Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33795520

RESUMO

Mutations in the melanocortin 4 receptor (MC4R) result in hyperphagia and obesity and are the most common cause of monogenic obesity in humans. Preclinical rodent studies have determined that the critical role of the MC4R in controlling feeding can be mapped in part to its expression in the paraventricular nucleus of the hypothalamus (paraventricular nucleus [PVN]), where it regulates the activity of anorexic neural circuits. Despite the critical role of PVN MC4R neurons in regulating feeding, the in vivo neuronal activity of these cells remains largely unstudied, and the network activity of PVN MC4R neurons has not been determined. Here, we utilize in vivo single-cell endomicroscopic and mathematical approaches to determine the activity and network dynamics of PVN MC4R neurons in response to changes in energy state and pharmacological manipulation of central melanocortin receptors. We determine that PVN MC4R neurons exhibit both quantitative and qualitative changes in response to fasting and refeeding. Pharmacological stimulation of MC4R with the therapeutic MC4R agonist setmelanotide rapidly increases basal PVN MC4R activity, while stimulation of melanocortin 3 receptor (MC3R) inhibits PVN MC4R activity. Finally, we find that distinct PVN MC4R neuronal ensembles encode energy deficit and energy surfeit and that energy surfeit is associated with enhanced network connections within PVN MC4R neurons. These findings provide valuable insight into the neural dynamics underlying hunger and energy surfeit.


Assuntos
Comportamento Alimentar/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Masculino , Camundongos , Microscopia de Fluorescência , Rede Nervosa , Imagem Óptica , Núcleo Hipotalâmico Paraventricular/citologia , Receptor Tipo 3 de Melanocortina/agonistas , Análise de Célula Única
2.
Am J Physiol Cell Physiol ; 324(3): C694-C706, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36717105

RESUMO

The inward rectifier potassium channel Kir7.1, encoded by the KCNJ13 gene, is a tetramer composed of two-transmembrane domain-spanning monomers, closer in homology to Kir channels associated with potassium transport such as Kir1.1, 1.2, and 1.3. Compared with other channels, Kir7.1 exhibits small unitary conductance and low dependence on external potassium. Kir7.1 channels also show a phosphatidylinositol 4,5-bisphosphate (PIP2) dependence for opening. Accordingly, retinopathy-associated Kir7.1 mutations mapped at the binding site for PIP2 resulted in channel gating defects leading to channelopathies such as snowflake vitreoretinal degeneration and Leber congenital amaurosis in blind patients. Lately, this channel's role in energy homeostasis was reported due to the direct interaction with the melanocortin type 4 receptor (MC4R) in the hypothalamus. As this channel seems to play a multipronged role in potassium homeostasis and neuronal excitability, we will discuss what is predicted from a structural viewpoint and its possible implications for hunger control.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização , Humanos , Mutação , Neurônios/metabolismo , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Domínios Proteicos
3.
Gen Comp Endocrinol ; 336: 114243, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36801393

RESUMO

Loss of agouti related neuropeptide (AgRP) does not lead to overt phenotypes in mammals unless AgRP neurons are ablated. In contrast, in zebrafish it has been shown that Agrp1 loss of function (LOF) leads to reduced growth in Agrp1 morphant as well as Agrp1 mutant larvae. Further, it has been shown that multiple endocrine axes are dysregulated upon Agrp1 LOF in Agrp1 morphant larvae. Here we show that adult Agrp1 LOF zebrafish show normal growth and reproductive behavior in spite of a significant reduction in multiple related endocrine axes namely reduced expression in pituitary growth hormone (gh) follicle stimulating hormone (fshb) as well as luteinizing hormone (lhb). We looked for compensatory changes in candidate gene expression but found no changes in growth hormone and gonadotropin hormone receptors that would explain the lack of phenotype. We further looked at expression in the hepatic and muscular insulin-like growth factor (Igf) axis which appears to be normal. Fecundity as well as ovarian histology also appear largely normal while we do see an increase in mating efficiency specifically in fed but not fasted AgRP1 LOF animals. This data shows that zebrafish can grow and reproduce normally in spite of significant central hormone changes and suggests a peripheral compensatory mechanism additional to previously reported central compensatory mechanisms in other zebrafish neuropeptide LOF lines.


Assuntos
Hormônio Foliculoestimulante , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Hormônio Foliculoestimulante/genética , Hormônio Luteinizante , Gonadotropinas , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Mamíferos/metabolismo
4.
J Biol Chem ; 295(48): 16370-16379, 2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-32943551

RESUMO

The melanocortin receptor accessory protein 2 (MRAP2) plays a pivotal role in the regulation of several G protein-coupled receptors that are essential for energy balance and food intake. MRAP2 loss-of-function results in obesity in mammals. MRAP2 and its homolog MRAP1 have an unusual membrane topology and are the only known eukaryotic proteins that thread into the membrane in both orientations. In this study, we demonstrate that the conserved polybasic motif that dictates the membrane topology and dimerization of MRAP1 does not control the membrane orientation and dimerization of MRAP2. We also show that MRAP2 dimerizes through its transmembrane domain and can form higher-order oligomers that arrange MRAP2 monomers in a parallel orientation. Investigating the molecular details of MRAP2 structure is essential for understanding the mechanism by which it regulates G protein-coupled receptors and will aid in elucidating the pathways involved in metabolic dysfunction.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Membrana Celular/metabolismo , Multimerização Proteica , Proteínas Adaptadoras de Transdução de Sinal/genética , Membrana Celular/genética , Células HEK293 , Humanos , Domínios Proteicos
5.
Nature ; 520(7545): 94-8, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25600267

RESUMO

The regulated release of anorexigenic α-melanocyte stimulating hormone (α-MSH) and orexigenic Agouti-related protein (AgRP) from discrete hypothalamic arcuate neurons onto common target sites in the central nervous system has a fundamental role in the regulation of energy homeostasis. Both peptides bind with high affinity to the melanocortin-4 receptor (MC4R); existing data show that α-MSH is an agonist that couples the receptor to the Gαs signalling pathway, while AgRP binds competitively to block α-MSH binding and blocks the constitutive activity mediated by the ligand-mimetic amino-terminal domain of the receptor. Here we show that, in mice, regulation of firing activity of neurons from the paraventricular nucleus of the hypothalamus (PVN) by α-MSH and AgRP can be mediated independently of Gαs signalling by ligand-induced coupling of MC4R to closure of inwardly rectifying potassium channel, Kir7.1. Furthermore, AgRP is a biased agonist that hyperpolarizes neurons by binding to MC4R and opening Kir7.1, independently of its inhibition of α-MSH binding. Consequently, Kir7.1 signalling appears to be central to melanocortin-mediated regulation of energy homeostasis within the PVN. Coupling of MC4R to Kir7.1 may explain unusual aspects of the control of energy homeostasis by melanocortin signalling, including the gene dosage effect of MC4R and the sustained effects of AgRP on food intake.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Potenciais de Ação , Proteína Relacionada com Agouti/metabolismo , Animais , Ingestão de Alimentos/genética , Metabolismo Energético , Feminino , Células HEK293 , Homeostase/genética , Humanos , Ligantes , Masculino , Melanocortinas/metabolismo , Camundongos , Receptor Tipo 4 de Melanocortina/genética , Transdução de Sinais/genética , alfa-MSH/metabolismo
6.
Addict Biol ; 26(1): e12880, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32064741

RESUMO

Eating disorders and substance use disorders frequently co-occur. Twin studies reveal shared genetic variance between liabilities to eating disorders and substance use, with the strongest associations between symptoms of bulimia nervosa and problem alcohol use (genetic correlation [rg ], twin-based = 0.23-0.53). We estimated the genetic correlation between eating disorder and substance use and disorder phenotypes using data from genome-wide association studies (GWAS). Four eating disorder phenotypes (anorexia nervosa [AN], AN with binge eating, AN without binge eating, and a bulimia nervosa factor score), and eight substance-use-related phenotypes (drinks per week, alcohol use disorder [AUD], smoking initiation, current smoking, cigarettes per day, nicotine dependence, cannabis initiation, and cannabis use disorder) from eight studies were included. Significant genetic correlations were adjusted for variants associated with major depressive disorder and schizophrenia. Total study sample sizes per phenotype ranged from ~2400 to ~537 000 individuals. We used linkage disequilibrium score regression to calculate single nucleotide polymorphism-based genetic correlations between eating disorder- and substance-use-related phenotypes. Significant positive genetic associations emerged between AUD and AN (rg = 0.18; false discovery rate q = 0.0006), cannabis initiation and AN (rg = 0.23; q < 0.0001), and cannabis initiation and AN with binge eating (rg = 0.27; q = 0.0016). Conversely, significant negative genetic correlations were observed between three nondiagnostic smoking phenotypes (smoking initiation, current smoking, and cigarettes per day) and AN without binge eating (rgs = -0.19 to -0.23; qs < 0.04). The genetic correlation between AUD and AN was no longer significant after co-varying for major depressive disorder loci. The patterns of association between eating disorder- and substance-use-related phenotypes highlights the potentially complex and substance-specific relationships among these behaviors.


Assuntos
Transtornos da Alimentação e da Ingestão de Alimentos/genética , Transtornos Relacionados ao Uso de Substâncias/genética , Alcoolismo/genética , Transtorno Depressivo Maior/genética , Estudo de Associação Genômica Ampla , Humanos , Desequilíbrio de Ligação , Fenótipo , Polimorfismo de Nucleotídeo Único , Fatores de Risco , Esquizofrenia/genética , Tabagismo/genética
7.
Proc Natl Acad Sci U S A ; 115(2): 427-432, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29279372

RESUMO

Subjects spending much time sitting have increased risk of obesity but the mechanism for the antiobesity effect of standing is unknown. We hypothesized that there is a homeostatic regulation of body weight. We demonstrate that increased loading of rodents, achieved using capsules with different weights implanted in the abdomen or s.c. on the back, reversibly decreases the biological body weight via reduced food intake. Importantly, loading relieves diet-induced obesity and improves glucose tolerance. The identified homeostat for body weight regulates body fat mass independently of fat-derived leptin, revealing two independent negative feedback systems for fat mass regulation. It is known that osteocytes can sense changes in bone strain. In this study, the body weight-reducing effect of increased loading was lost in mice depleted of osteocytes. We propose that increased body weight activates a sensor dependent on osteocytes of the weight-bearing bones. This induces an afferent signal, which reduces body weight. These findings demonstrate a leptin-independent body weight homeostat ("gravitostat") that regulates fat mass.


Assuntos
Tecido Adiposo/metabolismo , Peso Corporal/fisiologia , Homeostase/efeitos dos fármacos , Leptina/farmacologia , Obesidade/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Ingestão de Energia/efeitos dos fármacos , Ingestão de Energia/fisiologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Homeostase/fisiologia , Leptina/administração & dosagem , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/etiologia , Obesidade/genética , Osteócitos/metabolismo , Ratos Sprague-Dawley , Redução de Peso/efeitos dos fármacos , Redução de Peso/fisiologia
8.
J Biol Chem ; 293(46): 17739-17753, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30257863

RESUMO

Kir7.1 is an inwardly rectifying potassium channel with important roles in the regulation of the membrane potential in retinal pigment epithelium, uterine smooth muscle, and hypothalamic neurons. Regulation of G protein-coupled inwardly rectifying potassium (GIRK) channels by G protein-coupled receptors (GPCRs) via the G protein ßγ subunits has been well characterized. However, how Kir channels are regulated is incompletely understood. We report here that Kir7.1 is also regulated by GPCRs, but through a different mechanism. Using Western blotting analysis, we observed that multiple GPCRs tested caused a striking reduction in the complex glycosylation of Kir7.1. Further, GPCR-mediated reduction of Kir7.1 glycosylation in HEK293T cells did not alter its expression at the cell surface but decreased channel activity. Of note, mutagenesis of the sole Kir7.1 glycosylation site reduced conductance and open probability, as indicated by single-channel recording. Additionally, we report that the L241P mutation of Kir7.1 associated with Lebers congenital amaurosis (LCA), an inherited retinal degenerative disease, has significantly reduced complex glycosylation. Collectively, these results suggest that Kir7.1 channel glycosylation is essential for function, and this activity within cells is suppressed by most GPCRs. The melanocortin-4 receptor (MC4R), a GPCR previously reported to induce ligand-regulated activity of this channel, is the only GPCR tested that does not have this effect on Kir7.1.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Glicosilação , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Amaurose Congênita de Leber/genética , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/genética , Multimerização Proteica/fisiologia , Transporte Proteico/fisiologia , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Deleção de Sequência
9.
Proc Natl Acad Sci U S A ; 113(11): 3084-9, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26903647

RESUMO

Leptin is the primary adipostatic factor in mammals. Produced largely by adipocytes in proportion to total adipose mass, the hormone informs the brain regarding total energy stored as triglycerides in fat cells. The hormone acts on multiple circuits in the brain to regulate food intake, autonomic outflow, and endocrine function to maintain energy balance. In addition to regulating adipose mass, mammalian leptin also plays a role in the regulation of glucose homeostasis and as a gating factor in reproductive competence. Leptin-deficient mice and people exhibit early onset profound hyperphagia and obesity, diabetes, and infertility. Although leptin and the leptin receptor are found in fish, the hormone is not expressed in adipose tissue, but is found in liver and other tissues. Here, we show that adult zebrafish lacking a functional leptin receptor do not exhibit hyperphagia or increased adiposity, and exhibit normal fertility. However, leptin receptor-deficient larvae have increased numbers of ß-cells and increased levels of insulin mRNA. Furthermore, larval zebrafish have been shown to exhibit ß-cell hyperplasia in response to high fat feeding or peripheral insulin resistance, and we show here that leptin receptor is required for this response. Adult zebrafish also have increased levels of insulin mRNA and other alterations in glucose homeostasis. Thus, a role for leptin in the regulation of ß-cell mass and glucose homeostasis appears to be conserved across vertebrates, whereas its role as an adipostatic factor is likely to be a secondary role acquired during the evolution of mammals.


Assuntos
Adiposidade/fisiologia , Glucose/metabolismo , Células Secretoras de Insulina/fisiologia , Leptina/fisiologia , Receptores para Leptina/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Sequência de Aminoácidos , Animais , Tamanho Corporal , Peso Corporal , Contagem de Células , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Gorduras na Dieta , Fertilidade , Teste de Tolerância a Glucose , Glicogenólise , Glicólise , Homeostase , Hiperfagia/genética , Hiperfagia/fisiopatologia , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Larva , Leptina/genética , Fígado/metabolismo , Masculino , Dados de Sequência Molecular , Fenótipo , Fosfoenolpiruvato Carboxiquinase (ATP)/biossíntese , Fosfoenolpiruvato Carboxiquinase (ATP)/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores para Leptina/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais/fisiologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética
10.
Horm Behav ; 82: 87-100, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27156808

RESUMO

Melanocortin signaling is regulated by the binding of naturally occurring antagonists, agouti-signaling protein (ASIP) and agouti-related protein (AGRP) that compete with melanocortin peptides by binding to melanocortin receptors to regulate energy balance and growth. Using a transgenic model overexpressing ASIP, we studied the involvement of melanocortin system in the feeding behaviour, growth and stress response of zebrafish. Our data demonstrate that ASIP overexpression results in enhanced growth but not obesity. The differential growth is explained by increased food intake and feeding efficiency mediated by a differential sensitivity of the satiety system that seems to involve the cocaine- and amphetamine- related transcript (CART). Stress response was similar in both genotypes. Brain transcriptome of transgenic (ASIP) vs wild type (WT) fish was compared using microarrays. WT females and males exhibited 255 genes differentially expressed (DEG) but this difference was reduced to 31 after ASIP overexpression. Statistical analysis revealed 1122 DEG when considering only fish genotype but 1066 and 981 DEG when comparing ASIP males or females with their WT counterparts, respectively. Interaction between genotype and sex significantly affected the expression of 97 genes. Several neuronal systems involved in the control of food intake were identified which displayed a differential expression according to the genotype of the fish that unravelling the flow of melanocortinergic information through the central pathways that controls the energy balance. The information provided herein will help to elucidate new central systems involved in control of obesity and should be of invaluable use for sustaining fish production systems.


Assuntos
Proteína Agouti Sinalizadora/genética , Encéfalo/metabolismo , Peixe-Zebra/genética , Proteína Agouti Sinalizadora/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Animais Geneticamente Modificados , Ingestão de Alimentos/fisiologia , Metabolismo Energético/genética , Comportamento Alimentar/fisiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Masculino , Melanocortinas/antagonistas & inibidores , Vias Neurais/metabolismo , Peixe-Zebra/metabolismo
12.
Proc Natl Acad Sci U S A ; 110(17): 7050-5, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23569235

RESUMO

Haploinsufficiency of the melanocortin-4 receptor (MC4R) results in melanocortin obesity syndrome, the most common monogenic cause of severe early onset obesity in humans. The syndrome, which produces measurable hyperphagia, has focused attention on the role of MC4R in feeding behavior and macronutrient intake. Studies show that inhibition of MC4R signaling can acutely increase the consumption of high-fat foods. The current study examines the chronic feeding preferences of mice with deletion of one or both alleles of the MC4R to model the human syndrome. Using two-choice diet paradigms with high-fat or high-carbohydrate foods alongside normal chow, we show, paradoxically, that deletion of one allele has no effect, whereas deletion of both alleles of the MC4R actually decreases preference for palatable high-fat and high-sucrose foods, compared with wild-type mice. Nonetheless, we observed hyperphagic behavior from increased consumption of the low-fat standard chow when either heterozygous or homozygous mutant animals were presented with dietary variety. Thus, decreased MC4R signaling in melanocortin obesity syndrome consistently yields hyperphagia irrespective of the foods provided, but the hyperphagia appears driven by variety and/or novelty, rather than by a preference for high-fat or high-carbohydrate foodstuffs.


Assuntos
Ingestão de Alimentos/genética , Preferências Alimentares/fisiologia , Hiperfagia/genética , Obesidade/etiologia , Receptor Tipo 4 de Melanocortina/genética , Animais , Peso Corporal , Dieta Hiperlipídica , Carboidratos da Dieta , Ingestão de Alimentos/fisiologia , Deleção de Genes , Hiperfagia/complicações , Masculino , Camundongos , Camundongos Knockout , Receptor Tipo 4 de Melanocortina/fisiologia
13.
Proc Natl Acad Sci U S A ; 109(23): E1489-98, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22573815

RESUMO

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.


Assuntos
Metabolismo Energético/fisiologia , Jejum/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Receptor Tipo 3 de Melanocortina/fisiologia , Absorciometria de Fóton , Tecido Adiposo Branco/metabolismo , Adiposidade/genética , Glândulas Suprarrenais/citologia , Análise de Variância , Animais , Fenômenos Biomecânicos , Western Blotting , Composição Corporal/fisiologia , Corticosterona/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Lipólise/fisiologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 3 de Melanocortina/deficiência , Triglicerídeos/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(1): 355-60, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21169216

RESUMO

Melanocortin-4 receptor (MC4R) is critical for energy homeostasis, and the paraventricular nucleus of the hypothalamus (PVN) is a key site of MC4R action. Most studies suggest that leptin regulates PVN neurons indirectly, by binding to receptors in the arcuate nucleus or ventromedial hypothalamus and regulating release of products like α-melanocyte-stimulating hormone (α-MSH), neuropeptide Y (NPY), glutamate, and GABA from first-order neurons onto the MC4R PVN cells. Here, we investigate mechanisms underlying regulation of activity of these neurons under various metabolic states by using hypothalamic slices from a transgenic MC4R-GFP mouse to record directly from MC4R neurons. First, we show that in vivo leptin levels regulate the tonic firing rate of second-order MC4R PVN neurons, with fasting increasing firing frequency in a leptin-dependent manner. We also show that, although leptin inhibits these neurons directly at the postsynaptic membrane, α-MSH and NPY potently stimulate and inhibit the cells, respectively. Thus, in contrast with the conventional model of leptin action, the primary control of MC4R PVN neurons is unlikely to be mediated by leptin action on arcuate NPY/agouti-related protein and proopiomelanocortin neurons. We also show that the activity of MC4R PVN neurons is controlled by the constitutive activity of the MC4R and that expression of the receptor mRNA and α-MSH sensitivity are both stimulated by leptin. Thus, leptin acts multinodally on arcuate nucleus/PVN circuits to regulate energy homeostasis, with prominent mechanisms involving direct control of both membrane conductances and gene expression in the MC4R PVN neuron.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Leptina/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Receptor Tipo 4 de Melanocortina/metabolismo , Transdução de Sinais/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Eletrofisiologia , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Melanocortinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Neurológicos , Neurônios/metabolismo , Neurônios/fisiologia , Neuropeptídeo Y/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Reação em Cadeia da Polimerase , alfa-MSH/metabolismo
15.
Methods Mol Biol ; 2796: 229-248, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38856905

RESUMO

Automated patch clamp recording is a valuable technique in drug discovery and the study of ion channels. It allows for the precise measurement and manipulation of channel currents, providing insights into their function and modulation by drugs or other compounds. The melanocortin 4 receptor (MC4-R) is a G protein-coupled receptor (GPCR) crucial to appetite regulation, energy balance, and body weight. MC4-R signaling is complex and involves interactions with other receptors and neuropeptides in the appetite-regulating circuitry. MC4-Rs, like other GPCRs, are known to modulate ion channels such as Kir7.1, an inward rectifier potassium channel, in response to ligand binding. This modulation is critical for controlling ion flow across the cell membrane, which can influence membrane potential, excitability, and neurotransmission. The MC4-R is the target for the anti-obesity drug Imcivree. However, this drug is known to lack optimal potency and also has side effects. Using high-throughput techniques for studying the MC4-R/Kir7.1 complex allows researchers to rapidly screen many compounds or conditions, aiding the development of drugs that target this system. Additionally, automated patch clamp recording of this receptor-channel complex and its ligands can provide valuable functional and pharmacological insights supporting the development of novel therapeutic strategies. This approach can be generalized to other GPCR-gated ion channel functional complexes, potentially accelerating the pace of research in different fields with the promise to uncover previously unknown aspects of receptor-ion channel interactions.


Assuntos
Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização , Receptor Tipo 4 de Melanocortina , Técnicas de Patch-Clamp/métodos , Animais , Humanos , Receptor Tipo 4 de Melanocortina/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Células HEK293
16.
bioRxiv ; 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38895219

RESUMO

Hereditary defects in the function of the Kir7.1 in the retinal pigment epithelium are associated with the ocular diseases retinitis pigmentosa, Leber congenital amaurosis, and snowflake vitreal degeneration. Studies also suggest that Kir7.1 may be regulated by a GPCR, the melanocortin-4 receptor, in certain hypothalamic neurons. We present the first structures of human Kir7.1 and describe the conformational bias displayed by two pathogenic mutations, R162Q and E276A, to provide an explanation for the basis of disease and illuminate the gating pathway. We also demonstrate the structural basis for the blockade of the channel by a small molecule ML418 and demonstrate that channel blockade in vivo activates MC4R neurons in the paraventricular nucleus of the hypothalamus (PVH), inhibiting food intake and inducing weight loss. Preliminary purification, and structural and pharmacological characterization of an in tandem construct of MC4R and Kir7.1 suggests that the fusion protein forms a homotetrameric channel that retains regulation by liganded MC4R molecules.

17.
bioRxiv ; 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38895206

RESUMO

Most antipsychotic drugs (APDs) induce hyperphagia and weight gain. However, the neural mechanisms are poorly understood, partly due to challenges replicating their metabolic effects in rodents. Here, we report a new mouse model that recapitulates overeating induced by clozapine, a widely prescribed APD. Our study shows that clozapine boosts food intake by inhibiting melanocortin 4 receptor (MC4R) expressing neurons in the paraventricular nucleus of the hypothalamus. Interestingly, neither clozapine nor risperidone, another commonly used APD, affects receptor-ligand binding or the canonical Gαs signaling of MC4R. Instead, they inhibit neuronal activity by enhancing the coupling between MC4R and Kir7.1, leading to the open state of the inwardly rectifying potassium channel. Deletion of Kir7.1 in Mc4r-Cre neurons prevents clozapine-induced weight gain, while treatment with a selective Kir7.1 blocker mitigates overeating in clozapine-fed mice. Our findings unveil a molecular pathway underlying the effect of APDs on feeding behavior and suggest its potential as a therapeutic target.

18.
J Clin Invest ; 134(14)2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39007271

RESUMO

The melanocortin-3 receptor (MC3R) regulates GABA release from agouti-related protein (AgRP) nerve terminals and thus tonically suppresses multiple circuits involved in feeding behavior and energy homeostasis. Here, we examined the role of the MC3R and the melanocortin system in regulating the response to various anorexigenic agents. The genetic deletion or pharmacological inhibition of the MC3R, or subthreshold doses of an MC4R agonist, improved the dose responsiveness to glucagon-like peptide 1 (GLP1) agonists, as assayed by inhibition of food intake and weight loss. An enhanced anorectic response to the acute satiety factors peptide YY (PYY3-36) and cholecystokinin (CCK) and the long-term adipostatic factor leptin demonstrated that increased sensitivity to anorectic agents was a generalized result of MC3R antagonism. We observed enhanced neuronal activation in multiple hypothalamic nuclei using Fos IHC following low-dose liraglutide in MC3R-KO mice (Mc3r-/-), supporting the hypothesis that the MC3R is a negative regulator of circuits that control multiple aspects of feeding behavior. The enhanced anorectic response in Mc3r-/- mice after administration of GLP1 analogs was also independent of the incretin effects and malaise induced by GLP1 receptor (GLP1R) analogs, suggesting that MC3R antagonists or MC4R agonists may have value in enhancing the dose-response range of obesity therapeutics.


Assuntos
Liraglutida , Camundongos Knockout , Receptor Tipo 3 de Melanocortina , Receptor Tipo 4 de Melanocortina , Animais , Camundongos , Receptor Tipo 4 de Melanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/agonistas , Liraglutida/farmacologia , Receptor Tipo 3 de Melanocortina/genética , Receptor Tipo 3 de Melanocortina/metabolismo , Receptor Tipo 3 de Melanocortina/agonistas , Masculino , Depressores do Apetite/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Colecistocinina/metabolismo , Camundongos Endogâmicos C57BL , Ingestão de Alimentos/efeitos dos fármacos , Leptina/metabolismo , Peptídeo YY/metabolismo , Peptídeo YY/genética , Hipotálamo/metabolismo
19.
J Med Chem ; 67(4): 2690-2711, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38345933

RESUMO

Melanocortin 4 receptor (MC4-R) antagonists are actively sought for treating cancer cachexia. We determined the structures of complexes with PG-934 and SBL-MC-31. These peptides differ from SHU9119 by substituting His6 with Pro6 and inserting Gly10 or Arg10. The structures revealed two subpockets at the TM7-TM1-TM2 domains, separated by N2857.36. Two peptide series based on the complexed peptides led to an antagonist activity and selectivity SAR study. Most ligands retained the SHU9119 potency, but several SBL-MC-31-derived peptides significantly enhanced MC4-R selectivity over MC1-R by 60- to 132-fold. We also investigated MC4-R coupling to the K+ channel, Kir7.1. Some peptides activated the channel, whereas others induced channel closure independently of G protein coupling. In cell culture studies, channel activation correlated with increased feeding, while a peptide with Kir7.1 inhibitory activity reduced eating. These results highlight the potential for targeting the MC4-R:Kir7.1 complex for treating positive and restrictive eating disorders.


Assuntos
Peptídeos , Receptor Tipo 4 de Melanocortina , Humanos , Peptídeos/farmacologia , Ligantes , Desenho de Fármacos , Receptor Tipo 3 de Melanocortina , Receptores de Melanocortina
20.
Biochem Biophys Res Commun ; 439(1): 115-20, 2013 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-23954636

RESUMO

Mammalian ghrelin is a stomach-derived peptide that stimulates secretion of growth hormone and food intake. Zebrafish is an excellent model system for forward genetic studies, and many aspects of energy homeostasis characterized in mammals appear to be conserved in the zebrafish. In this study, we investigated the expression and regulation of zebrafish ghrelin by metabolic status. Quantitative RT-PCR revealed that zebrafish ghrelin is highly enriched in anterior gut associated tissues. Using in situ hybridization with adult zebrafish tissues, we found that zebrafish ghrelin mRNA was not expressed in intestine tissue, but rather in clusters of endocrine pancreas cells distinct from insulin-expressing islets. Fasting specifically upregulated pancreatic ghrelin but not brain ghrelin expression by 3- to 4-fold and refeeding restored ghrelin transcript to control levels seen in the fed group within 5 h. These results demonstrate that although ghrelin is expressed in a different site in zebrafish, it is responsive to metabolic state in a similar manner as mammalian ghrelin, suggesting a role in the regulation of feeding in teleosts, and thus validate the utility of zebrafish as a genetic model system for the analysis of the ghrelin system and energy homeostasis.


Assuntos
Encéfalo/metabolismo , Células Endócrinas/metabolismo , Regulação da Expressão Gênica , Grelina/metabolismo , Pâncreas/metabolismo , Peixe-Zebra/metabolismo , Animais , Jejum , Feminino , Masculino , RNA Mensageiro/metabolismo , Distribuição Tecidual , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA