Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
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.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R856-R865, 2018 10 01.
Article En | MEDLINE | ID: mdl-30133304

The pancreatic hormone amylin is released from beta cells following nutrient ingestion and contributes to the control of body weight and glucose homeostasis. Amylin reduces food intake by activating neurons in the area postrema (AP). Amylin was also shown to synergize with the adipokine leptin, with combination therapy producing greater weight loss and food intake reduction than either hormone alone. Although amylin and leptin were initially thought to interact downstream of the AP in the hypothalamus, recent findings show that the two hormones can act on the same AP neurons, suggesting a more direct relationship. The objective of this study was to determine whether amylin action depends on functional leptin signaling. We tested the ability of amylin to induce satiation and to activate its primary target neurons in the AP in two rodent models of LepR deficiency, the db/db mouse and the Zucker diabetic fatty (ZDF) rat. When compared with wild-type (WT) mice, db/db mice exhibited reduced amylin-induced satiation, reduced amylin-induced Fos in the AP, and a lower expression of calcitonin receptor (CTR) protein, the core component of all amylin receptors. ZDF rats also showed no reduction in food intake following amylin treatment; however, unlike the db/db mice, levels of amylin-induced Fos and CTR in the AP were no different than WT rats. Our results suggest that LepR expression is required for the full anorexic effect of amylin; however, the neuronal activation in the AP seems to depend on the type of LepR mutation.


Amylin Receptor Agonists/pharmacology , Appetite Depressants/pharmacology , Area Postrema/drug effects , Feeding Behavior/drug effects , Islet Amyloid Polypeptide/pharmacology , Leptin/metabolism , Receptors, Leptin/metabolism , Satiety Response/drug effects , Animals , Area Postrema/metabolism , Genotype , Male , Mutation , Phenotype , Proto-Oncogene Proteins c-fos/metabolism , Rats, Zucker , Receptors, Calcitonin/agonists , Receptors, Calcitonin/metabolism , Receptors, Islet Amyloid Polypeptide/drug effects , Receptors, Islet Amyloid Polypeptide/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/drug effects , Receptors, Leptin/genetics , Signal Transduction/drug effects
3.
Neuroscience ; 356: 44-51, 2017 07 25.
Article En | MEDLINE | ID: mdl-28528968

G-protein-coupled receptors (GPCRs) are shown to be involved in Alzheimer's disease (AD) pathogenesis. However, because GPCRs include a large family of membrane receptors, it is unclear which specific GPCR or pathway with rational ligands can become effective therapeutic targets for AD. Amylin receptor (AmR) is a GPCR that mediates several activities, such as improving glucose metabolism, relaxing cerebrovascular structure, modulating inflammatory reactions and potentially enhancing neural regeneration. Recent studies show that peripheral treatments with amylin or its clinical analog, pramlintide, reduced several components of AD pathology, including amyloid plaques, tauopathy, neuroinflammation and other components in the brain, corresponding with improved learning and memory in AD mouse models. Because amylin shares a similar secondary structure with amyloid-ß peptide (Aß), I propose that the AmR/GPCR pathway is disturbed by a large amount of Aß in the AD brain, leading to tau phosphorylation, neuroinflammation and neuronal death in the pathological cascade. Amylin-type peptides, readily crossing the blood-brain barrier (BBB), are the rational ligands to enhance this GPCR pathway and may exhibit utility as novel therapeutic agents for treating AD.


Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Islet Amyloid Polypeptide/therapeutic use , Receptors, Islet Amyloid Polypeptide/metabolism , Animals , Humans , Islet Amyloid Polypeptide/metabolism , Receptors, Islet Amyloid Polypeptide/drug effects
4.
J Pharmacol Exp Ther ; 362(1): 24-30, 2017 07.
Article En | MEDLINE | ID: mdl-28438778

KBP-042 is a dual amylin and calcitonin receptor agonist that increases glucose tolerance and insulin action and reduces body weight in rat models of obesity and prediabetes. The objective of the present study was to 1) evaluate KBP-042 as a treatment of late-stage type 2 diabetes in a rat model and 2) assess the value of adding KBP-042 to the standard of care, metformin, to consider KBP-042 as a relevant drug for treating patients with type 2 diabetes. Two studies were included: an intervention study and a prevention study. In the intervention study, treatment with 5 µg/kg KBP-042 was initiated in 11-week-old Zucker diabetic fatty (ZDF) rats, in which glucose tolerance, fasting glycemia, and glycated hemoglobin were assessed after 4 weeks. In the prevention study, either metformin (400 mg/kg), KBP-042 (5 µg/kg), or a combination of both were administered to ZDF rats for a total of 9 weeks. Glycemia, glucose tolerance, and insulin tolerance were tested. Furthermore, fasting plasma insulin and glucagon levels were evaluated. Finally, pancreatic content of insulin was assessed as a surrogate marker of beta-cell mass. It was found that KBP-042 was efficient in lowering fasting plasma glucose as well as improving glucose tolerance, both as prevention and intervention of disease progression. Furthermore, KBP-042 was efficient in combination with metformin and had additional effects compared with either therapy alone. In conclusion, KBP-042 is a highly relevant therapeutic candidate against type 2 diabetes, effective both as an add-on therapy to metformin and as a stand-alone therapy.


Calcitonin/analogs & derivatives , Diabetes Mellitus, Type 2/drug therapy , Glycated Hemoglobin/analysis , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Receptors, Calcitonin/agonists , Receptors, Islet Amyloid Polypeptide/drug effects , Animals , Blood Glucose/metabolism , Calcitonin/pharmacology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/prevention & control , Drug Therapy, Combination , Glucagon/blood , Glucose Tolerance Test , Hyperglycemia/blood , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Pancreas/drug effects , Pancreas/metabolism , Rats , Rats, Zucker
5.
Curr Opin Endocrinol Diabetes Obes ; 20(1): 8-13, 2013 Feb.
Article En | MEDLINE | ID: mdl-23183359

PURPOSE OF REVIEW: This review focuses on recent advances in receptor signaling, neurobiology, and pharmacological interactions of amylin with nutritive status, as well as other metabolism-related regulatory signals. RECENT FINDINGS: Manipulation of components of the amylin receptor complex revealed important roles for the accessory proteins of amylin receptors in energy balance. In-vitro findings point to potential novel sites of action and postreceptor signaling pathways activated by amylin. Neurobiological studies elucidated how amylin activation of hindbrain neural circuitry modulates hypothalamic signaling and responsiveness to leptin. The notion of 'amylin resistance' was addressed in several models (drug or diet-induced hyper-amylinemia). Finally, progress in the design and delivery of amylinomimetics is briefly discussed. SUMMARY: Collectively, these mechanistic studies deepen our understanding of the role of endogenous amylin in the regulation of appetite and adiposity, and hopefully will help guide research efforts towards the development of more effective amylin-based therapies for metabolic diseases.


Adiposity/drug effects , Appetite Depressants/pharmacology , Appetite Regulation/drug effects , Hypothalamus/drug effects , Islet Amyloid Polypeptide/pharmacology , Obesity/metabolism , Signal Transduction/drug effects , Adiposity/physiology , Amyloid/metabolism , Animals , Appetite Regulation/physiology , Energy Metabolism , Humans , Islet Amyloid Polypeptide/deficiency , Leptin/metabolism , Mice , Mice, Inbred C57BL , Neurobiology/trends , Obesity/drug therapy , Pharmacology/trends , Rats , Rats, Inbred Strains , Receptors, Islet Amyloid Polypeptide/drug effects , Receptors, Islet Amyloid Polypeptide/metabolism
6.
Cell Mol Life Sci ; 69(12): 1947-65, 2012 Jun.
Article En | MEDLINE | ID: mdl-22193913

Amylin is an important control of nutrient fluxes because it reduces energy intake, modulates nutrient utilization by inhibiting postprandial glucagon secretion, and increases energy disposal by preventing compensatory decreases of energy expenditure in weight-reduced individuals. The best investigated function of amylin which is cosecreted with insulin is to reduce eating by promoting meal-ending satiation. This effect is thought to be mediated by a stimulation of specific amylin receptors in the area postrema. Secondary brain sites to mediate amylin action include the nucleus of the solitary tract and the lateral parabrachial nucleus, which convey the neural signal to the lateral hypothalamic area and other hypothalamic nuclei. Amylin may also signal adiposity because plasma levels of amylin are increased in adiposity and because higher amylin concentrations in the brain result in reduced body weight gain and adiposity, while amylin receptor antagonists increase body adiposity. The central mechanisms involved in amylin's effect on energy expenditure are much less known. A series of recent experiments in animals and humans indicate that amylin is a promising option for anti-obesity therapy especially in combination with other hormones. The most extensive dataset is available for the combination therapy of amylin and leptin. Ongoing research focuses on the mechanisms of these interactions.


Energy Metabolism/physiology , Homeostasis , Islet Amyloid Polypeptide/physiology , Adiposity/drug effects , Adiposity/physiology , Animals , Anti-Obesity Agents/pharmacology , Brain Stem/drug effects , Brain Stem/physiology , Energy Metabolism/drug effects , Female , Gastric Emptying/drug effects , Gastric Emptying/physiology , Humans , Insulin/physiology , Islet Amyloid Polypeptide/drug effects , Leptin/physiology , Male , Mice , Obesity/drug therapy , Obesity/physiopathology , Rats , Receptors, Islet Amyloid Polypeptide/drug effects , Receptors, Islet Amyloid Polypeptide/physiology , Satiation/drug effects , Satiation/physiology
7.
Am J Physiol Regul Integr Comp Physiol ; 302(3): R340-51, 2012 Feb 01.
Article En | MEDLINE | ID: mdl-22129618

Peripheral amylin inhibits eating via the area postrema (AP). Because amylin activates the extracellular-signal regulated kinase 1/2 (ERK) pathway in some tissues, and because ERK1/2 phosphorylation (pERK) leads to acute neuronal responses, we postulated that it may be involved in amylin's eating inhibitory effect. Amylin-induced ERK phosphorylation (pERK) was investigated by immunohistochemistry in brain sections containing the AP. pERK-positive AP neurons were double-stained for the calcitonin 1a/b receptor, which is part of the functional amylin-receptor. AP sections were also phenotyped using dopamine-ß-hydroxylase (DBH) as a marker of noradrenergic neurons. The effect of fourth ventricular administration of the ERK cascade blocker U0126 on amylin's eating inhibitory action was tested in feeding trials. The number of pERK-positive neurons in the AP was highest ∼10-15 min after amylin treatment; the effect appeared to be dose-dependent (5-20 µg/kg amylin). A portion of pERK-positive neurons in the AP carried the amylin-receptor and 22% of the pERK-positive neurons were noradrenergic. Pretreatment of rats with U0126 decreased the number of pERK-positive neurons in the AP after amylin injection. U0126 also attenuated the ability of amylin to reduce eating, at least when the animals had been fasted 24 h prior to the feeding trial. Overall, our results suggest that amylin directly stimulates pERK in AP neurons in a time- and dose-dependent manner. Part of the AP neurons displaying pERK were noradrenergic. At least under fasting conditions, pERK was shown to be a necessary part in the signaling cascade mediating amylin's anorectic effect.


Anorexia/physiopathology , Appetite Regulation/drug effects , Appetite Regulation/physiology , Islet Amyloid Polypeptide/pharmacology , MAP Kinase Signaling System/physiology , Animals , Area Postrema/drug effects , Area Postrema/pathology , Area Postrema/physiopathology , Butadienes/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fourth Ventricle/drug effects , Fourth Ventricle/pathology , Fourth Ventricle/physiopathology , MAP Kinase Signaling System/drug effects , Male , Nitriles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Islet Amyloid Polypeptide/drug effects , Receptors, Islet Amyloid Polypeptide/physiology , Time Factors
...