eriodictyol decreases cholesterol levels, particularly LDL levels, accompanied by the suppression of HMGCR and HMGCS levels and upregulation of LDLR levels in the liver.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Sacarose Alimentar/efeitos adversos , Flavanonas/farmacologia , Extratos Vegetais/farmacologia , Receptores de LDL/metabolismo , Chá/química , Animais , Glicemia/metabolismo , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Sacarose Alimentar/administração & dosagem , Hidroximetilglutaril-CoA Redutases/genética , Hidroximetilglutaril-CoA Redutases/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/genética , Regulação para Cima
9.
Eur J Nutr ; 54(7): 1055-67, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25315197

RESUMO

PURPOSE: This study was conducted to investigate the effect of fish oil (FO) and krill oil (KO) supplementation on glucose tolerance in obese New Zealand white rabbits. METHODS: The experiments were carried out with 24 male rabbits randomly divided into four groups: KO-castrated, treated with KO; FO-castrated, treated with FO; C-castrated, non-treated; NC-non-castrated, non-treated. At the end of treatment period (2 months), an intravenous glucose tolerance test (IVGTT) was performed in all rabbits. RESULTS: Fasting blood glucose concentrations in FO and KO animals were significantly lower than in group C. The blood glucose concentrations in FO- and KO-treated animals returned to initial values after 30 and 60 min of IVGTT, respectively. In liver, carnitine palmitoyltransferase 2 (Cpt2) and 3-hydroxy-3-methyl-glutaryl-CoA synthase 2 (Hmgcs2) genes were significantly increased in FO-fed rabbits compared with the C group. Acetyl-CoA carboxylase alpha (Acaca) expression was significantly reduced in both KO- and FO-fed rabbits. In skeletal muscle, Hmgcs2 and Cd36 were significantly higher in KO-fed rabbits compared with the C group. Acaca expression was significantly lower in KO- and FO-fed rabbits compared with the C group. CONCLUSION: The present results indicate that FO and KO supplementation decreases fasting blood glucose and improves glucose tolerance in obese New Zealand white rabbits. This could be ascribed to the ameliorated insulin sensitivity and insulin secretion and modified gene expressions of some key enzymes involved in ß-oxidation and lipogenesis in liver and skeletal muscle.


Assuntos
Metabolismo dos Carboidratos/efeitos dos fármacos , Suplementos Nutricionais , Óleos de Peixe/administração & dosagem , Obesidade/sangue , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo , Animais , Glicemia/metabolismo , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Euphausiacea , Peixes , Regulação da Expressão Gênica , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Insulina/sangue , Resistência à Insulina , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Coelhos
10.
PLoS One ; 9(11): e112118, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25390336

RESUMO

Monocarboxylates such as pyruvate, lactate and ketone bodies are crucial for energy supply of all tissues, especially during energy restriction. The transport of monocarboxylates across the plasma membrane of cells is mediated by monocarboxylate transporters (MCTs). Out of 14 known mammalian MCTs, six isoforms have been functionally characterized to transport monocarboxylates and short chain fatty acids (MCT1-4), thyroid hormones (MCT8, -10) and aromatic amino acids (MCT10). Knowledge on the regulation of the different MCT isoforms is rare. In an attempt to get more insights in regulation of MCT expression upon energy deprivation, we carried out a comprehensive analysis of tissue specific expression of five MCT isoforms upon 48 h of fasting in mice. Due to the crucial role of peroxisome proliferator-activated receptor (PPAR)-α as a central regulator of energy metabolism and as known regulator of MCT1 expression, we included both wildtype (WT) and PPARα knockout (KO) mice in our study. Liver, kidney, heart, small intestine, hypothalamus, pituitary gland and thyroid gland of the mice were analyzed. Here we show that the expression of all examined MCT isoforms was markedly altered by fasting compared to feeding. Expression of MCT1, MCT2 and MCT10 was either increased or decreased by fasting dependent on the analyzed tissue. MCT4 and MCT8 were down-regulated by fasting in all examined tissues. However, PPARα appeared to have a minor impact on MCT isoform regulation. Due to the fundamental role of MCTs in transport of energy providing metabolites and hormones involved in the regulation of energy homeostasis, we assumed that the observed fasting-induced adaptations of MCT expression seem to ensure an adequate energy supply of tissues during the fasting state. Since, MCT isoforms 1-4 are also necessary for the cellular uptake of drugs, the fasting-induced modifications of MCT expression have to be considered in future clinical care algorithms.


Assuntos
Jejum , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ácido 3-Hidroxibutírico/sangue , Animais , Ácidos Graxos não Esterificados/sangue , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Hipotálamo/metabolismo , Intestino Delgado/metabolismo , Rim/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , PPAR alfa/deficiência , PPAR alfa/genética , PPAR alfa/metabolismo , Hipófise/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Glândula Tireoide/metabolismo , Tiroxina/sangue
11.
World J Microbiol Biotechnol ; 29(3): 523-31, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23138457

RESUMO

A hydroxymethylglutaryl-CoA synthase gene, designated as GlHMGS (GenBank accession No. JN391469) involved in ganoderic acid (GA) biosynthesis pathway was cloned from Ganoderma lucidum. The full-length cDNA of GlHMGS (GenBank accession No. JN391468) was found to contain an open reading frame of 1,413 bp encoding a polypeptide of 471 amino acid residues. The deduced amino acid sequence of GlHMGS shared high homology with other known hydroxymethylglutaryl-CoA synthase (HMGS) enzymes. In addition, functional complementation of GlHMGS in a mutant yeast strain YSC1021 lacking HMGS activity demonstrated that the cloned cDNA encodes a functional HMGS. A 1,561 bp promoter sequence was isolated and its putative regulatory elements and potential specific transcription factor binding sites were analyzed. GlHMGS expression profile analysis revealed that salicylic acid, abscisic acid and methyl jasmonate up-regulated GlHMGS transcript levels over the control. Further expression analysis revealed that the developmental stage and carbon source had significant effects on GlHMGS transcript levels. GlHMGS expression peaked on day 16 before decreasing with prolonged culture time. The highest mRNA level was observed when the carbon source was maltose. Overexpression of GlHMGS enhanced GA content in G. lucidum. This study provides useful information for further studying this gene and on its function in the ganoderic acid biosynthetic pathway in G. lucidum.


Assuntos
Regulação Fúngica da Expressão Gênica , Hidroximetilglutaril-CoA Sintase , Reishi/enzimologia , Triterpenos/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Vias Biossintéticas , Biotecnologia/métodos , Clonagem Molecular , DNA Complementar/genética , Medicamentos de Ervas Chinesas , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Dados de Sequência Molecular , Filogenia , Regiões Promotoras Genéticas , Reishi/genética , Reishi/crescimento & desenvolvimento , Reishi/metabolismo , Análise de Sequência de DNA , Regulação para Cima
12.
Nutrition ; 26(11-12): 1176-80, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20137896

