Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R250-R259, 2021 08 01.
Article En | MEDLINE | ID: mdl-34259025

The peptide hormone amylin reduces food intake and body weight and is an attractive candidate target for novel pharmacotherapies to treat obesity. However, the short half-life of native amylin and amylin analogs like pramlintide limits these compounds' potential utility in promoting sustained negative energy balance. Here, we evaluate the ability of the novel long-acting amylin/calcitonin receptor agonist ZP5461 to reduce feeding and body weight in rats, and also test the role of calcitonin receptors (CTRs) in the dorsal vagal complex (DVC) of the hindbrain in the energy balance effects of chronic ZP5461 administration. Acute dose-response studies indicate that systemic ZP5461 (0.5-3 nmol/kg) robustly suppresses energy intake and body weight gain in chow- and high-fat diet (HFD)-fed rats. When HFD-fed rats received chronic systemic administration of ZP5461 (1-2 nmol/kg), the compound initially produced reductions in energy intake and weight gain but failed to produce sustained suppression of intake and body weight. Using virally mediated knockdown of DVC CTRs, the ability of chronic systemic ZP5461 to promote early reductions in intake and body weight gain was determined to be mediated in part by activation of DVC CTRs, implicating the DVC as a central site of action for ZP5461. Future studies should address other dosing regimens of ZP5461 to determine whether an alternative dose/frequency of administration would produce more sustained body weight suppression.


Amylin Receptor Agonists/pharmacology , Appetite Depressants/pharmacology , Eating/drug effects , Feeding Behavior/drug effects , Receptors, Calcitonin/agonists , Receptors, Islet Amyloid Polypeptide/drug effects , Rhombencephalon/drug effects , Vagus Nerve/drug effects , Weight Gain/drug effects , Animals , Dose-Response Relationship, Drug , Energy Intake/drug effects , Male , Rats, Sprague-Dawley , Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism , Receptors, Islet Amyloid Polypeptide/genetics , Receptors, Islet Amyloid Polypeptide/metabolism , Rhombencephalon/metabolism , Signal Transduction , Time Factors , Vagus Nerve/metabolism
2.
Int J Obes (Lond) ; 44(12): 2405-2418, 2020 12.
Article En | MEDLINE | ID: mdl-32999409

BACKGROUND: The prevalence of adolescent obesity has increased dramatically, becoming a serious public health concern. While previous evidence suggests that in utero- and early postnatal overnutrition increases adult-onset obesity risk, the neurobiological mechanisms underlying this outcome are not well understood. Non-neuronal cells play an underestimated role in the physiological responses to metabolic/nutrient signals. Hypothalamic glial-mediated inflammation is now considered a contributing factor in the development and perpetuation of obesity; however, attention on the role of gliosis and microglia activation in other nuclei is still needed. METHODS/RESULTS: Here, we demonstrate that early life consumption of high-fat/sucrose diet (HFSD) is sufficient to increase offspring body weight, hyperleptinemia and potentially maladaptive cytoarchitectural changes in the brainstem dorsal-vagal-complex (DVC), an essential energy balance processing hub, across postnatal development. Our data demonstrate that pre- and postnatal consumption of HFSD result in increased body weight, hyperleptinemia and dramatically affects the non-neuronal landscape, and therefore the plasticity of the DVC in the developing offspring. CONCLUSIONS: Current findings are very provocative, considering the importance of the DVC in appetite regulation, suggesting that HFSD-consumption during early life may contribute to subsequent obesity risk via DVC cytoarchitectural changes.


Brain Stem/physiopathology , Neuronal Plasticity , Obesity/physiopathology , Overnutrition/physiopathology , Animals , Body Weight , Diet, High-Fat , Dietary Sucrose/administration & dosage , Female , Glucagon-Like Peptide 1/blood , Insulin/blood , Leptin/blood , Male , Maternal Nutritional Physiological Phenomena , Rats , Rats, Sprague-Dawley , Weight Gain
3.
Transl Psychiatry ; 10(1): 90, 2020 03 09.
Article En | MEDLINE | ID: mdl-32152264

Previous studies identify a role for hypothalamic glia in energy balance regulation; however, a narrow hypothalamic focus provides an incomplete understanding of how glia throughout the brain respond to and regulate energy homeostasis. We examined the responses of glia in the dorsal vagal complex (DVC) to the adipokine leptin and high fat diet-induced obesity. DVC astrocytes functionally express the leptin receptor; in vivo pharmacological studies suggest that DVC astrocytes partly mediate the anorectic effects of leptin in lean but not diet-induced obese rats. Ex vivo calcium imaging indicated that these changes were related to a lower proportion of leptin-responsive cells in the DVC of obese versus lean animals. Finally, we investigated DVC microglia and astroglia responses to leptin and energy balance dysregulation in vivo: obesity decreased DVC astrogliosis, whereas the absence of leptin signaling in Zucker rats was associated with extensive astrogliosis in the DVC and decreased hypothalamic micro- and astrogliosis. These data uncover a novel functional heterogeneity of astrocytes in different brain nuclei of relevance to leptin signaling and energy balance regulation.


Hypothalamus , Leptin , Animals , Energy Metabolism , Hypothalamus/metabolism , Leptin/metabolism , Neuroglia/metabolism , Rats , Rats, Zucker
4.
Sci Transl Med ; 12(533)2020 03 04.
Article En | MEDLINE | ID: mdl-32132220

The glucagon-like peptide-1 receptor (GLP-1R) agonist liraglutide is approved for the treatment of obesity; however, there is still much to be learned regarding the neuronal sites of action that underlie its suppressive effects on food intake and body weight. Peripherally administered liraglutide in rats acts in part through central GLP-1Rs in both the hypothalamus and the hindbrain. Here, we extend findings supporting a role for hindbrain GLP-1Rs in mediating the anorectic effects of liraglutide in male rats. To dissociate the contribution of GLP-1Rs in the area postrema (AP) and the nucleus tractus solitarius (NTS), we examined the effects of liraglutide in both NTS AAV-shRNA-driven Glp1r knockdown and AP-lesioned animals. Knockdown of NTS GLP-1Rs, but not surgical lesioning of the AP, attenuated the anorectic and body weight-reducing effects of acutely delivered liraglutide. In addition, NTS c-Fos responses were maintained in AP-lesioned animals. Moreover, NTS Glp1r knockdown was sufficient to attenuate the intake- and body weight-reducing effects of chronic daily administered liraglutide over 3 weeks. Development of improved obesity pharmacotherapies requires an understanding of the cellular phenotypes targeted by GLP-1R agonists. Fluorescence in situ hybridization identified Glp1r transcripts in NTS GABAergic neurons, which when inhibited using chemogenetics, attenuated the food intake- and body weight-reducing effects of liraglutide. This work demonstrates the contribution of NTS GLP-1Rs to the anorectic potential of liraglutide and highlights a phenotypically distinct (GABAergic) population of neurons within the NTS that express the GLP-1R and are involved in the mediation of liraglutide signaling.


Appetite Depressants , GABAergic Neurons , Glucagon-Like Peptide-1 Receptor , Liraglutide , Animals , Appetite Depressants/pharmacology , Eating , GABAergic Neurons/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , In Situ Hybridization, Fluorescence , Liraglutide/pharmacology , Male , Rats , Rats, Sprague-Dawley , Solitary Nucleus/metabolism
5.
Sci Rep ; 9(1): 8447, 2019 06 11.
Article En | MEDLINE | ID: mdl-31186439

A growing appreciation of the overlapping neuroendocrine mechanisms controlling energy balance has highlighted combination therapies as a promising strategy to enhance sustained weight loss. Here, we investigated whether amylin- and glucagon-like-peptide-1 (GLP-1)-based combination therapies produce greater food intake- and body weight-suppressive effects compared to monotherapies in both lean and diet-induced obese (DIO) rats. In chow-maintained rats, systemic amylin and GLP-1 combine to reduce meal size. Furthermore, the amylin and GLP-1 analogs salmon calcitonin (sCT) and liraglutide produce synergistic-like reductions in 24 hours energy intake and body weight. The administration of sCT with liraglutide also led to a significant enhancement in cFos-activation in the dorsal-vagal-complex (DVC) compared to mono-therapy, suggesting an activation of distinct, yet overlapping neural substrates in this critical energy balance hub. In DIO animals, long-term daily administration of this combination therapy, specifically in a stepwise manner, results in reduced energy intake and greater body weight loss over time when compared to chronic mono- and combined-treated groups, without affecting GLP-1 receptor, preproglucagon or amylin-receptor gene expression in the DVC.


Calcitonin/metabolism , Glucagon-Like Peptide 1/metabolism , Islet Amyloid Polypeptide/metabolism , Obesity/drug therapy , Weight Loss/drug effects , Animals , Anti-Obesity Agents/metabolism , Anti-Obesity Agents/pharmacology , Blood Glucose/drug effects , Body Weight/drug effects , Calcitonin/genetics , Calcitonin/pharmacology , Diet, High-Fat/adverse effects , Eating/drug effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/pharmacology , Islet Amyloid Polypeptide/genetics , Islet Amyloid Polypeptide/pharmacology , Male , Obesity/metabolism , Obesity/pathology , Rats , Receptors, Glucagon/genetics
6.
Neuropsychopharmacology ; 44(10): 1742-1751, 2019 09.
Article En | MEDLINE | ID: mdl-30959513

The overlap in neurobiological circuitry mediating the physiological and behavioral response to satiation and noxious/stressful stimuli are not well understood. The interaction between serotonin (5-HT) and glucagon-like peptide-1 (GLP-1) could play a role as upstream effectors involved in mediating associations between anorectic and noxious/stressful stimuli. We hypothesize that 5-HT acts as an endogenous modulator of the central GLP-1 system to mediate satiation and malaise in rats. Here, we investigate whether interactions between central 5-HT and GLP-1 signaling are behaviorally and physiologically relevant for the control of food intake and pica (i.e., behavioral measure of malaise). Results show that the anorexia and body weight changes induced by administration of exogenous hindbrain 5-HT are dependent on central GLP-1 receptor signaling. Furthermore, anatomical evidence shows mRNA expression of 5-HT2C and 5-HT3 receptors on GLP-1-producing preproglucagon (PPG) neurons in the medial nucleus tractus solitarius by fluorescent in situ hybridization, suggesting that PPG neurons are likely to express both of these receptors. Behaviorally, the hypophagia induced by the pharmacological activation of both of these receptors is also dependent on GLP-1 signaling. Finally, 5-HT3, but not 5-HT2C receptors, are required for the anorectic effects of the interoceptive stressor LiCl, suggesting the hypophagia induced by these 5-HT receptors may be driven by different mechanisms. Our findings highlight 5-HT as a novel endogenous modulator of the central GLP-1 system and suggest that the central interaction between 5-HT and GLP-1 is involved in the control of food intake in rats.


Feeding Behavior/drug effects , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Receptor, Serotonin, 5-HT2C/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Serotonin/pharmacology , Stress, Psychological/metabolism , Weight Loss/drug effects , Animals , Anorexia/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Lithium Chloride/pharmacology , Male , Neurons/drug effects , Neurons/metabolism , Ondansetron/pharmacology , Peptide Fragments/pharmacology , Pica/metabolism , Proglucagon , Rats , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Serotonin/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology , Serotonin 5-HT3 Receptor Agonists/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Signal Transduction , Solitary Nucleus/metabolism
7.
Diabetes Obes Metab ; 21(4): 866-875, 2019 04.
Article En | MEDLINE | ID: mdl-30456866

AIMS: To examine whether the glucagon-like peptide-1 receptor agonist liraglutide could be used in juvenile male and female rats as an anti-obesity/diabetic pharmaceutical to prevent not only adolescent obesity/hyperglycaemia, but also early-adult onset obesity. MATERIAL AND METHODS: Pregnant dams were fed either standard chow or a high-fat, high-sucrose diet (HFSD) from gestational day 2, throughout pregnancy and lactation. Offspring were weaned onto the respective maternal diet. Juveniles received daily subcutaneous injection of liraglutide (50 µg/kg, from postnatal day [PND]30 to PND40 and 200 µg/kg from PND40 to PND60) or vehicle. Food intake, body weight and glycaemic levels were evaluated across the experimental period. RESULTS: Chronic liraglutide administration in juveniles prevented body weight gain in males and retained a normoglycaemic profile in both male and female rats. CONCLUSION: These preclinical data suggest that maternal and early-life consumption of an HFSD increases caloric intake, body weight gain and hyperglycaemia, a collective set of unwanted metabolic effects that appear to be treatable in juveniles with liraglutide pharmacotherapy intervention.


Blood Glucose/drug effects , Body Weight/drug effects , Eating/drug effects , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Liraglutide/pharmacology , Obesity/metabolism , Animals , Blood Glucose/metabolism , Diet, High-Fat , Dietary Sucrose , Female , Gestational Weight Gain , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats
8.
Diabetes Obes Metab ; 20(5): 1223-1234, 2018 05.
Article En | MEDLINE | ID: mdl-29327400

AIMS: While pharmacological glucagon-like peptide-1 receptor (GLP-1R) agonists are FDA-approved for treating type 2 diabetes mellitus (T2DM) and obesity, a major side effect is nausea/malaise. We recently developed a conjugate of vitamin B12 (B12) bound to the GLP-1R agonist exendin-4 (Ex4), which displays enhanced proteolytic stability and retention of GLP-1R agonism. Here, we evaluate whether the conjugate (B12-Ex4) can improve glucose tolerance without producing anorexia and malaise. MATERIALS AND METHODS: We evaluated the effects of systemic B12-Ex4 and unconjugated Ex4 on food intake and body weight change, oral glucose tolerance and nausea/malaise in male rats, and on intraperitoneal glucose tolerance in mice. To evaluate whether differences in the profile of effects of B12-Ex4 vs unconjugated Ex4 are the result of altered CNS penetrance, rats received systemic injections of fluorescein-Ex4 (Flex), Cy5-B12 or Cy5-B12-Ex4 and brain penetrance was evaluated using confocal microscopy. Uptake of systemically administered Cy5-B12-Ex4 in insulin-containing pancreatic beta cells was also examined. RESULTS: B12-Ex4 conjugate improves glucose tolerance, but does not elicit the malaise and anorexia produced by unconjugated Ex4. While Flex robustly penetrates into the brain (dorsal vagal complex, paraventricular hypothalamus), Cy5-B12 and Cy5-B12-Ex4 fluorescence were not observed centrally, supporting an absence of CNS penetrance, in line with observed reduction in CNS-associated Ex4 side effects. Cy5-B12-Ex4 colocalizes with insulin in the pancreas, suggesting direct pancreatic action as a potential mechanism underlying the hypoglycaemic effects of B12-Ex4. CONCLUSION: These novel findings highlight the potential clinical utility of B12-Ex4 conjugates as possible future T2DM therapeutics with reduced incidence of adverse effects.


Appetite Regulation/drug effects , Exenatide/analogs & derivatives , Glucagon-Like Peptide-1 Receptor/agonists , Glucose Intolerance/drug therapy , Hypoglycemic Agents/therapeutic use , Vitamin B 12/analogs & derivatives , Animals , Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Drug Stability , Energy Intake/drug effects , Energy Metabolism/drug effects , Exenatide/adverse effects , Exenatide/pharmacokinetics , Exenatide/therapeutic use , Female , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , HEK293 Cells , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice, Inbred C57BL , Nausea/chemically induced , Nausea/prevention & control , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tissue Distribution , Vitamin B 12/adverse effects , Vitamin B 12/pharmacokinetics , Vitamin B 12/therapeutic use
9.
Physiol Behav ; 177: 122-128, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28433470

Glucagon-like peptide-1 (GLP-1) is an incretin hormone released from intestinal L-cells in response to food entering into the gastrointestinal tract. GLP-1-based pharmaceuticals improve blood glucose regulation and reduce feeding. Specific macronutrients, when ingested, may trigger GLP-1 secretion and enhance the effects of systemic sitagliptin, a pharmacological inhibitor of DPP-IV (an enzyme that rapidly degrades GLP-1). In particular, macronutrient constituents found in dairy foods may act as potent secretagogues for GLP-1, and acute preclinical trials show that ingestion of dairy protein may represent a promising adjunct behavioral therapy in combination with sitagliptin. To test this hypothesis further, chow-maintained or high-fat diet (HFD)-induced obese rats received daily IP injections of sitagliptin (6mg/kg) or saline in combination with a twice-daily 8ml oral gavage of milk protein concentrate (MPC; 80/20% casein/whey; 0.5kcal/ml), soy protein (non-dairy control; 0.5kcal/ml) or 0.9% NaCl for two months. Food intake and body weight were recorded every 24-48h; blood glucose regulation was examined at baseline and at 3 and 6.5weeks via a 2h oral glucose tolerance test (OGTT; 25% glucose; 2g/kg). MPC and soy protein significantly suppressed cumulative caloric intake in HFD but not chow-maintained rats. AUC analyses for OGTT show suppression in glycemia by sitagliptin with MPC or soy in chow- and HFD-maintained rats, suggesting that chronic ingestion of dairy or soy proteins may augment endogenous GLP-1 signaling and the glycemic- and food intake-suppressive effects of DPP-IV inhibition.


Anti-Obesity Agents/pharmacology , Dietary Proteins/administration & dosage , Dietary Supplements , Obesity/diet therapy , Obesity/drug therapy , Sitagliptin Phosphate/pharmacology , Animal Feed , Animals , Diet, High-Fat , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Energy Intake/drug effects , Energy Intake/physiology , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , Male , Obesity/metabolism , Rats, Sprague-Dawley
10.
Physiol Behav ; 173: 9-14, 2017 05 01.
Article En | MEDLINE | ID: mdl-28119159

The beneficial glycemic and food intake-suppressive effects of glucagon-like peptide-1 (GLP-1) have made this neuroendocrine system a leading target for pharmacological approaches to the treatment of diabetes and obesity. One strategy to increase the activity of endogenous GLP-1 is to prevent the rapid degradation of the hormone by the enzyme dipeptidyl peptidase-IV (DPP-IV). However, despite the expression of both DPP-IV and GLP-1 in the brain, and the clear importance of central GLP-1 receptor (GLP-1R) signaling for glycemic and energy balance control, the metabolic effects of central inhibition of DPP-IV activity are unclear. To test whether hindbrain DPP-IV inhibition suppresses blood glucose, feeding, and body weight gain, the effects of 4th intracerebroventricular (ICV) administration of the FDA-approved DPP-IV inhibitor sitagliptin were evaluated. Results indicate that hindbrain delivery of sitagliptin improves glycemic control in a GLP-1R-dependent manner, suggesting that this effect is due at least in part to increased endogenous brainstem GLP-1 activity after sitagliptin administration. Furthermore, 4th ICV injection of sitagliptin reduced 24h body weight gain and energy intake, with a selective suppression of high-fat diet, but not chow, intake. These data reveal a novel role for hindbrain GLP-1R activation in glycemic control and also demonstrate that DPP-IV inhibition in the caudal brainstem promotes negative energy balance.


Blood Glucose/physiology , Dipeptidyl Peptidase 4/metabolism , Energy Metabolism/physiology , Rhombencephalon/metabolism , Animals , Area Under Curve , Blood Glucose/drug effects , Body Weight/drug effects , Body Weight/physiology , Diet, High-Fat/methods , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Fasting , Glucose Tolerance Test , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley , Rhombencephalon/drug effects , Sitagliptin Phosphate/pharmacology
...