Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Gene Ther ; 21(7): 638-44, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24784449

ABSTRACT

Leptin targets specific receptors (OB-R) expressed in the hypothalamus to regulate energy balance. Leptin decreases food intake in normal weight individuals, but this effect is blunted in obese subjects who are characterized by a state of leptin resistance. The prevention of leptin resistance is one of the major goals of obesity research. Recently, we identified endospanin 1 as a negative regulator of OB-R, which by interacting with OB-R retains the receptor inside the cell. We show here that in obese mice endospanin 1 is upregulated in the hypothalamic arcuate nucleus (ARC), the major brain structure involved in body weight regulation, suggesting that endospanin 1 is implicated in obesity development and/or the installation of leptin resistance. In contrast, silencing of endospanin 1 with lentiviral vectors in the ARC of obese mice fully restores leptin responsiveness when combined with a switch to ad libitum fed chow diet. The recovery of central leptin sensitivity is accompanied by sustained body weight loss and amelioration of blood lipid parameters and steatosis. Collectively, our results define endospanin 1 as a novel therapeutic target against obesity.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Carrier Proteins/metabolism , Leptin/metabolism , Obesity/metabolism , Animals , Carrier Proteins/genetics , Diet, High-Fat , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Obese , STAT3 Transcription Factor/metabolism , Weight Loss
2.
Int J Obes (Lond) ; 37(3): 375-81, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22546775

ABSTRACT

BACKGROUND AND AIMS: Diet-induced obesity (DIO) is an excellent model for examining human obesity comprising both genotypic and environmental (diet) factors. Decreased responsiveness to peripheral satiety signaling may be responsible for the hyperphagia in this model. In this study, we investigated responses to nutrient-induced satiation in outbred DIO and DIO-resistant (DR) rats fed a high-energy/high-fat (HE/HF) diet as well as intestinal satiety peptide content, intestinal nutrient-responsive receptor abundance and vagal anorectic receptor expression. METHODS: Outbred DIO and DR rats fed a HE/HF diet were tested for short-term feeding responses following nutrient (glucose and intralipid (IL)) gastric loads. Gene and protein expressions of intestinal satiety peptides and fatty acid-responsive receptors were examined from isolated proximal intestinal epithelial cells and cholecystokinin-1 receptor (CCK-1R) and leptin receptor (LepR) mRNA from the nodose ganglia of DIO and DR animals. RESULTS: DIO rats were less responsive to IL- (P<0.05) but not glucose-induced suppression of food intake compared with DR rats. DIO rats exhibited decreased CCK, peptide YY (PYY) and glucagon-like peptide-1 (GLP-1; P<0.05 for each) protein expression compared with DR rats. Also, DIO rats expressed more G-protein-coupled receptor 40 (GPR40; P<0.0001), GPR41 (P<0.001) and GPR120 (P<0.01) relative to DR rats. Finally, there were no differences in mRNA expression for CCK-1R and LepR in the nodose ganglia of DIO and DR rats. CONCLUSIONS: Development of DIO may be partly due to decreased fat-induced satiation through low levels of endogenous satiety peptides, and changes in intestinal nutrient receptors.


Subject(s)
Enteroendocrine Cells/metabolism , Gastric Mucosa/metabolism , Gastrointestinal Tract/metabolism , Obesity/metabolism , Satiation , Sensory Receptor Cells/metabolism , Animals , Diet, High-Fat , Eating , Energy Intake , Gastric Mucosa/pathology , Gastrointestinal Tract/pathology , Male , Obesity/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction
3.
Br J Nutr ; 107(5): 621-30, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21781379

ABSTRACT

The chemosensory components shared by both lingual and intestinal epithelium play a critical role in food consumption and the regulation of intestinal functions. In addition to nutrient signals, other luminal contents, including micro-organisms, are important in signalling across the gastrointestinal mucosa and initiating changes in digestive functions. A potential role of gut microbiota in influencing food intake, energy homeostasis and weight gain has been suggested. However, whether gut microbiota modulates the expression of nutrient-responsive receptors and transporters, leading to altered food consumption, is unknown. Thus, we examined the preference for nutritive (sucrose) and non-nutritive (saccharin) sweet solutions in germ-free (GF, C57BL/6J) mice compared with conventional (CV, C57BL/6J) control mice using a two-bottle preference test. Then, we quantified mRNA and protein expression of the sweet signalling protein type 1 taste receptor 3 (T1R3) and α-gustducin and Na glucose luminal transporter-1 (SGLT-1) of the intestinal epithelium of both CV and GF mice. Additionally, we measured gene expression of T1R2, T1R3 and α-gustducin in the lingual epithelium. We found that, while the preference for sucrose was similar between the groups, GF mice consumed more of the high concentration (8 %) of sucrose solution than CV mice. There was no difference in either the intake of or the preference for saccharin. GF mice expressed significantly more T1R3 and SGLT-1 mRNA and protein in the intestinal epithelium compared with CV mice; however, lingual taste receptor mRNA expression was similar between the groups. We conclude that the absence of intestinal microbiota alters the expression of sweet taste receptors and GLUT in the proximal small intestine, which is associated with increased consumption of nutritive sweet solutions.


Subject(s)
Dietary Sucrose/administration & dosage , Food Preferences , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Receptors, G-Protein-Coupled/metabolism , Sodium-Glucose Transporter 1/metabolism , Up-Regulation , Animals , Appetite Regulation , Down-Regulation , Duodenum , Germ-Free Life , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Jejunum , Male , Mice , Mice, Inbred C57BL , Mouth Mucosa/metabolism , Mouth Mucosa/microbiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Saccharin/administration & dosage , Sodium-Glucose Transporter 1/genetics , Tongue
4.
Physiol Behav ; 99(1): 109-17, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-19887078

ABSTRACT

CCK-1 receptor deficient Otsuka Long Evans Tokushima Fatty (OLETF) rats are hyperphagic, which leads to subsequent obesity and diabetes. Additionally, they have increased sham intake and enhanced preference for sucrose solutions relative to control, Long Evans Tokushima Otsuka (LETO) rats. To determine the effects of oil on ingestion, we first measured real feeding of various concentrations of oil emulsions (12.5, 25, 50, 75, and 100%) in rats that were fed ad libitum. Secondly, to isolate the orosensory compontent of oils from post-ingestive consequences, as well as determine the contribution of energy status, we measured sham feeding in OLETF and LETO rats using one-bottle acceptance tests while non-deprived and overnight food deprived. Finally, to assess the orosensory effects of nutritive and non-nutritive oils, we used two-bottle preference tests in sham fed OLETF and LETO rats. We found that real feeding resulted in increased intake of high oil concentrations for OLETF rats relative to LETO rats. Similarly, OLETF rats consumed significantly more of higher concentration corn oils than LETO while non-deprived sham feeding. Conversely, OLETF rats overconsumed low concentration corn oil compared to LETO during overnight deprived sham-feeding tests. In two-bottle sham-feeding preference tests, both non-deprived OLETF and LETO rats preferred corn to mineral oil. Collectively, these results show that increased oil intake in OLETF rats is driven by both peripheral deficits to satiation and altered orosensory sensitivity.


Subject(s)
Food Preferences/physiology , Oils/administration & dosage , Receptor, Cholecystokinin A/deficiency , Taste/drug effects , Taste/physiology , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Feeding Behavior/physiology , Food Deprivation/physiology , Gastric Emptying/physiology , Glucose Tolerance Test , Rats , Rats, Inbred OLETF , Rats, Transgenic , Sucrose/administration & dosage , Sweetening Agents/administration & dosage
5.
Brain Res ; 1308: 79-86, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-19857467

ABSTRACT

Deficits in satiation signals are strongly suspected of accompanying obesity and contributing to its pathogenesis in both humans and rats. One such satiation signal is cholecystokinin (CCK), whose effects on food intake are diminished in animals adapted to a high fat diet. In this study, we tested the hypothesis that diet-induced obese prone (OP) rats exhibit altered feeding and vagal responses to systemic (IP) administration of CCK-8 compared to diet-induced obese resistant (OR) rats. We found that CCK (4.0 microg/kg) suppressed food intake significantly more in OP than OR rats. To determine whether enhanced suppression of feeding is accompanied by altered vagal sensory responsiveness, we examined dorsal hindbrain expression of Fos-like immunoreactivity (Fos-Li) following IP CCK injection in OP and OR rats. After 4.0 microg/kg CCK, there were significantly more Fos-positive nuclei in the NTS of OP compared to OR rats. Treatment with 8.0 microg/kg CCK resulted in no significant difference in food intake or in Fos-Li between OP and OR rats. Also, we found that OP rats were hyperphagic on a regular chow diet and gained more weight compared to OR rats. Finally OP rats had decreased relative fat pad mass compared to OR rats. Collectively, these results show that OP rats exhibit a different behavioral and vagal neuronal responses to CCK than OR rats.


Subject(s)
Cholecystokinin/administration & dosage , Eating/drug effects , Feeding Behavior/drug effects , Neurons/drug effects , Rhombencephalon/drug effects , Adiposity/physiology , Analysis of Variance , Animals , Body Weight/physiology , Cell Count , Cholecystokinin/metabolism , Diet , Hyperphagia/metabolism , Immunohistochemistry , Neurons/metabolism , Obesity/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Rhombencephalon/metabolism , Satiety Response/drug effects , Time Factors , Vagus Nerve/drug effects , Vagus Nerve/metabolism
6.
Brain Res ; 1266: 37-44, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19232331

ABSTRACT

Previous work has shown that blockade of NMDAR by non-competitive (MK-801) and competitive (AP5) antagonists increase food intake by acting in the dorsal hindbrain. NMDAR are heteromeric complexes composed of NR1, NR2 and NR3 subunits. Competitive NR2B antagonists potently increase feeding when injected into the hindbrain. NR2 immunoreactivity is present in the hindbrain, vagal afferents and enteric neurons. NMDA receptors expressed on peripheral vagal afferent processes in the GI tract modulate responsiveness to GI stimuli. Therefore, it is possible that peripheral as well as central vagal NMDA receptors participate in control of food intake. To examine this possibility, we recorded intake of rodent chow, a palatable liquid food (15% sucrose), and non-nutrient (0.2% saccharin) solutions following intraperitoneal (i.p.) administration of D-CPPene, a competitive NMDA receptor antagonist that is selective for binding to the NR2B/A channel subunit. To assess participation of peripheral NMDA receptors in postoral satiation signals, we examined the ability of D-CPPene to attenuate reduction of feeding and hindbrain Fos expression following IP CCK administration. IP D-CPPene (2, 3 mg/kg) produced a significant increase in sucrose and chow intake but not saccharin. Pretreatment with D-CPPene (2 mg/kg) reversed CCK (2 microg/kg)-induced inhibition of sucrose intake, and attenuated CCK-induced Fos-Li in the dorsal hindbrain. These results confirm that antagonism of hindbrain NMDA receptors increases food intake. In addition our results suggest that NMDA receptors outside the hindbrain, perhaps in the periphery, participate in vagally mediated, CCK-induced reduction of food intake and NTS neuron activation.


Subject(s)
Appetite Regulation/physiology , Eating , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Rhombencephalon/physiology , Sincalide/pharmacology , Analysis of Variance , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/pharmacology , Appetite Regulation/drug effects , Excitatory Amino Acid Antagonists/administration & dosage , Injections, Intraperitoneal , Male , Piperazines/administration & dosage , Piperazines/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Saccharin , Satiation/physiology , Sincalide/administration & dosage , Sucrose
SELECTION OF CITATIONS
SEARCH DETAIL
...