RESUMO

OBJECTIVE: Increased oxidative stress plays an important role in cardiovascular diseases including hypertension and stroke. Evidence has indicated that ketone bodies could exert antioxidative effects. We explored the role of renal mitochondrial 3-hydroxy-3-methylglutaryl-coenzyme A synthase (HMGCS2) expression, a key control site of ketogenesis, in stroke-prone spontaneously hypertensive rats (SHRSPs) and their ancestral hypertensive but stroke-resistant spontaneously hypertensive rats (SHRs). METHODS: Two groups of SHRSPs were fed a standard chow or standard chow supplemented with clofibrate (an agonist of HMGCS2 promoter), respectively, and SHRs fed with a standard chow were used as controls. The renal levels of HMGCS2, Akt, and phosphorylated protein kinase B (Akt) were measured by western blotting. Malondialdehyde, catalase, superoxide dismutase, and glutathione peroxidase were detected by assay kits. RESULTS: Compared with SHRs, lower HMGCS2 protein expression, enhanced phosphorylated Akt signal, higher malondialdehyde levels, and higher catalase activity were observed in kidney tissues in SHRSPs (P < 0.05). No differences in superoxide dismutase and glutathione peroxidase activities were observed between SHRSPs and SHRs. Clofibrate treatment significantly upregulated renal HMGCS2 expressions, inhibited phosphorylation of Akt, and decreased malondialdehyde levels and catalase activities in SHRSP kidney tissues (P < 0.05). CONCLUSION: These results demonstrated the difference in HMGCS2 expression and oxidative stress in kidney tissues between SHRSPs and their SHR controls. The enhanced oxidative stress was partly due to the lower HMGCS2 expression regulated possibly by the Akt signaling pathway.


Assuntos
Anti-Hipertensivos/farmacologia , Hidroximetilglutaril-CoA Sintase/metabolismo , Hipertensão/enzimologia , Rim/enzimologia , Mitocôndrias/enzimologia , Estresse Oxidativo/efeitos dos fármacos , Acidente Vascular Cerebral/fisiopatologia , Animais , Anti-Hipertensivos/uso terapêutico , Catalase/metabolismo , Clofibrato/farmacologia , Clofibrato/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Hidroximetilglutaril-CoA Sintase/genética , Hipertensão/metabolismo , Hipertensão/prevenção & controle , Rim/metabolismo , Masculino , Malondialdeído/metabolismo , PPAR gama/agonistas , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/prevenção & controle , Regulação para Cima/efeitos dos fármacos
13.
J Mol Neurosci ; 38(2): 159-65, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18792811

RESUMO

Depleted uranium results from the enrichment of natural uranium for energetic purpose. Its potential dispersion in the environment would set human populations at risk of being contaminated through ingestion. Uranium can build up in the brain and induce behavior disorders. As a major constituent of the myelin sheath, cholesterol is essential to brain function, and several neurological pathologies result from a disruption of cholesterol metabolism. To assess the effect of a chronic contamination with depleted uranium on cerebral cholesterol metabolism, rats were exposed to depleted uranium for 9 months through drinking water at 40 mg/l. The study focuses on gene expression. Cholesterol-catabolizing enzyme CYP46A1 displayed a 39% increase of its messenger RNA (mRNA) level. 3-Hydroxy-3-methylglutamyl CoA synthase gene expression rose from 91%. Concerning cholesterol transport, mRNA levels of scavenger receptor-B1 and adenosine triphosphate-binding cassette transporter A1 increased by 34% and that of apolipoprotein E by 75%. Concerning regulation, gene expression of nuclear receptors peroxisome proliferator-activated receptors alpha and gamma increased by 46% and 36% respectively, whereas that of retinoid-X-receptor decreased by 29%. In conclusion, a chronic internal contamination with depleted uranium does not affect the health status of rats but induces molecular changes in the dynamic equilibrium of the cerebral cholesterol pool.


Assuntos
Córtex Cerebral , Colesterol/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Acil Coenzima A/genética , Acil Coenzima A/metabolismo , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Colestanotriol 26-Mono-Oxigenase/genética , Colestanotriol 26-Mono-Oxigenase/metabolismo , Colesterol 24-Hidroxilase , Humanos , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de LDL/genética , Receptores de LDL/metabolismo , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Urânio/administração & dosagem , Urânio/farmacologia
14.
J Appl Microbiol ; 105(6): 1973-81, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19120644

RESUMO

AIMS: To identify factors associated with the Staphylococcus aureus pine-oil disinfectant-reduced-susceptibility (PD(RS)) mechanism and to describe one possible PD(RS) model. METHODS AND RESULTS: Comparative genomic sequencing (CGS) and microarray analysis were utilized to detect mutations and transcriptome alterations that occur in a S. aureus PD(RS) mutant. Mutant analysis, antimicrobial gradient plates, growth studies and 3-hydroxy-3-methylglutaryl coenzyme A synthase assays were then performed to confirm the biological consequences of the 'omics' alterations detected in a PD(RS) mutant. CGS uncovered three mutations in a PD(RS) mutant in a(n): alcohol dehydrogenase (adh), catabolite control protein A (ccpA) and an NADPH-flavin oxidoreductase (frp). These mutations lead to increased growth rates; increased transcription of an NAD-dependent D-lactate dehydrogenase gene (ddh); and increased flux through the mevalonate pathway. PD(RS) mutants demonstrated reduced susceptibility to bacitracin and farnesol, and one PD(RS) mutant displayed upregulation of bacA, a bacitracin-resistance gene. Collectively, this evidence demonstrates altered undecaprenol metabolism in PD(RS) mutants. CONCLUSIONS: The PD(RS) mechanism proposed results from increased catabolic capabilities and increased flux through the mevalonate pathway as well as altered bactoprenol physiology. SIGNIFICANCE AND IMPACT OF THE STUDY: A novel mechanism that bacteria utilize to overcome the killing effects of PD formulations is proposed that is unique from the PD(RS) mechanism of the enterobacteraciae.


Assuntos
Desinfetantes/farmacologia , Farmacorresistência Bacteriana/genética , Pinus/química , Óleos de Plantas/farmacologia , Staphylococcus aureus/genética , Proteínas de Bactérias/genética , Análise Mutacional de DNA , Farmacorresistência Bacteriana/fisiologia , Hidroximetilglutaril-CoA Sintase/metabolismo , Lactato Desidrogenases/genética , Lactato Desidrogenases/metabolismo , Análise em Microsséries , Testes de Sensibilidade Microbiana , Proteínas Repressoras/genética , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia
15.
Planta ; 221(6): 844-56, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15770484

RESUMO

3-Hydroxy-3-methylglutaryl-coenzyme-A (HMG-CoA) synthase (HMGS; EC 2.3.3.10) synthesizes HMG-CoA, a substrate for mevalonate biosynthesis in the isoprenoid pathway. It catalyzes the condensation of acetyl-CoA with acetoacetyl-CoA (AcAc-CoA) to yield S-HMG-CoA and HS-CoA. In Brassica juncea (Indian mustard), HMGS is encoded by four isogenes (BjHMGS1-BjHMGS4). We have already enzymatically characterized recombinant BjHMGS1 expressed in Escherichia coli, and have identified its residues that are significant in catalysis. To further study HMGS mRNA expression that is developmentally regulated in flowers and seedlings, we have examined its mRNA distribution by in situ hybridization and reverse transcriptase-polymerase chain reaction (RT-PCR). We observed predominant localization of HMGS mRNA in the stigmas and ovules of flower buds and in the piths of seedling hypocotyls. RT-PCR analysis revealed that BjHMGS1 and BjHMGS2 but not BjHMGS3 and BjHMGS4were expressed in floral buds. To investigate the subcellular localization of BjHMGS1, we fused BjHMGS1 translationally in-frame either to the N- or C-terminus of green fluorescent protein (GFP). BjHMGS1-GFP and GFP-BjHMGS1 fusions were used in particle gun bombardment of onion epidermal cells and tobacco BY-2 cells. The GFP-BjHMGS1 construct was also used in agroinfiltration of tobacco leaves. Both GFP-fusion proteins were observed transiently expressed in the cytosol on confocal microscopy of onion epidermal cells, tobacco BY-2 cells, and agroinfiltrated tobacco leaves. Further, subcellular fractionation of total proteins from transgenic plants expressing GFP-BjHMGS1 derived from Agrobacterium-mediated transformation confirmed that BjHMGS1 is a cytosolic enzyme. We suggest that the presence of BjHMGS isoforms is likely related to the specialization of each in different cellular and metabolic processes rather than to a different intracellular compartmentation of the enzyme.


Assuntos
Hidroximetilglutaril-CoA Sintase/metabolismo , Mostardeira/enzimologia , RNA Mensageiro/metabolismo , Sequência de Aminoácidos , Células Cultivadas , Regulação Enzimológica da Expressão Gênica , Regulação da Expressão Gênica de Plantas , Hidroximetilglutaril-CoA Sintase/genética , Isoenzimas , Dados de Sequência Molecular , Cebolas , Plantas Geneticamente Modificadas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Nicotiana
16.
J Biol Chem ; 274(20): 14508-13, 1999 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-10318878

RESUMO

Ying Yang 1 (YY1) is shown to bind to the proximal promoters of the genes encoding 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) synthase, farnesyl diphosphate (FPP) synthase, and the low density lipoprotein (LDL) receptor. To investigate the potential effect of YY1 on the expression of SREBP-responsive genes, HepG2 cells were transiently transfected with luciferase reporter constructs under the control of promoters derived from either HMG-CoA synthase, FPP synthase, or the LDL receptor genes. The luciferase activity of each construct increased when HepG2 cells were incubated in lipid-depleted media or when the cells were cotransfected with a plasmid encoding mature sterol regulatory element-binding protein (SREBP)-1a. In each case, the increase in luciferase activity was attenuated by coexpression of wild-type YY1 but not by coexpression of mutant YY1 proteins that are known to be defective in either DNA binding or in modulating transcription of other known YY1-responsive genes. In contrast, incubation of cells in lipid-depleted media resulted in induction of an HMG-CoA reductase promoter-luciferase construct by a process that was unaffected by coexpression of wild-type YY1. Electromobility shift assays were used to demonstrate that the proximal promoters of the HMG-CoA synthase, FPP synthase, and the LDL receptor contain YY1 binding sites and that YY1 displaced nuclear factor Y from the promoter of the HMG-CoA synthase gene. We conclude that YY1 inhibits the transcription of specific SREBP-dependent genes and that, in the case of the HMG-CoA synthase gene, this involves displacement of nuclear factor Y from the promoter. We hypothesize that YY1 plays a regulatory role in the transcriptional regulation of specific SREBP-responsive genes.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica , Dedos de Zinco , Alquil e Aril Transferases/metabolismo , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Geraniltranstransferase , Humanos , Hidroximetilglutaril-CoA Sintase/metabolismo , Regiões Promotoras Genéticas , Receptores de LDL/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1 , Proteína de Ligação a Elemento Regulador de Esterol 2 , Fator de Transcrição YY1
17.
J Biol Chem ; 269(46): 28523-6, 1994 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-7961793

RESUMO

A somatic cell mutant of the Chinese hamster ovary (CHO)-K1 (called Mev-1), auxotrophic for mevalonate by virtue of a complete lack of detectable cytosolic 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) synthase activity, was transfected with a plasmid containing the cDNA for ketogenic mitochondrial HMG-CoA synthase under the control of SV40 early promoter. The resulting stable cell line (Mev-SM) was able to grow in the absence of mevalonate. Analysis of Western blot showed that the new cell line strongly expressed mitochondrial HMG-CoA synthase protein. Immunocytochemical studies using specific antibodies against mitochondrial HMG-CoA synthase showed that the protein was located exclusively inside the mitochondria. The prototroph cell line Mev-SM can incorporate labeled acetate into cholesterol in the absence of mevalonate. These results show that the new cell line may circumvent the lack of cytosolic HMG-CoA synthase activity by producing cholesterol-convertible HMG-CoA inside the mitochondria.


Assuntos
Hidroximetilglutaril-CoA Sintase/genética , Cetonas/metabolismo , Ácido Mevalônico/metabolismo , Mitocôndrias/enzimologia , Sequência de Aminoácidos , Animais , Células CHO , Linhagem Celular , Colesterol/biossíntese , Cricetinae , DNA Complementar , Hidroximetilglutaril-CoA Sintase/metabolismo , Cloreto de Magnésio/farmacologia , Microscopia Imunoeletrônica , Mitocôndrias/ultraestrutura , Dados de Sequência Molecular , Mutação , Transfecção
19.
Biochem Biophys Res Commun ; 153(1): 422-7, 1988 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-2897853

RESUMO

We reported previously that, in the perfused rat liver, oleic acid increased the specific activity of cytosolic enzymes of cholesterol biosynthesis. In this study, we examined the effects of oral administration of olive oil on the activities of HMG-CoA synthase, AcAc-CoA thiolase, AcAc-CoA ligase and HMG-CoA reductase. Olive oil feeding increased the specific activity of hepatic HMG-CoA synthase by 50%, AcAc-CoA thiolase by 2-fold, and AcAc-CoA ligase by 3-fold. Olive oil had no effect on HMG-CoA reductase activity. These data suggest that the enzymes that supply the HMG-CoA required for hepatic cholesterogenesis are regulated in parallel by a physiological substrate, fatty acid, independent of HMG-CoA reductase under these conditions.


Assuntos
Acetil-CoA C-Acetiltransferase/metabolismo , Acetiltransferases/metabolismo , Colesterol/biossíntese , Coenzima A Ligases/metabolismo , Gorduras na Dieta/farmacologia , Ácidos Graxos/farmacologia , Hidroximetilglutaril-CoA Sintase/metabolismo , Fígado/metabolismo , Oxo-Ácido-Liases/metabolismo , Óleos de Plantas/farmacologia , Animais , Intubação Gastrointestinal , Fígado/efeitos dos fármacos , Masculino , Azeite de Oliva , Ratos , Ratos Endogâmicos
20.
Biochem J ; 240(2): 541-7, 1986 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-2880580

RESUMO

In order to determine the regulation mechanisms of ergosterol biosynthesis in yeast, we developed growth conditions leading to high or limiting ergosterol levels in wild type and sterol-auxotrophic mutant strains. An excess of sterol is obtained in anaerobic sterol-supplemented cultures of mutant and wild type strains. A low sterol level is obtained in aerobic growth conditions in mutant strains cultured with optimal sterol supplementation and in wild type strain deprived of pantothenic acid, as well as in anaerobic cultures without sterol supplementation. Measurements of the specific activities of acetoacetyl-CoA thiolase, HMG-CoA (3-hydroxy-3-methylglutaryl-CoA) synthase and HMG-CoA reductase (the first three enzymes of the pathway), show that in cells deprived of ergosterol, acetoacetyl-CoA thiolase and HMG-CoA synthase are generally increased. In an excess of ergosterol, in anaerobiosis, the same enzymes are strongly decreased. A 5-10-fold decrease is observed for acetoacetyl-CoA thiolase and HMG-CoA synthase. In contrast, HMG-CoA reductase is only slightly affected by these conditions. These results show that ergosterol could regulate its own synthesis, at least partially, by repression of the first two enzymes of the pathway. Our results also show that exogenous sterols, even if strongly incorporated by auxotrophic mutant cells, cannot suppress enzyme activities in aerobic growth conditions. Measurement of specific enzyme activities in mutant cells also revealed that farnesyl pyrophosphate thwarts the enhancement of the activities of the two first enzymes.


Assuntos
Ergosterol/biossíntese , Saccharomyces cerevisiae/enzimologia , Acetil-CoA C-Acetiltransferase/metabolismo , Anaerobiose , Hidroximetilglutaril-CoA Redutases/metabolismo , Hidroximetilglutaril-CoA Sintase/metabolismo , Hidroximetilglutaril-CoA-Redutases NADP-Dependentes , Mutação , Ácido Pantotênico/farmacologia , Saccharomyces cerevisiae/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA