Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 197: 108746, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34371079

ABSTRACT

Increasing evidence indicates that the melanocortin and mesolimbic dopamine (DA) systems interact to regulate feeding and body weight. Because melanocortin-3 receptors (MC3R) are highly expressed in the ventral tegmental area (VTA), we tested whether VTA neurons expressing these receptors (VTA MC3R neurons) control feeding and body weight in vivo. We also tested whether there were sex differences in the ability of VTA MC3R neurons to control feeding, as MC3R -/- mice show sex-dependent alterations in reward feeding and DA levels, and there are clear sex differences in multiple DA-dependent behaviors and disorders. Designer receptors exclusively activated by designer drugs (DREADD) were used to acutely activate and inhibit VTA MC3R neurons and changes in food intake and body weight were measured. Acutely altering the activity of VTA MC3R neurons decreased feeding in an activity- and sex-dependent manner, with acute activation decreasing feeding, but only in females, and acute inhibition decreasing feeding, but only in males. These differences did not appear to be due to sex differences in the number of VTA MC3R neurons, the ability of hM3Dq to activate VTA MC3R neurons, or the proportion of VTA MC3R neurons expressing tyrosine hydroxylase (TH). These studies demonstrate an important role for VTA MC3R neurons in the control of feeding and reveal important sex differences in behavior, whereby opposing changes in neuronal activity in male and female mice cause similar changes in behavior.


Subject(s)
Motor Activity/physiology , Neurons/physiology , Receptor, Melanocortin, Type 3/physiology , Ventral Tegmental Area/physiology , Animals , Body Weight , Designer Drugs/pharmacology , Dopamine/metabolism , Feeding Behavior , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Receptor, Melanocortin, Type 3/genetics , Reward , Sex Characteristics , Ventral Tegmental Area/cytology
2.
Domest Anim Endocrinol ; 74: 106507, 2021 01.
Article in English | MEDLINE | ID: mdl-32841887

ABSTRACT

The melanocortin-3 receptor (MC3R) is a G protein-coupled receptor and potentially important in production traits. Three naturally occurring mutations (M54L, G104S, and L151R) in chicken MC3R (cMC3R) were reported previously to be associated with production traits. Here, we inserted the full-length cMC3R coding sequence into pcDNA3.1(+) and generated the 3 mutations by site-directed mutagenesis. The total and cell surface expression of the receptors was measured by flow cytometry. We analyzed the pharmacological characteristics, including binding and cyclic adenosine monophosphate (cAMP) and mitogen-activated protein kinase (MAPK) signaling, using 6 ligands ([Nle4, D-Phe7]-α-melanocyte stimulating hormone (MSH), α-, ß-, γ-, and D-Trp8-γ-MSHs, and agouti-related peptide). All mutants had similar total and cell surface expression as the wild-type (WT) cMC3R. M54L had similar pharmacological properties as the WT cMC3R. G104S did not exhibit any specific binding but had minimal response to α-, ß-, γ-, and D-Trp8-γ-MSH, although it generated 24% WT response when stimulated by NDP-MSH. Although L151R had normal binding, the responses to agonists were reduced to approximately 25% of that of the WT. In MAPK signaling, all 3 mutants showed significantly increased agonist-stimulated phosphorylation of extracellular signal-regulated protein kinases 1/2, indicating the existence of biased signaling at G104S and L151R. In summary, our studies demonstrated that although all 3 mutations are significantly associated with production traits, only G104S and L151R had severe defects in receptor pharmacology. How M54L might cause production trait differences remains to be investigated.


Subject(s)
Chickens/genetics , Mutation/genetics , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 3/physiology , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cyclic AMP/metabolism , Gene Expression , HEK293 Cells , Humans , MAP Kinase Signaling System/physiology , Melanocyte-Stimulating Hormones/metabolism , Protein Binding , Receptor, Melanocortin, Type 3/chemistry , Signal Transduction
3.
J Physiol ; 597(12): 3217-3232, 2019 06.
Article in English | MEDLINE | ID: mdl-31054267

ABSTRACT

KEY POINTS: Alpha-melanocyte stimulating hormone (α-MSH) is an anorexigenic peptide. Injection of the α-MSH analog MTII into the ventral tegmental area (VTA) decreases food and sucrose intake and food reward. Melanocortin-3 receptors (MC3R) are highly expressed in the VTA, suggesting that the effects of intra-VTA α-MSH may be mediated by α-MSH changing the activity of MC3R-expressing VTA neurons. α-MSH increased the firing rate of MC3R VTA neurons in acute brain slices from mice, although it did not affect the firing rate of non-MC3R VTA neurons. The α-MSH induced increase in MC3R neuron firing rate is probably activity-dependent, and was independent of fast synaptic transmission and intracellular Ca2+ levels. These results help us to better understand how α-MSH acts in the VTA to affect feeding and other dopamine-dependent behaviours. ABSTRACT: The mesocorticolimbic dopamine system, the brain's reward system, regulates multiple behaviours, including food intake and food reward. There is substantial evidence that the melanocortin system of the hypothalamus, an important neural circuit controlling feeding and body weight, interacts with the mesocorticolimbic dopamine system to affect feeding, food reward and body weight. For example, melanocortin-3 receptors (MC3Rs) are expressed in the ventral tegmental area (VTA) and our laboratory previously showed that intra-VTA injection of the MC3R agonist, MTII, decreases home-cage food intake and operant responding for sucrose pellets. However, the cellular mechanisms underlying the effects of intra-VTA alpha-melanocyte stimulating hormone (α-MSH) on feeding and food reward are unknown. To determine how α-MSH acts in the VTA to affect feeding, we performed electrophysiological recordings in acute brain slices from mice expressing enhanced yellow fluorescent protein in MC3R neurons to test how α-MSH affects the activity of VTA MC3R neurons. α-MSH significantly increased the firing rate of VTA MC3R neurons without altering the activity of non-MC3R expressing VTA neurons. In addition, the α-MSH-induced increase in MC3R neuron activity was independent of fast synaptic transmission and intracellular Ca2+ levels. Finally, we show that the effect of α-MSH on MC3R neuron firing rate is probably activity-dependent. Overall, these studies provide an important advancement in the understanding of how α-MSH acts in the VTA to affect feeding and food reward.


Subject(s)
Receptor, Melanocortin, Type 3/physiology , Ventral Tegmental Area/physiology , alpha-MSH/physiology , Animals , Female , In Vitro Techniques , Male , Mice, Transgenic , Neurons/physiology
4.
Sci Adv ; 4(8): eaat0866, 2018 08.
Article in English | MEDLINE | ID: mdl-30140740

ABSTRACT

Like most homeostatic systems, adiposity in mammals is defended between upper and lower boundary conditions. While leptin and melanocortin-4 receptor (MC4R) signaling are required for defending energy set point, mechanisms controlling upper and lower homeostatic boundaries are less well understood. In contrast to the MC4R, deletion of the MC3R does not produce measurable hyperphagia or hypometabolism under normal conditions. However, we demonstrate that MC3R is required bidirectionally for controlling responses to external homeostatic challenges, such as caloric restriction or calorie-rich diet. MC3R is also required for regulated excursion from set point, or rheostasis, during pregnancy. Further, we demonstrate a molecular mechanism: MC3R provides regulatory inputs to melanocortin signaling, acting presynaptically on agouti-related protein neurons to regulate γ-aminobutyric acid release onto anorexigenic MC4R neurons, exerting boundary control on the activity of MC4R neurons. Thus, the MC3R is a critical regulator of boundary controls on melanocortin signaling, providing rheostatic control on energy storage.


Subject(s)
Energy Metabolism , Feeding Behavior , Homeostasis , Inhibitory Postsynaptic Potentials/physiology , Neurons/physiology , Receptor, Melanocortin, Type 3/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL
5.
Child Obes ; 14(4): 218-226, 2018.
Article in English | MEDLINE | ID: mdl-29688747

ABSTRACT

BACKGROUND: Two common missense variants in the melanocortin-3 receptor (MC3R) gene, Thr6Lys (T6K) and Val81Ile (V81I), are presumably correlated with pediatric obesity. This meta-analysis aimed to examine and synthesize evidence on the association between these two common MC3R polymorphisms and the development of childhood obesity. METHODS: A combination of words relevant to the research question was searched on PubMed, EMBASE, Scopus, and the Cochrane database. Results were restricted to human studies, specifically child and adolescent populations. Articles were excluded based on accessibility of full online texts and availability of pertinent data. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using a random effects model to determine the association of the polymorphisms with obesity. RESULTS: Searches on the databases using the keywords identified 65 potentially relevant reports. Among them, 32 studies were excluded due to irrelevance, and 28 studies excluded due to lack of access, insufficient data, and investigation of other variants. A final set of five studies included in this meta-analysis found that the risk of overweight/obesity increased by 46.1% per K allele and 21.7% per I allele. Only homozygous genotypes for T6K were associated with a 3.10-fold (95% CI: 1.29-7.43) increased risk of overweight/obesity in children. Data were insufficient to examine if homozygosity for both rare alleles further increases risk. CONCLUSIONS: Our results supported a recessive inheritance model for MC3R gene as a potential cause of childhood obesity. High clinical heterogeneity existed among studies and thus requires more research of larger participation for future integration of data.


Subject(s)
Genetic Predisposition to Disease/genetics , Mutation, Missense/genetics , Pediatric Obesity/genetics , Receptor, Melanocortin, Type 3/genetics , Threonine/genetics , Valine/genetics , Child , Humans , Pediatric Obesity/physiopathology , Polymorphism, Genetic , Receptor, Melanocortin, Type 3/physiology
6.
Neuropsychopharmacology ; 41(9): 2241-51, 2016 08.
Article in English | MEDLINE | ID: mdl-26852738

ABSTRACT

The central melanocortin (MC) system mediates its effects on food intake via MC3 (MC3R) and MC4 receptors (MC4R). Although the role of MC4R in meal size determination, satiation, food preference, and motivation is well established, the involvement of MC3R in the modulation of food intake has been less explored. Here, we investigated the role of MC3R on the incentive motivation for food, which is a crucial component of feeding behavior. Dopaminergic neurons within the ventral tegmental area (VTA) have a crucial role in the motivation for food. We here report that MC3Rs are expressed on VTA dopaminergic neurons and that pro-opiomelanocortinergic (POMC) neurons in the arcuate nucleus of the hypothalamus (Arc) innervate these VTA dopaminergic neurons. Our findings show that intracerebroventricular or intra-VTA infusion of the selective MC3R agonist γMSH increases responding for sucrose under a progressive ratio schedule of reinforcement, but not free sucrose consumption in rats. Furthermore, ex vivo electrophysiological recordings show increased VTA dopaminergic neuronal activity upon γMSH application. Consistent with a dopamine-mediated effect of γMSH, the increased motivation for sucrose after intra-VTA infusion of γMSH was blocked by pretreatment with the dopamine receptor antagonist α-flupenthixol. Taken together, we demonstrate an Arc POMC projection onto VTA dopaminergic neurons that modulates motivation for palatable food via activation of MC3R signaling.


Subject(s)
Dopaminergic Neurons/physiology , Feeding Behavior , Motivation , Receptor, Melanocortin, Type 3/physiology , Reward , Ventral Tegmental Area/physiology , Action Potentials/drug effects , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Dopaminergic Neurons/cytology , Eating/drug effects , Food , Male , Neurons/cytology , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Rats, Wistar , Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/metabolism , Signal Transduction , Sucrose/administration & dosage , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , gamma-MSH/administration & dosage
7.
Bull Exp Biol Med ; 160(1): 40-4, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26593415

ABSTRACT

The peptides structurally corresponding in to cytoplasmic loops of G protein-coupled receptors (GPCR) are able to control functional activity of homologous receptors and the corresponding signaling pathways. Modification of these peptides with hydrophobic radicals enhances their biological activity due to penetration of lipophilic derivatives through the membrane and anchoring near their targets, GPCR. We synthesized an N-palmitoylated peptide Palm-Val-[Lys-Asn-Lys-Asn-Leu-His-Ser-Pro-(Nle)-Tyr-Phe-Phe71-82]-amide-Palm-Val-(71-82) structurally corresponding to cytoplasmic loop 1 of melanocortin 4 receptor (M4R). We found that in micromolar concentrations it very effectively suppresses stimulation of basal adenylate cyclase activity and basal level of GppNHp binding of heterotrimeric G proteins produced by THIQ and α-melanocyte stimulating hormone (α-MSH), agonists of M4R homologous to the peptide, in synaptosomal membranes of rat brain. The peptide Palm-Val-(71-82) also reduced, albeit to a significantly less extent, stimulation of adenylate cyclase and G-proteins by M3R agonist of γ-MSH, due to high homology of the peptide primary structure to M3R cytoplasmic loop 1. The synthesized peptide with activity of M4R/M3R antagonist can be used for the development of regulators of M4R and M3R and the corresponding biochemical and physiological processes.


Subject(s)
Receptor, Melanocortin, Type 4/antagonists & inhibitors , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Brain Chemistry , Guanylyl Imidodiphosphate/pharmacology , Lipoylation , Male , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Protein Processing, Post-Translational , Rats , Rats, Wistar , Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/physiology , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/chemistry , Signal Transduction/physiology , Synaptosomes/drug effects , Synaptosomes/metabolism , Tetrahydroisoquinolines/pharmacology , Triazoles/pharmacology , alpha-MSH/pharmacology , gamma-MSH/pharmacology
8.
J Hypertens ; 33(6): 1201-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25668357

ABSTRACT

BACKGROUND: Although we previously demonstrated that activation of central nervous system (CNS) melanocortin3/4 receptors (MC3/4R) play a key role in blood pressure (BP) regulation, especially in spontaneously hypertensive rats (SHRs), the importance of hindbrain MC4R is still unclear. METHOD: In the present study, we examined the cardiovascular and metabolic effects of chronic inhibition of MC3/4R in the hindbrain of SHRs and normotensive Wistar-Kyoto (WKY) rats. Male WKY rats (n = 6) and SHRs (n = 7) were implanted with telemetry probes to measure BP and heart rate (HR) 24 h/day, and an intracerebroventricular cannula was placed into the fourth ventricle. After 10 days of recovery and 5 days of control measurements, the MC3/4R antagonist (SHU-9119) was infused into the fourth ventricle (1 nmol/h) to antagonize hindbrain MC4R for 10 days, followed by a 5-day recovery period. RESULTS: Chronic hindbrain MC3/4R antagonism significantly increased food intake and body weight in WKY rats (17 ±â€Š1 to 35 ±â€Š2 g/day and 280 ±â€Š8 to 353 ±â€Š8 g) and SHRs (19 ±â€Š2 to 35 ±â€Š2 g/day and 323 ±â€Š7 to 371 ±â€Š11 g), and markedly increased fasting insulin and leptin levels while causing no changes in blood glucose levels (99 ±â€Š4 to 87 ±â€Š4 and 89 ±â€Š5 to 89 ±â€Š4 mg/dl, respectively, for WKY rats and SHRs). Chronic SHU-9119 infusion reduced mean arterial pressure and HR similarly in WKY rats (-8 ±â€Š1 mmHg and -47 ±â€Š3 b.p.m.) and SHRs (-11 ±â€Š3 mmHg and -44 ±â€Š3 b.p.m.). CONCLUSION: These results suggest that although hindbrain MC4R activity contributes to appetite and HR regulation, it does not play a major role in mediating the elevated BP in SHRs.


Subject(s)
Appetite/physiology , Blood Pressure/physiology , Heart Rate/physiology , Hypertension/physiopathology , Receptor, Melanocortin, Type 4/physiology , Rhombencephalon/physiology , Animals , Body Weight , Eating , Insulin/blood , Leptin/metabolism , Male , Melanocyte-Stimulating Hormones/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/physiology , Receptor, Melanocortin, Type 4/antagonists & inhibitors
9.
J Physiol ; 593(7): 1633-47, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25398524

ABSTRACT

Obesity and hypertension are commonly associated, and activation of the sympathetic nervous system is considered to be a major contributor, at least in part due to the central actions of leptin. However, while leptin increases sympathetic nerve activity (SNA) in males, whether leptin is equally effective in females is unknown. Here, we show that intracerebroventricular (i.c.v.) leptin increases lumbar (LSNA) and renal (RSNA) SNA and baroreflex control of LSNA and RSNA in α-chloralose anaesthetized female rats, but only during pro-oestrus. In contrast, i.c.v. leptin increased basal and baroreflex control of splanchnic SNA (SSNA) and heart rate (HR) in rats in both the pro-oestrus and dioestrus states. The effects of leptin on basal LSNA, RSNA, SSNA and HR were similar in males and pro-oestrus females; however, i.c.v. leptin increased mean arterial pressure (MAP) only in males. Leptin did not alter LSNA or HR in ovariectomized rats, but its effects were normalized with 4 days of oestrogen treatment. Bilateral nanoinjection of SHU9119 into the paraventricular nucleus of the hypothalamus (PVN), to block α-melanocyte-stimulating hormone (α-MSH) type 3 and 4 receptors, decreased LSNA in leptin-treated pro-oestrus but not dioestrus rats. Unlike leptin, i.c.v. insulin infusion increased basal and baroreflex control of LSNA and HR similarly in pro-oestrus and dioestrus rats; these responses did not differ from those in male rats. We conclude that, in female rats, leptin's stimulatory effects on SNA are differentially enhanced by oestrogen, at least in part via an increase in α-MSH activity in the PVN. These data further suggest that the actions of leptin and insulin to increase the activity of various sympathetic nerves occur via different neuronal pathways or cellular mechanisms. These results may explain the poor correlation in females of SNA with adiposity, or of MAP with leptin.


Subject(s)
Baroreflex/drug effects , Estrogens/pharmacology , Kidney/innervation , Leptin/pharmacology , Lumbosacral Region/innervation , Splanchnic Nerves/drug effects , Animals , Baroreflex/physiology , Estradiol/blood , Estradiol/pharmacology , Estrogens/blood , Estrous Cycle/drug effects , Estrous Cycle/physiology , Female , Insulin/pharmacology , Male , Melanocyte-Stimulating Hormones/pharmacology , Ovariectomy , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiology , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/physiology , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/physiology , Splanchnic Nerves/physiology
10.
J Mol Endocrinol ; 53(3): 319-30, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25228159

ABSTRACT

The melanocortin 3 receptor (MC3R) regulates several physiological functions, including feed efficiency, nutrient partitioning, fasting response, natriuresis, and immune reactions. Naturally occurring mutations in the MC3R gene have been shown to be associated with increased adiposity and lung diseases such as tuberculosis and cystic fibrosis. The DRY motif at the cytoplasmic end of transmembrane domain 3 (TM3) and the second intracellular loop 2 (ICL2) are known to be important for receptor function in several G protein-coupled receptors (GPCRs). To gain a better understanding of the functions of this domain in MC3R, we performed alanine-scanning mutagenesis on 18 residues. We showed that alanine mutation of 11 residues reduced the maximal binding and maximal cAMP production stimulated by agonists. Mutation of two residues did not change maximal binding but resulted in impaired signaling in the Gs-cAMP pathway. Mutation of five residues impaired signaling in the ERK1/2 pathway. We have also shown that alanine mutants of seven residues that were defective in the cAMP pathway were not defective in the ERK1/2 pathway, demonstrating biased signaling. In summary, we demonstrated that the cytoplasmic end of TM3 and the ICL2 were critical for MC3R function. We also reported for the first time biased signaling in MC3R.


Subject(s)
Receptor, Melanocortin, Type 3/chemistry , Receptor, Melanocortin, Type 3/physiology , Amino Acid Sequence , Amino Acid Substitution , Cells, Cultured , HEK293 Cells , Humans , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding/genetics , Protein Structure, Tertiary/physiology , Structure-Activity Relationship , Transfection
11.
Proc Natl Acad Sci U S A ; 109(23): E1489-98, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22573815

ABSTRACT

The melanocortin-3 receptor-deficient (MC3-R(-/-)) mouse exhibits mild obesity without hyperphagia or hypometabolism. MC3-R deletion is reported to increase adiposity, reduce lean mass and white adipose tissue inflammation, and increase sensitivity to salt-induced hypertension. We show here that the MC3-R(-/-) mouse exhibits defective fasting-induced white adipose tissue lipolysis, fasting-induced liver triglyceride accumulation, fasting-induced refeeding, and fasting-induced regulation of the adipostatic and hypothalamic-adrenal-pituitary axes. Close examination of the hypothalamic-pituitary-adrenal axis showed that MC3-R(-/-) mice exhibit elevated nadir corticosterone as well as a blunted fasting-induced activation of the axis. The previously described phenotypes of this animal and the reduced bone density reported here parallel those of Cushing syndrome. Thus, MC3-R is required for communicating nutritional status to both central and peripheral tissues involved in nutrient partitioning, and this defect explains much of the metabolic phenotype in the model.


Subject(s)
Energy Metabolism/physiology , Fasting/physiology , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/physiology , Receptor, Melanocortin, Type 3/physiology , Absorptiometry, Photon , Adipose Tissue, White/metabolism , Adiposity/genetics , Adrenal Glands/cytology , Analysis of Variance , Animals , Biomechanical Phenomena , Blotting, Western , Body Composition/physiology , Corticosterone/metabolism , Immunohistochemistry , In Situ Hybridization , Lipolysis/physiology , Liver/metabolism , Male , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptor, Melanocortin, Type 3/deficiency , Triglycerides/metabolism
12.
J. physiol. biochem ; 68(1): 71-76, mar. 2012.
Article in English | IBECS | ID: ibc-122379

ABSTRACT

No disponible


Mice genetically deficient in the melanocortin-3 receptor gene are characterized by normal body weight, increased body fat, mild hypophagia, reduced locomotor activity, and increased respiratory quotient compared with wild-type mice. In humans, the 6Lys-81Ile haplotype of melanocortin-3 receptor (MC3R) gene has been associated with childhood obesity, higher body fat percentage, and reduced fat oxidation compared to non-carriers. The aim of this study was to evaluate the association between MC3R 6Lys-81Ile haplotype with body composition and substrate oxidation in response to moderate exercise in obese children. Eight Chilean obese children (aged 8-12) carriers of MC3R 6Lys-81Ile haplotype were compared with eight age-gender-matched obese non-carriers. Children were identified through a previous cross-sectional study on genetic determinants of childhood obesity (..) (AU)


Subject(s)
Humans , Male , Female , Child , Receptor, Melanocortin, Type 3/physiology , Obesity/genetics , Body Composition/genetics , Exercise/physiology , Substrates for Biological Treatment , Case-Control Studies , Genetic Markers , Glucose/metabolism
13.
Genes Brain Behav ; 11(3): 291-302, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22353545

ABSTRACT

The central nervous melanocortin system forms a neural network that maintains energy homeostasis. Actions involving neural melanocortin-3 receptors (MC3Rs) regulate the expression rhythms in ingestive behaviors and metabolism anticipating nutrient intake. Here, we characterized the response of Mc3r knockout (Mc3r(-/-)) and wild type (WT) mice to a restricted feeding (RF) schedule where food access was limited to a 4-h period mid light cycle using a mechanical barrier. Mc3r(-/-) mice adapted poorly to the food restriction schedule. Anticipatory activity and the initial bout of intense feeding activity associated with granting food access were attenuated in Mc3r(-/-) mice, resulting in increased weight loss relative to controls. To investigate whether activity in specific hypothalamic nuclei contribute to the Mc3r(-/-) phenotype observed, we assessed hypothalamic FOS-immunoreactivity (FOS-IR) associated with food restriction. Food access markedly increased FOS-IR in the dorsomedial hypothalamus (DMH), but not in the suprachiasmatic or ventromedial hypothalamic nuclei (SCN and VMN, respectively) compared to ad libitum fed mice. Mc3r(-/-) mice displayed a significant reduction in FOS-IR in the DMH during feeding. Analysis of MC3R signaling in vitro indicated dose-dependent stimulation of the extracellular signal-regulated kinase (ERK) pathway by the MC3R agonist d-Trp(8)-γMSH. Treatment of WT mice with d-Trp(8)-γMSH administered intracerebroventricularly increased the number of pERK neurons 1.7-fold in the DMH. These observations provide further support for the involvement of the MC3Rs in regulating adaptation to food restriction. Moreover, MC3Rs may modulate the activity of neurons in the DMH, a region previously linked to the expression of the anticipatory response to RF.


Subject(s)
Adaptation, Physiological , Caloric Restriction , Feeding Behavior/physiology , Receptor, Melanocortin, Type 3/physiology , Starvation/metabolism , Adaptation, Physiological/genetics , Animals , Caloric Restriction/adverse effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/genetics , Starvation/genetics , Starvation/physiopathology
14.
Neuroendocrinology ; 95(4): 277-88, 2012.
Article in English | MEDLINE | ID: mdl-22327910

ABSTRACT

The worldwide obesity epidemic is increasing, yet at this time, no long-acting and specific pharmaceutical therapies are available. Peripheral hormonal signals communicate metabolic status to the hypothalamus by activating their corresponding receptors in the arcuate nucleus (ARC). In this brain region, a variety of G protein-coupled receptors (GPCRs) are expressed that are potentially involved in weight regulation, but so far, the detailed function of most hypothalamic GPCRs is only partially understood. An important and underappreciated feature of GPCRs is the capacity for regulation via di- and heterodimerization. Increasing evidence implicates that heterodimerization of GPCRs results in profound functional consequences. Recently, we could demonstrate that interaction of the melanocortin 3 receptor (MC3R) and the growth hormone secretagogue receptor (GHSR)-1a results in a modulation of function in both receptors. Although the physiological role of GPCR-GPCR interaction in the hypothalamus is yet to be elucidated, this concept promises new avenues for investigation and understanding of hypothalamic functions dependent on GPCR signaling. Since GPCRs are important targets for drugs to combat many diseases, identification of heterodimers may be a prerequisite for highly specific drugs. Therefore, a detailed understanding of the mechanisms and their involvement in weight regulation is necessary. Fundamental to this understanding is the interplay of GPCR-GPCR in the hypothalamic nuclei in energy metabolism. In this review, we summarize the current knowledge on melanocortin receptors and GHSR-1a in hypothalamic weight regulation, especially as they pertain to possible drug targets. Furthermore, we include available evidence for the participation and significance of GPCR dimerization.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/metabolism , Receptors, Ghrelin/metabolism , Animals , Appetite Regulation/physiology , Arcuate Nucleus of Hypothalamus/anatomy & histology , Arcuate Nucleus of Hypothalamus/physiology , Humans , Models, Biological , Paraventricular Hypothalamic Nucleus/anatomy & histology , Paraventricular Hypothalamic Nucleus/physiology , Protein Multimerization/physiology , Receptor, Melanocortin, Type 3/physiology , Receptor, Melanocortin, Type 4/physiology , Receptors, Ghrelin/physiology
15.
Am J Pathol ; 179(1): 259-69, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703408

ABSTRACT

Synthetic and natural melanocortin (MC) peptides afford inhibitory properties in inflammation and tissue injury, but characterization of receptor involvement is still elusive. We used the agonist AP214 to test MC-dependent anti-inflammatory effects. In zymosan peritonitis, treatment of mice with AP214 (400 to 800 µg/kg) inhibited cell infiltration, an effect retained in MC receptor type 1, or MC(1), mutant mice but lost in MC(3) null mice. In vitro, cytokine release from zymosan-stimulated macrophages was affected by AP214, with approximately 80%, 30%, and 40% reduction in IL-1ß, tumor necrosis factor-α, and IL-6, respectively. Inhibition of IL-1ß release was retained in MC(1) mutant cells but was lost in MC(3) null cells. Furthermore, AP214 augmented uptake of zymosan particles and human apoptotic neutrophils by wild-type macrophages: this proresolving property was lost in MC(3) null macrophages. AP214 displayed its pro-efferocytotic effect also in vivo. Finally, in a model of inflammatory arthritis, AP214 evoked significant reductions in the clinical score. These results indicate that AP214 elicits anti-inflammatory responses, with a preferential effect on IL-1ß release. Furthermore, we describe for the first time a positive modulation of an MC agonist on the process of efferocytosis. In all cases, endogenous MC(3) is the receptor that mediates these novel properties of AP214. These findings might clarify the tissue-protective properties of AP214 in clinical settings and may open further development for novel MC agonists.


Subject(s)
Arthritis, Experimental/drug therapy , Inflammation/drug therapy , Melanocortins/agonists , Peritonitis/drug therapy , Receptor, Melanocortin, Type 1/physiology , Receptor, Melanocortin, Type 3/physiology , alpha-MSH/analogs & derivatives , Animals , Apoptosis/drug effects , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Blotting, Western , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/metabolism , Peritonitis/metabolism , Peritonitis/pathology , Phagocytosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , alpha-MSH/pharmacology
16.
Physiol Behav ; 104(4): 546-54, 2011 Sep 26.
Article in English | MEDLINE | ID: mdl-21497617

ABSTRACT

The central nervous melanocortin system is a neural network linking nutrient-sensing systems with hypothalamic, limbic and hindbrain neurons regulating behavior and metabolic homeostasis. Primary melanocortin neurons releasing melanocortin receptor ligands residing in the hypothalamic arcuate nucleus are regulated by nutrient-sensing and metabolic signals. A smaller group of primary neurons releasing melanocortin agonists in the nucleus tractus solitarius in the brainstem are also regulated by signals of metabolic state. Two melanocortin receptors regulate energy homeostasis. Melanocortin-4 receptors regulate satiety and autonomic outputs controlling peripheral metabolism. The functions of melanocortin-3 receptors (MC3R) expressed in hypothalamic and limbic structures are less clear. Here we discuss published data and preliminary observations from our laboratory suggesting that neural MC3R regulate inputs into systems governing the synchronization of rhythms in behavior and metabolism with nutrient intake. Mice subjected to a restricted feeding protocol, where a limited number of calories are presented at a 24h interval, rapidly exhibit bouts of increased wakefulness and activity which anticipate food presentation. The full expression of these responses is dependent on MC3R. Moreover, MC3R knockout mice are unique in exhibiting a dissociation of weight loss from improved glucose homeostasis when subject to a restricted feeding protocol. While mice lacking MC3R fed ad libitum exhibit normal to moderate hyperinsulinemia, when subjected to a restricted protocol they develop hyperglycemia, glucose intolerance, and dyslipidemia. Collectively, our data suggest that the central nervous melanocortin system is a point convergence in the control of energy balance and the expression of rhythms anticipating nutrient intake.


Subject(s)
Energy Metabolism/physiology , Homeostasis/physiology , Receptor, Melanocortin, Type 3/physiology , Animals , Anticipation, Psychological/physiology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Hypothalamus/physiology , Melanocortins/physiology , Neural Pathways/physiology , Satiety Response/physiology , Signal Transduction/physiology
17.
Curr Opin Investig Drugs ; 11(10): 1092-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20882712

ABSTRACT

The melanocortin-3 receptor (MC3R), together with the related melanocortin-4 receptor (MC4R), are important regulators of energy homeostasis. Rodent studies have demonstrated that the two receptors have non-redundant roles in regulating energy balance. However, while mutations for the MC4R have been established as a cause of monogenic obesity, mutations in the MC3R gene remain controversially associated with human obesity pathogenesis. This editorial summarizes the current status of MC3R in rodent energy homeostasis and human obesity pathogenesis.


Subject(s)
Receptor, Melanocortin, Type 3/genetics , Adiposity/drug effects , Adiposity/genetics , Adiposity/physiology , Animals , Energy Metabolism/genetics , Energy Metabolism/physiology , Homeostasis/genetics , Homeostasis/physiology , Humans , Mice , Mice, Knockout , Mutation , Obesity/genetics , Obesity/physiopathology , Polymorphism, Genetic , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 3/physiology
18.
FASEB J ; 24(3): 862-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19837866

ABSTRACT

Melanocortin-3 receptors (Mc3rs) in the central nervous system are involved in expression of anticipatory rhythms and synchronizing clocks maintaining circadian rhythms during restricted feeding (RF) [mice housed under a 12-h light-dark cycle with lights on between zeitgeber time (ZT) 0 to ZT12 fed 60% of normal calories between ZT7 and ZT11]. Because the systems governing circadian rhythms are important for adaptation to RF, we investigated whether Mc3rs are required for metabolic adaption to RF. Mc3r(-/-) mice subjected to RF exhibited normal weight loss; however, they developed hyperinsulinemia, glucose intolerance, increased expression of lipogenic genes, and increased ketogenesis relative to controls. Rhythmic expression of transcription factors regulating liver clock activity and energy metabolism (Bmal1, Rev-erbalpha, Pgc1, Foxo1, Hnf4alpha, and Pck1) was severely compromised in Mc3r(-/-) mice during RF. Inhibition of neural melanocortin receptors by agouti-related peptide also attenuated rhythmicity in the hepatic expression of these genes during RF. Collectively, these data suggest that neural Mc3rs are important for adapting metabolism and maintaining rhythms of liver metabolism during periods when feeding is restricted to the light cycle.-Sutton, G. M., Begriche, K., Kumar, K. G., Gimble, J. M., Perez-Tilve, D., Nogueiras, R., McMillan, R. P., Hulver, M. W., Tschöp, M. H., Butler, A. A. Central nervous system melanocortin-3 receptors are required for synchronizing metabolism during entrainment to restricted feeding during the light cycle.


Subject(s)
Caloric Restriction , Central Nervous System/metabolism , Photoperiod , Receptor, Melanocortin, Type 3/physiology , Agouti-Related Protein/pharmacology , Animals , Central Nervous System/radiation effects , Electrophoresis, Polyacrylamide Gel , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Acids/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Glucose Intolerance/genetics , Glucose Tolerance Test , Hyperinsulinism/genetics , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Mutant Strains , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/genetics
19.
Obes Rev ; 10 Suppl 2: 14-24, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19849798

ABSTRACT

Obesity, insulin resistance and increased propensity for type 2 diabetes and cardiovascular disease result from an imbalance between energy intake and expenditure. The cloning of genes involved in energy homeostasis produced a simple feedback model for the homeostatic regulation of adipose mass. Serum leptin secreted from adipocytes signals nutrient sufficiency, curbing appetite and supporting energy expenditure. A rapid decline in leptin during nutrient scarcity instigates adaptive mechanisms, including increased appetite and reduced energy expenditure. Hypothalamic melanocortin neurons are important mediators of this response, integrating inputs of energy status from leptin with other peripheral signals. While this feedback response prolongs survival during fasting, other mechanisms allowing the prediction of nutrient availability also confer a selective advantage. This adaptation has been commonly studied in rodents using restricted feeding paradigms constraining food intake to limited periods at 24-h intervals. Restricted feeding rapidly elicits rhythmic bouts of activity and wakefulness anticipating food presentation. While the response exhibits features suggesting a clock-like mechanism, the neuromolecular mechanisms governing expression of food anticipatory behaviours are poorly understood. Here we discuss a model whereby melanocortin neurons regulating the homeostatic adaptation to variable caloric availability also regulate inputs into neural networks governing anticipatory rhythms in wakefulness, activity and metabolism.


Subject(s)
Appetite Regulation/physiology , Biological Clocks/physiology , Circadian Rhythm/physiology , Energy Metabolism/physiology , Melanocortins/physiology , Adipose Tissue/metabolism , Energy Intake/physiology , Homeostasis/physiology , Humans , Leptin/metabolism , Receptor, Melanocortin, Type 3/physiology , Signal Transduction
20.
Curr Top Med Chem ; 9(6): 554-63, 2009.
Article in English | MEDLINE | ID: mdl-19689365

ABSTRACT

Current biological and pharmacological evidence suggests that the melanocortin 4 and melanocortin 3 receptors which are seven transmembrane G-protein coupled receptors (GPCRs) are involved in various aspects of energy balance and feeding behaviors in animals including humans. The natural endogenous ligands for these receptors are products of the gene pro-opiomelanocortin (POMC), and include alpha-melanocyte stimulating hormone, gamma-melanocyte stimulating hormone and perhaps other modified products of POMC. Thus well designed agonists and antagonists of these ligands might serve as drugs for the treatment of feeding disorders. However, these melanotropin peptides also can have other biological activities that involve the MC3R and MC4R, and these other biological properties will need to be modulated in ligands that are likely to be useful drugs for feeding disorders. Current progress in these areas with special emphasis on the MC3R will be discussed along with possible new directions that might be fruitful in these important aspects of contemporary biology and medicine.


Subject(s)
Anti-Obesity Agents/chemistry , Anti-Obesity Agents/therapeutic use , Melanocyte-Stimulating Hormones/chemistry , Melanocyte-Stimulating Hormones/therapeutic use , Nutrition Disorders/drug therapy , Animals , Anti-Obesity Agents/pharmacology , Drug Evaluation, Preclinical , Feeding Behavior/drug effects , Feeding Behavior/physiology , Humans , Melanocyte-Stimulating Hormones/pharmacology , Nutrition Disorders/metabolism , Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/chemistry , Receptor, Melanocortin, Type 3/physiology , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/physiology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...