Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Obesity (Silver Spring) ; 32(4): 710-722, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38311801

RESUMO

OBJECTIVE: Intestinal gluconeogenesis (IGN), via the initiation of a gut-brain nervous circuit, accounts for the metabolic benefits linked to dietary proteins or fermentable fiber in rodents and has been positively correlated with the rapid amelioration of body weight after gastric bypass surgery in humans with obesity. In particular, the activation of IGN moderates the development of hepatic steatosis accompanying obesity. In this study, we investigated the specific effects of IGN on adipose tissue metabolism, independent of its induction by nutritional manipulation. METHODS: We used two transgenic mouse models of suppression or overexpression of G6pc1, the catalytic subunit of glucose-6 phosphatase, which is the key enzyme of endogenous glucose production specifically in the intestine. RESULTS: Under a hypercaloric diet, mice overexpressing IGN showed lower adiposity and higher thermogenic capacities than wild-type mice, featuring marked browning of white adipose tissue (WAT) and prevention of the whitening of brown adipose tissue (BAT). Sympathetic denervation restricted to BAT caused the loss of the antiobesity effects associated with IGN. Conversely, IGN-deficient mice exhibited an increase in adiposity under a standard diet, which was associated with decreased expression of markers of thermogenesis in both BAT and WAT. CONCLUSIONS: IGN is sufficient to activate the sympathetic nervous system and prevent the expansion and the metabolic alterations of BAT and WAT metabolism under a high-calorie diet, thereby preventing the development of obesity. These data increase knowledge of the mechanisms of weight reduction in gastric bypass surgery and pave the way for new approaches to prevent or cure obesity.


Assuntos
Tecido Adiposo Marrom , Gluconeogênese , Humanos , Animais , Camundongos , Tecido Adiposo Marrom/metabolismo , Gluconeogênese/genética , Obesidade/complicações , Tecido Adiposo Branco/metabolismo , Glucose/metabolismo , Sistema Nervoso Simpático/metabolismo , Termogênese , Metabolismo Energético
2.
Nat Rev Gastroenterol Hepatol ; 20(3): 183-194, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36470967

RESUMO

The intestine, like the liver and kidney, in various vertebrates and humans is able to carry out gluconeogenesis and release glucose into the blood. In the fed post-absorptive state, intestinal glucose is sensed by the gastrointestinal nervous system. The latter initiates a signal to the brain regions controlling energy homeostasis and stress-related behaviour. Intestinal gluconeogenesis (IGN) is activated by several complementary mechanisms, in particular nutritional situations (for example, when food is enriched in protein or fermentable fibre and after gastric bypass surgery in obesity). In these situations, IGN has several metabolic and behavioural benefits. As IGN is activated by nutrients capable of fuelling systemic gluconeogenesis, IGN could be a signal to the brain that food previously ingested is suitable for maintaining plasma glucose for a while. This process might account for the benefits observed. Finally, in this Perspective, we discuss how the benefits of IGN in fasting and fed states could explain why IGN emerged and was maintained in vertebrates by natural selection.


Assuntos
Gluconeogênese , Intestinos , Animais , Humanos , Gluconeogênese/fisiologia , Glucose/metabolismo , Homeostase/fisiologia , Fígado/metabolismo
3.
Sci Rep ; 12(1): 1415, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-35082330

RESUMO

Intestinal gluconeogenesis (IGN), gastric bypass (GBP) and gut microbiota positively regulate glucose homeostasis and diet-induced dysmetabolism. GBP modulates gut microbiota, whether IGN could shape it has not been investigated. We studied gut microbiota and microbiome in wild type and IGN-deficient mice, undergoing GBP or not, and fed on either a normal chow (NC) or a high-fat/high-sucrose (HFHS) diet. We also studied fecal and urine metabolome in NC-fed mice. IGN and GBP had a different effect on the gut microbiota of mice fed with NC and HFHS diet. IGN inactivation increased abundance of Deltaproteobacteria on NC and of Proteobacteria such as Helicobacter on HFHS diet. GBP increased abundance of Firmicutes and Proteobacteria on NC-fed WT mice and of Firmicutes, Bacteroidetes and Proteobacteria on HFHS-fed WT mice. The combined effect of IGN inactivation and GBP increased abundance of Actinobacteria on NC and the abundance of Enterococcaceae and Enterobacteriaceae on HFHS diet. A reduction was observed in the amounf of short-chain fatty acids in fecal (by GBP) and in both fecal and urine (by IGN inactivation) metabolome. IGN and GBP, separately or combined, shape gut microbiota and microbiome on NC- and HFHS-fed mice, and modify fecal and urine metabolome.


Assuntos
Derivação Gástrica/métodos , Microbioma Gastrointestinal/fisiologia , Gluconeogênese/fisiologia , Intestinos/metabolismo , Metaboloma , Estômago/metabolismo , Actinobacteria/classificação , Actinobacteria/genética , Actinobacteria/isolamento & purificação , Animais , DNA Bacteriano/genética , Enterobacteriaceae/classificação , Enterobacteriaceae/genética , Enterobacteriaceae/isolamento & purificação , Enterococcaceae/classificação , Enterococcaceae/genética , Enterococcaceae/isolamento & purificação , Ácidos Graxos Voláteis/metabolismo , Firmicutes/classificação , Firmicutes/genética , Firmicutes/isolamento & purificação , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Filogenia , Proteobactérias/classificação , Proteobactérias/genética , Proteobactérias/isolamento & purificação , Estômago/microbiologia , Estômago/cirurgia
4.
Endocrinology ; 162(7)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33999998

RESUMO

Tamoxifen is a selective estrogen receptor modulator used to activate the CREERT2 recombinase, allowing tissue-specific and temporal control of the somatic mutagenesis to generate transgenic mice. Studies integrating development and metabolism require a genetic modification induced by a neonatal tamoxifen administration. Here, we investigate the effects of a neonatal tamoxifen administration on energy homeostasis in adult male and female C57BL/6J mice. C57BL/6J male and female mouse pups received a single injection of tamoxifen 1 day after birth (NTT) and were fed a high-fat/high-sucrose diet at 6 weeks of age. We measured weight, body composition, glucose and insulin tolerance, basal metabolism, and tibia length and weight in adult mice. The neonatal tamoxifen administration exerted long-term, sex-dependent effects on energy homeostasis. NTT female mice became overweight and developed impaired glucose control in comparison to vehicle-treated littermates. NTT females exhibited 60% increased fat mass, increased food intake, decreased physical activity and energy expenditure, impaired glucose and insulin tolerance, and fasting hyperglycemia and hyperinsulinemia. In contrast, NTT male mice exhibited a modest amelioration of glucose and insulin tolerance and long-term decreased lean mass linked to decreased bone weight. These results suggest that the neonatal tamoxifen administration exerted a marked and sex-dependent influence on adult energy homeostasis and bone weight and must therefore be used with caution for the development of transgenic mouse models regarding studies on energy homeostasis and bone biology.


Assuntos
Animais Recém-Nascidos/metabolismo , Glicemia/metabolismo , Metabolismo Energético/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Fatores Sexuais , Tamoxifeno/farmacologia , Animais , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Feminino , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Condicionamento Físico Animal , Moduladores Seletivos de Receptor Estrogênico/farmacologia
5.
Gut ; 69(12): 2193-2202, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32205419

RESUMO

OBJECTIVE: Hepatic steatosis accompanying obesity is a major health concern, since it may initiate non-alcoholic fatty liver disease (NAFLD) and associated complications like cirrhosis or cancer. Intestinal gluconeogenesis (IGN) is a recently described function that contributes to the metabolic benefits of specific macronutrients as protein or soluble fibre, via the initiation of a gut-brain nervous signal triggering brain-dependent regulations of peripheral metabolism. Here, we investigate the effects of IGN on liver metabolism, independently of its induction by the aforementioned macronutrients. DESIGN: To study the specific effects of IGN on hepatic metabolism, we used two transgenic mouse lines: one is knocked down for and the other overexpresses glucose-6-phosphatase, the key enzyme of endogenous glucose production, specifically in the intestine. RESULTS: We report that mice with a genetic overexpression of IGN are notably protected from the development of hepatic steatosis and the initiation of NAFLD on a hypercaloric diet. The protection relates to a diminution of de novo lipogenesis and lipid import, associated with benefits at the level of inflammation and fibrosis and linked to autonomous nervous system. Conversely, mice with genetic suppression of IGN spontaneously exhibit increased hepatic triglyceride storage associated with activated lipogenesis pathway, in the context of standard starch-enriched diet. The latter is corrected by portal glucose infusion mimicking IGN. CONCLUSION: We conclude that IGN per se has the capacity of preventing hepatic steatosis and its eventual evolution toward NAFLD.


Assuntos
Trato Gastrointestinal/metabolismo , Gluconeogênese/fisiologia , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Obesidade/fisiopatologia , Animais , Quimiocina CCL2/metabolismo , Dieta Hiperlipídica , Interleucina-6/metabolismo , Fígado/inervação , Fígado/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Neurônios/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
6.
J Clin Med ; 9(1)2020 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-31936857

RESUMO

Little is known about the jejunal insulin signalling pathways in insulin resistance/diabetes states and their possible regulation by insulin/leptin. We study in jejunum the relation between insulin signalling and insulin resistance in morbidly obese subjects with low (MO-low-IR) or with high insulin resistance (MO-high-IR), and with type 2 diabetes treated with metformin (MO-metf-T2DM)), and the effect of insulin/leptin on intestinal epithelial cells (IEC). Insulin receptor substrate-1 (IRS1) and the catalytic p110ß subunit (p110ß) of phosphatidylinositol 3-kinase (PI3K) were higher in MO-high-IR than in MO-low-IR. The regulatory p85α subunit of PI3K (p85α)/p110ß ratio was lower in MO-high-IR and MO-metf-T2DM than in MO-low-IR. Akt-phosphorylation in Ser473 was reduced in MO-high-IR compared with MO-low-IR. IRS1 and p110-ß were associated with insulin and leptin levels. The improvement of body mass index (BMI) and HOMA-IR (homeostasis model assessment of insulin resistance index) after bariatric surgery was associated with a higher IRS1 and a lower p85α/p110ß ratio. IEC (intestinal epithelial cells) incubation with a high glucose + insulin dose produced an increase of p85α and p110ß. High dose of leptin produced an increase of IRS1, p85α and p110ß. In conclusion, despite the existence of insulin resistance, the jejunal expression of genes involved in insulin signalling was increased in MO-high-IR. Their expressions were regulated mainly by leptin. IRS1 and p85α/p110ß ratio was associated with the evolution of insulin resistance after bariatric surgery.

7.
Mol Metab ; 31: 14-23, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31918916

RESUMO

OBJECTIVE: Roux-en-Y gastric surgery (RYGB) promotes a rapid and sustained weight loss and amelioration of glucose control in obese patients. A high number of molecular hypotheses were previously tested using duodenal-jejunal bypass (DJB) performed in various genetic models of mice with knockouts for various hormones or receptors. The data were globally negative or inconsistent. Therefore, the mechanisms remained elusive. Intestinal gluconeogenesis is a gut function that has been suggested to contribute to the metabolic benefits of RYGB in obese patients. METHODS: We studied the effects of DJB on body weight and glucose control in obese mice fed a high fat-high sucrose diet. Wild type mice and mice with a genetic suppression of intestinal gluconeogenesis were studied in parallel using glucose- and insulin-tolerance tests. Fecal losses, including excretion of lipids, were studied from the feces recovered in metabolic cages. RESULTS: DJB induced a dramatic decrease in body weight and improvement in glucose control (glucose- and insulin-tolerance) in obese wild type mice fed a high calorie diet, for 25 days after the surgery. The DJB-induced decrease in food intake was transient and resumed to normal in 7-8 days, suggesting that decreased food intake could not account for the benefits. Total fecal losses were about 5 times and lipid losses 7 times higher in DJB-mice than in control (sham-operated and pair-fed) mice, and could account for the weight loss of mice. The results were comparable in mice with suppression of intestinal gluconeogenesis. There was no effect of DJB on food intake, body weight or fecal loss in lean mice fed a normal chow diet. CONCLUSIONS: DJB in obese mice fed a high calorie diet promotes dramatic fecal loss, which could account for the dramatic weight loss and metabolic benefits observed. This could dominate the effects of the mouse genotype/phenotype. Thus, fecal energy loss should be considered as an essential process contributing to the metabolic benefits of DJB in obese mice.


Assuntos
Derivação Gástrica , Obesidade/metabolismo , Obesidade/cirurgia , Animais , Peso Corporal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Redução de Peso
8.
Metabolites ; 9(12)2019 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-31756997

RESUMO

: Cells efficiently adjust their metabolism according to the abundance of nutrients and energy. The ability to switch cellular metabolism between anabolic and catabolic processes is critical for cell growth. Glucose-6 phosphate is the first intermediate of glucose metabolism and plays a central role in the energy metabolism of the liver. It acts as a hub to metabolically connect glycolysis, the pentose phosphate pathway, glycogen synthesis, de novo lipogenesis, and the hexosamine pathway. In this review, we describe the metabolic fate of glucose-6 phosphate in a healthy liver and the metabolic reprogramming occurring in two pathologies characterized by a deregulation of glucose homeostasis, namely type 2 diabetes, which is characterized by fasting hyperglycemia; and glycogen storage disease type I, where patients develop severe hypoglycemia during short fasting periods. In these two conditions, dysfunction of glucose metabolism results in non-alcoholic fatty liver disease, which may possibly lead to the development of hepatic tumors. Moreover, we also emphasize the role of the transcription factor carbohydrate response element-binding protein (ChREBP), known to link glucose and lipid metabolisms. In this regard, comparing these two metabolic diseases is a fruitful approach to better understand the key role of glucose-6 phosphate in liver metabolism in health and disease.

9.
Sci Rep ; 7: 44856, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28332577

RESUMO

Roux-en-Y gastric bypass (RYGB) induces remission or substantial improvement of type 2 diabetes mellitus (T2D) but underlying mechanisms are still unclear. The beneficial effects of dietary proteins on energy and glucose homeostasis are mediated by the antagonist effects of peptides toward mu-opioid receptors (MORs), which are highly expressed in the distal gut. We hypothesized that the beneficial effects of RYGB could depend at least in part on the interaction of peptides from food with intestinal MORs. Duodenal-jejunal bypass (DJB) was performed in obese and lean wild-type (WT) or MOR deficient (MOR-/-) mice. Food intake and body weight was monitored daily during 3 weeks. Glucose homeostasis was assessed from glucose and insulin tolerance tests. In obese WT and MOR-/- mice, DJB induced a rapid and sustained weight loss partly independent of food intake, and a rapid improvement in glycaemic parameters. Weight loss was a major determinant of the improvements observed. In lean WT and MOR-/- mice, DJB had no effect on weight loss but significantly enhanced glucose tolerance. We found that MORs are not essential in the metabolic beneficial effects of DJB, suggesting that protein sensing in the distal gut is not a link in the metabolic benefits of gastric surgery.


Assuntos
Proteínas Alimentares/metabolismo , Duodeno/fisiologia , Derivação Gástrica , Jejuno/fisiologia , Animais , Metabolismo Energético , Derivação Gástrica/métodos , Masculino , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Redução de Peso
10.
J Thorac Oncol ; 12(4): 724-733, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28034829

RESUMO

INTRODUCTION: Malignant mesothelioma is a deadly disease that is strongly associated with asbestos exposure. Peritoneal mesotheliomas account for 10% of all the cases. BRCA1 associated protein 1 (BAP1) is a deubiquitinating hydrolase that plays a key role in various cellular processes. Germline and somatic inactivation of BRCA1 associated protein 1 gene (BAP1) is frequent in pleural mesothelioma; however, little is known about its status in peritoneal mesothelioma. METHODS: Taking advantage of the extensive French National Network for the Diagnosis of Malignant Pleural Mesothelioma and Rare Peritoneal Tumors and the French National Network for the Treatment of Rare Peritoneal Surface Malignancies, we collected biological material and clinical and epidemiological data for 46 patients with peritoneal mesothelioma. The status of BAP1 was evaluated at the mutational and protein expression levels and combined with our previous data on copy number alterations assessed in the same samples. RESULTS: We detected mutations in 32% of the malignant peritoneal mesotheliomas analyzed. In addition, we have previously reported that copy number losses occurred in 42% of the samples included in this series. Overall, 73% of the malignant peritoneal mesotheliomas analyzed carried at least one inactivated BAP1 allele, but only 57% had a complete loss of its protein nuclear expression. Better overall survival was observed for patients with BAP1 mutations (p = 0.04), protein expression loss (p = 0.016), or at least one of these alterations (p = 0.007) independently of tumor histological subtype, age, and sex. CONCLUSIONS: As in pleural mesothelioma, inactivation of BAP1 is frequent in peritoneal mesotheliomas. We found that BAP1 protein nuclear expression is a good prognostic factor and a more reliable marker for the complete loss of BAP1 activity than mutation or copy number loss.


Assuntos
Variações do Número de Cópias de DNA , Neoplasias Pulmonares/patologia , Mesotelioma/patologia , Mutação , Neoplasias Peritoneais/patologia , Neoplasias Pleurais/patologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Adolescente , Adulto , Idoso , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Feminino , Seguimentos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Mesotelioma/genética , Mesotelioma/metabolismo , Mesotelioma Maligno , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/metabolismo , Neoplasias Pleurais/genética , Neoplasias Pleurais/metabolismo , Prognóstico , Taxa de Sobrevida , Adulto Jovem
12.
J Proteome Res ; 15(4): 1342-9, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26958868

RESUMO

The excessive endogenous glucose production (EGP) induced by glucagon participates in the development of type 2 diabetes. To further understand this hormonal control, we studied the short-term regulation by cyclic adenosine monophosphate (cAMP) of the glucose-6-phosphatase (G6Pase) enzyme, which catalyzes the last reaction of EGP. In gluconeogenic cell models, a 1-h treatment by the adenylate cyclase activator forskolin increased G6Pase activity and glucose production independently of any change in enzyme protein amount or G6P content. Using specific inhibitors or protein overexpression, we showed that the stimulation of G6Pase activity involved the protein kinase A (PKA). Results of site-directed mutagenesis, mass spectrometry analyses, and in vitro phosphorylation experiments suggested that the PKA stimulation of G6Pase activity did not depend on a direct phosphorylation of the enzyme. However, the temperature-dependent induction of both G6Pase activity and glucose release suggested a membrane-based mechanism. G6Pase is composed of a G6P transporter (G6PT) and a catalytic unit (G6PC). Surprisingly, we demonstrated that the increase in G6PT activity was required for the stimulation of G6Pase activity by forskolin. Our data demonstrate the existence of a post-translational mechanism that regulates G6Pase activity and reveal the key role of G6PT in the hormonal regulation of G6Pase activity and of EGP.


Assuntos
Antiporters/genética , AMP Cíclico/farmacologia , Células Epiteliais/efeitos dos fármacos , Glucagon/farmacologia , Glucose-6-Fosfatase/genética , Glucose/biossíntese , Proteínas de Transporte de Monossacarídeos/genética , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Animais , Antiporters/metabolismo , Células CACO-2 , Linhagem Celular , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Glucose-6-Fosfatase/metabolismo , Glucose-6-Fosfato/metabolismo , Células Hep G2 , Humanos , Proteínas de Transporte de Monossacarídeos/metabolismo , Mutagênese Sítio-Dirigida , Fosforilação , Biossíntese de Proteínas , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Transdução de Sinais
13.
J Inherit Metab Dis ; 38(3): 521-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25164786

RESUMO

Patients with glycogen storage diseases type 1 (GSD1) suffer from life-threatening hypoglycaemia, when left untreated. Despite an intensive dietary treatment, patients develop severe complications, such as liver tumors and renal failure, with aging. Until now, the animal models available for studying the GSD1 did not survive after weaning. To gain further insights into the molecular mechanisms of the disease and to evaluate potential treatment strategies, we have recently developed novel mouse models in which the catalytic subunit of glucose-6 phosphatase (G6pc) is deleted in each glucose-producing organ specifically. For that, B6.G6pc(ex3lox/ex3lox) mice were crossed with transgenic mice expressing a recombinase under the control of the serum albumin, the kidney androgen protein or the villin promoter, in order to obtain liver, kidney or intestine G6pc(-/-) mice, respectively. As opposed to total G6pc knockout mice, tissue-specific G6pc deficiency allows mice to maintain their blood glucose by inducing glucose production in the other gluconeogenic organs. Even though it is considered that glucose is produced mainly by the liver, liver G6pc(-/-) mice are perfectly viable and exhibit the same hepatic pathological features as GSD1 patients, including the late development of hepatocellular adenomas and carcinomas. Interestingly, renal G6pc(-/-) mice developed renal symptoms similar to the early human GSD1 nephropathy. This includes glycogen overload that leads to nephromegaly and morphological and functional alterations in the kidneys. Thus, our data suggest that renal G6Pase deficiency per se is sufficient to induce the renal pathology of GSD1. Therefore, these new mouse models should allow us to improve the strategies of treatment on both nutritional and pharmacological points of view.


Assuntos
Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/genética , Neoplasias Hepáticas/etiologia , Insuficiência Renal/etiologia , Animais , Modelos Animais de Doenças , Cães , Terapia Genética , Doença de Depósito de Glicogênio Tipo I/terapia , Humanos , Hipoglicemia/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Especificidade de Órgãos , Fatores de Tempo
14.
Diabetes ; 61(10): 2451-60, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22787137

RESUMO

The activation of glucose-6-phosphatase (G6Pase), a key enzyme of endogenous glucose production, is correlated with type 2 diabetes. Type 2 diabetes is characterized by sustained hyperglycemia leading to glucotoxicity. We investigated whether glucotoxicity mechanisms control the expression of the G6Pase catalytic unit (G6pc). We deciphered the transcriptional regulatory mechanisms of the G6pc promoter by glucotoxicity in a hepatoma cell line then in primary hepatocytes and in the liver of diabetic mice. High glucose exposure induced the production of reactive oxygen species (ROS) and, in parallel, induced G6pc promoter activity. In hepatocytes, glucose induced G6pc gene expression and glucose release. The decrease of ROS concentrations by antioxidants eliminated all the glucose-inductive effects. The induction of G6pc promoter activity by glucose was eliminated in the presence of small interfering RNA, targeting either the hypoxia-inducible factor (HIF)-1α or the CREB-binding protein (CBP). Glucose increased the interaction of HIF-1α with CBP and the recruitment of HIF-1 on the G6pc promoter. The same mechanism might occur in hyperglycemic mice. We deciphered a new regulatory mechanism induced by glucotoxicity. This mechanism leading to the induction of HIF-1 transcriptional activity may contribute to the increase of hepatic glucose production during type 2 diabetes.


Assuntos
Proteína de Ligação a CREB/metabolismo , Glucose-6-Fosfatase/metabolismo , Glucose/toxicidade , Hepatócitos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fígado/metabolismo , Animais , Proteína de Ligação a CREB/genética , Linhagem Celular , Células Cultivadas , Glucose-6-Fosfatase/genética , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Fígado/citologia , Fígado/efeitos dos fármacos , Camundongos , Regiões Promotoras Genéticas , Espécies Reativas de Oxigênio/metabolismo
15.
Biochimie ; 94(3): 695-703, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21983240

RESUMO

Glucose-6 phosphatase (G6Pase), a key enzyme of glucose homeostasis, catalyses the hydrolysis of glucose-6 phosphate (G6P) to glucose and inorganic phosphate. A deficiency in G6Pase activity causes type 1 glycogen storage disease (GSD-1), mainly characterised by hypoglycaemia. Genetic analyses of the two forms of this rare disease have shown that the G6Pase system consists of two proteins, a catalytic subunit (G6PC) responsible for GSD-1a, and a G6P translocase (G6PT), responsible for GSD-1b. However, since their identification, few investigations concerning their structural relationship have been made. In this study, we investigated the localisation and membrane organisation of the G6Pase complex. To this aim, we developed chimera proteins by adding a fluorescent protein to the C-terminal ends of both subunits. The G6PC and G6PT fluorescent chimeras were both addressed to perinuclear membranes as previously suggested, but also to vesicles throughout the cytoplasm. We demonstrated that both proteins strongly colocalised in perinuclear membranes. Then, we studied G6PT organisation in the membrane. We highlighted FRET between the labelled C and N termini of G6PT. The intramolecular FRET of this G6PT chimera was 27%. The coexpression of unlabelled G6PC did not modify this FRET intensity. Finally, the chimera constructs generated in this work enabled us for the first time to analyze the relationship between GSD-1 mutations and the intracellular localisation of both G6Pase subunits. We showed that GSD1 mutations did neither alter the G6PC or G6PT chimera localisation, nor the interaction between G6PT termini. In conclusion, our results provide novel information on the intracellular distribution and organisation of the G6Pase complex.


Assuntos
Glucose-6-Fosfatase/metabolismo , Subunidades Proteicas/metabolismo , Antiporters/química , Antiporters/genética , Antiporters/metabolismo , Transferência Ressonante de Energia de Fluorescência , Glucose-6-Fosfatase/química , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/enzimologia , Doença de Depósito de Glicogênio Tipo I/genética , Células HeLa , Células Hep G2 , Humanos , Immunoblotting , Imuno-Histoquímica , Membranas Intracelulares , Proteínas de Transporte de Monossacarídeos/química , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
J Hepatol ; 54(3): 529-37, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21109326

RESUMO

BACKGROUND AND AIMS: Glycogen storage disease type 1a (GSD1a) is an inherited disease caused by a deficiency in the catalytic subunit of the glucose-6 phosphatase enzyme (G6Pase). GSD1a is characterized by hypoglycaemia, hyperlipidemia, and lactic acidosis with associated hepatic (including hepatocellular adenomas), renal, and intestinal disorders. A total G6pc (catalytic subunit of G6Pase) knock-out mouse model has been generated that mimics the human pathology. However, these mice rarely live longer than 3 months and long-term liver pathogenesis cannot be evaluated. Herein, we report the long-term characterization of a liver-specific G6pc knock-out mouse model (L-G6pc(-/-)). METHODS: We generated L-G6pc(-/-) mice using an inducible CRE-lox strategy and followed up the development of hepatic tumours using magnetic resonance imaging. RESULTS: L-G6pc(-/-) mice are viable and exhibit normoglycemia in the fed state. They develop hyperlipidemia, lactic acidosis, and uricemia during the first month after gene deletion. However, these plasmatic parameters improved after 6 months. L-G6pc(-/-) mice develop hepatomegaly with glycogen accumulation and hepatic steatosis. Using an MRI approach, we could detect hepatic nodules with diameters of less than 1 mm, 9 months after induction of deficiency. Hepatic nodules (1 mm) were detected in 30-40% of L-G6pc(-/-) mice at 12 months. After 18 months, all L-G6pc(-/-) mice developed multiple hepatocellular adenomas of 1-10 mm diameter. CONCLUSIONS: This is the first report of a viable animal model of the hepatic pathology of GSD1a, including the late development of hepatocellular adenomas.


Assuntos
Adenoma de Células Hepáticas/etiologia , Glucose-6-Fosfatase/antagonistas & inibidores , Glucose-6-Fosfatase/genética , Neoplasias Hepáticas Experimentais/etiologia , Fígado/enzimologia , Adenoma de Células Hepáticas/enzimologia , Adenoma de Células Hepáticas/patologia , Animais , Sequência de Bases , Primers do DNA , Modelos Animais de Doenças , Fígado Gorduroso/enzimologia , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Feminino , Técnicas de Inativação de Genes , Marcação de Genes , Doença de Depósito de Glicogênio Tipo I/enzimologia , Doença de Depósito de Glicogênio Tipo I/etiologia , Doença de Depósito de Glicogênio Tipo I/genética , Hepatomegalia/enzimologia , Hepatomegalia/etiologia , Hepatomegalia/patologia , Humanos , Neoplasias Hepáticas Experimentais/enzimologia , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica
17.
J Biol Chem ; 281(42): 31268-78, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16893891

RESUMO

Gluconeogenesis is induced in both the liver and intestine by increased cAMP levels. However, hepatic and intestinal glucose production can have opposite effects on glucose homeostasis. Glucose release into the portal vein by the intestine increases glucose uptake and reduces food intake. In contrast, glucose production by the liver contributes to hyperglycemia in type II diabetes. Glucose-6-phosphatase (Glc6Pase) is the key enzyme of gluconeogenesis in both the liver and intestine. Here we specify the cAMP/protein kinase A regulation of the Glc6Pase gene in the intestine compared with the liver. Similarly to the liver, the molecular mechanism of cAMP/protein kinase A regulation involves cAMP-response element-binding protein, HNF4alpha, CAAT/enhancer-binding protein, and HNF1. In contrast to the situation in the liver, we find that different isoforms of CAAT/enhancer-binding protein and HNF1 contribute to the specific regulation of the Glc6Pase gene in the intestine. Moreover, we show that cAMP-response element binding modulator specifically contributes to the regulation of the Glc6Pase gene in the intestine but not in the liver. These results allow us to identify intestine-specific regulators of the Glc6Pase gene and to improve the understanding of the differences in the regulation of gluconeogenesis in the intestine compared with the liver.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/fisiologia , Modulador de Elemento de Resposta do AMP Cíclico/fisiologia , AMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica , Glucose-6-Fosfatase/biossíntese , Glucose-6-Fosfatase/genética , Fator 1-alfa Nuclear de Hepatócito/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Mucosa Intestinal/metabolismo , Peptídeo Intestinal Vasoativo/química , Animais , Sequência de Bases , Células CACO-2 , Humanos , Intestinos/enzimologia , Masculino , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley
18.
Cell Metab ; 2(5): 321-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16271532

RESUMO

Protein feeding is known to decrease hunger and subsequent food intake in animals and humans. It has also been suggested that glucose appearance into portal vein, as occurring during meal assimilation, may induce comparable effects. Here, we connect these previous observations by reporting that intestinal gluconeogenesis (i.e., de novo synthesis of glucose) is induced during the postabsorptive time (following food digestion) in rats specifically fed on protein-enriched diet. This results in glucose release into portal blood, counterbalancing the lowering of glycemia resulting from intestinal glucose utilization. Comparable infusions into the portal vein of control postabsorptive rats (fed on starch-enriched diet) decrease food consumption and activate the hypothalamic nuclei regulating food intake. Similar hypothalamic activation occurs on protein feeding. All these effects are absent after denervation of the portal vein. Thus, portal sensing of intestinal gluconeogenesis may be a novel mechanism connecting the macronutrient composition of diet to food intake.


Assuntos
Proteínas Alimentares , Ingestão de Alimentos , Gluconeogênese , Glucose-6-Fosfatase/biossíntese , Glutaminase/biossíntese , Intestino Delgado/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/biossíntese , Animais , Comportamento Animal , Carboidratos da Dieta , Indução Enzimática , Glucose/metabolismo , Hipotálamo/metabolismo , Veia Porta/inervação , Veia Porta/metabolismo , Período Pós-Prandial , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Fatores de Tempo
19.
Mol Endocrinol ; 19(1): 163-74, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15388792

RESUMO

Glucose-6-phosphatase (Glc6Pase) is the last enzyme of gluconeogenesis and is only expressed in the liver, kidney, and small intestine. In these tissues, the mRNA and its activity are increased when cAMP levels increased (e.g. in fasting or diabetes). We first report that a proximal region (within -200 bp relative to the transcription start site) and a distal region (-694/-500 bp) are both required for a potent cAMP and a protein kinase A (PKA) responsiveness of the Glc6Pase promoter. Using different molecular approaches, we demonstrate that hepatocyte nuclear factor (HNF4alpha), CAAT/enhancer-binding protein-alpha (C/EBPalpha), C/EBPbeta, and cAMP response element-binding protein (CREB) are involved in the potentiated PKA responsiveness: in the distal region, via one HNF4alpha- and one C/EBP-binding sites, and in the proximal region, via two HNF4alpha and two CREB-binding sites. We also show that HNF4alpha, C/EBPalpha, and C/EBPbeta are constitutively bound to the endogenous Glc6Pase gene, whereas CREB and CREB-binding protein (CBP) will be bound to the gene upon stimulation by cAMP. These data strongly suggest that the cAMP responsiveness of the Glc6Pase promoter requires a tight cooperation between a proximal and a distal region, which depends on the presence of several HNF4alpha-, C/EBP-, and CREB-binding sites, therefore involving an intricate association of hepatic and ubiquitous transcription factors.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Glucose-6-Fosfatase/genética , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT/genética , Linhagem Celular Tumoral , AMP Cíclico/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fator 4 Nuclear de Hepatócito , Humanos , Fosfoproteínas/genética , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos , Elementos de Resposta/genética , Fatores de Transcrição/genética
20.
Nucleic Acids Res ; 31(18): 5238-46, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12954759

RESUMO

Glucose-6-phosphatase (Glc6Pase), the last enzyme of gluconeogenesis, is only expressed in the liver, kidney and small intestine. The expression of the Glc6Pase gene exhibits marked specificities in the three tissues in various situations, but the molecular basis of the tissue specificity is not known. The presence of a consensus binding site of CDX proteins in the minimal Glc6Pase gene promoter has led us to consider the hypothesis that these intestine-specific CDX factors could be involved in the Glc6Pase-specific expression in the small intestine. We first show that the Glc6Pase promoter is active in both hepatic HepG2 and intestinal CaCo2 cells. Using gel shift mobility assay, mutagenesis and competition experiments, we show that both CDX1 and CDX2 can bind the minimal promoter, but only CDX1 can transactivate it. Consistently, intestinal IEC6 cells stably overexpressing CDX1 exhibit induced expression of the Glc6Pase protein. We demonstrate that a TATAAAA sequence, located in position -31/-25 relating to the transcription start site, exhibits separable functions in the preinitiation of transcription and the transactivation by CDX1. Disruption of this site dramatically suppresses both basal transcription and the CDX1 effect. The latter may be restored by inserting a couple of CDX- binding sites in opposite orientation similar to that found in the sucrase-isomaltase promoter. We also report that the specific stimulatory effect of CDX1 on the Glc6Pase TATA-box, compared to CDX2, is related to the fact that CDX1, but not CDX2, can interact with the TATA-binding protein. Together, these data strongly suggest that CDX proteins could play a crucial role in the specific expression of the Glc6Pase gene in the small intestine. They also suggest that CDX transactivation might be essential for intestine gene expression, irrespective of the presence of a functional TATA box.


Assuntos
Glucose-6-Fosfatase/genética , Proteínas de Homeodomínio/metabolismo , TATA Box/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Sequência de Bases , Sítios de Ligação/genética , Fator de Transcrição CDX2 , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Regulação Enzimológica da Expressão Gênica , Células HeLa , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Mutação , Plasmídeos/genética , Ligação Proteica , Ratos , Transativadores , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA