Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Obes Rev ; 23(7): e13478, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35610763
2.
Brain Res ; 1537: 191-200, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24076449

ABSTRACT

Insulin injections into the central nucleus of the amygdala (CeA) inhibit food intake but this response is lost quickly on feeding a high fat diet. The purpose of the studies described in this manuscript was to identify the potential mechanism for the development of this insulin resistance. High fat diets (HFD) induced PKCθ activation and blocked the stimulation of Akt but not mTOR phosphorylation in the amygdala in response to CeA insulin injections. Infusions of palmitic acid onto the CeA had identical effects to HFD on PKCθ expression and insulin signaling in the amygdala. CeA insulin also induced an increase in Akt phosphorylation in the hypothalamus but had no effect on hypothalamic mTOR phosphorylation. Feeding HFD but not CeA palmitate infusions reversed the hypothalamic Akt signaling response to CeA insulin. These data, which show the independence of Akt and mTOR signaling responses to insulin in the amygdala and the effect of insulin signaling in the CeA on hypothalamic Akt signaling, suggest that the amygdala might also have a significant role in regulating hypothalamic responses to dietary fat.


Subject(s)
Amygdala/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Signal Transduction , Animal Feed , Animals , Dietary Fats/metabolism , Insulin Resistance , Male , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/physiology
3.
Obesity (Silver Spring) ; 21(4): 755-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23712979

ABSTRACT

OBJECTIVE: To investigate the signaling mechanisms that might underlie the loss of anorectic response to insulin injections into the central nucleus of the amygdala (CeA) within 3 days of feeding a high fat diet. DESIGN AND METHODS: Protein samples from amygdala and hypothalamus of rats fed high or low fat diets were subjected to a phosphorylation screening assay. The effects of dietary fat intake on the expression and activation of protein kinase C theta (PKCθ) in brain regions was studied. Finally, lentiviral vectors were used to overexpress rat PKCθ unilaterally or bilaterally into the CeA of rats and the effects on food intake, body weight and insulin stimulation of Akt phosphorylation were studied. RESULTS: The level of pMARCKS (Myristoylated alanine-rich C-kinase substrate), a major substrate of PKCθ, was increased 116% in amygdala of high fat diet fed rats but reduced in the hypothalamus. High fat diets increased the level of PKCθ in a region specific manner in the brain and this PKCθ was activated by membrane association. Overexpressing rat PKCθ either unilaterally or bilaterally into the CeA inhibited insulin stimulation of Akt signaling and blocked the anorectic response to insulin injected into the amygdala. Bilaterally injected PKCθ rats gained more weight and body fat and had increased food intake when fed a high fat diet compared to the control rats that received a lentiviral-Green Fluorescent Protein construct. CONCLUSION: The data suggest that insulin may have a physiological role within the amygdala to regulate energy balance.


Subject(s)
Amygdala/metabolism , Body Weight , Energy Intake , Insulin/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Adipose Tissue/metabolism , Amygdala/cytology , Amygdala/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Diet, Fat-Restricted , Diet, High-Fat , Dietary Fats/administration & dosage , Genetic Vectors , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , Insulin Resistance , Isoenzymes/genetics , Lentivirus/genetics , Male , Phenotype , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C-theta , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
4.
Brain Res ; 1498: 85-94, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23295188

ABSTRACT

The increase in PKCθ expression in the amygdala of rats fed high fat diet (HFD) has been related to the loss of the anorectic response to insulin injections into the central nucleus of the amygdala (CeA) in these animals. PKCθ overexpression in the CeA increases food intake, body weight and body fat and inhibits insulin stimulation of Akt signaling. To study the effects of bilateral overexpression of PKCθ in the CeA of rats on peripheral metabolism, rats were injected into the CeA or 3rd ventricle with a lentiviral (LV)-PKCθ construct or LV-Green fluorescent protein (GFP) construct as a control and fed either LFD or HFD. Insulin and glucose tolerance tests were undertaken and hepatic AMPK activation, Pepck, Srebp1c gene expression and lipid levels assayed. CeA LV-PKCθ injected rats increased food intake, body weight and body fat and increased hepatic, but not serum, triglyceride levels compared to control rats that received a CeA-LV-GFP construct. Hepatic AMP-kinase activity was reduced but expression of Pepck increased while serum insulin decreased, glucose tolerance improved and the hypoglycemic response to insulin was enhanced in CeA LV-PKCθ injected rats. In contrast, rats that received LV-PKCθ injections into the 3rd Ventricle did not show any changes in food intake or body weight although serum, but not hepatic, triglyceride levels were increased and glucose tolerance was impaired. The data suggest that activation of PKCθ in the CeA and hypothalamus have different effects on energy balance and peripheral metabolism and that insulin signaling in the amygdala regulates peripheral metabolism.


Subject(s)
Central Amygdaloid Nucleus/enzymology , Glucose/metabolism , Homeostasis/physiology , Hypothalamus/enzymology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Adenylate Kinase/metabolism , Adipose Tissue/physiology , Animals , Body Weight/physiology , Diet, Fat-Restricted , Diet, High-Fat , Eating/physiology , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/genetics , Lentivirus/genetics , Liver/metabolism , Male , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Protein Kinase C/genetics , Protein Kinase C-theta , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
5.
Physiol Behav ; 106(2): 158-63, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22336738

ABSTRACT

We have previously shown that voluntary wheel running activity in mice is associated with an increase in the Endoplasmic Reticulum (ER) Unfolded Protein stress response in multiple regions of the brain. Mice that are given access to running wheels show large variations in individual running activity. In contrast, when food is placed on the lid of their cages, rather than within the cage, all mice must undertake significant physical activity in order to gain access to their food. Hence we investigated the effects of food location on food intake and growth of C57BL/6 mice and on the activity of the ER stress system in the brain as reflected in the expression of two marker genes, Xbp1 and Atf6. Mice that had food in cups within their cages and allowed access to running wheels showed the anticipated changes in food intake, body weight and ER stress in the hippocampus compared to mice with no access to running wheels. Locating the food on the lid had no effect on food intake but reduced weight gain significantly. Likewise, locating food on the lid increased the expression of both Xbp1 and Atf6 in the hippocampi in the absence of any running wheel activity. Voluntary wheel running activity was reduced in mice whose food was located on the cage lid and this running actually reduced the expression of the two marker ER stress genes. We conclude that the usual practice of providing food for mice on their cage lids provides a significant level of physical activity that alters the metabolic status and increases ER stress. As such, this may not be the optimal model for the majority of mouse studies that are reported in the literature and it may significantly alter the interpretation of the effect of wheel running activity on ER stress. The differential effects of food location on hippocampal Bdnf gene expression also suggest that BDNF does not directly regulate UPR activity but may be coordinately regulated in response to running activity.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Energy Metabolism/physiology , Feeding Behavior/physiology , Hippocampus/metabolism , Physical Exertion/physiology , Running/physiology , Activating Transcription Factor 6/biosynthesis , Animals , Body Weight , Brain-Derived Neurotrophic Factor/biosynthesis , DNA-Binding Proteins/biosynthesis , Eating , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/biosynthesis , Male , Mice , Mice, Inbred C57BL , Motor Activity , Regulatory Factor X Transcription Factors , Transcription Factors/biosynthesis , X-Box Binding Protein 1 , eIF-2 Kinase/biosynthesis
6.
Diabetes ; 60(1): 97-106, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980461

ABSTRACT

OBJECTIVE: The agouti-related protein (Agrp) is a powerful orexigenic peptide, but little is known about its transcriptional regulation. The objective of this study was to determine molecular mechanisms for the activation of hypothalamic Agrp and identify compounds that stimulate appetite. RESEARCH DESIGN AND METHODS: We used promoter analyses methods, hypothalamic cell culture and transfection, immunohistochemistry, luciferase-expressing transgenic mice, in vivo bioluminescence, anitisense RNA, mouse feeding studies, indirect calorimetry, real-time PCR, and Western blots. RESULTS: We found that the Krüppel-like factor 4 (Klf4) is a potent activator of Agrp by binding to a specific CACCC-box in its minimal promoter. We also found that an extract of tarragon, termed PMI-5011, activated hypothalamic Klf4 and Agrp. In vivo, PMI-5011 increased Agrp promoter activity in luciferase-expressing transgenic mice, increased hypothalamic Klf4 and Agrp expression, increased hypothalamic Orexin and melanin-concentrating hormone, increased food intake, reduced circulating insulin and leptin levels, attenuated energy expenditure, and enhanced body weight but only when using a high-fat diet. CONCLUSIONS: These data show that Klf4 augmented hypothalamic Agrp by binding to a specific CACCC-box onto its minimal promoter. In addition, the tarragon extract PMI-5011 activated Klf4 and orexigenic neuropeptides and reduced peripheral insulin and leptin levels leading to positive energy balance.


Subject(s)
Agouti-Related Protein/genetics , Appetite/physiology , Animal Feed , Animals , Body Composition , Calorimetry, Indirect/methods , DNA Primers , Energy Intake , Energy Metabolism , Feeding Behavior , Insulin/blood , Kruppel-Like Factor 4 , Leptin/blood , Luciferases/genetics , Mice , Mice, Transgenic , Polymerase Chain Reaction/methods , Promoter Regions, Genetic , RNA/genetics , RNA/isolation & purification , Transfection
7.
Brain Res ; 1349: 48-55, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20599830

ABSTRACT

Studies have demonstrated defects of DA and 5HT neurotransmission in dietary fat induced obese animals. In the present study, we used a perfusion system to assay the release of DA and 5HT from striatal slices preloaded with [(3)H]-DA or [(3)H]-5HT. The release of both DA and 5HT from striatal slices of rats fed a high fat diet for 10 days, but not 3 days, was reduced when compared to striatal slices taken from rats fed a low fat diet. Enterostatin, an endogenous pentapeptide inhibits dietary fat intake when administered peripherally and centrally in animals. The central mechanism for the action of enterostatin is not yet determined even though several mechanisms have been suggested. We have shown that enterostatin enhanced [(3)H]-DA release, but not [(3)H]-5HT release from striatal slices of rats that had been adapted to high fat diet for 10 days. The enterostatin-induced increase in [(3)H]-DA release was blocked by nomifensine. Enterostatin did not alter [(3)H]-DA or [(3)H]-5HT release from striatal slices of rats adapted to high fat or low fat diet feeding for 3 days. These findings suggest that enterostatin may inhibit dietary fat intake by blocking dopamine reuptake transport to increase central striatal DA release from rats that have acquired diminished dopamine signal after an adaptive period of fat consumption.


Subject(s)
Colipases/pharmacology , Corpus Striatum/drug effects , Dietary Fats/pharmacology , Dopamine/metabolism , Enzyme Precursors/pharmacology , Serotonin/metabolism , Analysis of Variance , Animals , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Tritium/metabolism
8.
Brain Res ; 1317: 13-23, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20045396

ABSTRACT

The unfolded protein response (UPR) is a dynamic cellular mechanism for reducing endoplasmic reticulum (ER) stress. ER stress occurs from a variety of causes such as nutritional deprivation or over-nutrition, expression of misfolded or mutant proteins and increased synthesis of secretory protein. Obesity induced by over-nutrition has been associated with ER stress. Although exercise has a beneficial effect in opposing the development of obesity and neurodegenerative diseases, there have been no studies on the effect of exercise on ER stress in the brain induced by over-nutrition. We have taken advantage of the substantial individual differences in voluntary running activity among inbred C57BL/6 mice to investigate the relation between ER stress within regions of the brain and voluntary running activity in mice fed on either a low fat or high fat diet while maintained individually in cages with running wheels. Mice were divided into three groups depending on their voluntary running level and compared with a sedentary group. ER stress was assayed by real-time PCR and Western blots of the UPR pathway markers Xbp1, PERK, eIF2alpha, Hspa5 and ATF6. Three weeks of HFD had little effect on ER stress in the brain of the sedentary group compared to animals fed the LFD. Higher voluntary running activity was associated with increased ER stress in the hypothalamus, hippocampus and cortex. The responses were largest in the hypothalamus. The increase in the UPR response in response to exercise did not induce apoptotic signals and may thus contribute to the protective effect of exercise in preventing neurodegenerative disease.


Subject(s)
Brain/physiology , Diet , Endoplasmic Reticulum/physiology , Physical Conditioning, Animal/physiology , Stress, Physiological/physiology , Animals , Body Composition , Body Weight , Cerebral Cortex/physiology , Dietary Fats , Eating , Endoplasmic Reticulum Chaperone BiP , Hippocampus/physiology , Hypothalamus/physiology , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , Volition
9.
Am J Physiol Regul Integr Comp Physiol ; 298(2): R385-93, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923360

ABSTRACT

The amygdala is rich in melanocortin 4 receptors. Because the reduction in dietary fat intake after enterostatin is injected in the central nucleus of the amygdala (CeA) is blocked by a melanocortin 4 receptor antagonist, we investigated the role of melanocortin activity in the CeA in regulating food intake and macronutrient choice. Sprague-Dawley rats, fitted with CeA cannulas, were fed either chow, a high-fat (HF) diet, or adapted to a two-choice HF or low-fat (LF) diet. Injections of the MC4R agonist melanotan II (MTII) in the CeA had a dose-dependent inhibitory effect on food intake that lasted for at least 24 h. This response was greater in rats fed a HF diet. The inverse agonist agouti-related protein (AgRP) and antagonist SHU-9119 increased food intake in a dose-dependent manner, with the hyperphagia lasting for 60 h. In rats adapted to a two-choice HF/LF diet, MTII decreased HF consumption but had no effect on LF consumption, resulting in a long-lasting decrease in total calorie intake (-35.5% after 24 h, P < 0.05). Total calorie intake increased in both AgRP- and SHU-9119-treated rats (32 and 109% after 24 h, respectively) as the result of increased intake of HF diet. There was no modification of LF consumption with AgRP treatment and a transient nonsignificant decrease with SHU-9119 treatment. Amygdala brain-derived neurotrophic factor expression was increased by AgRP in fed rats. These results identify the amygdala as a site of action for the melanocortin system to control food intake and dietary preferences.


Subject(s)
Amygdala/physiology , Appetite Regulation/physiology , Dietary Fats , Melanocortins/physiology , Agouti-Related Protein/pharmacology , Amygdala/metabolism , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Dose-Response Relationship, Drug , Eating/drug effects , Food Preferences/drug effects , Male , Melanocortins/pharmacology , Peptides, Cyclic/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
10.
Am J Physiol Regul Integr Comp Physiol ; 297(5): R1302-11, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726717

ABSTRACT

Intracerebroventricular insulin decreases food intake (FI). The central bed nucleus of the amygdala (CeA), as other regions of the brain regulating feeding behavior, expresses insulin receptors. Our objectives were to show an insulin anorectic response in the amygdala, study the effect of high-fat diets on this response, and map the neural network activated by CeA insulin using c-Fos immunohistochemistry. Sprague-Dawley (SD) rats fitted with unilateral CeA cannulas were adapted to a low-fat (LFD) diet before they were fed a high-fat diet (HFD). Their feeding response to CeA saline or insulin (8 mU) was tested after 24 h, 72 h, or 7 days of being on a HFD. In a second experiment, SD rats were fed the HFD for 3, 7, or 49 days and were then refed with the LFD. They were tested for their insulin response before and after an HFD and every 3 days for the following weeks. Insulin tolerance tests were performed in a parallel group of rats. The CeA insulin stimulation c-Fos expression was studied to identify the distribution of activated neuronal populations. Feeding an HFD for 72 h or more induced a CeA, but not peripheral, insulin resistance, which was slowly reversed by LFD refeeding. The duration of HFD feeding determined the time frame for reversal of the insulin resistance. CeA insulin increased c-Fos in multiple brain regions, including the arcuate nucleus/paraventricular nucleus region of the hypothalamus. We conclude that the amygdala may be an important site for insulin regulation of food intake and may have a significant role in determining susceptibility to HFD-induced obesity.


Subject(s)
Amygdala/physiopathology , Anorexia/chemically induced , Anorexia/physiopathology , Dietary Fats/pharmacology , Hypoglycemic Agents/adverse effects , Insulin/adverse effects , Amygdala/metabolism , Animals , Anorexia/metabolism , Blood Glucose/metabolism , Diet, Fat-Restricted , Disease Models, Animal , Dose-Response Relationship, Drug , Eating/physiology , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance/physiology , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
11.
Peptides ; 30(10): 1866-73, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19563849

ABSTRACT

Enterostatin is a peptide that regulates dietary fat intake in rodents and inhibits insulin secretion from pancreatic beta cells. Microarray studies of the genomic response of both a human hepatoma cell line (HepG2 cells) and a mouse hypothalamic cell line (GT1-7 cells) to enterostatin suggested that it might regulate protein trafficking. Using semi-quantitative real-time PCR and Western blot analysis, we confirmed that enterostatin upregulated Scamp2 and down regulated Dynamin2 in these cell lines. The receptor for enterostatin is the F1-ATPase beta subunit. We transfected HepG2 cells with either a green fluorescent protein (GFP) tagged F1-ATPase beta subunit or a red fluorescent protein (RFP) tagged F1-ATPase alpha subunit to study the effects of enterostatin on translocation of its own receptor protein. Enterostatin induced movement of GFP-beta subunit to the cell periphery area but did not have any effect on the localization of RFP-alpha subunit protein in HepG2. As Scamp2 is involved in glucose uptake in mouse Beta-TC6 insulinoma cells we tested enterostatin's effect in Beta-TC6 cells. Glucose stimulated insulin release was inhibited by enterostatin as reported previously. Using siRNA to Scamp2 did not change glucose stimulated insulin release but siRNA to Dynamin2 and dominant negative Dynamin2 (Dyn K44A) inhibited glucose stimulated insulin release and abolished the response to enterostatin. This suggests enterostatin inhibits glucose stimulated insulin release in pancreatic beta cells through down regulation of Dynamin2. This study also suggests that enterostatin might have a more generalized effect on protein trafficking in various cells.


Subject(s)
Colipases/metabolism , Enzyme Precursors/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Protein Transport/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Dynamin II/genetics , Dynamin II/metabolism , Glucose/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/cytology , Mice , Microarray Analysis , Protein Subunits/genetics , Protein Subunits/metabolism , Proton-Translocating ATPases/genetics , Proton-Translocating ATPases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology
12.
Peptides ; 30(2): 181-90, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19059445

ABSTRACT

Enterostatin, a gut-brain peptide, inhibits dietary fat intake in rats. The purpose of this study was to identify the intracellular signaling pathways that are responsive to enterostatin and that modulate the effects of enterostatin on the expression of Agouti-related protein (AgRP). We used the hypothalamic GT1-7 neuronal cell line to identify the effects of enterostatin on cyclic AMP and ERK signaling using conventional immunoassays or Western blots to assay the activity of these pathways. Enterostatin enhanced the level of cyclic AMP, PKA(RIIbeta) and phospho-CREB and increased pERK levels in GT 1-7 cells. The effects on pERK were rapid (7.5 min) and dose-dependent. These signaling responses were blocked by an antibody to the enterostatin receptor (beta subunit of F1-ATPase), by the pERK inhibitor U0126 and by the P2Y receptor antagonist Suramin. Enterostatin showed a biphasic effect on AgRP mRNA, initially increasing but subsequently decreasing the levels. The cyclic AMP activator Sp-cAMP increased AgRP mRNA expression. Transfection of a wild type ERK construct reduced AgRP mRNA levels. Enterostatin inhibited expression of Krüppel-like factor 4 (KLF4), a transcriptional regulator of AgRP. KLF4 gene expression was increased by Sp-cAMP but decreased by wild-type ERK expression. U0126 blocked the effect of enterostatin on KLF4 expression. We conclude that enterostatin binding to its receptor activates the pERK pathway to inhibit AgRP gene expression but may enhance AgRP expression through activation of the cyclic AMP pathway. These pathways probably mediate the enterostatin inhibition of dietary fat intake.


Subject(s)
Agouti-Related Protein/genetics , Colipases/pharmacology , Cyclic AMP/metabolism , Enzyme Precursors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Signal Transduction , Adenylate Kinase/metabolism , Agouti-Related Protein/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Neurons/metabolism , Transfection
13.
Chin Med ; 2: 10, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17868447

ABSTRACT

BACKGROUND: Chinese herbal extract Number Ten (NT) is a dietary herbal formulation prepared from rhubarb, ginger, astragalus, red sage and tumeric. This study tested the effectiveness of NT in reducing body weight gain in rats. METHODS: Sixty female Wistar rats were fed a high fat diet and acclimated to gavage feeding. The rats were divided into five treatment groups: (1) Control (n = 15); (2) NT-H (n = 15), 1.5 g/day; (3) NT-L (n = 10), 0.75 g/day; (4) Pr-fed (n = 10), pair fed to NT-H; (5) d-FF (n = 10), d-fenfluramine 2 mg/kg. Ten rats per group were sacrificed on day 56. Weight, food intake, clinical chemistry and body composition were evaluated. Five animals in the control and 1.5 g/day NT groups were left untreated during a two week recovery period. RESULTS: The 0.75 g/day NT, 1.5 g/day NT, d-fenfluramine and pair fed groups gained 24.6%, 33.3%, 12.3% and 33.3% less than the control respectively (P < 0.0006). Leptin decreased 27.5% to 46.2% in the treatment groups vs. control (P < 0.009). Parametrial fat decreased 14.1% to 55.5% in the NT and pair fed groups vs. control (P < 0.006). The NT groups had soft stools, loss of hair around the mouth and coloration to the urine and stool without evidence of blood or bilirubin (attributed to chromogens in NT). There were no differences between groups in the clinical chemistry. CONCLUSION: This study demonstrated the efficacy of NT in reducing weight gain in rodents.

14.
Obesity (Silver Spring) ; 15(3): 624-31, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17372312

ABSTRACT

OBJECTIVE: To investigate whether there is a difference in sensitivity to a serotonin agonist, meta-chlorophenylpiperazine (mCPP), or cholecystokinin (CCK-8), an intestinal hormone that inhibits food intake, between the Osborne-Mendel (OM) rat, which becomes obese eating a high-fat diet, and the S5B/Pl (S5B) rat, which is resistant to dietary-induced obesity. RESEARCH METHODS AND PROCEDURES: OM and S5B rats were adapted to either a high-saturated-fat diet (56% energy as fat) or a low-fat diet (10% energy as fat) or to both for 14 days and then treated with several doses of mCPP or CCK-8. RESULTS: Treatment with mCPP reduced food intake in both strains of rats. The dose-response curve showed that the OM rats had an increased sensitivity to the serotonergic agonist. Animals eating the high-fat diet had less response to mCPP; and in the S5B rats, the response was significantly reduced. After treatment with CCK-8, there was a similar dose-related suppression of food intake in both the OM and S5B rats. DISCUSSION: These data are consistent with the hypothesis that the serotonin system in the S5B rat has a greater activity that could act to inhibit fat intake. The response to CCK was not significantly affected by strain or diet.


Subject(s)
Cholecystokinin/pharmacology , Eating/drug effects , Piperazines/pharmacology , Animals , Diet, Atherogenic , Diet, Fat-Restricted , Dose-Response Relationship, Drug , Gastrointestinal Agents/pharmacology , Male , Rats , Rats, Inbred Strains , Serotonin 5-HT2 Receptor Agonists
15.
Peptides ; 28(3): 643-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17113194

ABSTRACT

Enterostatin injected into the amygdala selectively reduces dietary fat intake by an action that involves a serotonergic component in the paraventricular nucleus. We have investigated the role of melanocortin signaling in the response to enterostatin by studies in melanocortin 4 receptor (MC4R) knock out mice and by the use of the MC4R and MC3R antagonist SHU9119, and by neurochemical phenotyping of enterostatin activated cells. We also determined the effect of enterostatin in vivo on the expression of AgRP in the hypothalamus and amygdala of rats and in culture on a GT1-7 neuronal cell line. Enterostatin had no effect on food intake in MC4R knock out mice. SHU9119 i.c.v. blocked the feeding response to amygdala enterostatin in rats. Amygdala enterostatin induced fos activation in alpha-melanocyte stimulating hormone (alpha-MSH) neurons in the arcuate nucleus. Enterostatin also reduced the expression of AgRP in the hypothalamus and amygdala and in GT1-7 cells. These data suggest enterostatin inhibits dietary fat intake through a melanocortin signaling pathway.


Subject(s)
Colipases/pharmacology , Dietary Fats/administration & dosage , Eating/drug effects , Protein Precursors/pharmacology , Receptor, Melanocortin, Type 4/physiology , Agouti-Related Protein , Amygdala/drug effects , Amygdala/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/physiology , Base Sequence , Cell Line , DNA Primers/genetics , Eating/physiology , Enzyme Precursors , Female , Gene Expression/drug effects , Hypothalamus/drug effects , Hypothalamus/physiology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Melanocyte-Stimulating Hormones/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pro-Opiomelanocortin/genetics , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/physiology , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/deficiency , Receptor, Melanocortin, Type 4/genetics , Signal Transduction/drug effects , alpha-MSH/metabolism
16.
Nutr Neurosci ; 10(5-6): 273-8, 2007.
Article in English | MEDLINE | ID: mdl-18284036

ABSTRACT

Chronic hyperglycemia and hyperlipidemia exert deleterious effects on beta-cell function and impair glucose-induced insulin release, referred to as glucotoxicity and lipotoxticity. These abnormalities are associated with decreased glucose-induced ATP production; ATP serves as an important signal for insulin secretion. To investigate the mechanism of the impaired ATP formation, we examined the effects of elevated glucose and fatty acids levels on ATP synthase beta-subunit expression, ATP content and insulin secretion in INS-1 insulinoma beta-cells. ATP synthase beta-subunit expression was measured by western blot, ATP content was monitored by ATP luminescence and insulin secretion detected by radio immunoassay. Our result indicated that chronic exposure to high doses of fatty acids together with high levels glucose produced a marked decrease in ATP synthase beta-subunit protein expression. Reduction of ATP synthase beta-subunit protein expression occurred with a decreased intracellular ATP concentration and insulin secretion at high fatty acid concentrations. These results indicate that high glucose together with fatty acids impair the production of ATP in beta-cells through the suppression of mitochondrial ATP synthesis. We conclude that ATP synthase beta-subunit may have an important role in the glucolipotoxicity of islet cells and suggest that ATP synthase beta-subunit might be a target of lipotoxicity in beta-cells.


Subject(s)
Fatty Acids/administration & dosage , Glucose/administration & dosage , Insulin-Secreting Cells/enzymology , Mitochondrial Proton-Translocating ATPases/analysis , Adenosine Triphosphate/analysis , Animals , Cell Line, Tumor , Fatty Acids, Nonesterified/administration & dosage , Insulin/metabolism , Insulin Secretion , Insulinoma , Mitochondrial Proton-Translocating ATPases/physiology , Rats
17.
Peptides ; 27(12): 3292-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16996647

ABSTRACT

Stimulation of mu opioid receptors preferentially increases the intake of a high fat diet. In this paper we investigated whether there was a difference in the expression of mu opioid receptors between animals susceptible (Osborne-Mendel) or resistant (S5B/Pl) to obesity induced by eating a high fat diet. Immunohistochemical studies demonstrated that Osborne-Mendel rats eating a chow diet had an increased number of mu opioid receptors in the arcuate nucleus when compared to S5B/Pl rats. These immunohistochemical findings were supported by Real Time-PCR which demonstrated that the mRNA level of mu opioid receptors was also increased in the hypothalamus of Osborne-Mendel rats compared to S5B/Pl rats. Low doses of the mu opioid receptor agonist DAMGO [d-Ala(2)-N-Me-Phe(4)-Glycol(5)]-enkephalin administered to Osborne-Mendel rats caused a significant increase in the preference for a diet high in fat. The same doses of DAMGO switched the diet preference of S5B/Pl rats to high fat but did not significantly increase food intake. The combination of these findings suggests that the increased levels of hypothalamic mu opioid receptors in Osborne-Mendel rats may contribute to their preference for a diet high in fat and increase their susceptibility to becoming obese.


Subject(s)
Diet , Genetic Predisposition to Disease , Obesity/metabolism , Receptors, Opioid, mu/genetics , Animals , Male , Obesity/genetics , Rats , Receptors, Opioid, mu/biosynthesis
18.
Phytother Res ; 20(9): 806-13, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16835875

ABSTRACT

An aqueous extract of leaves from Rubus suavissimus S. Lee (Rosaceae) or sweet leaf tea was tested for antiangiogenic activity in a human tissue-based fibrin-thrombin clot angiogenesis assay. Further fractionation of this crude extract was performed and the antiangiogenic effect of individual fractions was assessed. The extract was also tested for its oral bioavailability by using the serum of normal rats gavaged with the extract in the assay. At a 0.1% w/v concentration, the extract inhibited initiation of the angiogenic response and subsequent neovessel growth from samples that had already initiated an angiogenic response. Two subfractions of the extract showed significant inhibition of angiogenesis at 0.1% w/w. Gallic acid was elucidated as one of the active angiogenesis inhibitors in one fraction. A 1 mm concentration of gallic acid totally inhibited angiogenesis. In the form of leaf extract, a one-tenth concentration produced the same total inhibition as pure gallic acid. The 10-fold difference in potency suggests the presence of other active compounds contributing to the overall antiangiogenic effect of the extract. The oral absorption of this extract was tested by using serum from rats given the extract orally (gavage) in the angiogenesis assay system. The serum of rats orally administered the sweet leaf tea extract at doses of 0.1% w/w and 0.3% w/w did not significantly inhibit angiogenesis. However, the serum of rats injected intraperitoneally at a dose of 0.1% w/w caused a 41% inhibition of angiogenesis compared with saline injected controls. These preliminary results warrant further bioassay directed identification of other responsible compounds.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Gallic Acid/pharmacology , Plant Extracts/pharmacology , Rosaceae/chemistry , Veins/drug effects , Absorption , Administration, Oral , Angiogenesis Inhibitors/pharmacokinetics , Animals , Gallic Acid/analysis , Humans , Plant Leaves/chemistry , Rats , Tissue Culture Techniques
19.
Arch Physiol Biochem ; 112(1): 23-30, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16754200

ABSTRACT

The role of adrenoceptor subtypes was studied in rat brown adipose tissue (BAT). The type II 5'-deiodinase (5'DII) was activated in response to simultaneous stimulation by beta3- and alpha1-adrenergic agonists, BRL 37344 or CGP 12177, and cirazoline, in brown adipocytes. Inhibition of the alpha1- and beta-adrenergic phenylephrine-stimulated 5'DII activity was obtained by the alpha1-adrenergic antagonists in the order of prazosin >/= wb 4101 > 5-methylurapidil. In comparison, the binding of [3H]prazosin to rat BAT plasma membranes was inhibited by alpha1-adrenergic antagonists in the order of prazosin > WB 4101 = benoxathian > 5-methylurapidil. Although the order of the alpha1-adrenergic competition seemed to be rather typical for the alpha1B-adrenergic receptors, a molecular analysis on adrenoceptor mRNAs should be made to confirm the exact alpha1-adrenergic subtypes at the level of brown adipocytes, since the possibility of a mixture of different receptor subtypes in brown fat cells and/or tissue may interact with the pharmacological characterization. Thus, specific alpha1- and beta-adrenoceptor subtypes participate in the regulation of 5'DII activity in the rat brown adipocytes, and therefore, an impaired alpha1- and beta-adrenergic co-work may be involved in a defective BAT function, e.g., in obese Zucker rats, too. An interesting possibility is that the decreased number of alpha1-adrenoceptors in the BAT of obese Zucker rats is due to the decrease in the alpha1B-adrenoceptor subtype which would further be involved especially in the regulation of BAT 5'DII activity.


Subject(s)
Adipose Tissue, Brown/metabolism , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Iodide Peroxidase/metabolism , Receptors, Adrenergic, alpha/metabolism , Receptors, Adrenergic, beta/metabolism , Adipose Tissue, Brown/enzymology , Animals , Cell Membrane/metabolism , Drug Synergism , Ethanolamines/pharmacology , Female , Imidazoles/pharmacology , Iodide Peroxidase/drug effects , Male , Oxathiins/pharmacology , Piperazines/pharmacology , Prazosin/metabolism , Prazosin/pharmacology , Propanolamines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Zucker , Triiodothyronine/blood
20.
Obesity (Silver Spring) ; 14(3): 345-56, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16648603

ABSTRACT

Voluntary physical activity and exercise training can favorably influence brain plasticity by facilitating neurogenerative, neuroadaptive, and neuroprotective processes. At least some of the processes are mediated by neurotrophic factors. Motor skill training and regular exercise enhance executive functions of cognition and some types of learning, including motor learning in the spinal cord. These adaptations in the central nervous system have implications for the prevention and treatment of obesity, cancer, depression, the decline in cognition associated with aging, and neurological disorders such as Parkinson's disease, Alzheimer's dementia, ischemic stroke, and head and spinal cord injury. Chronic voluntary physical activity also attenuates neural responses to stress in brain circuits responsible for regulating peripheral sympathetic activity, suggesting constraint on sympathetic responses to stress that could plausibly contribute to reductions in clinical disorders such as hypertension, heart failure, oxidative stress, and suppression of immunity. Mechanisms explaining these adaptations are not as yet known, but metabolic and neurochemical pathways among skeletal muscle, the spinal cord, and the brain offer plausible, testable mechanisms that might help explain effects of physical activity and exercise on the central nervous system.


Subject(s)
Central Nervous System/physiology , Exercise/physiology , Adaptation, Physiological , Behavior/physiology , Cognition/physiology , Energy Metabolism/physiology , Humans , Neuronal Plasticity/physiology , Stress, Physiological/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...