Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Int J Obes (Lond) ; 45(1): 170-183, 2021 01.
Article in English | MEDLINE | ID: mdl-33037328

ABSTRACT

OBJECTIVES: Altered enteroendocrine cell (EEC) function in obesity and type 2 diabetes is not fully understood. Understanding the transcriptional program that controls EEC differentiation is important because some EEC types harbor significant therapeutic potential for type 2 diabetes. METHODS: EEC isolation from jejunum of obese individuals with (ObD) or without (Ob) type 2 diabetes was obtained with a new method of cell sorting. EEC transcriptional profiles were established by RNA-sequencing in a first group of 14 Ob and 13 ObD individuals. EEC lineage and densities were studied in the jejunum of a second independent group of 37 Ob, 21 ObD and 22 non obese (NOb) individuals. RESULTS: The RNA seq analysis revealed a distinctive transcriptomic signature and a decreased differentiation program in isolated EEC from ObD compared to Ob individuals. In the second independent group of ObD, Ob and NOb individuals a decreased GLP-1 cell lineage and GLP-1 maturation from proglucagon, were observed in ObD compared to Ob individuals. Furthermore, jejunal density of GLP-1-positive cells was significantly reduced in ObD compared to Ob individuals. CONCLUSIONS: These results highlight that the transcriptomic signature of EEC discriminate obese subjects according to their diabetic status. Furthermore, type 2 diabetes is associated with reduced GLP-1 cell differentiation and proglucagon maturation leading to low GLP-1-cell density in human obesity. These mechanisms could account for the decrease plasma GLP-1 observed in metabolic diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Enteroendocrine Cells/metabolism , Jejunum/cytology , Obesity , Adult , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Enteroendocrine Cells/cytology , Female , Humans , Male , Middle Aged , Obesity/complications , Obesity/epidemiology , Obesity/metabolism
2.
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
3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(2): 199-211, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29196159

ABSTRACT

Scavenger receptor Class B type 1 (SR-B1) is a lipid transporter and sensor. In intestinal epithelial cells, SR-B1-dependent lipid sensing is associated with SR-B1 recruitment in raft-like/ detergent-resistant membrane domains and interaction of its C-terminal transmembrane domain with plasma membrane cholesterol. To clarify the initiating events occurring during lipid sensing by SR-B1, we analyzed cholesterol trafficking and raft-like domain composition in intestinal epithelial cells expressing wild-type SR-B1 or the mutated form SR-B1-Q445A, defective in membrane cholesterol binding and signal initiation. These features of SR-B1 were found to influence both apical cholesterol efflux and intracellular cholesterol trafficking from plasma membrane to lipid droplets, and the lipid composition of raft-like domains. Lipidomic analysis revealed likely participation of d18:0/16:0 sphingomyelin and 16:0/0:0 lysophosphatidylethanolamine in lipid sensing by SR-B1. Proteomic analysis identified proteins, whose abundance changed in raft-like domains during lipid sensing, and these included molecules linked to lipid raft dynamics and signal transduction. These findings provide new insights into the role of SR-B1 in cellular cholesterol homeostasis and suggest molecular links between SR-B1-dependent lipid sensing and cell cholesterol and lipid droplet dynamics.


Subject(s)
Cholesterol/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Lysophospholipids/metabolism , Membrane Microdomains/metabolism , Scavenger Receptors, Class B/metabolism , Sphingomyelins/metabolism , Caco-2 Cells , Humans , Lipid Droplets/metabolism , Signal Transduction/physiology
4.
J Biol Chem ; 291(31): 16328-38, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27255710

ABSTRACT

The worldwide prevalence of metabolic diseases is increasing, and there are global recommendations to limit consumption of certain nutrients, especially saturated lipids. Insulin resistance, a common trait occurring in obesity and type 2 diabetes, is associated with intestinal lipoprotein overproduction. However, the mechanisms by which the intestine develops insulin resistance in response to lipid overload remain unknown. Here, we show that insulin inhibits triglyceride secretion and intestinal microsomal triglyceride transfer protein expression in vivo in healthy mice force-fed monounsaturated fatty acid-rich olive oil but not in mice force-fed saturated fatty acid-rich palm oil. Moreover, when mouse intestine and human Caco-2/TC7 enterocytes were treated with the saturated fatty acid, palmitic acid, the insulin-signaling pathway was impaired. We show that palmitic acid or palm oil increases ceramide production in intestinal cells and that treatment with a ceramide analogue partially reproduces the effects of palmitic acid on insulin signaling. In Caco-2/TC7 enterocytes, ceramide effects on insulin-dependent AKT phosphorylation are mediated by protein kinase C but not by protein phosphatase 2A. Finally, inhibiting de novo ceramide synthesis improves the response of palmitic acid-treated Caco-2/TC7 enterocytes to insulin. These results demonstrate that a palmitic acid-ceramide pathway accounts for impaired intestinal insulin sensitivity, which occurs within several hours following initial lipid exposure.


Subject(s)
Ceramides/biosynthesis , Enterocytes/metabolism , Insulin/metabolism , Intestinal Mucosa/metabolism , Palmitic Acid/pharmacology , Signal Transduction , Animals , Caco-2 Cells , Humans , Mice , Palm Oil , Palmitic Acid/metabolism , Phosphorylation/drug effects , Plant Oils/pharmacology , Proto-Oncogene Proteins c-akt/metabolism
5.
Exp Cell Res ; 340(2): 172-9, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26431584

ABSTRACT

Cytosolic lipid droplets (LDs) are observed in enterocytes of jejunum during lipid absorption. One important function of the intestine is to secrete chylomicrons, which provide dietary lipids throughout the body, from digested lipids in meals. The current hypothesis is that cytosolic LDs in enterocytes constitute a transient pool of stored lipids that provides lipids during interprandial period while lowering chylomicron production during the post-prandial phase. This smoothens the magnitude of peaks of hypertriglyceridemia. Here, we review the composition and functions of lipids and associated proteins of enterocyte LDs, the known physiological functions of LDs as well as the role of LDs in pathological processes in the context of the intestine.


Subject(s)
Biological Transport/physiology , Chylomicrons/metabolism , Enterocytes/metabolism , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Animals , Humans , Triglycerides/metabolism
6.
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
7.
J Endocrinol ; 252(1): 31-44, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34647524

ABSTRACT

Changes in dietary habits have occurred concomitantly with a rise of type 2 diabetes (T2D) and obesity. Intestine is the first organ facing nutrient ingestion and has to adapt its metabolism with these dietary changes. HNF-4γ, a transcription factor member of the nuclear receptor superfamily and mainly expressed in intestine, has been suggested to be involved in susceptibility to T2D. Our aim was to investigate the role of HNF-4γ in metabolic disorders and related mechanisms. Hnf4g-/- mice were fed high-fat/high-fructose (HF-HF) diet for 6 weeks to induce obesity and T2D. Glucose homeostasis, energy homeostasis in metabolic cages, body composition and stool energy composition, as well as gene expression analysis in the jejunum were analyzed. Despite an absence of decrease in calorie intake, of increase in locomotor activity or energy expenditure, Hnf4g-/- mice fed with HF-HF are protected against weight gain after 6 weeks of HF-HF diet. We showed that Hnf4g-/- mice fed HF-HF display an increase in fecal calorie loss, mainly due to intestinal lipid malabsorption. Gene expression of lipid transporters, Fatp4 and Scarb1 and of triglyceride-rich lipoprotein secretion proteins, Mttp and ApoB are decreased in gut epithelium of Hnf4g-/- mice fed HF-HF, showing the HNF-4γ role in intestine lipid absorption. Furthermore, plasma GLP-1 and jejunal GLP-1 content are increased in Hnf4g-/- mice fed HF-HF, which could contribute to the glucose intolerance protection. The loss of HNF-4γ leads to a protection against a diet-induced weight gain and to a deregulated glucose homeostasis, associated with lipid malabsorption.


Subject(s)
Hepatocyte Nuclear Factor 4/genetics , Intestinal Absorption/genetics , Lipid Metabolism/genetics , Obesity/genetics , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Female , Fructose/adverse effects , Gene Deletion , Glucose Intolerance/etiology , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Intestines/metabolism , Malabsorption Syndromes/genetics , Malabsorption Syndromes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Triglycerides/metabolism , Weight Gain/genetics
8.
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
9.
Article in English | MEDLINE | ID: mdl-31647994

ABSTRACT

The mechanisms leading to the low-grade inflammation observed during obesity are not fully understood. Seeking the initiating events, we tested the hypothesis that the intestine could be damaged by repeated lipid supply and therefore participate in inflammation. In mice, 1-5 palm oil gavages increased intestinal permeability via decreased expression and mislocalization of junctional proteins at the cell-cell contacts; altered the intestinal bacterial species by decreasing the abundance of Akkermansia muciniphila, segmented filamentous bacteria, and Clostridium leptum; and increased inflammatory cytokine expression. This was further studied in human intestinal epithelial Caco-2/TC7 cells using the two main components of palm oil, i.e., palmitic and oleic acid. Saturated palmitic acid impaired paracellular permeability and junctional protein localization, and induced inflammatory cytokine expression in the cells, but unsaturated oleic acid did not. Inhibiting de novo ceramide synthesis prevented part of these effects. Altogether, our data show that short exposure to palm oil or palmitic acid induces intestinal dysfunctions targeting barrier integrity and inflammation. Excessive palm oil consumption could be an early player in the gut alterations observed in metabolic diseases.


Subject(s)
Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Metabolic Syndrome/pathology , Palm Oil/adverse effects , Palmitic Acid/adverse effects , Administration, Oral , Animals , Caco-2 Cells , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Feces/microbiology , Gastrointestinal Microbiome/immunology , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Metabolic Syndrome/immunology , Mice , Palm Oil/administration & dosage , Palm Oil/chemistry , Palmitic Acid/administration & dosage , Permeability , Tight Junctions/drug effects
10.
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
11.
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
12.
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
13.
Prion ; 10(2): 143-52, 2016 03 03.
Article in English | MEDLINE | ID: mdl-27216988

ABSTRACT

The cellular prion protein PrP(c) plays important roles in proliferation, cell death and survival, differentiation and adhesion. The participation of PrP(c) in tumor growth and metastasis was pointed out, but the underlying mechanisms were not deciphered completely. In the constantly renewing intestinal epithelium, our group demonstrated a dual localization of PrP(c), which is targeted to cell-cell junctions in interaction with Src kinase and desmosomal proteins in differentiated enterocytes, but is predominantly nuclear in dividing cells. While the role of PrP(c) in the dynamics of intercellular junctions was confirmed in other biological systems, we unraveled its function in the nucleus only recently. We identified several nuclear PrP(c) partners, which comprise γ-catenin, one of its desmosomal partners, ß-catenin and TCF7L2, the main effectors of the canonical Wnt pathway, and YAP, one effector of the Hippo pathway. PrP(c) up-regulates the activity of the ß-catenin/TCF7L2 complex and its invalidation impairs the proliferation of intestinal progenitors. We discuss how PrP(c) could participate to oncogenic processes through its interaction with Wnt and Hippo pathway effectors, which are controlled by cell-cell junctions and Src family kinases and dysregulated during tumorigenesis. This highlights new potential mechanisms that connect PrP(c) expression and subcellular redistribution to cancer.


Subject(s)
Cell Nucleus/pathology , Intercellular Junctions/pathology , Neoplasms/pathology , PrPC Proteins/metabolism , Signal Transduction , Animals , Cell Nucleus/metabolism , Cell Proliferation , Desmosomes/metabolism , Desmosomes/pathology , Epithelial-Mesenchymal Transition , Hippo Signaling Pathway , Humans , Intercellular Junctions/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Neoplasms/metabolism , PrPC Proteins/analysis , Protein Interaction Maps , Protein Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway
14.
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
15.
Diabetes ; 53(3): 672-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14988251

ABSTRACT

Type 2 diabetic patients present high triglyceride and low HDL levels, significant determinants for the risk of atherosclerosis. Transgenic mice overproducing human apolipoprotein (apo)A-II, one of the two major apos of HDLs, display the same lipid disorders. Here, we investigated the possible regulation of apoA-II gene expression by glucose. In primary rat hepatocytes and in HepG2 cells, the transcription of the human apoA-II gene was upregulated by glucose. This response was mediated by a hormone-responsive element within the enhancer of the apoA-II promoter and was dependent on hepatocyte nuclear factor-4alpha. Accordingly, in transgenic mice, the human apoA-II gene is stimulated by a high-carbohydrate diet after fasting and at weaning. By contrast, the apoA-II mRNA level is not modified in streptozotocin-induced diabetic rats. In transgenic mice overexpressing the human apoA-II gene, plasma human apoA-II concentration was positively correlated with blood glucose levels. These mice displayed a marked delay in plasma glucose tolerance as compared with control mice. We hypothesize that the following pathogenic pathway might occur in the course of type 2 diabetes: increased apoA-II level causes a rise in plasma triglyceride level and glucose intolerance, resulting in hyperglycemia, which in turn might further increase apoA-II gene transcription.


Subject(s)
Apolipoprotein A-II/genetics , Gene Expression Regulation/drug effects , Glucose/pharmacology , Transcription, Genetic/drug effects , Animals , Base Sequence , Blood Glucose/metabolism , DNA Primers , Humans , Liver/physiology , Mice , Mice, Transgenic , Polymerase Chain Reaction/methods , RNA, Messenger/genetics
16.
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
17.
Biochem J ; 378(Pt 1): 219-27, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14632628

ABSTRACT

The transcription factor PDX-1 (pancreatic and duodenal homeobox-1) is essential for pancreatic development and the maintainence of expression of islet beta-cell-specific genes. In an previous study [Rafiq, Kennedy and Rutter (1998) J. Biol. Chem. 273, 23241-23247] we demonstrated that PDX-1 may be activated at elevated glucose concentrations by translocation from undefined binding sites in the cytosol and nuclear membrane into the nucleoplasm. In the present study, we show that PDX-1 interacts directly and specifically in vitro with the nuclear import receptor family member, importin beta1, and that this interaction is mediated by the PDX-1 homeodomain (amino acids 146-206). Demonstrating the functional importance of the PDX-1-importin beta1 interaction, microinjection of MIN6 beta-cells with anti-(importin beta1) antibodies blocked both the nuclear translocation of PDX-1, and the activation by glucose (30 mM versus 3 mM) of the pre-proinsulin promoter. However, treatment with extracts from pancreatic islets incubated at either low or high glucose concentrations had no impact on the ability of PDX-1 to interact with importin beta1 in vitro. Furthermore, importin beta1 also interacted with SREBP1c (sterol-regulatory-element-binding protein 1c) in vitro, and microinjection of importin beta1 antibodies blocked the activation by glucose of SREBP1c target genes. Since the subcellular distribution of SREBP1c is unaffected by glucose, these findings suggest that a redistribution of importin beta1 is unlikely to explain the glucose-stimulated nuclear uptake of PDX-1. Instead, we conclude that the uptake of PDX-1 into the nucleoplasm, as glucose concentrations increase, may be mediated by release of the factor both from sites of retention in the cytosol and from non-productive complexes with importin beta1 at the nuclear membrane.


Subject(s)
Cell Nucleus/metabolism , Glucose/pharmacology , Homeodomain Proteins , Islets of Langerhans/metabolism , Trans-Activators/metabolism , Transcription Factors , beta Karyopherins/physiology , Active Transport, Cell Nucleus , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Cells, Cultured , DNA-Binding Proteins/metabolism , Islets of Langerhans/drug effects , Male , Protein Structure, Tertiary , Rats , Rats, Wistar , Sterol Regulatory Element Binding Protein 1 , Trans-Activators/analysis , Trans-Activators/chemistry , Transcription, Genetic
18.
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.

19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL