Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Pathol ; 246(2): 217-230, 2018 10.
Article in English | MEDLINE | ID: mdl-29984492

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Inflammation/metabolism , Intestinal Absorption/drug effects , Jejunum/drug effects , Lipids/administration & dosage , Obesity/metabolism , Acute-Phase Proteins , Adult , Aged , Caco-2 Cells , Carrier Proteins/blood , Case-Control Studies , Cholera Toxin/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Female , Haptoglobins , Humans , Inflammation/complications , Inflammation/physiopathology , Jejunum/metabolism , Jejunum/physiopathology , MARVEL Domain Containing 2 Protein/metabolism , Male , Membrane Glycoproteins/blood , Micelles , Middle Aged , Obesity/complications , Obesity/physiopathology , Occludin/metabolism , Permeability , Protein Precursors , Tight Junctions/metabolism , Young Adult
2.
J Biol Chem ; 288(43): 31080-92, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-23986439

ABSTRACT

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.


Subject(s)
Cell Differentiation/physiology , Glucose Transporter Type 2/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Mutation, Missense , Polymorphism, Single Nucleotide , Amino Acid Substitution , Animals , Biological Transport, Active/genetics , Cell Line, Tumor , Glucose/genetics , Glucose/metabolism , Glucose Transporter Type 2/genetics , Humans , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/cytology , Mice , Rats , Signal Transduction , Xenopus laevis
3.
Am J Physiol Endocrinol Metab ; 298(5): E1078-87, 2010 May.
Article in English | MEDLINE | ID: mdl-20179244

ABSTRACT

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.


Subject(s)
Eating/physiology , Glucose Transporter Type 2/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Analysis of Variance , Animals , Biological Transport/physiology , Body Weight/physiology , Cell Count , Energy Metabolism , Feeding Behavior/physiology , Ghrelin/blood , Glucose Transporter Type 2/genetics , Homeostasis/physiology , Immunohistochemistry , Insulin/blood , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leptin/blood , Mice , Mice, Transgenic , Neuropeptides/genetics , Neuropeptides/metabolism , Orexins , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Statistics, Nonparametric , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism
4.
Mol Metab ; 39: 101007, 2020 09.
Article in English | MEDLINE | ID: mdl-32360426

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Intestinal Mucosa/metabolism , Obesity/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Adiposity/genetics , Adult , Aged , Aged, 80 and over , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers , Cell Line , Comorbidity , Cytokines/metabolism , Epithelial Cells/metabolism , Female , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Jejunum/metabolism , Lipid Metabolism , MAP Kinase Signaling System , Male , Mice , Middle Aged , Models, Biological , Obesity/etiology , Obesity/pathology , Permeability , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism
5.
J Cell Physiol ; 213(3): 834-43, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17786952

ABSTRACT

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.


Subject(s)
Cell Polarity , Enterocytes/metabolism , Hexoses/metabolism , Monosaccharide Transport Proteins/metabolism , Sucrose/metabolism , Sweetening Agents/metabolism , Animals , Caco-2 Cells , Cloning, Molecular , Fructose/metabolism , Glucose/metabolism , Glucose Transporter Type 2/chemistry , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Green Fluorescent Proteins/metabolism , Humans , Jejunum/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monosaccharide Transport Proteins/genetics , Oligo-1,6-Glucosidase/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Sucrase/genetics , Transfection
6.
Mol Metab ; 6(1): 61-72, 2017 01.
Article in English | MEDLINE | ID: mdl-28123938

ABSTRACT

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.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 2/metabolism , Glucose/metabolism , Animals , Blood Glucose/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/physiology , Homeostasis , Intestinal Absorption , Intestinal Mucosa/metabolism , Mice , Mice, Knockout , Sodium-Glucose Transporter 1/metabolism , Tissue Distribution
7.
Diabetes ; 54(10): 3056-62, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16186415

ABSTRACT

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.


Subject(s)
Carbohydrate Metabolism , Intestinal Absorption , Animals , Cell Membrane/metabolism , Diabetes Mellitus , Diet , Dietary Carbohydrates/pharmacokinetics , Energy Metabolism , Glycemic Index , Homeostasis , Humans , Sodium-Glucose Transporter 1/physiology , Stress, Physiological
8.
Med Sci (Paris) ; 32(5): 461-9, 2016 May.
Article in French | MEDLINE | ID: mdl-27225918

ABSTRACT

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.


Subject(s)
Dysbiosis/complications , Gastrointestinal Microbiome/physiology , Inflammation/etiology , Intestinal Mucosa/metabolism , Obesity/etiology , Animals , Dysbiosis/immunology , Dysbiosis/metabolism , Humans , Inflammation/metabolism , Inflammation/microbiology , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/microbiology , Obesity/immunology , Obesity/metabolism , Obesity/microbiology , Permeability
9.
Biochem J ; 375(Pt 1): 167-74, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-12820898

ABSTRACT

In intestinal cells, levels of the fructose transporter GLUT5 are increased by glucose and to a greater extent by fructose. We investigated the mechanism by which fructose increases GLUT5 expression. In Caco-2 cells, fructose and glucose increased activity of the -2500/+41 GLUT5 promoter to the same extent. cAMP also activated the GLUT5 promoter. However, if a protein kinase A inhibitor was used to block cAMP signalling, extensive GLUT5 mRNA degradation was observed, with no change in basal transcription levels demonstrating the involvement of cAMP in GLUT5 mRNA stability. Indeed, the half-life of GLUT5 mRNA was correlated ( R2=0.9913) with cellular cAMP levels. Fructose increased cAMP concentration more than glucose, accounting for the stronger effect of fructose when compared with that of glucose on GLUT5 production. We identified several complexes between GLUT5 3'-UTR RNA (where UTR stands for untranslated region) and cytosolic proteins that might participate in mRNA processing. Strong binding of a 140 kDa complex I was observed in sugar-deprived cells, with levels of binding lower in the presence of fructose and glucose by factors of 12 and 6 respectively. This may account for differences in the effects of fructose and glucose. In contrast, the amounts of two complexes of 96 and 48 kDa increased equally after stimulation with either glucose or fructose. Finally, PABP (polyadenylated-binding protein)-interacting protein 2, a destabilizing partner of PABP, was identified as a component of GLUT5 3'-UTR RNA-protein complexes. We conclude that the post-transcriptional regulation of GLUT5 by fructose involves increases in mRNA stability mediated by the cAMP pathway and Paip2 (PABP-interacting protein 2) binding.


Subject(s)
Cyclic AMP/metabolism , Fructose/pharmacology , Monosaccharide Transport Proteins/genetics , RNA Stability , RNA-Binding Proteins/physiology , 3' Untranslated Regions/metabolism , Caco-2 Cells , Carbohydrate Metabolism , Cell Differentiation , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose/pharmacology , Glucose Transporter Type 5 , Humans , Monosaccharide Transport Proteins/metabolism , RNA Stability/drug effects , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Second Messenger Systems , Transcriptional Activation
10.
J Nutr Sci ; 4: e22, 2015.
Article in English | MEDLINE | ID: mdl-26157580

ABSTRACT

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.

11.
Cell Metab ; 22(1): 113-24, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26094890

ABSTRACT

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.


Subject(s)
Enterocytes/pathology , Inflammation/complications , Insulin/immunology , Jejunum/pathology , Obesity/complications , T-Lymphocytes/pathology , Adult , CD8 Antigens/immunology , Cells, Cultured , Enterocytes/immunology , Female , Glucose Transporter Type 2/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Insulin Resistance , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Jejunum/cytology , Jejunum/immunology , Male , Middle Aged , Obesity/immunology , Obesity/pathology , Signal Transduction , T-Lymphocytes/immunology
12.
FEBS Lett ; 549(1-3): 77-82, 2003 Aug 14.
Article in English | MEDLINE | ID: mdl-12914929

ABSTRACT

Skeletal muscle, a primary insulin target tissue, expresses the GLUT5 fructose transporter. Although insulin has no acute effect on GLUT5 expression and function in muscle, we show here that long-term (24 h) insulin treatment of L6 muscle cells induces a dose-dependent increase in GLUT5 protein (by up to two-fold), leading to a concomitant increase in fructose uptake. The increase in GLUT5 expression and function was suppressed by inhibitors of gene transcription and protein synthesis, suggesting that insulin promotes de novo carrier synthesis. Transfection of the GLUT5 gene promoter fused to luciferase into L6 cells revealed that insulin induced a 1.8-fold increase in GLUT5 promoter activity. Our findings indicate that insulin is capable of increasing the abundance and functional activity of GLUT5 in skeletal muscle cells and that this is most likely mediated via activation of the GLUT5 promoter.


Subject(s)
Insulin/pharmacology , Monosaccharide Transport Proteins/biosynthesis , Monosaccharide Transport Proteins/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Fructose/pharmacokinetics , Glucose Transporter Type 5 , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Promoter Regions, Genetic/drug effects , Rats , Transcription, Genetic/drug effects
13.
Prog Mol Biol Transl Sci ; 108: 113-27, 2012.
Article in English | MEDLINE | ID: mdl-22656375

ABSTRACT

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.


Subject(s)
Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/pharmacology , Food Preferences , Gene Expression Regulation/drug effects , Genetic Diseases, Inborn/physiopathology , Humans
14.
Mol Biol Cell ; 23(2): 324-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22114352

ABSTRACT

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.


Subject(s)
Actins/metabolism , Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Actins/ultrastructure , Animals , Enterocytes/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/ultrastructure , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/ultrastructure , Microscopy, Electron, Transmission , Microvilli/metabolism , Microvilli/ultrastructure , Myosin Heavy Chains/metabolism , Protein Structure, Tertiary
15.
Diabetes ; 60(10): 2598-607, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21852673

ABSTRACT

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.


Subject(s)
Cell Membrane/metabolism , Dietary Fats/administration & dosage , Enterocytes/metabolism , Glucose Transporter Type 2/metabolism , Obesity, Morbid/metabolism , Adult , Animals , Diabetes Mellitus, Type 2/metabolism , Dietary Carbohydrates/administration & dosage , Dietary Carbohydrates/pharmacology , Glucose Transporter Type 2/genetics , Humans , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Obese , Middle Aged , Young Adult
17.
Am J Physiol Endocrinol Metab ; 296(5): E985-92, 2009 May.
Article in English | MEDLINE | ID: mdl-19223655

ABSTRACT

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.


Subject(s)
Dietary Sucrose/metabolism , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Mutation , Animals , Carbohydrate Metabolism , Glycogen Storage Disease/genetics , Glycogen Storage Disease/metabolism , Humans , Insulin-Secreting Cells/metabolism
18.
Annu Rev Nutr ; 28: 35-54, 2008.
Article in English | MEDLINE | ID: mdl-18393659

ABSTRACT

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.


Subject(s)
Cell Membrane/metabolism , Energy Metabolism/physiology , Glucose Transporter Type 2/physiology , Glucose/metabolism , Intestinal Absorption/physiology , Calcium/metabolism , Carbohydrate Metabolism , Enterocytes/drug effects , Enterocytes/metabolism , Glucose Transporter Type 2/metabolism , Humans , Nutritional Support
19.
Am J Physiol Gastrointest Liver Physiol ; 294(1): G155-64, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17962360

ABSTRACT

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.


Subject(s)
Fructose/metabolism , Glucose Transporter Type 5/metabolism , Intestinal Absorption , Intestinal Mucosa/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/metabolism , Sepsis/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Fragmentation , Disease Models, Animal , Down-Regulation , Enterocytes/metabolism , Injections, Intravenous , Intestinal Absorption/drug effects , Intestines/drug effects , Intestines/enzymology , Intestines/ultrastructure , Lipopolysaccharides/administration & dosage , Male , Microvilli/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Rabbits , Sepsis/chemically induced , Sepsis/enzymology , Sepsis/pathology , Signal Transduction , Time Factors
20.
Diabetes ; 57(3): 555-62, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18057092

ABSTRACT

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.


Subject(s)
Enterocytes/drug effects , Enterocytes/metabolism , Glucose Transporter Type 2/metabolism , Insulin Resistance/physiology , Insulin/pharmacology , Animals , Caco-2 Cells , Carbohydrate Metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Enterocytes/ultrastructure , Gene Expression Regulation , Glucose Clamp Technique , Humans , Mice , Microvilli/metabolism , Protein Transport/drug effects , Protein Transport/physiology
SELECTION OF CITATIONS
SEARCH DETAIL