Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
FASEB J ; 28(9): 4100-10, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24928195

ABSTRACT

The importance of B-isoform of leptin receptor (LEPR-B) signaling in the hypothalamus, pancreas, or liver has been well characterized, but in the intestine, a unique site of entry for dietary nutrition into the body, it has been relatively ignored. To address this question, we characterized a mouse model deficient for LEPR-B specifically in intestinal epithelial cells (IECs). (IEC)LEPR-B-knockout (KO) and wild-type (WT) mice were generated by Cre-Lox strategy and fed a normal or high-fat diet (HFD). The analyses of the animals involved histology and immunohistochemistry of intestinal mucosa, indirect calorimetric measurements, whole-body composition, and expression and activities of nutrient transporters. (IEC)LEPR-B-KO mice exhibited a 2-fold increase in length of jejunal villi and have normal growth on a normal diet but were less susceptible (P<0.01) to HFD-induced obesity. No differences occurred in energy intake and expenditure between (IEC)LEPR-B-WT and -KO mice, but (IEC)LEPR-B-KO mice fed an HFD showed increased excreted fats (P<0.05). Activities of the Na(+)/glucose cotransporter SGLT-1 and GLUT2 were unaffected in LEPR-B-KO jejunum, while GLUT5-mediated fructose transport and PepT1-mediated peptide transport were substantially reduced (P<0.01). These data demonstrate that intestinal LEPR-B signaling is important for the onset of diet-induced obesity. They suggest that intestinal LEPR-B could be a potential per os target for prevention against obesity.


Subject(s)
Diet, High-Fat/adverse effects , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 2/metabolism , Intestinal Mucosa/metabolism , Obesity/etiology , Receptors, Leptin/physiology , Symporters/metabolism , Animals , Blotting, Western , Body Composition , Body Weight , Cell Proliferation , Cells, Cultured , Energy Intake , Female , Glucose Transport Proteins, Facilitative/genetics , Glucose Transporter Type 2/genetics , Glucose Transporter Type 5 , Immunoenzyme Techniques , Intestinal Mucosa/pathology , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Transporter 1 , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Symporters/genetics
3.
Diabetes ; 63(5): 1624-36, 2014 May.
Article in English | MEDLINE | ID: mdl-24430437

ABSTRACT

Aberrations in gut microbiota are associated with metabolic disorders, including obesity. However, whether shifts in the microbiota profile during obesity are a characteristic of the phenotype or a consequence of obesogenic feeding remains elusive. Therefore, we aimed to determine differences in the gut microbiota of obese-prone (OP) and obese-resistant (OR) rats and examined the contribution of this microbiota to the behavioral and metabolic characteristics during obesity. We found that OP rats display a gut microbiota distinct from OR rats fed the same high-fat diet, with a higher Firmicutes-to-Bacteroidetes ratio and significant genera differences. Transfer of OP but not OR microbiota to germ-free (GF) mice replicated the characteristics of the OP phenotype, including reduced intestinal and hypothalamic satiation signaling, hyperphagia, increased weight gain and adiposity, and enhanced lipogenesis and adipogenesis. Furthermore, increased gut permeability through conventionalization resulted in inflammation by proinflammatory nuclear factor (NF)-κB/inhibitor of NF-κB kinase subunit signaling in adipose tissue, liver, and hypothalamus. OP donor and GF recipient animals harbored specific species from Oscillibacter and Clostridium clusters XIVa and IV that were completely absent from OR animals. In conclusion, susceptibility to obesity is characterized by an unfavorable microbiome predisposing the host to peripheral and central inflammation and promoting weight gain and adiposity during obesogenic feeding.


Subject(s)
Gastrointestinal Tract/microbiology , Hypothalamus/metabolism , Microbiota/physiology , Obesity/microbiology , Signal Transduction/physiology , Animals , Eating/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Rats
4.
J Nutr Biochem ; 24(10): 1663-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24041374

ABSTRACT

The gastrointestinal (GI) tract is a specialized sensory system that detects and responds to constant changes in nutrient- and bacterial-derived intestinal signals, thus contributing to controls of food intake. Chronic exposure to dietary fat causes morphological, physiological and metabolic changes leading to disruptions in the regulatory feeding pathways promoting more efficient fat absorption and utilization, blunted satiation signals and excess adiposity. Accumulating evidence demonstrates that impaired gastrointestinal signals following long-term high fat consumption are, at least partially, responsible for increased caloric intake. This review focuses on the role of dietary fat in modulating oral and post-oral chemosensory signaling elements responsible for lipid detection and responses, including changes in sensitivity to satiation signals, such as GLP-1, PYY and CCK and their impact on food intake and weight gain. Furthermore, the influence of the gut microbiota on mechanisms controlling energy regulation in the face of excessive fat exposure will be explored. The profound influence of dietary fats on altering complex regulatory feeding pathways can result in dysregulation of body weight and development of obesity, while restoration or manipulation of satiation signaling may prove an effective tool in prevention and treatment of obesity.


Subject(s)
Dietary Fats/administration & dosage , Gastrointestinal Tract/physiology , Obesity/metabolism , Signal Transduction/drug effects , Adiposity , Animals , CD36 Antigens/physiology , Cholecystokinin/metabolism , Energy Intake/physiology , Gastrointestinal Tract/microbiology , Ghrelin/physiology , Glucagon-Like Peptide 1/drug effects , Glucagon-Like Peptide 1/metabolism , Humans , Intestinal Absorption/drug effects , Microbiota/physiology , Peptide YY/drug effects , Peptide YY/metabolism , Receptors, G-Protein-Coupled/physiology , Satiation/physiology , Taste/physiology , Weight Gain
5.
Diabetes ; 62(7): 2410-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23423571

ABSTRACT

Gastrointestinal mechanisms involved in the suppression of appetite are compromised in obesity. Glucagon-like peptide-1 (GLP-1) is released in response to nutrients, suppresses food intake, and has been shown to play a role in regulation of energy balance. It is not known whether obese-prone (OP) rats exhibit dysfunctional GLP-1 signaling that could contribute to decreased nutrient-induced satiation and hyperphagia. Therefore, we examined the effects of exogenous intraperitoneal administration of the GLP-1R agonist, exendin-4 (Ex-4), on food intake in OP and obese-resistant (OR) rats during chow or high-energy/high-fat (HE/HF) feeding. All doses of Ex-4 effectively suppressed intake in OP and OR rats fed chow; however, during HE/HF-feeding, OP rats suppressed intake significantly less than OR rats at all Ex-4 doses tested. This was associated with downregulation of GLP-1R mRNA expression in the vagal nodose ganglia of OP rats. Furthermore, HE/HF-fed OP rats had significantly lower plasma GLP-1 levels, decreased protein levels of GLP-1 in the intestinal epithelium, and reduced number of L cells in the distal ileum. These results demonstrate that HE/HF-feeding, coupled with OP phenotype, results in reduced endogenous GLP-1 and GLP-1R activation, indicating that impaired GLP-1 signaling during obesity may exacerbate hyperphagia and weight gain.


Subject(s)
Diet, High-Fat , Eating/drug effects , Hypoglycemic Agents/pharmacology , Obesity/metabolism , Peptides/pharmacology , Receptors, Glucagon/agonists , Venoms/pharmacology , Adiposity/drug effects , Animals , Body Weight/drug effects , Down-Regulation/drug effects , Exenatide , Glucagon-Like Peptide-1 Receptor , Ileum/metabolism , Intestinal Mucosa/metabolism , Male , Nodose Ganglion/drug effects , Nodose Ganglion/metabolism , Obesity/genetics , Rats , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism
6.
Gastroenterology ; 144(4): 771-80, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23313268

ABSTRACT

BACKGROUND & AIMS: Glucose is absorbed into intestine cells via the sodium glucose transporter 1 (SGLT-1) and glucose transporter 2 (GLUT2); various peptides and hormones control this process. Apelin is a peptide that regulates glucose homeostasis and is produced by proximal digestive cells; we studied whether glucose modulates apelin secretion by enterocytes and the effects of apelin on intestinal glucose absorption. METHODS: We characterized glucose-related luminal apelin secretion in vivo and ex vivo by mass spectroscopy and immunologic techniques. The effects of apelin on (14)C-labeled glucose transport were determined in jejunal loops and in mice following apelin gavage. We determined levels of GLUT2 and SGLT-1 proteins and phosphorylation of AMPKα2 by immunoblotting. The net effect of apelin on intestinal glucose transepithelial transport was determined in mice. RESULTS: Glucose stimulated luminal secretion of the pyroglutaminated apelin-13 isoform ([Pyr-1]-apelin-13) in the small intestine of mice. Apelin increased specific glucose flux through the gastric epithelial barrier in jejunal loops and in vivo following oral glucose administration. Conversely, pharmacologic apelin blockade in the intestine reduced the increased glycemia that occurs following oral glucose administration. Apelin activity was associated with phosphorylation of AMPKα2 and a rapid increase of the GLUT2/SGLT-1 protein ratio in the brush border membrane. CONCLUSIONS: Glucose amplifies its own transport from the intestinal lumen to the bloodstream by increasing luminal apelin secretion. In the lumen, active apelin regulates carbohydrate flux through enterocytes by promoting AMPKα2 phosphorylation and modifying the ratio of SGLT-1:GLUT2. The glucose-apelin cycle might be pharmacologically handled to regulate glucose absorption and assess better control of glucose homeostasis.


Subject(s)
Carbohydrates/pharmacokinetics , Glucose/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Analysis of Variance , Animals , Biological Transport/drug effects , Biological Transport/physiology , Blotting, Western , Chromatography, Liquid/methods , Disease Models, Animal , Glucose/pharmacology , Glucose Transporter Type 2/metabolism , Immunohistochemistry , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Random Allocation , Reference Values , Sodium-Glucose Transporter 1/metabolism
7.
FASEB J ; 27(4): 1701-10, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23349551

ABSTRACT

The gut microbiota is implicated in host metabolism and energy regulation. Germ-free (GF) C57BL/6 mice display decreased adiposity, an effect associated with increased intestinal fasting-induced adipose factor (FIAF) and decreased hepatic lipogenesis. However, whether the altered metabolism observed in the absence of gut microbiota extends to other species, commonly used to examine energy metabolism, is unknown. Thus, we used the GF Fischer 344 rat to examine adiposity and associated alterations in intestinal nutrient chemoreceptors, gut peptide levels, and FIAF expression, as well as markers of hepatic and adipose lipogenesis and adipogenesis. We found that GF rats displayed similar body weights and adiposity relative to controls. GF state was associated with up-regulation of intestinal and hepatic FIAF, decreased expression of hepatic FAS, ACC-1, and SREBP, and increased pAMPK and pACC. However, GF rats displayed reduced adipocyte FIAF, increased lipogenic enzymes, and decreased pAMPK, accompanied by an increase in adipocyte size. These findings show that, despite increased intestinal FIAF and reduced hepatic lipogenesis, adiposity is preserved in the Fisher 344 GF rat, unlike the C57Bl/6J GF mouse, with a shift in increased adipocyte lipogenesis. This also demonstrates that adipose, rather than intestinal, FIAF may have a more prominent role in adiposity.


Subject(s)
Adipose Tissue/metabolism , Adiposity/physiology , Germ-Free Life/physiology , Intestinal Mucosa/metabolism , Metagenome/physiology , Adipocytes/metabolism , Animals , Body Weight/physiology , Dietary Fats/metabolism , Energy Metabolism/physiology , Intestines/microbiology , Liver/metabolism , Male , Mice , Rats , Rats, Inbred F344 , Up-Regulation/physiology
8.
PLoS One ; 7(6): e39748, 2012.
Article in English | MEDLINE | ID: mdl-22768116

ABSTRACT

Germ-free (GF) mice lacking intestinal microbiota are significantly leaner than normal (NORM) control mice despite consuming more calories. The contribution of microbiota on the recognition and intake of fats is not known. Thus, we investigated the preference for, and acceptance of, fat emulsions in GF and NORM mice, and associated changes in lingual and intestinal fatty acid receptors, intestinal peptide content, and plasma levels of gut peptides. GF and NORM C57Bl/6J mice were given 48-h two-bottle access to water and increasing concentrations of intralipid emulsions. Gene expression of the lingual fatty acid translocase CD36 and protein expression of intestinal satiety peptides and fatty-acid receptors from isolated intestinal epithelial cells were determined. Differences in intestinal enteroendocrine cells along the length of the GI tract were quantified. Circulating plasma satiety peptides reflecting adiposity and biochemical parameters of fat metabolism were also examined. GF mice had an increased preference and intake of intralipid relative to NORM mice. This was associated with increased lingual CD36 (P<0.05) and decreased intestinal expression of fatty acid receptors GPR40 (P<0.0001), GPR41 (P<0.0001), GPR43 (P<0.05), and GPR120 (P<0.0001) and satiety peptides CCK (P<0.0001), PYY (P<0.001), and GLP-1 (P<0.001). GF mice had fewer enteroendocrine cells in the ileum (P<0.05), and more in the colon (P<0.05), relative to NORM controls. Finally, GF mice had lower levels of circulating leptin and ghrelin (P<0.001), and altered plasma lipid metabolic markers indicative of energy deficits. Increased preference and caloric intake from fats in GF mice are associated with increased oral receptors for fats coupled with broad and marked decreases in expression of intestinal satiety peptides and fatty-acid receptors.


Subject(s)
Intestinal Mucosa/metabolism , Intestines/microbiology , Lipid Metabolism , Metagenome , Mouth/metabolism , Signal Transduction , Animals , Blood Glucose/metabolism , Body Weight , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Count , Emulsions , Energy Intake , Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Gastrointestinal Hormones/blood , Gene Expression Regulation , Germ-Free Life , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Oils , Peptides/metabolism
9.
Am J Physiol Gastrointest Liver Physiol ; 299(1): G179-85, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20448142

ABSTRACT

L-glutamine is the primary metabolic fuel for enterocytes. Glutamine from the diet is transported into the absorptive cells by two sodium-dependent neutral amino acid transporters present at the apical membrane: ASCT2/SLC1A5 and B(0)AT1/SLC6A19. We have demonstrated that leptin is secreted into the stomach lumen after a meal and modulates the transport of sugars after binding to its receptors located at the brush border of the enterocytes. The present study was designed to address the effect of luminal leptin on Na(+)-dependent glutamine (Gln) transport in rat intestine and identify the transporters involved. We found that 0.2 nM leptin inhibited uptake of Gln and phenylalanine (Phe) (substrate of B(0)AT1) using everted intestinal rings. In Ussing chambers, 10 mM Gln absorption followed as Na(+)-induced short-circuit current was inhibited by leptin in a dose-dependent manner (maximum inhibition at 10 nM; I(C50) = approximately 0.1 nM). Phe absorption was also decreased by leptin. Western blot analysis after 3-min incubation of the intestinal loops with 10 mM Gln, showed marked increase of ASCT2 and B(0)AT1 protein in the brush-border membrane that was reduced by rapid preincubation of the intestinal lumen with 1 nM leptin. Similarly, the increase in ASCT2 and B(0)AT1 gene expression induced by 60-min incubation of the intestine with 10 mM Gln was strongly reduced after a short preincubation period with leptin. Altogether these data demonstrate that, in rat, leptin controls the active Gln entry through reduction of both B(0)AT1 and ASCT2 proteins traffic to the apical plasma membrane and modulation of their gene expression.


Subject(s)
Amino Acid Transport System ASC/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Glutamine/metabolism , Intestine, Small/metabolism , Leptin/metabolism , Amino Acid Transport System ASC/genetics , Amino Acid Transport Systems, Neutral/genetics , Animals , Biological Transport , Gene Expression Regulation , In Vitro Techniques , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Male , Membrane Potentials , Minor Histocompatibility Antigens , Peptides/pharmacology , Phenylalanine/metabolism , Protein Transport , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Leptin/antagonists & inhibitors , Receptors, Leptin/metabolism , Recombinant Proteins/metabolism , Serous Membrane/metabolism , Sodium/metabolism , Time Factors
10.
PLoS One ; 4(11): e7935, 2009 Nov 30.
Article in English | MEDLINE | ID: mdl-19956534

ABSTRACT

BACKGROUND AND AIMS: The small intestine is the major site of absorption of dietary sugars. The rate at which they enter and exit the intestine has a major effect on blood glucose homeostasis. In this study, we determine the effects of luminal leptin on activity/expression of GLUT2 and GLUT5 transporters in response to sugars intake and analyse their physiological consequences. METHODOLOGY: Wistar rats, wild type and AMPKalpha(2) (-/-) mice were used. In vitro and in vivo isolated jejunal loops were used to quantify transport of fructose and galactose in the absence and the presence of leptin. The effects of fructose and galactose on gastric leptin release were determined. The effects of leptin given orally without or with fructose were determined on the expression of GLUT2/5, on some gluconeogenesis and lipogenic enzymes in the intestine and the liver. PRINCIPAL FINDINGS: First, in vitro luminal leptin activating its receptors coupled to PKCbetaII and AMPKalpha, increased insertion of GLUT2/5 into the brush-border membrane leading to enhanced galactose and fructose transport. Second in vivo, oral fructose but not galactose induced in mice a rapid and potent release of gastric leptin in gastric juice without significant changes in plasma leptin levels. Moreover, leptin given orally at a dose reproducing comparable levels to those induced by fructose, stimulated GLUT5-fructose transport, and potentiated fructose-induced: i) increase in blood glucose and mRNA levels of key gluconeogenesis enzymes; ii) increase in blood triglycerides and reduction of mRNA levels of intestinal and hepatic Fasting-induced adipocyte factor (Fiaf) and iii) increase in SREBP-1c, ACC-1, FAS mRNA levels and dephosphorylation/activation of ACC-1 in liver. CONCLUSION/SIGNIFICANCE: These data identify for the first time a positive regulatory control loop between gut leptin and fructose in which fructose triggers release of gastric leptin which, in turn, up-regulates GLUT5 and concurrently modulates metabolic functions in the liver. This loop appears to be a new mechanism (possibly pathogenic) by which fructose consumption rapidly becomes highly lipogenic and deleterious.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 5/metabolism , Leptin/metabolism , Liver/metabolism , Animals , Blood Glucose/metabolism , Fructose/metabolism , Gluconeogenesis , Homeostasis , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...