Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 160
1.
Nature ; 619(7968): 143-150, 2023 Jul.
Article En | MEDLINE | ID: mdl-37380764

Caloric restriction that promotes weight loss is an effective strategy for treating non-alcoholic fatty liver disease and improving insulin sensitivity in people with type 2 diabetes1. Despite its effectiveness, in most individuals, weight loss is usually not maintained partly due to physiological adaptations that suppress energy expenditure, a process known as adaptive thermogenesis, the mechanistic underpinnings of which are unclear2,3. Treatment of rodents fed a high-fat diet with recombinant growth differentiating factor 15 (GDF15) reduces obesity and improves glycaemic control through glial-cell-derived neurotrophic factor family receptor α-like (GFRAL)-dependent suppression of food intake4-7. Here we find that, in addition to suppressing appetite, GDF15 counteracts compensatory reductions in energy expenditure, eliciting greater weight loss and reductions in non-alcoholic fatty liver disease (NAFLD) compared to caloric restriction alone. This effect of GDF15 to maintain energy expenditure during calorie restriction requires a GFRAL-ß-adrenergic-dependent signalling axis that increases fatty acid oxidation and calcium futile cycling in the skeletal muscle of mice. These data indicate that therapeutic targeting of the GDF15-GFRAL pathway may be useful for maintaining energy expenditure in skeletal muscle during caloric restriction.


Energy Metabolism , Growth Differentiation Factor 15 , Muscle, Skeletal , Weight Loss , Animals , Humans , Mice , Appetite Depressants/metabolism , Appetite Depressants/pharmacology , Appetite Depressants/therapeutic use , Caloric Restriction , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Eating/drug effects , Energy Metabolism/drug effects , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/pharmacology , Growth Differentiation Factor 15/therapeutic use , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/therapy , Receptors, Adrenergic, beta/metabolism , Weight Loss/drug effects
2.
Mol Metab ; 58: 101444, 2022 04.
Article En | MEDLINE | ID: mdl-35091058

OBJECTIVE: The behavioral mechanisms and the neuronal pathways mediated by amylin and its long-acting analog sCT (salmon calcitonin) are not fully understood and it is unclear to what extent sCT and amylin engage overlapping or distinct neuronal subpopulations to reduce food intake. We here hypothesize that amylin and sCT recruit different neuronal population to mediate their anorectic effects. METHODS: Viral approaches were used to inhibit calcitonin gene-related peptide (CGRP) lateral parabrachial nucleus (LPBN) neurons and assess their role in amylin's and sCT's ability to decrease food intake in mice. In addition, to test the involvement of LPBN CGRP neuropeptidergic signaling in the mediation of amylin and sCT's effects, a LPBN site-specific knockdown was performed in rats. To deeper investigate whether the greater anorectic effect of sCT compared to amylin is due do the recruitment of additional neuronal pathways related to malaise multiple and distinct animal models tested whether amylin and sCT induce conditioned avoidance, nausea, emesis, and conditioned affective taste aversion. RESULTS: Our results indicate that permanent or transient inhibition of CGRP neurons in LPBN blunts sCT-, but not amylin-induced anorexia and neuronal activation. Importantly, sCT but not amylin induces behaviors indicative of malaise including conditioned affective aversion, nausea, emesis, and conditioned avoidance; the latter mediated by CGRPLPBN neurons. CONCLUSIONS: Together, the present study highlights that although amylin and sCT comparably decrease food intake, sCT is distinctive from amylin in the activation of anorectic neuronal pathways associated with malaise.


Appetite Depressants , Islet Amyloid Polypeptide , Animals , Anorexia/chemically induced , Appetite Depressants/adverse effects , Appetite Depressants/metabolism , Calcitonin , Calcitonin Gene-Related Peptide/metabolism , Islet Amyloid Polypeptide/metabolism , Mice , Nausea/metabolism , Neurons/metabolism , Rats , Vomiting
3.
Article En | MEDLINE | ID: mdl-34534676

Amylin is a 37-amino acid polypeptide that has been found to be involved in feeding regulation in some mammals, birds, and goldfish. We cloned amylin of Siberian sturgeon and detected its distribution pattern in 15 tissues. The expression levels in the periprandial period (pre-and post-feeding), the changes in the food intake, and the expression levels of related appetite factors after the intraperitoneal injection of amylin were detected. The expression of amylin was found to be the highest in the hypothalamus. Compared with 1 h pre-feeding, the expression levels of amylin in the hypothalamus and duodenum were increased significantly 1 h post-feeding. Compared with the control group (saline), intraperitoneal injection of 50 ng/g, 100 ng/g, and 200 ng/g of amylin significantly inhibited food intake at 1 h post injection, but not at 3 h and 6 h. The injection of 50 ng/g, 100 ng/g, and 200 ng/g amylin significantly inhibited the cumulative feed. After 1 h of 50 ng/g amylin injection, the levels of MC4R and somatostatin in the hypothalamus increased significantly, while the levels of amylin and NPY decreased significantly. The levels of CCK in the valvular intestine were increased significantly. Insulin in the duodenum was also increased significantly, but there was no significant change in ghrelin in the duodenum. These results show that amylin inhibits feeding in Siberian sturgeon by down-regulating the appetite-stimulating factor NPY and up-regulating the appetite-suppressing factors somatostatin, MC4R, CCK, and insulin. This study provides a theoretical basis for studying the feeding function and action mechanisms of amylin in Siberian sturgeon.


Fish Proteins/metabolism , Fishes/metabolism , Islet Amyloid Polypeptide/metabolism , Amino Acid Sequence , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/metabolism , Appetite Regulation/drug effects , Appetite Regulation/genetics , Appetite Regulation/physiology , Base Sequence , Cloning, Molecular , Duodenum/metabolism , Eating/drug effects , Eating/genetics , Eating/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Fish Proteins/administration & dosage , Fish Proteins/genetics , Fishes/genetics , Fishes/physiology , Gene Expression/drug effects , Hypothalamus/metabolism , Injections, Intraperitoneal , Islet Amyloid Polypeptide/administration & dosage , Islet Amyloid Polypeptide/genetics , Phylogeny , Sequence Homology, Amino Acid , Tissue Distribution
4.
Drug Test Anal ; 14(1): 101-109, 2022 Jan.
Article En | MEDLINE | ID: mdl-34405558

In recent years, overseas anti-obesity drugs including amfepramone have flowed into China through the internet or personal import by travelers. Amfepramone is controlled in China and is not available as a pharmaceutical product. It is obtainable either through the internet or imported by individuals across the border. The abuse of amfepramone is causing serious health problems. A method for the detection and quantification of amfepramone and its metabolite cathinone in human hair was developed and fully validated using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Approximately 10 mg of hair was weighed and pulverized with extraction solvent (a mixture of methanol: acetonitrile: 2 mM ammonium formate [pH 5.3] [25:29:46, v/v/v]). The limit of detection (LOD) and the limit of quantitation (LOQ) were 5 and 10 pg/mg, respectively. The method was linear over a concentration range from 10 to 10,000 pg/mg. The accuracy varied from -9.3% to 2.3%, with acceptable intra- and inter-day precision. The validated method was successfully applied to 17 authentic cases. The amfepramone concentrations ranged from 11.7 to 209 pg/mg, with a median of 30.2 pg/mg, and the hair cathinone concentrations ranged from 11.9 to 507 pg/mg, with a median of 54.0 pg/mg. This is the first report of amfepramone concentrations in human hair from amfepramone users. Cathinone can be incorporated into hair after amfepramone use.


Alkaloids/analysis , Diethylpropion/analysis , Hair/chemistry , Substance Abuse Detection/methods , Adult , Appetite Depressants/analysis , Appetite Depressants/metabolism , Chromatography, Liquid/methods , Diethylpropion/metabolism , Female , Humans , Limit of Detection , Male , Middle Aged , Tandem Mass Spectrometry/methods
5.
Gut ; 71(5): 928-937, 2022 05.
Article En | MEDLINE | ID: mdl-34083384

OBJECTIVE: Colonic enteroendocrine cells (EECs) store and release potent anorectic hormones that are key regulators of satiety. EECs express multiple nutrient sensing receptors, particularly for medium-chain fatty acids (MCFAs): GPR84 and FFAR4. Here we show a non-surgical approach with targeted colonic delivery of MCFA, which induces EEC and neuronal activation leading to anorectic effects. DESIGN: A randomised, double-blind, placebo-controlled, cross-over study was performed in obese adults given combined GPR84 and FFAR4 agonists in colonic release capsules before meals. We measured serum hormones, energy intake and appetite perception. Cell type, activation by agonists and hormone/serotonin release were determined in human colonic explants. Mouse colonic afferent nerve responses to nutrients/mediators were recorded electrophysiologically. RESULTS: Subjects receiving GPR84 and FFAR4 agonists had reduced overall calorific intake and increased postprandial levels of PYY versus placebo. Receptors including GPR84 and FFAR4 were coexpressed on human colonic EEC. Activation of GPR84 exclusively induced intracellular pERK, whereas FFAR4 selectively activated pCaMKII. Coactivation of GPR84 and FFAR4 induced both phosphoproteins, and superadditive release of GLP-1 and PYY. Nutrients and hormones convergently activated murine colonic afterent nerves via GLP-1, Y2 and 5-HT3 receptors. CONCLUSIONS: Colonic GPR84 and FFAR4 agonists reduce energy intake and increase postprandial PYY in obese adults. Human colonic EECs coexpress these receptors, which activate cells via parallel intracellular pathways and synergistically evoke hormone release. Further synergism occurs in sensory nerve responses to MCFA and EEC mediators. Thus, synergistic activation of colonic endocrine cells via nutrient receptors is an important target for metabolic regulation. TRAIL REGISTRATION NUMBER: NCT04292236.


Appetite Depressants , Animals , Appetite , Appetite Depressants/metabolism , Appetite Depressants/pharmacology , Cross-Over Studies , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Mice , Nutrients , Obesity/metabolism , Receptors, G-Protein-Coupled/metabolism
6.
Cell Rep ; 37(7): 109997, 2021 11 16.
Article En | MEDLINE | ID: mdl-34788630

The anorexigenic effect of serotonergic compounds has largely been attributed to activation of serotonin 2C receptors (Htr2cs). Using mouse genetic models in which Htr2c can be selectively deleted or restored (in Htr2c-null mice), we investigate the role of Htr2c in forebrain Sim1 neurons. Unexpectedly, we find that Htr2c acts in these neurons to promote food intake and counteract the anorectic effect of serotonergic appetite suppressants. Furthermore, Htr2c marks a subset of Sim1 neurons in the paraventricular nucleus of the hypothalamus (PVH). Chemogenetic activation of these neurons in adult mice suppresses hunger, whereas their silencing promotes feeding. In support of an orexigenic role of PVH Htr2c, whole-cell patch-clamp experiments demonstrate that activation of Htr2c inhibits PVH neurons. Intriguingly, this inhibition is due to Gαi/o-dependent activation of ATP-sensitive K+ conductance, a mechanism of action not identified previously in the mammalian nervous system.


GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Animals , Anorexia , Appetite Depressants/metabolism , Appetite Depressants/pharmacology , Energy Metabolism/physiology , Feeding Behavior/physiology , Hunger/physiology , Hypothalamus/metabolism , Hypothalamus/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Neurons/physiology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/physiology , Potassium/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Serotonin/metabolism , Serotonin/pharmacology , Serotonin Agents
7.
Peptides ; 132: 170366, 2020 10.
Article En | MEDLINE | ID: mdl-32634450

Amylin is a peptide hormone that is mainly known to be produced by pancreatic ß-cells in response to a meal but amylin is also produced by brain cells in discrete brain areas albeit in a lesser amount. Amylin receptor (AMY) is composed of the calcitonin core-receptor (CTR) and one of the 3 receptor activity modifying protein (RAMP), thus forming AMY1-3; RAMP enhances amylin binding properties to the CTR. However, amylin receptor agonist such as salmon calcitonin is able to bind CTR alone. Peripheral amylin's main binding site is located in the area postrema (AP) which then propagate the signal to the nucleus of the solitary tract and lateral parabrachial nucleus (LPBN) and it is then transmitted to the forebrain areas such as central amygdala and bed nucleus of the stria terminalis. Amylin's activation of these different brain areas mediates eating and other metabolic pathways controlling energy expenditure and glucose homeostasis. Peripheral amylin can also bind in the arcuate nucleus of the hypothalamus where it acts independently of the AP to activate POMC and NPY neurons. Amylin activation of NPY neurons has been shown to be transmitted to LPBN neurons to act on eating while amylin POMC signaling affects energy expenditure and locomotor activity. While a large amount of experiments have already been conducted, future studies will have to further investigate how amylin is taken up by forebrain areas and deepen our understanding of amylin action on peripheral metabolism.


Appetite Depressants/metabolism , Brain/metabolism , Eating/physiology , Islet Amyloid Polypeptide/metabolism , Animals , Humans , Pancreatic Hormones/metabolism , Signal Transduction
8.
PLoS Biol ; 18(2): e3000629, 2020 02.
Article En | MEDLINE | ID: mdl-32097406

Human biology has evolved to keep body fat within a range that supports survival. During the last 25 years, obesity biologists have uncovered key aspects of physiology that prevent fat mass from becoming too low. In contrast, the mechanisms that counteract excessive adipose expansion are largely unknown. Evidence dating back to the 1950s suggests the existence of a blood-borne molecule that defends against weight gain. In this article, we discuss the research supporting an "unidentified factor of overfeeding" and models that explain its role in body weight control. If it exists, revealing the identity of this factor could end a long-lasting enigma of energy balance regulation and facilitate a much-needed breakthrough in the pharmacological treatment of obesity.


Appetite Depressants/metabolism , Body Weight/physiology , Hormones/metabolism , Adipose Tissue/metabolism , Animals , Appetite Depressants/blood , Hormones/blood , Humans , Hyperphagia/genetics , Hyperphagia/metabolism , Obesity/genetics , Obesity/metabolism , Parabiosis , Weight Gain/physiology
9.
Mini Rev Med Chem ; 20(9): 768-778, 2020.
Article En | MEDLINE | ID: mdl-30961494

This review refers to the all-inclusive details of Lorcaserin Hydrochloride on comprehensive information about the synthesis, analytical methods, pharmacodynamics, pharmacokinetics, drug interactions and adverse effects. Lorcaserin Hydrochloride is chemically (R)-8-Chloro-1-methyl-2,3,4,5- tetrahydro-1H-3-benzazepine hydrochloride. Lorcaserin HCl is a novel, synthetic, centrally-acting selective serotonin C (5-HT2c) receptor, l agonist, which results in increased satiety and decreased food consumption in patients. Headache, dizziness and nausea are the most common side effects associated with this drug. Lorcaserin HCl has two major metabolites, one conjugated with glucuronide called N-carbamoyl glucuronide which is excreted in urine and the second Lorcaserin N-sulfamate, which is circulated in the blood. Lorcaserin HCl is synthesized using four different schemes of which a six-step method that resulted in 92.3% yield with 99.8% of purity is employed for scale-up production. It is analyzed quantitatively in the plasma and brain tissue matrix of rats by Ultra Performance Liquid chromatographic (UPLC) method using MS-MS (Mass Spectrometric) detection.


Benzazepines/chemistry , Serotonin 5-HT2 Receptor Agonists/chemistry , Appetite Depressants/adverse effects , Appetite Depressants/chemistry , Appetite Depressants/metabolism , Appetite Depressants/therapeutic use , Benzazepines/adverse effects , Benzazepines/metabolism , Benzazepines/therapeutic use , Clinical Trials as Topic , Drug Interactions , Half-Life , Headache/etiology , Humans , Obesity/drug therapy , Obesity/pathology , Serotonin 5-HT2 Receptor Agonists/adverse effects , Serotonin 5-HT2 Receptor Agonists/metabolism , Serotonin 5-HT2 Receptor Agonists/therapeutic use
10.
J Am Coll Nutr ; 39(4): 345-351, 2020.
Article En | MEDLINE | ID: mdl-31369353

Nesfatin-1 is a peptide derived from nucleobindin-2 and involved in the regulation of food intake and hyperglycemia. Nesfatin-1 is a recently described anorexigenic peptide, which may be involved in weight loss, malnutrition, and the regulation of appetite. Nesfatin-1 has an effect on the regulation of glucose homeostasis as well as that of food intake. The aim of this article is to bring a different perspective to the readers on the effects of nesfatin-1 on food intake and hyperglycemia. The central injection of nesfatin-1 may produce anorexigenic effects. The circulating level of nesfatin-1 is thought to be regulated by nutritional status. Long-term changes in body weight can affect nesfatin-1 levels. In overweight and obese individuals, nesfatin-1 levels may increase. Nesfatin-1 is synthesized in the hypothalamic appetite control regions. Nesfatin-1 levels may decrease in individuals with diabetes but may increase in those with impaired glucose tolerance. Nesfatin-1 may have a reducing effect on glucose levels. In addition, an increase in glucose levels may lead to an increase in the release of nesfatin-1 from pancreatic cells. Injection of nesfatin-1 can prevent hepatic glucose formation and stimulate glucose uptake. Reduction of hypothalamic nesfatin-1 levels increases hepatic glucose flow and decreases glucose uptake from peripheral tissues. In the light of all this information, nesfatin-1 may be considered to be an important regulator in the metabolic process. Nesfatin-1 appears to be able to contribute to the treatment of obesity and diabetes because of its anorexigenic and antihyperglycemic effects. Key teaching pointsNesfatin-1 is a anorexigenic peptide.Nesfatin-1 is derived from Nucleobindin-2.Nucleobindin-2 mRNA is produced in different areas of the brain.Nesfatin-1 is an inhibitory factor on appetite and a regulator of energy balance that reduces the increase in body weight.


Appetite Depressants/metabolism , Eating/drug effects , Hyperglycemia/metabolism , Nucleobindins/metabolism , Animals , Appetite/drug effects , Body Weight/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Glucose Intolerance/metabolism , Homeostasis/drug effects , Humans , Hypothalamus/metabolism , Nucleobindins/biosynthesis , Nutritional Status/drug effects
11.
Nutr Res ; 71: 56-64, 2019 11.
Article En | MEDLINE | ID: mdl-31757635

Walnuts are a nutrient dense food, but most health research is on English walnuts (EW). Black walnuts (BW) contain a different antioxidant and fatty acid profile, and more protein, compared to EW. The purpose of the study was to compare postprandial responses following the consumption of 3 breakfast meals containing either butter (control), BW, or EW. We hypothesized that walnut-containing meals would mitigate post-meal increases in glucose, insulin, triglycerides, and lipid peroxidation while increasing TAC compared to the traditional meal without nuts. Furthermore, we hypothesized that the BW meal would exhibit greater TAC and subjective fullness while mitigating postprandial increases in lipid peroxidation better than the EW. This was a randomized, double-blind control crossover study in 30 healthy adults with three testing visits. At each visit, subjects consumed either the control, BW, or EW meal. Blood draws and visual analog scale appetite ratings were obtained at fasting, 30, 60, 120, and 180 min postprandially. The BW and EW meals resulted in greater suppression of appetite vs. control (P < .01 and P = .03, respectively), and the BW meal also increased fullness more than EW and control (P < .01 and P < .001, respectively). Finally, the BW meal also had a greater suppression of lipid peroxidation vs. control (P = .01). There were no other treatment differences in the other measures of appetite or for glycemia, triglycerides, or total antioxidant capacity. Substituting butter in a breakfast meal with BW or EW increased fullness; however, the BW meal was superior for suppressing overall appetite while also lowering postprandial lipid peroxidation.


Appetite Depressants/pharmacology , Juglans/metabolism , Lipid Peroxidation/drug effects , Postprandial Period , Satiety Response/drug effects , Adolescent , Adult , Appetite Depressants/administration & dosage , Appetite Depressants/metabolism , Double-Blind Method , Female , Humans , Male , Middle Aged , Young Adult
12.
J Nutr ; 149(3): 362-365, 2019 03 01.
Article En | MEDLINE | ID: mdl-30722047

Oleoylethanolamide is a well-recognized anorectic compound which also has noteworthy effects on food-reward, influencing the acetylcholine (ACh)/dopamine (DA) balance in the cholinergic system. After its administration, oleoylethanolamide is quickly degraded into oleic acid and ethanolamine. The effect of oleic acid on the gut-brain axis has been extensively investigated, whereas ethanolamine has received scarce attention. However, there is scattered evidence from old and recent research that has underlined the influence of ethanolamine on the cholinergic system. In the present article, we propose a model by which the released ethanolamine contributes to the overall balance between DA and ACh after oleoylethanolamide administration.


Acetylcholine/metabolism , Dopamine/metabolism , Endocannabinoids/pharmacology , Ethanolamine/metabolism , Feeding Behavior/drug effects , Oleic Acids/pharmacology , Animals , Appetite Depressants/metabolism , Appetite Depressants/pharmacology , Endocannabinoids/metabolism , Humans , Mice , Models, Biological , Oleic Acids/metabolism , Rats
13.
Neuropeptides ; 74: 95-102, 2019 Apr.
Article En | MEDLINE | ID: mdl-30739813

Central administration of corticotropin-releasing factor (CRF), a 41-amino acid peptide, is associated with potent anorexigenic effects in rodents and chickens. However, the mechanism underlying this effect remains unclear. Hence, the objective of the current study was to elucidate the hypothalamic mechanisms that mediate CRF-induced anorexia in 4 day-old Cobb-500 chicks. After intracerebroventricular (ICV) injection of 0.02 nmol of CRF, CRF-injected chicks ate less than vehicle chicks while no effect on water intake was observed at 30 min post-injection. In subsequent experiments, the hypothalamus samples were processed at 60 min post-injection. The CRF-injected chicks had more c-Fos immunoreactive cells in the arcuate nucleus (ARC), dorsomedial nucleus (DMN), ventromedial hypothalamus (VMH), and paraventricular nucleus (PVN) of the hypothalamus than vehicle-treated chicks. CRF injection was associated with decreased whole hypothalamic mRNA abundance of neuropeptide Y receptor sub-type 1 (NPYR1). In the ARC, CRF-injected chicks expressed more CRF and CRF receptor sub-type 2 (CRFR2) mRNA but less agouti-related peptide (AgRP), NPY, and NPYR1 mRNA than vehicle-injected chicks. CRF-treated chicks expressed greater amounts of CRFR2 and mesotocin mRNA than vehicle chicks in the PVN and VMH, respectively. In the DMN, CRF injection was associated with reduced NPYR1 mRNA. In conclusion, the results provide insights into understanding CRF-induced hypothalamic actions and suggest that the anorexigenic effect of CRF involves increased CRFR2-mediated signaling in the ARC and PVN that overrides the effects of NPY and other orexigenic factors.


Anorexia/metabolism , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Animals , Anorexia/chemically induced , Appetite/drug effects , Appetite Depressants/administration & dosage , Appetite Depressants/metabolism , Chickens , Female , Hypothalamus/drug effects , Male , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects
14.
J Endocrinol ; 240(2): R47-R72, 2019 02 01.
Article En | MEDLINE | ID: mdl-30475219

Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder in the elderly population. Numerous epidemiological and experimental studies have demonstrated that patients who suffer from obesity or type 2 diabetes mellitus have a higher risk of cognitive dysfunction and AD. Several recent studies demonstrated that food intake-lowering (anorexigenic) peptides have the potential to improve metabolic disorders and that they may also potentially be useful in the treatment of neurodegenerative diseases. In this review, the neuroprotective effects of anorexigenic peptides of both peripheral and central origins are discussed. Moreover, the role of leptin as a key modulator of energy homeostasis is discussed in relation to its interaction with anorexigenic peptides and their analogs in AD-like pathology. Although there is no perfect experimental model of human AD pathology, animal studies have already proven that anorexigenic peptides exhibit neuroprotective properties. This phenomenon is extremely important for the potential development of new drugs in view of the aging of the human population and of the significantly increasing incidence of AD.


Alzheimer Disease/prevention & control , Energy Metabolism/drug effects , Leptin/metabolism , Oligopeptides/pharmacology , Pyrrolidonecarboxylic Acid/analogs & derivatives , Alzheimer Disease/metabolism , Animals , Appetite Depressants/metabolism , Appetite Depressants/pharmacology , Disease Models, Animal , Homeostasis/drug effects , Humans , Neuroprotective Agents/pharmacology , Oligopeptides/metabolism , Pyrrolidonecarboxylic Acid/metabolism , Pyrrolidonecarboxylic Acid/pharmacology
15.
Cell Metab ; 28(4): 619-630.e5, 2018 10 02.
Article En | MEDLINE | ID: mdl-30146485

To meet the challenge to human health posed by obesity, a better understanding of the regulation of feeding is essential. Medications targeting 5-hydroxytryptamine (5-HT; serotonin) 2C receptors (htr2c; 5-HT2CR) improve obesity. Here we probed the functional significance of 5-HT2CRs specifically within the brainstem nucleus of the solitary tract (5-HT2CRNTS) in feeding behavior. Selective activation of 5-HT2CRNTS decreased feeding and was sufficient to mediate acute food intake reductions elicited by the 5-HT2CR agonist obesity medication lorcaserin. Similar to pro-opiomelanocortin neurons expressed within the hypothalamic arcuate nucleus (POMCARC), a subset of POMCNTS neurons co-expressed 5-HT2CRs and were activated by 5-HT2CR agonists. Knockdown of POMCNTS prevented the acute appetite-suppressive effect of lorcaserin, whereas POMCARC knockdown prevented the full anorectic effect. These data identify 5-HT2CRNTS as a sufficient subpopulation of 5-HT2CRs in reducing food intake when activated and reveal that 5-HT2CR agonist obesity medications require POMC within the NTS and ARC to reduce food intake.


Appetite Depressants/therapeutic use , Benzazepines/therapeutic use , Eating/physiology , Obesity/drug therapy , Serotonin 5-HT2 Receptor Agonists/therapeutic use , Solitary Nucleus/metabolism , Analysis of Variance , Animals , Appetite Depressants/metabolism , Appetite Regulation/drug effects , Arcuate Nucleus of Hypothalamus/cytology , Benzazepines/metabolism , Cell Line, Tumor , Feeding Behavior/physiology , Male , Mice , Mice, Knockout , Neurons/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Agonists/metabolism , Statistics, Nonparametric , Transfection
16.
Diabetes Obes Metab ; 20(8): 1817-1828, 2018 Aug.
Article En | MEDLINE | ID: mdl-29687585

Type 2 diabetes mellitus is now a worldwide health problem with increasing prevalence. Mounting efforts have been made to treat, prevent and predict this chronic disease. In recent years, increasing evidence from mice and clinical studies suggests that bone-derived molecules modulate glucose metabolism. This review aims to summarize our current understanding of the interplay between bone and glucose metabolism and to highlight potential new means of therapeutic intervention. The first molecule recognized as a link between bone and glucose metabolism is osteocalcin (OCN), which functions in its active form, that is, undercarboxylated OCN (ucOC). ucOC acts in promoting insulin expression and secretion, facilitating insulin sensitivity, and favouring glucose and fatty acid uptake and utilization. A second bone-derived molecule, lipocalin2, functions in suppressing appetite in mice through its action on the hypothalamus. Osteocytes, the most abundant cells in bone matrix, are suggested to act on the browning of white adipose tissue and energy expenditure through secretion of bone morphogenetic protein 7 and sclerostin. The involvement of bone resorption in glucose homeostasis has also been examined. However, there is evidence indicating the implication of the receptor activator of nuclear factor κ-B ligand, neuropeptide Y, and other known and unidentified bone-derived factors that function in glucose homeostasis. We summarize recent advances and the rationale for treating, preventing and predicting diabetes by skeleton intervention.


Bone and Bones/drug effects , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Models, Biological , Prediabetic State/drug therapy , Animals , Appetite Depressants/metabolism , Appetite Depressants/pharmacology , Appetite Depressants/therapeutic use , Appetite Regulation/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/prevention & control , Energy Metabolism/drug effects , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin Secretion/drug effects , Lipocalin-2/genetics , Lipocalin-2/metabolism , Lipocalin-2/pharmacology , Lipocalin-2/therapeutic use , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Neuropeptide Y/therapeutic use , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteocalcin/pharmacology , Osteocalcin/therapeutic use , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Prediabetic State/metabolism , Prediabetic State/pathology , Prediabetic State/prevention & control , RANK Ligand/genetics , RANK Ligand/metabolism , RANK Ligand/pharmacology , RANK Ligand/therapeutic use , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Secretagogues/metabolism , Secretagogues/pharmacology , Secretagogues/therapeutic use
17.
Eur J Nutr ; 57(3): 965-979, 2018 Apr.
Article En | MEDLINE | ID: mdl-28161724

PURPOSE: Increasing secretion and production of glucagon-like peptide-1 (GLP-1) by continuous ingestion of certain food components has been expected to prevent glucose intolerance and obesity. In this study, we examined whether a physiological dose (5% weight in diet) of digestion-resistant maltodextrin (RMD) has a GLP-1-promoting effect in rats fed a high-fat and high-sucrose (HFS) diet. METHODS: Rats were fed a control diet or the HFS (30% fat, 40% sucrose wt/wt) diet supplemented with 5% RMD or fructooligosaccharides (FOS) for 8 weeks or for 8 days in separated experiments. Glucose tolerance, energy intake, plasma and tissue GLP-1 concentrations, and cecal short-chain fatty acids concentrations were assessed. RESULTS: After 4 weeks of feeding, HFS-fed rats had significantly higher glycemic response to oral glucose than control rats, but rats fed HFS + RMD/FOS did not (approx. 50% reduction vs HFS rats). HFS + RMD/FOS-fed rats had higher GLP-1 responses (~twofold) to oral glucose, than control rats. After 8 weeks, visceral adipose tissue weight was significantly higher in HFS-fed rats than control rats, while HFS + RMD/FOS rats had a trend of reduced gain (~50%) of the tissue weight. GLP-1 contents and luminal propionate concentrations in the large intestine increased (>twofold) by adding RMD/FOS to HFS. Eight days feeding of RMD/FOS-supplemented diets reduced energy intake (~10%) and enhanced cecal GLP-1 production (~twofold), compared to HFS diet. CONCLUSIONS: The physiological dose of a prebiotic fiber promptly (within 8 days) promotes GLP-1 production in rats fed an obesogenic diet, which would help to prevent excess energy intake and fat accumulation.


Appetite Depressants/therapeutic use , Dysbiosis/prevention & control , Glucagon-Like Peptide 1/agonists , Obesity/prevention & control , Oligosaccharides/therapeutic use , Polysaccharides/therapeutic use , Prebiotics , Adiposity , Animals , Appetite Depressants/metabolism , Cecum/metabolism , Cecum/microbiology , Cecum/pathology , Diet, Western/adverse effects , Digestion , Dysbiosis/metabolism , Dysbiosis/microbiology , Dysbiosis/pathology , Energy Intake , Fatty Acids, Volatile/metabolism , Fermentation , Gastrointestinal Contents/chemistry , Gastrointestinal Contents/microbiology , Gene Expression Regulation , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intra-Abdominal Fat/pathology , Male , Obesity/metabolism , Obesity/microbiology , Obesity/pathology , Oligosaccharides/metabolism , Organ Size , Polysaccharides/metabolism , Rats, Sprague-Dawley
18.
Arch Physiol Biochem ; 124(2): 156-166, 2018 May.
Article En | MEDLINE | ID: mdl-28857634

The current study aimed to investigate the effect of Citrullus colocynthis (C. colocynthis) hydro-alcoholic extract on blood haemostasis in control and high-fat diet (HFD) induced obese rats. In control rats, the extract significantly enhanced bleeding time and plasma levels of tPA and significantly decreased plasma levels PAI-1 and serum levels of thromboxane B2 leading to inhibition of platelets aggregation. In HFD induced obese rats, similar effects were seen and the extract was also able to reverse HFD induced increases in fibrinogen and VWF. Searching for the mechanism, C. colocynthis acts by (1) inhibiting of food intake, (2) inhibiting the activity of pancreatic lipase, (3) decreasing levels of TNF-α and IL-6 and (4) decreasing circulatory levels of the prothrombotic adipokine, leptin and enhanced circulatory levels of the antithrombic adipokines and adiopnectin. In conclusion, C. colocynthis has antiplatelets and profibrinolytic activity in both control and HFD induced obese rats.


Anti-Obesity Agents/therapeutic use , Antifibrinolytic Agents/therapeutic use , Citrullus colocynthis/chemistry , Dietary Supplements , Obesity/prevention & control , Plant Extracts/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Animals , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/metabolism , Antifibrinolytic Agents/adverse effects , Antifibrinolytic Agents/metabolism , Appetite Depressants/adverse effects , Appetite Depressants/metabolism , Appetite Depressants/therapeutic use , Biomarkers/analysis , Biomarkers/blood , Biomarkers/urine , Bleeding Time , Diet, High-Fat/adverse effects , Dietary Supplements/adverse effects , Feces/chemistry , Fibrinolysis , Fruit/chemistry , Lipid Metabolism , Male , Obesity/blood , Obesity/etiology , Obesity/metabolism , Plant Extracts/adverse effects , Plant Extracts/metabolism , Plasminogen Activator Inhibitor 1/blood , Platelet Aggregation , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/metabolism , Rats, Sprague-Dawley , Thromboxane B2/blood , Tissue Plasminogen Activator/blood
19.
Neuropharmacology ; 130: 62-70, 2018 03 01.
Article En | MEDLINE | ID: mdl-29191753

Melanin-Concentrating Hormone (MCH) is one of the most relevant orexigenic factors specifically located in the lateral hypothalamic area (LHA), with its physiological relevance demonstrated in studies using several genetically manipulated mice models. However, the central mechanisms controlling MCH-induced hyperphagia remain largely uncharacterized. Here, we show that central injection of MCH in mice deficient for kappa opoid receptor (k-OR) failed to stimulate feeding. To determine the hypothalamic area responsible for this MCH/k-OR interaction, we performed virogenetic studies and found that downregulation of k-OR by adeno-associated viruses (shOprk1-AAV) in LHA, but not in other hypothalamic nuclei, was sufficient to block MCH-induced food intake. Next, we sought to investigate the molecular signaling pathway within the LHA that mediates acute central MCH stimulation of food intake. We found that MCH activates k-OR and that increased levels of phosphorylated extracellular signal regulated kinase (ERK) are associated with downregulation of phospho-S6 Ribosomal Protein. This effect was prevented when a pharmacological inhibitor of k-OR was co-administered with MCH. Finally, the specific activation of the direct upstream regulator of S6 (p70S6K) in the LHA attenuated MCH-stimulated food consumption. Our results reveal that lateral hypothalamic k-OR system modulates the orexigenic action of MCH via the p70S6K/S6 pathway.


Eating/drug effects , Hypothalamic Hormones/administration & dosage , Melanins/administration & dosage , Pituitary Hormones/administration & dosage , Receptors, Opioid, kappa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/metabolism , Dependovirus , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , MAP Kinase Signaling System/drug effects , Male , Melanins/metabolism , Mice , Mice, Inbred C57BL , Pituitary Hormones/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/metabolism , Ribosomal Protein S6 Kinases/drug effects , Ribosomal Protein S6 Kinases/metabolism
20.
Nat Med ; 23(12): 1444-1453, 2017 Dec.
Article En | MEDLINE | ID: mdl-29106398

Asprosin is a recently discovered fasting-induced hormone that promotes hepatic glucose production. Here we demonstrate that asprosin in the circulation crosses the blood-brain barrier and directly activates orexigenic AgRP+ neurons via a cAMP-dependent pathway. This signaling results in inhibition of downstream anorexigenic proopiomelanocortin (POMC)-positive neurons in a GABA-dependent manner, which then leads to appetite stimulation and a drive to accumulate adiposity and body weight. In humans, a genetic deficiency in asprosin causes a syndrome characterized by low appetite and extreme leanness; this is phenocopied by mice carrying similar mutations and can be fully rescued by asprosin. Furthermore, we found that obese humans and mice had pathologically elevated concentrations of circulating asprosin, and neutralization of asprosin in the blood with a monoclonal antibody reduced appetite and body weight in obese mice, in addition to improving their glycemic profile. Thus, in addition to performing a glucogenic function, asprosin is a centrally acting orexigenic hormone that is a potential therapeutic target in the treatment of both obesity and diabetes.


Appetite Regulation/genetics , Hypothalamus/metabolism , Microfilament Proteins/physiology , Peptide Fragments/physiology , Peptide Hormones/physiology , Adolescent , Adult , Animals , Appetite Depressants/metabolism , Female , Fibrillin-1 , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Neurons/metabolism , Peptide Fragments/genetics , Peptide Hormones/genetics , Rats , Signal Transduction , Young Adult
...