Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 168(5): 758-774, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235194

ABSTRACT

Because human energy metabolism evolved to favor adiposity over leanness, the availability of palatable, easily attainable, and calorically dense foods has led to unprecedented levels of obesity and its associated metabolic co-morbidities that appear resistant to traditional lifestyle interventions. However, recent progress identifying the molecular signaling pathways through which the brain and the gastrointestinal system communicate to govern energy homeostasis, combined with emerging insights on the molecular mechanisms underlying successful bariatric surgery, gives reason to be optimistic that novel precision medicines that mimic, enhance, and/or modulate gut-brain signaling can have unprecedented potential for stopping the obesity and type 2 diabetes pandemics.


Subject(s)
Brain/physiology , Energy Metabolism , Gastrointestinal Tract/physiology , Animals , Appetite Regulation , Brain/anatomy & histology , Gastrointestinal Tract/anatomy & histology , Gastrointestinal Tract/innervation , Homeostasis , Humans , Neural Pathways , Pleasure , Satiation
2.
Cell ; 166(4): 867-880, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27518562

ABSTRACT

We report that astrocytic insulin signaling co-regulates hypothalamic glucose sensing and systemic glucose metabolism. Postnatal ablation of insulin receptors (IRs) in glial fibrillary acidic protein (GFAP)-expressing cells affects hypothalamic astrocyte morphology, mitochondrial function, and circuit connectivity. Accordingly, astrocytic IR ablation reduces glucose-induced activation of hypothalamic pro-opio-melanocortin (POMC) neurons and impairs physiological responses to changes in glucose availability. Hypothalamus-specific knockout of astrocytic IRs, as well as postnatal ablation by targeting glutamate aspartate transporter (GLAST)-expressing cells, replicates such alterations. A normal response to altering directly CNS glucose levels in mice lacking astrocytic IRs indicates a role in glucose transport across the blood-brain barrier (BBB). This was confirmed in vivo in GFAP-IR KO mice by using positron emission tomography and glucose monitoring in cerebral spinal fluid. We conclude that insulin signaling in hypothalamic astrocytes co-controls CNS glucose sensing and systemic glucose metabolism via regulation of glucose uptake across the BBB.


Subject(s)
Astrocytes/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Signal Transduction , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Blood-Brain Barrier , Endoplasmic Reticulum/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Homeostasis , Mice , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
3.
J Biol Chem ; 293(34): 13349-13350, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143599

ABSTRACT

Mice harboring a particular allele of the human brain-derived neurotropic factor (BDNFM/M mice) develop extreme obesity and insulin resistance when fed a high-fat diet. The underlying mechanisms of this genetic risk factor for obesity are unclear. In the current issue of JBC, Yang et al. report that pharmacological inhibition of integral membrane protein CD36 significantly reduces body weight gain and improves glucose tolerance in BDNFM/M mice. Targeting CD36 may therefore be a promising strategy to improve metabolic dysfunctions and normalize risk factors in obese individuals.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , CD36 Antigens/metabolism , Metabolic Diseases/physiopathology , Mutation , Obesity/physiopathology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Humans , Mice
4.
J Biol Chem ; 293(6): 2091-2101, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29263093

ABSTRACT

We previously found that 17ß-estradiol (E2) stimulates apolipoprotein A-IV (apoA-IV) gene expression in the nucleus of the solitary tract (NTS) of lean ovariectomized (OVX) rodents. Here we report that in the NTS of high-fat diet-induced obese (DIO) rats, the apoA-IV mRNA level is significantly reduced and that the estrogenic effects on apoA-IV gene expression and food intake are impaired. E2 regulates apoA-IV gene expression through its nuclear receptor α (ERα), which requires co-activators, such as steroid receptor coactivator-1 (SRC-1), to facilitate the transcription of targeted genes. Interestingly, SRC-1 gene expression is significantly reduced in DIO OVX rats. SRC-1 is colocalized with apoA-IV in the cells of the NTS and E2 treatment enhances the recruitment of ERα and SRC-1 to the estrogen response element at the apoA-V promoter, implying the participation of SRC-1 in E2's stimulatory effect on apoA-IV gene expression. Using small hairpin RNA (shRNA), which was validated in cultured neuronal cells, we found that SRC-1 gene knockdown specifically in the NTS significantly diminished E2's anorectic action, leading to increased food intake and body weight. More importantly, the stimulatory effect of E2 on apoA-IV gene expression in the NTS was significantly attenuated in SRC-1 knockdown rats. These results collectively demonstrate the critical roles of NTS SRC-1 in mediating E2's actions on food intake and apoA-IV gene expression and suggest that reduced levels of endogenous SRC-1 and apoA-IV expression are responsible for the impaired E2's anorectic action in obese females.


Subject(s)
Apolipoproteins A/genetics , Estradiol/metabolism , Estrogens/metabolism , Nuclear Receptor Coactivator 1/genetics , Obesity/genetics , Solitary Nucleus/metabolism , Animals , Apolipoproteins A/metabolism , Diet, High-Fat/adverse effects , Eating , Female , Gene Silencing , Humans , Nuclear Receptor Coactivator 1/metabolism , Obesity/metabolism , Obesity/physiopathology , Ovariectomy , Rats , Rats, Long-Evans
5.
Nature ; 503(7474): 59-66, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24201279

ABSTRACT

Although a prominent role for the brain in glucose homeostasis was proposed by scientists in the nineteenth century, research throughout most of the twentieth century focused on evidence that the function of pancreatic islets is both necessary and sufficient to explain glucose homeostasis, and that diabetes results from defects of insulin secretion, action or both. However, insulin-independent mechanisms, referred to as 'glucose effectiveness', account for roughly 50% of overall glucose disposal, and reduced glucose effectiveness also contributes importantly to diabetes pathogenesis. Although mechanisms underlying glucose effectiveness are poorly understood, growing evidence suggests that the brain can dynamically regulate this process in ways that improve or even normalize glycaemia in rodent models of diabetes. Here we present evidence of a brain-centred glucoregulatory system (BCGS) that can lower blood glucose levels via both insulin-dependent and -independent mechanisms, and propose a model in which complex and highly coordinated interactions between the BCGS and pancreatic islets promote normal glucose homeostasis. Because activation of either regulatory system can compensate for failure of the other, defects in both may be required for diabetes to develop. Consequently, therapies that target the BCGS in addition to conventional approaches based on enhancing insulin effects may have the potential to induce diabetes remission, whereas targeting just one typically does not.


Subject(s)
Brain/metabolism , Diabetes Mellitus/metabolism , Glucose/metabolism , Homeostasis , Islets of Langerhans/metabolism , Animals , Blood Glucose/metabolism , Humans , Insulin/metabolism
7.
J Lipid Res ; 59(9): 1649-1659, 2018 09.
Article in English | MEDLINE | ID: mdl-29991652

ABSTRACT

Meta-inflammation of hypothalamic areas governing energy homeostasis has recently emerged as a process of potential pathophysiological relevance for the development of obesity and its metabolic sequelae. The current model suggests that diet-induced neuronal injury triggers microgliosis and astrocytosis, conditions which ultimately may induce functional impairment of hypothalamic circuits governing feeding behavior, systemic metabolism, and body weight. Epidemiological data indicate that low circulating HDL levels, besides conveying cardiovascular risk, also correlate strongly with obesity. We simulated that condition by using a genetic loss of function mouse model (apoA-I-/-) with markedly reduced HDL levels to investigate whether HDL may directly modulate hypothalamic inflammation. Astrogliosis was significantly enhanced in the hypothalami of apoA-I-/- compared with apoA-I+/+ mice and was associated with compromised mitochondrial function. apoA-I-/- mice exhibited key components of metabolic disease, like increased fat mass, fasting glucose levels, hepatic triglyceride content, and hepatic glucose output compared with apoA-I+/+ controls. Administration of reconstituted HDL (CSL-111) normalized hypothalamic inflammation and mitochondrial function markers in apoA-I-/- mice. Treatment of primary astrocytes with apoA-I resulted in enhanced mitochondrial activity, implying that circulating HDL levels are likely important for astrocyte function. HDL-based therapies may consequently avert reactive gliosis in hypothalamic astrocytes by improving mitochondrial bioenergetics and thereby offering potential treatment and prevention for obesity and metabolic disease.


Subject(s)
Apolipoprotein A-I/metabolism , Gliosis/metabolism , Gliosis/pathology , Hypothalamus/pathology , Lipoproteins, HDL/blood , Adenosine Triphosphate/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Biomarkers/metabolism , Gliosis/blood , Glycolysis , Male , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Oxidative Phosphorylation , Phenotype
8.
Am J Physiol Regul Integr Comp Physiol ; 313(5): R535-R548, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28768657

ABSTRACT

Apolipoprotein AIV (ApoAIV) and cholecystokinin (CCK) are well-known satiating signals that are stimulated by fat consumption. Peripheral ApoAIV and CCK interact to prolong satiating signals. In the present study, we hypothesized that ApoAIV and CCK control energy homeostasis in response to high-fat diet feeding. To test this hypothesis, energy homeostasis in ApoAIV and CCK double knockout (ApoAIV/CCK-KO), ApoAIV knockout (ApoAIV-KO), and CCK knockout (CCK-KO) mice were monitored. When animals were maintained on a low-fat diet, ApoAIV/CCK-KO, ApoAIV-KO, and CCK-KO mice had comparable energy intake and expenditure, body weight, fat mass, fat absorption, and plasma parameters relative to the controls. In contrast, these KO mice exhibited impaired lipid transport to epididymal fat pads in response to intraduodenal infusion of dietary lipids. Furthermore, ApoAIV-KO mice had upregulated levels of CCK receptor 2 (CCK2R) in the small intestine while ApoAIV/CCK-KO mice had upregulated levels of CCK2R in the brown adipose tissue. After 20 wk of a high-fat diet, ApoAIV-KO and CCK-KO mice had comparable body weight and fat mass, as well as lower energy expenditure at some time points. However, ApoAIV/CCK-KO mice exhibited reduced body weight and adiposity relative to wild-type mice, despite having normal food intake. Furthermore, ApoAIV/CCK-KO mice displayed normal fat absorption and locomotor activity, as well as enhanced energy expenditure. These observations suggest that mice lacking ApoAIV and CCK have reduced body weight and adiposity, possibly due to impaired lipid transport and elevated energy expenditure.


Subject(s)
Apolipoproteins A/metabolism , Cholecystokinin/metabolism , Dietary Fats/metabolism , Homeostasis/physiology , Adiposity/genetics , Adiposity/physiology , Animals , Apolipoproteins A/deficiency , Body Weight/physiology , Cholecystokinin/deficiency , Diet, Fat-Restricted/methods , Eating/physiology , Energy Intake/physiology , Energy Metabolism/genetics , Mice, Knockout
9.
Front Neuroendocrinol ; 38: 1-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25527432

ABSTRACT

The gastrointestinal hormone peptide tyrosine tyrosine 3-36 (PYY(3-36)) has attained broad recognition with respect to its involvement in energy homeostasis and the control of food intake. It is mainly secreted by distal intestinal enteroendocrine L-cells in response to eating and exerts neurally mediated, paracrine and endocrine effects on various target organs. In addition to its gastrointestinal effects, PYY(3-36) has long been known to inhibit food intake. Recent closer examination of the effects of PYY(3-36) revealed that this gut-derived peptide also influences a wide spectrum of behavioral and cognitive functions that are pivotal for basic processes of perception and judgment, including central information processing, salience learning, working memory, and behavioral responding to novelty. Here, we review the effects of PYY(3-36) that go beyond food intake and provide a conceptual framework suggesting that several apparently unrelated behavioral actions of PYY(3-36) may actually reflect different manifestations of modulating the central dopamine system.


Subject(s)
Dopamine/metabolism , Eating/physiology , Homeostasis/physiology , Peptide YY/metabolism , Animals , Gastrointestinal Absorption/physiology , Humans
10.
Am J Physiol Regul Integr Comp Physiol ; 311(5): R979-R987, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27581811

ABSTRACT

The mechanisms involved in the weight loss seen after vertical sleeve gastrectomy (VSG) are not clear. The rat stomach has two morphologically and functionally distinct proximal and distal parts. The rat model for VSG involves complete removal of the proximal part and 80% removal of the distal part along the greater curvature. The purpose of this study was to understand the potential independent contributions of removal of these distinct gastric sections to VSG outcomes. We prepared four surgical groups of male Long-Evans rats: VSG, sham surgery (control), selective proximal section removal (PR), and selective distal section removal (DR). Gastric emptying rate (GER) was highest after VSG compared with all other groups. However, PR, in turn, had significantly greater GER compared with both DR and sham groups. The surgery-induced weight loss followed the same pattern with VSG causing the greatest weight loss and PR having greater weight loss compared with DR and sham groups. The results were robust for rats fed regular chow or a high-fat diet. Body mass analysis revealed that the weight loss was due to the loss of fat mass, and there was no change in lean mass after the surgeries. In conclusion, removal of the proximal stomach contributes to most, but not all, of the physiological impact of VSG.


Subject(s)
Gastrectomy , Obesity/physiopathology , Obesity/surgery , Stomach/physiopathology , Stomach/surgery , Weight Loss/physiology , Animals , Gastric Emptying , Male , Obesity/diagnosis , Rats , Rats, Long-Evans , Treatment Outcome
11.
Am J Physiol Gastrointest Liver Physiol ; 309(8): G680-7, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26294669

ABSTRACT

Both glucagon-like peptide-1 (GLP-1) and apolipoprotein A-IV (apoA-IV) are produced from the gut and enhance postprandial insulin secretion. This study investigated whether apoA-IV regulates nutrient-induced GLP-1 secretion and whether apoA-IV knockout causes compensatory GLP-1 release. Using lymph-fistula-mice, we first determined lymphatic GLP-1 secretion by administering apoA-IV before an intraduodenal Ensure infusion. apoA-IV changed neither basal nor Ensure-induced GLP-1 secretion relative to saline administration. We then assessed GLP-1 in apoA-IV-/- and wild-type (WT) mice administered intraduodenal Ensure. apoA-IV-/- mice had comparable lymph flow, lymphatic triglyceride, glucose, and protein outputs as WT mice. Intriguingly, apoA-IV-/- mice had higher lymphatic GLP-1 concentration and output than WT mice 30 min after Ensure administration. Increased GLP-1 was also observed in plasma of apoA-IV-/- mice at 30 min. apoA-IV-/- mice had comparable total gut GLP-1 content relative to WT mice under fasting, but a lower GLP-1 content 30 min after Ensure administration, suggesting that more GLP-1 was secreted. Moreover, an injection of apoA-IV protein did not reverse the increased GLP-1 secretion in apoA-IV-/- mice. Finally, we assessed gene expression of GLUT-2 and the lipid receptors, including G protein-coupled receptor (GPR) 40, GPR119, and GPR120 in intestinal segments. GLUT-2, GPR40 and GPR120 mRNAs were unaltered by apoA-IV knockout. However, ileal GPR119 mRNA was significantly increased in apoA-IV-/- mice. GPR119 colocalizes with GLP-1 in ileum and stimulates GLP-1 secretion by sensing OEA, lysophosphatidylcholine, and 2-monoacylglycerols. We suggest that increased ileal GPR119 is a potential mechanism by which GLP-1 secretion is enhanced in apoA-IV-/- mice.


Subject(s)
Apolipoproteins A/metabolism , Glucagon-Like Peptide 1/metabolism , Animals , Apolipoproteins A/genetics , Gene Expression Regulation/physiology , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/genetics , Ileum/metabolism , Incretins/metabolism , Lymph/chemistry , Lymph/metabolism , Lymphatic System/metabolism , Male , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
12.
Proc Natl Acad Sci U S A ; 109(24): 9641-6, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22619326

ABSTRACT

Apolipoprotein A-IV (apoA-IV) is secreted by the small intestine in response to fat absorption. Here we demonstrate a potential role for apoA-IV in regulating glucose homeostasis. ApoA-IV-treated isolated pancreatic islets had enhanced insulin secretion under conditions of high glucose but not of low glucose, suggesting a direct effect of apoA-IV to enhance glucose-stimulated insulin release. This enhancement involves cAMP at a level distal to Ca(2+) influx into the ß cells. Knockout of apoA-IV results in compromised insulin secretion and impaired glucose tolerance compared with WT mice. Challenging apoA-IV(-/-) mice with a high-fat diet led to fasting hyperglycemia and more severe glucose intolerance associated with defective insulin secretion than occurred in WT mice. Administration of exogenous apoA-IV to apoA-IV(-/-) mice improved glucose tolerance by enhancing insulin secretion in mice fed either chow or a high-fat diet. Finally, we demonstrate that exogenous apoA-IV injection decreases blood glucose levels and stimulates a transient increase in insulin secretion in KKAy diabetic mice. These results suggest that apoA-IV may provide a therapeutic target for the regulation of glucose-stimulated insulin secretion and treatment of diabetes.


Subject(s)
Apolipoproteins A/physiology , Glucose/metabolism , Homeostasis , Insulin/metabolism , Animals , Apolipoproteins A/genetics , Glucose Tolerance Test , Insulin Secretion , Mice , Mice, Knockout
13.
J Neurosci ; 33(11): 4825-33, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23486953

ABSTRACT

Obesity is associated with increased levels of angiotensin-II (Ang-II), which activates angiotensin type 1a receptors (AT1a) to influence cardiovascular function and energy homeostasis. To test the hypothesis that specific AT1a within the brain control these processes, we used the Cre/lox system to delete AT1a from the paraventricular nucleus of the hypothalamus (PVN) of mice. PVN AT1a deletion did not affect body mass or adiposity when mice were maintained on standard chow. However, maintenance on a high-fat diet revealed a gene by environment interaction whereby mice lacking AT1a in the PVN had increased food intake and decreased energy expenditure that augmented body mass and adiposity relative to controls. Despite this increased adiposity, PVN AT1a deletion reduced systolic blood pressure, suggesting that this receptor population mediates the positive correlation between adiposity and blood pressure. Gene expression studies revealed that PVN AT1a deletion decreased hypothalamic expression of corticotrophin-releasing hormone and oxytocin, neuropeptides known to control food intake and sympathetic nervous system activity. Whole-cell patch-clamp recordings confirmed that PVN AT1a deletion eliminates responsiveness of PVN parvocellular neurons to Ang-II, and suggest that Ang-II responsiveness is increased in obese wild-type mice. Central inflammation is associated with metabolic and cardiovascular disorders and PVN AT1a deletion reduced indices of hypothalamic inflammation. Collectively, these studies demonstrate that PVN AT1a regulate energy balance during environmental challenges that promote metabolic and cardiovascular pathologies. The implication is that the elevated Ang-II that accompanies obesity serves as a negative feedback signal that activates PVN neurons to alleviate weight gain.


Subject(s)
Diet, High-Fat/adverse effects , Obesity , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Angiotensin, Type 1/deficiency , Action Potentials/drug effects , Action Potentials/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adrenal Glands/metabolism , Analysis of Variance , Angiotensin II/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Composition/genetics , Body Weight/genetics , Brain/pathology , Calorimetry , Cardiovascular System/physiopathology , Disease Models, Animal , Drinking/genetics , Eating/genetics , Energy Metabolism/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , In Vitro Techniques , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Obesity/etiology , Obesity/metabolism , Obesity/prevention & control , Patch-Clamp Techniques , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sodium Chloride/metabolism , Telemetry
14.
Diabetologia ; 57(2): 383-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24190582

ABSTRACT

AIMS/HYPOTHESIS: Genome-wide association studies have revealed an association of the transcription factor ETS variant gene 5 (ETV5) with human obesity. However, its role in glucose homeostasis and energy balance is unknown. METHODS: Etv5 knockout (KO) mice were monitored weekly for body weight (BW) and food intake. Body composition was measured at 8 and 16 weeks of age. Glucose metabolism was studied, and glucose-stimulated insulin secretion was measured in vivo and in vitro. RESULTS: Etv5 KO mice are smaller and leaner, and have a reduced BW and lower fat mass than their wild-type controls on a chow diet. When exposed to a high-fat diet, KO mice are resistant to diet-induced BW gain. Despite a greater insulin sensitivity, KO mice have profoundly impaired glucose tolerance associated with impaired insulin secretion. Morphometric analysis revealed smaller islets and a reduced beta cell size in the pancreatic islets of Etv5 KO mice. Knockdown of ETV5 in an insulin-secreting cell line or beta cells from human donors revealed intact mitochondrial and Ca(2+) channel activity, but reduced insulin exocytosis. CONCLUSION/INTERPRETATION: This work reveals a critical role for ETV5 in specifically regulating insulin secretion both in vitro and in vivo.


Subject(s)
C-Peptide/metabolism , DNA-Binding Proteins/metabolism , Exocytosis/physiology , Glucose/metabolism , Homeostasis/physiology , Insulin/metabolism , Transcription Factors/metabolism , Animals , Body Composition , Body Weight , Diet, High-Fat , Eating , Genome-Wide Association Study , Glucose Tolerance Test , Insulin Resistance , Insulin Secretion , Mice , Mice, Knockout
15.
Am J Physiol Endocrinol Metab ; 307(9): E784-92, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25159330

ABSTRACT

Meal-fed (MF) rats with access to food for only 4 consecutive hours during the light cycle learn to eat large meals to maintain energy balance. MF animals develop behavioral and endocrine changes that permit glucose tolerance despite increased meal size. We hypothesized that enhanced activity of the enteroinsular axis mediates glucose homeostasis during MF. Cohorts of rats were allocated to MF or ad libitum (AL) regimens for 2-4 wk. Insulin secretion and glucose tolerance were determined after oral carbohydrate and intraperitoneal (ip) and intravenous (iv) glucose. MF rats ate less than AL in the first week but maintained a comparable weight trajectory thereafter. MF rats had decreased glucose excursions after a liquid mixed meal (AUC: MF 75 ± 7, AL 461 ± 28 mmol·l⁻¹·min, P < 0.001), with left-shifted insulin secretion (AUC(0-15): MF 31.0 ± 4.9, AL 9.6 ± 4.4 pM·min, P < 0.02), which peaked before a significant rise in blood glucose. Both groups had comparable fasting glucagon levels, but postprandial responses were lower with MF. However, neither intestinal expression of proGIP and proglucagon mRNA nor plasma incretin levels differed between MF and AL groups. There were no differences in the insulin response to ip or iv glucose between MF and AL rats. These findings demonstrate that MF improves oral glucose tolerance and is associated with significant changes in postprandial islet hormone secretion. Because MF enhanced ß-cell function during oral but not parenteral carbohydrate administration, and was not accounted for by changes in circulating incretins, these results support a neural mechanism of adaptive insulin secretion.


Subject(s)
Allostasis , Feeding Behavior , Glucose Intolerance/drug therapy , Insulin/metabolism , Islets of Langerhans/metabolism , Meals , Neurosecretory Systems/physiopathology , Animals , Behavior, Animal , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Glucagon/blood , Glucagon/genetics , Glucagon/metabolism , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose Intolerance/physiopathology , Hyperglycemia/prevention & control , Ileum/metabolism , Incretins/blood , Incretins/metabolism , Insulin/blood , Insulin Secretion , Intestinal Mucosa/metabolism , Islets of Langerhans/innervation , Jejunum/metabolism , Male , Organ Specificity , Postprandial Period , Proglucagon/genetics , Proglucagon/metabolism , Rats, Long-Evans
16.
Am J Physiol Endocrinol Metab ; 307(11): E1065-72, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25315695

ABSTRACT

Vertical sleeve gastrectomy (VSG) is currently one of the most effective treatments for obesity. Despite recent developments, the underlying mechanisms that contribute to the metabolic improvements following bariatric surgery remain unresolved. VSG reduces postprandial intestinal triglyceride (TG) production, but whether the effects of VSG on intestinal metabolism are related to metabolic outcomes has yet to be established. The lipid synthesis enzyme acyl CoA:monoacylglycerol acyltransferase-2 (Mogat2; MGAT2) plays a crucial role in the assimilation of dietary fat in the intestine and in regulation of adiposity stores as well. Given the phenotypic similarities between VSG-operated and MGAT2-deficient animals, we reasoned that this enzyme could also have a key role in mediating the metabolic benefits of VSG. However, VSG reduced body weight and fat mass and improved glucose metabolism similarly in whole body MGAT2-deficient (Mogat2(-/-)) mice and wild-type littermates. Furthermore, along with an increase in energy expenditure, surgically naive Mogat2(-/-) mice had altered macronutrient preference, shifting preference away from fat and toward carbohydrates, and increased locomotor activity. Collectively, these data suggest that the beneficial effects of VSG on body weight and glucose metabolism are independent of MGAT2 activity and rather that they are separate from the effects of MGAT2 deficiency. Because MGAT2 inhibitors are proposed as a pharmacotherapeutic option for obesity, our data suggest that, in addition to increasing energy expenditure, shifting macronutrient preference away from fat could be another important mechanism by which these compounds could contribute to weight loss.


Subject(s)
Gastrectomy , N-Acetylglucosaminyltransferases/deficiency , Animals , Body Composition , Body Weight , Diet, Fat-Restricted , Eating , Food Preferences , Male , Mice , Mice, Knockout , Obesity/genetics
17.
Am J Physiol Endocrinol Metab ; 306(4): E424-32, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24368666

ABSTRACT

Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) are effective weight loss surgeries that also improve glucose metabolism. Rapid, early rises of circulating insulin and glucagon-like peptide-1 (GLP-1) concentrations following food ingestion are characteristic of these procedures. The purpose of the current study was to test the hypothesis that postprandial hormone release is due to increased nutrient emptying from the stomach. Radioscintigraphy and chemical and radiolabeled tracers were used to examine gastric emptying in rat models of VSG and RYGB surgery. Intraduodenal nutrient infusions were used to assess intestinal GLP-1 secretion and nutrient sensitivity in VSG rats compared with shams. Five minutes after a nutrient gavage, the stomachs of RYGB and VSG rats were completely emptied, whereas only 6.1% of the nutrient mixture had emptied from sham animals. Gastric pressure was increased in VSG animals, and rats with this procedure did not inhibit gastric emptying normally in response to increasing caloric loads of dextrose or corn oil, and they did not respond to neural or endocrine effectors of gastric motility. Finally, direct infusion of liquid nutrients into the duodenum caused significantly greater GLP-1 release in VSG compared with shams, indicating that increases in GLP-1 secretion after VSG are the result of both greater gastric emptying rates and altered responses at the level of the intestine. These findings demonstrate greatly accelerated gastric emptying in rat models of RYGB and VSG. In VSG this is likely due to increased gastric pressure and reduced responses to inhibitory feedback from the intestine.


Subject(s)
Gastrectomy/methods , Gastric Emptying/physiology , Gastric Mucosa/metabolism , Glucagon-Like Peptide 1/metabolism , Postprandial Period/physiology , Animals , Body Weight/drug effects , Body Weight/physiology , Exenatide , Gastric Emptying/drug effects , Gastric Mucosa/drug effects , Hypoglycemic Agents/pharmacology , Male , Peptides/pharmacology , Rats , Rats, Long-Evans , Stomach/drug effects , Venoms/pharmacology
18.
Stress ; 17(6): 484-93, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25238021

ABSTRACT

Behavioral modifications for the treatment of obesity, including caloric restriction, have notoriously low long-term success rates relative to bariatric weight-loss surgery. The reasons for the difference in sustained weight loss are not clear. One possibility is that caloric restriction alone activates the stress-responsive hypothalamo-pituitary-adrenocortical (HPA) axis, undermining the long-term maintenance of weight loss, and that this is abrogated after bariatric surgery. Accordingly, we compared the HPA response to weight loss in five groups of male rats: (1) high-fat diet-induced obese (DIO) rats treated with Roux-en-Y gastric bypass surgery (RYGB, n = 7), (2) DIO rats treated with vertical sleeve gastrectomy (VSG, n = 11), (3) DIO rats given sham surgery and subsequently restricted to the food intake of the VSG/RYGB groups (Pair-fed, n = 11), (4) ad libitum-fed DIO rats given sham surgery (Obese, n = 11) and (5) ad libitum chow-fed rats given sham surgery (Lean, n = 12). Compared with Lean controls, food-restricted rats exhibited elevated morning (nadir) non-stress plasma corticosterone concentration and increased hypothalamic corticotropin-releasing hormone and vasopressin mRNA expression, indicative of basal HPA activation. This was largely prevented when weight loss was achieved by bariatric surgery. DIO increased HPA activation by acute (novel environment) stress and this was diminished by bariatric surgery-, but not pair-feeding-, induced weight loss. These results indicate that the HPA axis is differentially affected by weight loss from caloric restriction versus bariatric surgery, and this may contribute to the differing long-term effectiveness of these two weight-loss approaches.


Subject(s)
Caloric Restriction , Gastrectomy , Gastric Bypass , Hypothalamo-Hypophyseal System/physiopathology , Obesity/diet therapy , Obesity/surgery , Pituitary-Adrenal System/physiopathology , Weight Loss , Animals , Corticosterone/blood , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Diet, High-Fat , Disease Models, Animal , Hypothalamo-Hypophyseal System/metabolism , Male , Obesity/blood , Obesity/etiology , Obesity/physiopathology , Pituitary-Adrenal System/metabolism , RNA, Messenger/metabolism , Rats, Long-Evans , Stress, Physiological , Time Factors , Vasopressins/genetics , Vasopressins/metabolism
19.
J Lipid Res ; 54(5): 1430-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23434611

ABSTRACT

Ginsenoside Rb1 (Rb1), a natural compound extracted from ginseng, exerts anti-obesity activity and improves insulin sensitivity in high-fat diet (HFD)-induced obese rats. The objective of the current study was to evaluate the protective effect of Rb1 on fatty liver in HFD-induced obese rats and to elucidate underlying mechanisms. After chronic intraperitoneal administration, Rb1 (10 mg/kg) significantly ameliorated hepatic fat accumulation in HFD-induced obese rats, as demonstrated by reduced liver weight, hepatic triglyceride content, and histological evaluation of liver sections by hematoxylin and eosin and Oil Red O staining. Using primary cultured rat hepatic cells, we found that the rate of fatty acid oxidation and the activity of carnitine palmitoyltransferase 1 (CPT1), a key enzyme in fatty acid ß-oxidation, were significantly elevated in Rb1-treated hepatocytes compared with those of vehicle-treated cells. HPLC analysis revealed that Rb1 increased the cellular AMP/ATP ratio, which is associated with elevated activation of hepatic AMP-activated protein kinase (AMPK) and phosphorylated acetyl-CoA carboxylase. Consistent with the activation of AMPK, Rb1 stimulated the expression of genes encoding fatty acid oxidative enzymes and proteins, and suppressed the expression of genes encoding enzymes or proteins that function in lipogenesis, assessed by quantitative PCR. We conclude that Rb1 has a potent ability to reduce hepatic fat accumulation and might be useful as a therapeutic agent for fatty liver disorder.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Ginsenosides/administration & dosage , Obesity/drug therapy , Obesity/metabolism , Animals , Diet, High-Fat , Fat Body/drug effects , Fat Body/metabolism , Fatty Liver/drug therapy , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Liver/drug effects , Liver/metabolism , Male , Obesity/pathology , Rats
20.
Am J Physiol Regul Integr Comp Physiol ; 305(1): R68-77, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23616105

ABSTRACT

Overconsumption of a high-fat diet promotes weight gain that can result in obesity and associated comorbidities, including Type 2 diabetes mellitus. Consumption of a high-fat diet also alters gut-brain communication. Glucagon-like peptide 1 (GLP-1) is an important gastrointestinal signal that modulates both short- and long-term energy balance and is integral in maintenance of glucose homeostasis. In the current study, we investigated whether high-fat diets (40% or 81% kcal from fat) modulated the ability of the GLP-1 receptor (GLP-1r) agonists exendin-4 (Ex4) and liraglutide to reduce food intake and body weight. We observed that rats maintained on high-fat diets had a delayed acute anorexic response to peripheral administration of Ex4 or liraglutide compared with low-fat diet-fed rats (17% kcal from fat). However, once suppression of food intake in response to Ex4 or liraglutide started, the effect persisted for a longer time in the high-fat diet-fed rats compared with low-fat diet-fed rats. In contrast, centrally administered Ex4 suppressed food intake similarly between high-fat diet-fed and low-fat diet-fed rats. Chronic consumption of a high-fat diet did not change the pharmacokinetics of Ex4 but increased intestinal Glp1r expression and decreased hindbrain Glp1r expression. Taken together, these findings demonstrate that dietary composition alters the temporal profile of the anorectic response to exogenous GLP-1r agonists.


Subject(s)
Body Weight/physiology , Diet, High-Fat , Eating/physiology , Malnutrition/physiopathology , Receptors, Glucagon/agonists , Receptors, Glucagon/physiology , Animals , Body Weight/drug effects , Diet, Fat-Restricted , Eating/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Exenatide , Glucagon-Like Peptide 1/adverse effects , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Glucose/metabolism , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Liraglutide , Male , Malnutrition/chemically induced , Models, Animal , Peptides/adverse effects , Peptides/pharmacology , Rats , Rats, Long-Evans , Venoms/adverse effects , Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL