Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Metab ; 39: 101007, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32360426

RESUMO

OBJECTIVE: Obesity is characterized by systemic and low-grade tissue inflammation. In the intestine, alteration of the intestinal barrier and accumulation of inflammatory cells in the epithelium are important contributors of gut inflammation. Recent studies demonstrated the role of the aryl hydrocarbon receptor (AhR) in the maintenance of immune cells at mucosal barrier sites. A wide range of ligands of external and local origin can activate this receptor. We studied the causal relationship between AhR activation and gut inflammation in obesity. METHODS: Jejunum samples from subjects with normal weight and severe obesity were phenotyped according to T lymphocyte infiltration in the epithelium from lamina propria and assayed for the mRNA level of AhR target genes. The effect of an AhR agonist was studied in mice and Caco-2/TC7 cells. AhR target gene expression, permeability to small molecules and ions, and location of cell-cell junction proteins were recorded under conditions of altered intestinal permeability. RESULTS: We showed that a low AhR tone correlated with a high inflammatory score in the intestinal epithelium in severe human obesity. Moreover, AhR activation protected junctional complexes in the intestinal epithelium in mice challenged by an oral lipid load. AhR ligands prevented chemically induced damage to barrier integrity and cytokine expression in Caco-2/TC7 cells. The PKC and p38MAPK signaling pathways were involved in this AhR action. CONCLUSIONS: The results of these series of human, mouse, and cell culture experiments demonstrate the protective effect of AhR activation in the intestine targeting particularly tight junctions and cytokine expression. We propose that AhR constitutes a valuable target to protect intestinal functions in metabolic diseases, which can be achieved in the future via food or drug ligands.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Mucosa Intestinal/metabolismo , Obesidade/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Adiposidade/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Biomarcadores , Linhagem Celular , Comorbidade , Citocinas/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Jejuno/metabolismo , Metabolismo dos Lipídeos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Biológicos , Obesidade/etiologia , Obesidade/patologia , Permeabilidade , Receptores de Hidrocarboneto Arílico/genética , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
2.
J Pathol ; 246(2): 217-230, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29984492

RESUMO

Obesity and its metabolic complications are characterized by subclinical systemic and tissue inflammation. In rodent models of obesity, inflammation and metabolic impairments are linked with intestinal barrier damage. However, whether intestinal permeability is altered in human obesity remains to be investigated. In a cohort of 122 severely obese and non-obese patients, we analyzed intestinal barrier function combining in vivo and ex vivo investigations. We found tight junction impairments in the jejunal epithelium of obese patients, evidenced by a reduction of occludin and tricellulin. Serum levels of zonulin and LPS binding protein, two markers usually associated with intestinal barrier alterations, were also increased in obese patients. Intestinal permeability per se was assessed in vivo by quantification of urinary lactitol/mannitol (L/M) and measured directly ex vivo on jejunal samples in Ussing chambers. In the fasting condition, L/M ratio and jejunal permeability were not significantly different between obese and non-obese patients, but high jejunal permeability to small molecules (0.4 kDa) was associated with systemic inflammation within the obese cohort. Altogether, these results suggest that intestinal barrier function is subtly compromised in obese patients. We thus tested whether this barrier impairment could be exacerbated by dietary lipids. To this end, we challenged jejunal samples with lipid micelles and showed that a single exposure increased permeability to macromolecules (4 kDa). Jejunal permeability after the lipid load was two-fold higher in obese patients compared to non-obese controls and correlated with systemic and intestinal inflammation. Moreover, lipid-induced permeability was an explicative variable of type 2 diabetes. In conclusion, intestinal barrier defects are present in human severe obesity and exacerbated by a lipid challenge. This paves the way to the development of novel therapeutic approaches to modulate intestinal barrier function or personalize nutrition therapy to decrease lipid-induced jejunal leakage in metabolic diseases. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Inflamação/metabolismo , Absorção Intestinal/efeitos dos fármacos , Jejuno/efeitos dos fármacos , Lipídeos/administração & dosagem , Obesidade/metabolismo , Proteínas de Fase Aguda , Adulto , Idoso , Células CACO-2 , Proteínas de Transporte/sangue , Estudos de Casos e Controles , Toxina da Cólera/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/fisiopatologia , Feminino , Haptoglobinas , Humanos , Inflamação/complicações , Inflamação/fisiopatologia , Jejuno/metabolismo , Jejuno/fisiopatologia , Proteína 2 com Domínio MARVEL/metabolismo , Masculino , Glicoproteínas de Membrana/sangue , Micelas , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/fisiopatologia , Ocludina/metabolismo , Permeabilidade , Precursores de Proteínas , Junções Íntimas/metabolismo , Adulto Jovem
3.
Mol Metab ; 6(1): 61-72, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28123938

RESUMO

OBJECTIVE: Intestinal glucose absorption is orchestrated by specialized glucose transporters such as SGLT1 and GLUT2. However, the role of GLUT2 in the regulation of glucose absorption remains to be fully elucidated. METHODS: We wanted to evaluate the role of GLUT2 on glucose absorption and glucose homeostasis after intestinal-specific deletion of GLUT2 in mice (GLUT2ΔIEC mice). RESULTS: As anticipated, intestinal GLUT2 deletion provoked glucose malabsorption as visualized by the delay in the distribution of oral sugar in tissues. Consequences of intestinal GLUT2 deletion in GLUT2ΔIEC mice were limiting body weight gain despite normal food intake, improving glucose tolerance, and increasing ketone body production. These features were reminiscent of calorie restriction. Other adaptations to intestinal GLUT2 deletion were reduced microvillus length and altered gut microbiota composition, which was associated with improved inflammatory status. Moreover, a reduced density of glucagon-like peptide-1 (GLP-1) positive cells was compensated by increased GLP-1 content per L-cell, suggesting a preserved enteroendocrine function in GLUT2ΔIEC mice. CONCLUSIONS: Intestinal GLUT2 modulates glucose absorption and constitutes a control step for the distribution of dietary sugar to tissues. Consequently, metabolic and gut homeostasis are improved in the absence of functional GLUT2 in the intestine, thus mimicking calorie restriction.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Transportador de Glucose Tipo 2/metabolismo , Glucose/metabolismo , Animais , Glicemia/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/fisiologia , Homeostase , Absorção Intestinal , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Transportador 1 de Glucose-Sódio/metabolismo , Distribuição Tecidual
4.
Med Sci (Paris) ; 32(5): 461-9, 2016 May.
Artigo em Francês | MEDLINE | ID: mdl-27225918

RESUMO

The increasing incidence of obesity and associated metabolic complications is a worldwide public health issue. The role of the gut in the pathophysiology of obesity, with an important part for microbiota, is becoming obvious. In rodent models of diet-induced obesity, the modifications of gut microbiota are associated with an alteration of the intestinal permeability increasing the passage of food or bacterial antigens, which contribute to low-grade inflammation and insulin resistance. In human obesity, intestinal permeability modification, and its role in the crosstalk between gut microbiota changes and inflammation at systemic and tissular levels, are still poorly documented. Hence, further characterization of the triggering mechanisms of such inflammatory responses in obese subjects could enable the development of personalized intervention strategies that will help to reduce the risk of obesity-associated diseases.


Assuntos
Disbiose/complicações , Microbioma Gastrointestinal/fisiologia , Inflamação/etiologia , Mucosa Intestinal/metabolismo , Obesidade/etiologia , Animais , Disbiose/imunologia , Disbiose/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Mucosa Intestinal/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Obesidade/imunologia , Obesidade/metabolismo , Obesidade/microbiologia , Permeabilidade
6.
J Nutr Sci ; 4: e22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26157580

RESUMO

The enterohormone glucagon-like peptide-1 (GLP-1) is required to amplify glucose-induced insulin secretion that facilitates peripheral glucose utilisation. Alteration in GLP-1 secretion during obesity has been reported but is still controversial. Due to the high adaptability of intestinal cells to environmental changes, we hypothesised that the density of GLP-1-producing cells could be modified by nutritional factors to prevent the deterioration of metabolic condition in obesity. We quantified L-cell density in jejunum samples collected during Roux-en-Y gastric bypass in forty-nine severely obese subjects analysed according to their fat consumption. In mice, we deciphered the mechanisms by which a high-fat diet (HFD) makes an impact on enteroendocrine cell density and function. L-cell density in the jejunum was higher in obese subjects consuming >30 % fat compared with low fat eaters. Mice fed a HFD for 8 weeks displayed an increase in GLP-1-positive cells in the jejunum and colon accordingly to GLP-1 secretion. The regulation by the HFD appears specific to GLP-1-producing cells, as the number of PYY (peptide YY)-positive cells remained unchanged. Moreover, genetically obese ob/ob mice did not show alteration of GLP-1-positive cell density in the jejunum or colon, suggesting that obesity per se is not sufficient to trigger the mechanism. The higher L-cell density in HFD-fed mice involved a rise in L-cell terminal differentiation as witnessed by the increased expression of transcription factors downstream of neurogenin3 (Ngn3). We suggest that the observed increase in GLP-1-positive cell density triggered by high fat consumption in humans and mice might favour insulin secretion and therefore constitute an adaptive response of the intestine to balance diet-induced insulin resistance.

7.
Cell Metab ; 22(1): 113-24, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26094890

RESUMO

In obesity, insulin resistance is linked to inflammation in several tissues. Although the gut is a very large lymphoid tissue, inflammation in the absorptive small intestine, the jejunum, where insulin regulates lipid and sugar absorption is unknown. We analyzed jejunal samples of 185 obese subjects stratified in three metabolic groups: without comorbidity, suffering from obesity-related comorbidity, and diabetic, versus 33 lean controls. Obesity increased both mucosa surface due to lower cell apoptosis and innate and adaptive immune cell populations. The preferential CD8αß T cell location in epithelium over lamina propria appears a hallmark of obesity. Cytokine secretion by T cells from obese, but not lean, subjects blunted insulin signaling in enterocytes relevant to apical GLUT2 mislocation. Statistical links between T cell densities and BMI, NAFLD, or lipid metabolism suggest tissue crosstalk. Obesity triggers T-cell-mediated inflammation and enterocyte insulin resistance in the jejunum with potential broader systemic implications.


Assuntos
Enterócitos/patologia , Inflamação/complicações , Insulina/imunologia , Jejuno/patologia , Obesidade/complicações , Linfócitos T/patologia , Adulto , Antígenos CD8/imunologia , Células Cultivadas , Enterócitos/imunologia , Feminino , Transportador de Glucose Tipo 2/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Resistência à Insulina , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Jejuno/citologia , Jejuno/imunologia , Masculino , Pessoa de Meia-Idade , Obesidade/imunologia , Obesidade/patologia , Transdução de Sinais , Linfócitos T/imunologia
8.
J Biol Chem ; 288(43): 31080-92, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23986439

RESUMO

The structure-function relationships of sugar transporter-receptor hGLUT2 coded by SLC2A2 and their impact on insulin secretion and ß cell differentiation were investigated through the detailed characterization of a panel of mutations along the protein. We studied naturally occurring SLC2A2 variants or mutants: two single-nucleotide polymorphisms and four proposed inactivating mutations associated to Fanconi-Bickel syndrome. We also engineered mutations based on sequence alignment and conserved amino acids in selected domains. The single-nucleotide polymorphisms P68L and T110I did not impact on sugar transport as assayed in Xenopus oocytes. All the Fanconi-Bickel syndrome-associated mutations invalidated glucose transport by hGLUT2 either through absence of protein at the plasma membrane (G20D and S242R) or through loss of transport capacity despite membrane targeting (P417L and W444R), pointing out crucial amino acids for hGLUT2 transport function. In contrast, engineered mutants were located at the plasma membrane and able to transport sugar, albeit with modified kinetic parameters. Notably, these mutations resulted in gain of function. G20S and L368P mutations increased insulin secretion in the absence of glucose. In addition, these mutants increased insulin-positive cell differentiation when expressed in cultured rat embryonic pancreas. F295Y mutation induced ß cell differentiation even in the absence of glucose, suggesting that mutated GLUT2, as a sugar receptor, triggers a signaling pathway independently of glucose transport and metabolism. Our results describe the first gain of function mutations for hGLUT2, revealing the importance of its receptor versus transporter function in pancreatic ß cell development and insulin secretion.


Assuntos
Diferenciação Celular/fisiologia , Transportador de Glucose Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Substituição de Aminoácidos , Animais , Transporte Biológico Ativo/genética , Linhagem Celular Tumoral , Glucose/genética , Glucose/metabolismo , Transportador de Glucose Tipo 2/genética , Humanos , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Camundongos , Ratos , Transdução de Sinais , Xenopus laevis
9.
Prog Mol Biol Transl Sci ; 108: 113-27, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22656375

RESUMO

Carbohydrates represent more than 50% of the energy sources present in most human diets. Sugar intake is regulated by metabolic, neuronal, and hedonic factors, and gene polymorphisms are involved in determining sugar preference. Nutrigenomic adaptations to carbohydrate availability have been evidenced in metabolic diseases, in the persistence of lactose digestion, and in amylase gene copy number. Furthermore, dietary oligosaccharides, fermentable by gut flora, can modulate the microbiotal diversity to the benefit of the host. Genetic diseases linked to mutations in the disaccharidase genes (sucrase-isomaltase, lactase) and in sugar transporter genes (sodium/glucose cotransporter 1, glucose transporters 1 and 2) severely impact carbohydrate intake. These diseases are revealed upon exposure to food containing the offending sugar, and withdrawal of this sugar from the diet prevents disease symptoms, failure to thrive, and premature death. Tailoring the sugar composition of diets to optimize wellness and to prevent the chronic occurrence of metabolic diseases is a future goal that may yet be realized through continued development of nutrigenetics and nutrigenomics approaches.


Assuntos
Carboidratos da Dieta/administração & dosagem , Carboidratos da Dieta/farmacologia , Preferências Alimentares , Regulação da Expressão Gênica/efeitos dos fármacos , Doenças Genéticas Inatas/fisiopatologia , Humanos
10.
Mol Biol Cell ; 23(2): 324-36, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22114352

RESUMO

Actin-bundling proteins are identified as key players in the morphogenesis of thin membrane protrusions. Until now, functional redundancy among the actin-bundling proteins villin, espin, and plastin-1 has prevented definitive conclusions regarding their role in intestinal microvilli. We report that triple knockout mice lacking these microvillar actin-bundling proteins suffer from growth delay but surprisingly still develop microvilli. However, the microvillar actin filaments are sparse and lack the characteristic organization of bundles. This correlates with a highly inefficient apical retention of enzymes and transporters that accumulate in subapical endocytic compartments. Myosin-1a, a motor involved in the anchorage of membrane proteins in microvilli, is also mislocalized. These findings illustrate, in vivo, a precise role for local actin filament architecture in the stabilization of apical cargoes into microvilli. Hence, the function of actin-bundling proteins is not to enable microvillar protrusion, as has been assumed, but to confer the appropriate actin organization for the apical retention of proteins essential for normal intestinal physiology.


Assuntos
Actinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Actinas/ultraestrutura , Animais , Enterócitos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/ultraestrutura , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/ultraestrutura , Microscopia Eletrônica de Transmissão , Microvilosidades/metabolismo , Microvilosidades/ultraestrutura , Cadeias Pesadas de Miosina/metabolismo , Estrutura Terciária de Proteína
11.
Diabetes ; 60(10): 2598-607, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21852673

RESUMO

OBJECTIVE: In healthy rodents, intestinal sugar absorption in response to sugar-rich meals and insulin is regulated by GLUT2 in enterocyte plasma membranes. Loss of insulin action maintains apical GLUT2 location. In human enterocytes, apical GLUT2 location has not been reported but may be revealed under conditions of insulin resistance. RESEARCH DESIGN AND METHODS: Subcellular location of GLUT2 in jejunal enterocytes was analyzed by confocal and electron microscopy imaging and Western blot in 62 well-phenotyped morbidly obese subjects and 7 lean human subjects. GLUT2 locations were assayed in ob/ob and ob/+ mice receiving oral metformin or in high-fat low-carbohydrate diet-fed C57Bl/6 mice. Glucose absorption and secretion were respectively estimated by oral glucose tolerance test and secretion of [U-(14)C]-3-O-methyl glucose into lumen. RESULTS: In human enterocytes, GLUT2 was consistently located in basolateral membranes. Apical GLUT2 location was absent in lean subjects but was observed in 76% of obese subjects and correlated with insulin resistance and glycemia. In addition, intracellular accumulation of GLUT2 with early endosome antigen 1 (EEA1) was associated with reduced MGAT4a activity (glycosylation) in 39% of obese subjects on a low-carbohydrate/high-fat diet. Mice on a low-carbohydrate/high-fat diet for 12 months also exhibited endosomal GLUT2 accumulation and reduced glucose absorption. In ob/ob mice, metformin promoted apical GLUT2 and improved glucose homeostasis. Apical GLUT2 in fasting hyperglycemic ob/ob mice tripled glucose release into intestinal lumen. CONCLUSIONS: In morbidly obese insulin-resistant subjects, GLUT2 was accumulated in apical and/or endosomal membranes of enterocytes. Functionally, apical GLUT2 favored and endosomal GLUT2 reduced glucose transepithelial exchanges. Thus, altered GLUT2 locations in enterocytes are a sign of intestinal adaptations to human metabolic pathology.


Assuntos
Membrana Celular/metabolismo , Gorduras na Dieta/administração & dosagem , Enterócitos/metabolismo , Transportador de Glucose Tipo 2/metabolismo , Obesidade Mórbida/metabolismo , Adulto , Animais , Diabetes Mellitus Tipo 2/metabolismo , Carboidratos da Dieta/administração & dosagem , Carboidratos da Dieta/farmacologia , Transportador de Glucose Tipo 2/genética , Humanos , Hipoglicemiantes/uso terapêutico , Metformina/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Pessoa de Meia-Idade , Adulto Jovem
12.
Am J Physiol Endocrinol Metab ; 298(5): E1078-87, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20179244

RESUMO

The sugar transporter GLUT2, present in several tissues of the gut-brain axis, has been reported to be involved in the control of food intake. GLUT2 is a sugar transporter sustaining energy production in the cell, but it can also function as a receptor for extracellular glucose. A glucose-signaling pathway is indeed triggered, independently of glucose metabolism, through its large cytoplasmic loop domain. However, the contribution of the receptor function over the transporter function of GLUT2 in the control of food intake remains to be determined. Thus, we generated transgenic mice that express a GLUT2-loop domain, blocking the detection of glucose but leaving GLUT2-dependent glucose transport unaffected. Inhibiting GLUT2-mediated glucose detection augmented daily food intake by a mechanism that increased the meal size but not the number of meals. Peripheral hormones (ghrelin, insulin, leptin) were unaffected, leading to a focus on central aspects of feeding behavior. We found defects in c-Fos activation by glucose in the arcuate nucleus and changes in the amounts of TRH and orexin neuropeptide mRNA, which are relevant to poorly controlled meal size. Our data provide evidence that glucose detection by GLUT2 contributes to the control of food intake by the hypothalamus. The sugar transporter receptor, i.e., "transceptor" GLUT2, may constitute a drug target to treat eating disorders and associated metabolic diseases, particularly by modulating its receptor function without affecting vital sugar provision by its transporter function.


Assuntos
Ingestão de Alimentos/fisiologia , Transportador de Glucose Tipo 2/metabolismo , Glucose/metabolismo , Hipotálamo/metabolismo , Análise de Variância , Animais , Transporte Biológico/fisiologia , Peso Corporal/fisiologia , Contagem de Células , Metabolismo Energético , Comportamento Alimentar/fisiologia , Grelina/sangue , Transportador de Glucose Tipo 2/genética , Homeostase/fisiologia , Imuno-Histoquímica , Insulina/sangue , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leptina/sangue , Camundongos , Camundongos Transgênicos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Orexinas , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Estatísticas não Paramétricas , Hormônio Liberador de Tireotropina/genética , Hormônio Liberador de Tireotropina/metabolismo
13.
Am J Physiol Endocrinol Metab ; 296(5): E985-92, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19223655

RESUMO

Cloned 20 years ago, GLUT2 is a facilitative glucose transporter in the liver, pancreas, intestine, kidney, and brain. It ensures large bidirectional fluxes of glucose in and out the cell due to its low affinity and high capacity. It also transports other dietary sugars, such as fructose and galactose, within the range of physiological concentrations. Sugars and hormones regulate its gene expression. The contribution of GLUT2 to human metabolic diseases previously appeared modest. However, in the past decade, three major features of the GLUT2 protein have been revealed. First, GLUT2 mutations cause the severe but rare Fanconi-Bickel syndrome, mainly characterized by glycogenosis. Recently, a GLUT2 polymorphism has been associated with preferences for sugary food. Second, the GLUT2 location at the cell surface is regulated; this governs cellular activities dependent on glucose in the intestine and possibly those in the liver and pancreas. For instance, GLUT2 translocation from an intracellular pool to the apical membrane after a sugar meal transiently increases sugar uptake by enterocytes (reviewed in 32). Third, GLUT2 functions as a membrane receptor of sugar. Independently of glucose metabolism, GLUT2 detects the presence of extracellular sugar and transduces a signal to modulate cell functions, including beta-cell insulin secretion, renal reabsorption, and intestinal absorption according to the sugar environment. These recent developments are examined here in heath and metabolic disease, highlighting various unanswered questions.


Assuntos
Sacarose Alimentar/metabolismo , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Mutação , Animais , Metabolismo dos Carboidratos , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo
14.
Annu Rev Nutr ; 28: 35-54, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18393659

RESUMO

Intestinal glucose absorption comprises two components. One is classical active absorption mediated by the Na+/glucose cotransporter. The other is a diffusive component, formerly attributed to paracellular flow. Recent evidence, however, indicates that the diffusive component is mediated by the transient insertion of glucose transporter type 2 (GLUT2) into the apical membrane. This apical GLUT2 pathway of intestinal sugar absorption is present in species from insect to human, providing a major route at high sugar concentrations. The pathway is regulated by rapid trafficking of GLUT2 to the apical membrane induced by glucose during assimilation of a meal. Apical GLUT2 is therefore a target for multiple short-term and long-term nutrient-sensing mechanisms. These include regulation by a newly recognized pathway of calcium absorption through the nonclassical neuroendocrine l-type channel Cav1.3 operating during digestion, activation of intestinal sweet taste receptors by natural sugars and artificial sweeteners, paracrine and endocrine hormones, especially insulin and GLP-2, and stress. Permanent apical GLUT2, resulting in increased sugar absorption, is a characteristic of experimental diabetes and of insulin-resistant states induced by fructose and fat. The nutritional consequences of apical and basolateral GLUT2 regulation are discussed in the context of Western diet, processed foods containing artificial sweeteners, obesity, and diabetes.


Assuntos
Membrana Celular/metabolismo , Metabolismo Energético/fisiologia , Transportador de Glucose Tipo 2/fisiologia , Glucose/metabolismo , Absorção Intestinal/fisiologia , Cálcio/metabolismo , Metabolismo dos Carboidratos , Enterócitos/efeitos dos fármacos , Enterócitos/metabolismo , Transportador de Glucose Tipo 2/metabolismo , Humanos , Apoio Nutricional
15.
Diabetes ; 57(3): 555-62, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18057092

RESUMO

OBJECTIVES: A physiological adaptation to a sugar-rich meal is achieved by increased sugar uptake to match dietary load, resulting from a rapid transient translocation of the fructose/glucose GLUT2 transporter to the brush border membrane (BBM) of enterocytes. The aim of this study was to define the contributors and physiological mechanisms controlling intestinal sugar absorption, focusing on the action of insulin and the contribution of GLUT2-mediated transport. RESEARCH DESIGN AND METHODS: The studies were performed in the human enterocytic colon carcinoma TC7 subclone (Caco-2/TC7) cells and in vivo during hyperinsulinemic-euglycemic clamp experiments in conscious mice. Chronic high-fructose or high-fat diets were used to induce glucose intolerance and insulin resistance in mice. RESULTS AND CONCLUSIONS: In Caco-2/TC7 cells, insulin action diminished the transepithelial transfer of sugar and reduced BBM and basolateral membrane (BLM) GLUT2 levels, demonstrating that insulin can target sugar absorption by controlling the membrane localization of GLUT2 in enterocytes. Similarly, in hyperinsulinemic-euglycemic clamp experiments in sensitive mice, insulin abolished GLUT2 (i.e., the cytochalasin B-sensitive component of fructose absorption), decreased BBM GLUT2, and concomitantly increased intracellular GLUT2. Acute insulin treatment before sugar intake prevented the insertion of GLUT2 into the BBM. Insulin resistance in mice provoked a loss of GLUT2 trafficking, and GLUT2 levels remained permanently high in the BBM and low in the BLM. We propose that, in addition to its peripheral effects, insulin inhibits intestinal sugar absorption to prevent excessive blood glucose excursion after a sugar meal. This protective mechanism is lost in the insulin-resistant state induced by high-fat or high-fructose feeding.


Assuntos
Enterócitos/efeitos dos fármacos , Enterócitos/metabolismo , Transportador de Glucose Tipo 2/metabolismo , Resistência à Insulina/fisiologia , Insulina/farmacologia , Animais , Células CACO-2 , Metabolismo dos Carboidratos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Enterócitos/ultraestrutura , Regulação da Expressão Gênica , Técnica Clamp de Glucose , Humanos , Camundongos , Microvilosidades/metabolismo , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia
16.
Am J Physiol Gastrointest Liver Physiol ; 294(1): G155-64, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17962360

RESUMO

Lipopolysaccharide (LPS) endotoxin is a causative agent of sepsis. The aim of this study was to examine LPS effects on intestinal fructose absorption and to decipher mechanisms. Sepsis was induced by intravenous injection of LPS in rabbits. The ultrastructural study and DNA fragmentation patterns were identical in the intestine of LPS and sham animals. LPS treatment reduced fructose absorption altering both mucosal-to-serosal transepithelial fluxes and uptake into brush border membrane vesicles (BBMVs). Cytochalasin B was ineffective on fructose uptake, indicating that GLUT5, but not GLUT2, transport activity was targeted. GLUT5 protein levels in BBMvs were lower in LPS than in sham-injected rabbits. Thus lower fructose transport resulted from lower levels of GLUT5 protein. LPS treatment decreased GLUT5 levels by proteasome-dependent degradation. Specific inhibitors of PKC, PKA, and MAP kinases (p38MAPK, JNK, MEK1/2) protected fructose uptake from adverse LPS effect. Moreover, a TNF-alpha antagonist blocked LPS action on fructose uptake. We conclude that intestinal fructose transport inhibition by LPS is associated with diminished GLUT5 numbers in the brush border membrane of enterocytes triggered by activation of several interrelated signaling cascades and proteasome degradation.


Assuntos
Frutose/metabolismo , Transportador de Glucose Tipo 5/metabolismo , Absorção Intestinal , Mucosa Intestinal/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Quinases/metabolismo , Sepse/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fragmentação do DNA , Modelos Animais de Doenças , Regulação para Baixo , Enterócitos/metabolismo , Injeções Intravenosas , Absorção Intestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Intestinos/enzimologia , Intestinos/ultraestrutura , Lipopolissacarídeos/administração & dosagem , Masculino , Microvilosidades/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Coelhos , Sepse/induzido quimicamente , Sepse/enzimologia , Sepse/patologia , Transdução de Sinais , Fatores de Tempo
17.
PLoS One ; 2(12): e1288, 2007 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18074013

RESUMO

BACKGROUND: Mammals must sense the amount of sugar available to them and respond appropriately. For many years attention has focused on intracellular glucose sensing derived from glucose metabolism. Here, we studied the detection of extracellular glucose concentrations in vivo by invalidating the transduction pathway downstream from the transporter-detector GLUT2 and measured the physiological impact of this pathway. METHODOLOGY/PRINCIPAL FINDINGS: We produced mice that ubiquitously express the largest cytoplasmic loop of GLUT2, blocking glucose-mediated gene expression in vitro without affecting glucose metabolism. Impairment of GLUT2-mediated sugar detection transiently protected transgenic mice against starvation and streptozotocin-induced diabetes, suggesting that both low- and high-glucose concentrations were not detected. Transgenic mice favored lipid oxidation, and oral glucose was slowly cleared from blood due to low insulin production, despite massive urinary glucose excretion. Kidney adaptation was characterized by a lower rate of glucose reabsorption, whereas pancreatic adaptation was associated with a larger number of small islets. CONCLUSIONS/SIGNIFICANCE: Molecular invalidation of sugar sensing in GLUT2-loop transgenic mice changed multiple aspects of glucose homeostasis, highlighting by a top-down approach, the role of membrane glucose receptors as potential therapeutic targets.


Assuntos
Transportador de Glucose Tipo 2/metabolismo , Glucose/metabolismo , Homeostase , Animais , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 2/genética , Metabolismo dos Lipídeos , Camundongos , Camundongos Transgênicos , Oxirredução , Pâncreas/fisiologia
18.
J Cell Physiol ; 213(3): 834-43, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17786952

RESUMO

Sugar consumption and subsequent sugar metabolism are known to regulate the expression of genes involved in intestinal sugar absorption and delivery. Here we investigate the hypothesis that sugar-sensing detectors in membranes facing the intestinal lumen or the bloodstream can also modulate intestinal sugar absorption. We used wild-type and GLUT2-null mice, to show that dietary sugars stimulate the expression of sucrase-isomaltase (SI) and L-pyruvate kinase (L-PK) by GLUT2-dependent mechanisms, whereas the expression of GLUT5 and SGLT1, did not rely on the presence of GLUT2. By providing sugar metabolites, sugar transporters, including GLUT2, fuelled a sensing pathway. In Caco2/TC7 enterocytes, we could disconnect the sensing triggered by detector from that produced by metabolism, and found that GLUT2 generated a metabolism-independent pathway to stimulate the expression of SI and L-PK. In cultured enterocytes, both apical and basolateral fructose could increase the expression of GLUT5, conversely, basolateral sugar administration could stimulate the expression of GLUT2. Finally, we located the sweet-taste receptors T1R3 and T1R2 in plasma membranes, and we measured their cognate G alpha Gustducin mRNA levels. Furthermore, we showed that a T1R3 inhibitor altered the fructose-induced expression of SGLT1, GLUT5, and L-PK. Intestinal gene expression is thus controlled by a combination of at least three sugar-signaling pathways triggered by sugar metabolites and membrane sugar receptors that, according to membrane location, determine sugar-sensing polarity. This provides a rationale for how intestine adapts sugar delivery to blood and dietary sugar provision.


Assuntos
Polaridade Celular , Enterócitos/metabolismo , Hexoses/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Sacarose/metabolismo , Edulcorantes/metabolismo , Animais , Células CACO-2 , Clonagem Molecular , Frutose/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 2/química , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Transportador de Glucose Tipo 5/genética , Transportador de Glucose Tipo 5/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Jejuno/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Transporte de Monossacarídeos/genética , Oligo-1,6-Glucosidase/genética , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo , Sacarase/genética , Transfecção
19.
Diabetes ; 54(10): 3056-62, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16186415

RESUMO

Understanding the mechanisms that determine postprandial fluctuations in blood glucose concentration is central for effective glycemic control in the management of diabetes. Intestinal sugar absorption is one such mechanism, and studies on its increase in experimental diabetes led us to propose a new model of sugar absorption. In the apical GLUT2 model, the glucose transported by the Na(+)/glucose cotransporter SGLT1 promotes insertion of GLUT2 into the apical membrane within minutes, so that the mechanism operates during assimilation of a meal containing high-glycemic index carbohydrate to provide a facilitated component of absorption up to three times greater than by SGLT1. Here we review the evidence for the apical GLUT2 model and describe how apical GLUT2 is a target for multiple short-term nutrient-sensing mechanisms by dietary sugars, local and endocrine hormones, cellular energy status, stress, and diabetes. These mechanisms suggest that apical GLUT2 is a potential therapeutic target for novel dietary or pharmacological approaches to control intestinal sugar delivery and thereby improve glycemic control.


Assuntos
Metabolismo dos Carboidratos , Absorção Intestinal , Animais , Membrana Celular/metabolismo , Diabetes Mellitus , Dieta , Carboidratos da Dieta/farmacocinética , Metabolismo Energético , Índice Glicêmico , Homeostase , Humanos , Transportador 1 de Glucose-Sódio/fisiologia , Estresse Fisiológico
20.
J Biol Chem ; 280(20): 20094-101, 2005 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15767253

RESUMO

Glucose-6-phosphatase (G6Pase) catalyzes the release of glucose from glucose 6-phosphate. This enzyme was mainly studied in the liver, but while detected in the small intestine little is known about the regulation of its intestinal expression. This study describes the mechanisms of the glucose-dependent regulation of G6Pase expression in intestinal cells. Results obtained in vivo and in Caco-2/TC7 enterocytes showed that glucose increases the G6Pase mRNA level. In Caco-2/TC7 cells, glucose stabilized G6Pase mRNA and activated the transcription of the gene, meaning that glucose-dependent G6Pase expression involved both transcriptional and post-transcriptional mechanisms. Reporter-gene studies showed that, although the -299/+57 region of the human G6Pase promoter was sufficient to trigger the glucose response in the hepatoma cell line HepG2, the -1157/-1133 fragment was required for maximal activation of glucose-6-phosphatase gene transcription in Caco-2/TC7 cells. This fragment binds the aryl receptor nuclear translocator (ARNT), cAMP-responsive element-binding protein, and upstream stimulatory factor transcription factors. The DNA binding activity of these transcription factors was increased in nuclear extracts of differentiated cells from the intestinal villus of mice fed sugar-rich diets as compared with mice fed a no-sugar diet. A direct implication of ARNT in the activation of G6Pase gene transcription by glucose has been observed in Caco-2/TC7 cells using RNA interference experiments. These results support a physiological role for G6Pase in the control of nutrient absorption in the small intestine.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glucose-6-Fosfatase/genética , Glucose/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Fatores de Transcrição/metabolismo , Animais , Translocador Nuclear Receptor Aril Hidrocarboneto , Sequência de Bases , Sítios de Ligação/genética , Células CACO-2 , Linhagem Celular , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Expressão Gênica/efeitos dos fármacos , Genes Reporter , Glucose/farmacologia , Humanos , Intestinos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Receptores de Hidrocarboneto Arílico/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...