Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 224
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Med ; 30(1): 34, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38448811

RESUMEN

BACKGROUND: Imbalance in energy regulation is a major cause of insulin resistance and diabetes. Melanocortin-4 receptor (MC4R) signaling at specific sites in the central nervous system has synergistic but non-overlapping functions. However, the mechanism by which MC4R in the arcuate nucleus (ARC) region regulates energy balance and insulin resistance remains unclear. METHODS: The MC4Rflox/flox mice with proopiomelanocortin (POMC) -Cre mice were crossed to generate the POMC-MC4Rflox/+ mice. Then POMC-MC4Rflox/+ mice were further mated with MC4Rflox/flox mice to generate the POMC-MC4Rflox/flox mice in which MC4R is selectively deleted in POMC neurons. Bilateral injections of 200 nl of AAV-sh-Kir2.1 (AAV-sh-NC was used as control) were made into the ARC of the hypothalamus. Oxygen consumption, carbon dioxide production, respiratory exchange ratio and energy expenditure were measured by using the CLAMS; Total, visceral and subcutaneous fat was analyzed using micro-CT. Co-immunoprecipitation assays (Co-IP) were used to analyze the interaction between MC4R and Kir2.1 in GT1-7 cells. RESULTS: POMC neuron-specific ablation of MC4R in the ARC region promoted food intake, impaired energy expenditure, leading to increased weight gain and impaired systemic glucose homeostasis. Additionally, MC4R ablation reduced the activation of POMC neuron, and is not tissue-specific for peripheral regulation, suggesting the importance of its central regulation. Mechanistically, sequencing analysis and Co-IP assay demonstrated a direct interaction of MC4R with Kir2.1. Knockdown of Kir2.1 in POMC neuron-specific ablation of MC4R restored the effect of MC4R ablation on energy expenditure and systemic glucose homeostasis, indicating by reduced body weight and ameliorated insulin resistance. CONCLUSION: Hypothalamic POMC neuron-specific knockout of MC4R affects energy balance and insulin sensitivity by regulating Kir2.1. Kir2.1 represents a new target and pathway that could be targeted in obesity.


Asunto(s)
Resistencia a la Insulina , Animales , Ratones , Glucosa , Hipotálamo , Resistencia a la Insulina/genética , Neuronas , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/genética
2.
Nutrients ; 16(4)2024 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-38398881

RESUMEN

This study aimed to determine the impact of a fiber supplement on body weight and composition in individuals with obesity with specific genetic polymorphisms. It involved 112 adults with obesity, each with at least one minor allele in the FTO, LEP, LEPR, or MC4R polymorphism. Participants were randomized to receive either a fiber supplement (glucomannan, inulin, and psyllium) or a placebo for 180 days. The experimental group showed significant reductions in body weight (treatment difference: -4.9%; 95% CI: -6.9% to -2.9%; p < 0.01) and BMI (treatment difference: -1.4 kg/m2; 95% CI: -1.7 to -1.2; p < 0.01) compared to placebo. Further significant decreases in fat mass (treatment difference: -13.0%; 95% CI: -14.4 to -11.7; p < 0.01) and visceral fat rating (treatment difference: -1.3; 95% CI: -1.6 to -1.0; p < 0.01) were noted. Homozygous minor allele carriers experienced greater decreases in body weight (treatment difference: -3.2%; 95% CI: -4.9% to -1.6%; p < 0.01) and BMI (treatment difference: -1.2 kg/m2; 95% CI: -2.0 to -0.4; p < 0.01) compared to heterozygous allele carriers. These carriers also had a more significant reduction in fat mass (treatment difference: -9.8%; 95% CI: -10.6 to -9.1; p < 0.01) and visceral fat rating (treatment difference: -0.9; 95% CI: -1.3 to -0.5; p < 0.01). A high incidence of gastrointestinal events was reported in the experimental group (74.6%), unlike the placebo group, which reported no side effects. Dietary supplementation with glucomannan, inulin, and psyllium effectively promotes weight loss and improves body composition in individuals with obesity, particularly those with specific genetic polymorphisms.


Asunto(s)
Inulina , Mananos , Psyllium , Adulto , Humanos , Psyllium/uso terapéutico , Polimorfismo de Nucleótido Simple , Obesidad/tratamiento farmacológico , Obesidad/genética , Obesidad/epidemiología , Peso Corporal/genética , Pérdida de Peso/genética , Suplementos Dietéticos , Índice de Masa Corporal , Receptor de Melanocortina Tipo 4/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética
3.
Front Endocrinol (Lausanne) ; 14: 1288282, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38116320

RESUMEN

Introduction: Physical activity is recommended as an alternative treatment for depression. Myokines, which are secreted from skeletal muscles during physical activity, play an important role in the skeletal muscle-brain axis. Musclin, a newly discovered myokine, exerts physical endurance, however, the effects of musclin on emotional behaviors, such as depression, have not been evaluated. This study aimed to access the anti-depressive effect of musclin and clarify the connection between depression-like behavior and hypothalamic neuropeptides in mice. Methods: We measured the immobility time in the forced swim (FS) test, the time spent in open arm in the elevated-plus maze (EPM) test, the mRNA levels of hypothalamic neuropeptides, and enumerated the c-Fos-positive cells in the paraventricular nucleus (PVN), arcuate nucleus (ARC), and nucleus tractus solitarii (NTS) in mice with the intraperitoneal (i.p.) administration of musclin. Next, we evaluated the effects of a selective corticotropin-releasing factor (CRF) type 1 receptor antagonist, selective CRF type 2 receptor antagonist, melanocortin receptor (MCR) agonist, and selective melanocortin 4 receptor (MC4R) agonist on changes in behaviors induced by musclin. Finally we evaluated the antidepressant effect of musclin using mice exposed to repeated water immersion (WI) stress. Results: We found that the i.p. and i.c.v. administration of musclin decreased the immobility time and relative time in the open arms (open %) in mice and increased urocortin 2 (Ucn 2) levels but decreased proopiomelanocortin levels in the hypothalamus. The numbers of c-Fos-positive cells were increased in the PVN and NTS but decreased in the ARC of mice with i.p. administration of musclin. The c-Fos-positive cells in the PVN were also found to be Ucn 2-positive. The antidepressant and anxiogenic effects of musclin were blocked by central administration of a CRF type 2 receptor antagonist and a melanocortin 4 receptor agonist, respectively. Peripheral administration of musclin also prevented depression-like behavior and the decrease in levels of hypothalamic Ucn 2 induced by repeated WI stress. Discussion: These data identify the antidepressant effects of musclin through the activation of central Ucn 2 signaling and suggest that musclin and Ucn 2 can be new therapeutic targets and endogenous peptides mediating the muscle-brain axis.


Asunto(s)
Hormona Liberadora de Corticotropina , Urocortinas , Ratones , Masculino , Animales , Hormona Liberadora de Corticotropina/genética , Urocortinas/farmacología , Depresión/prevención & control , Receptor de Melanocortina Tipo 4 , Hipotálamo/metabolismo , Núcleo Solitario/metabolismo , Antidepresivos/farmacología , Proteínas Proto-Oncogénicas c-fos
4.
CNS Neurosci Ther ; 29(2): 646-658, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36510669

RESUMEN

AIMS: Central melanocortin 4 receptor (MC4R) has been reported to induce anhedonia via eliciting dysfunction of excitatory synapses. It is evident that metabolic signals are closely related to chronic stress-induced depression. Here, we investigated that a neural circuit is involved in melanocortin signaling contributing to susceptibility to stress. METHODS: Chronic social defeat stress (CSDS) was used to develop depressive-like behavior. Electrophysiologic and chemogenetic approaches were performed to evaluate the role of paraventricular thalamus (PVT) glutamatergic to nucleus accumbens shell (NAcsh) circuit in stress susceptibility. Pharmacological and genetic manipulations were applied to investigate the molecular mechanisms of melanocortin signaling in the circuit. RESULTS: CSDS increases the excitatory neurotransmission in NAcsh through MC4R signaling. The enhanced excitatory synaptic input in NAcsh is projected from PVT glutamatergic neurons. Moreover, chemogenetic manipulation of PVTGlu -NAcsh projection mediates the susceptibility to stress, which is dependent on MC4R signaling. Overall, these results reveal that the strengthened excitatory neurotransmission in NAcsh originates from PVT glutamatergic neurons, facilitating the susceptibility to stress through melanocortin signaling. CONCLUSIONS: Our results make a strong case for harnessing a thalamic circuit to reorganize excitatory synaptic transmission in relieving stress susceptibility and provide insights gained on metabolic underpinnings of protection against stress-induced depressive-like behavior.


Asunto(s)
Núcleo Accumbens , Receptor de Melanocortina Tipo 4 , Núcleo Accumbens/metabolismo , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Tálamo , Neuronas/metabolismo , Transmisión Sináptica
5.
Sci Rep ; 12(1): 10105, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35710868

RESUMEN

Melanocortin 4 receptor gene-knockout (MC4R-KO) mice are known to develop obesity with a high-fat diet. Meanwhile, daisaikoto, one of Kampo medicines, is a drug that is expected to have therapeutic effects on obesity. Here, we report the efficacy of daisaikoto in MC4R-KO mice. Eight-week-old MC4R-KO male mice (n = 12) were divided into three groups as follows: the SD group, which is fed with a standard diet; the HFD group, fed a high-fat diet; and the DSK group, fed with a high-fat diet containing 10% of daisaikoto. After the four-week observation period, mice in each group were sacrificed and samples were collected. The body weights at 12 weeks were significantly higher in the HFD group than in the other groups, indicating that daisaikoto significantly reduced body weight gain and fat deposition of the liver. The metabolome analysis indicated that degradation of triglycerides and fatty acid oxidation in the liver were enhanced by daisaikoto administration. In MC4R-KO mice, the cytoplasm and uncoupling protein 1 expression of brown adipose tissue was decreased; however, it was reversed in the DSK group. In conclusion, daisaikoto has potentially improved fatty liver and obesity, making it a useful therapeutic agent for obesity and fatty liver.


Asunto(s)
Tejido Adiposo Pardo , Hígado Graso , Tejido Adiposo/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Medicamentos Herbarios Chinos , Hígado Graso/tratamiento farmacológico , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/tratamiento farmacológico , Obesidad/genética , Obesidad/metabolismo , Receptor de Melanocortina Tipo 4
6.
Arq. ciências saúde UNIPAR ; 26(2): 159-174, maio-ago. 2022.
Artículo en Portugués | LILACS | ID: biblio-1372969

RESUMEN

A obesidade é definida pelo excesso de gordura corporal acumulada no tecido adiposo quando o indivíduo atinge valores de IMC igual ou superior a 30 Kg/m2. Constitui um dos principais fatores de risco para várias doenças não transmissíveis (DNTs) como por exemplo, diabetes mellitus tipo 2 (DM2), doenças cardiovasculares, hipertensão arterial, acidente vascular cerebral e até mesmo o câncer. Embora a obesidade esteja diretamente relacionada com o consumo calórico excessivo em relação ao gasto energético diário, sua etiologia pode estar associada aos baixos níveis de atividade física, às alterações neuroendócrinas e aos fatores genéticos. Considerando o componente genético, esta pode ser classificada como sindrômicas e estar associada às alterações cromossômicas estruturais ou numéricas, ou como não sindrômica, quando relacionada, principalmente, com os polimorfismos de nucleotídeos simples (SNPs) em alelos que atuam como herança monogênica, ou ainda com a interação vários genes (poligênica multifatorial). Apesar de existirem muitas etiologias diferentes, normalmente a obesidade é tratada a partir da mesma abordagem, desconsiderando a fisiologia que a desencadeou. Dessa forma, o objetivo do presente trabalho foi abordar a obesidade genética não sindrômica por meio a) da descrição breve de perspectiva histórica sobre seu entendimento; b) da exposição dos principais mecanismos moleculares envolvidos com o controle de peso; c) da compilação dos principais genes e SNPs relacionados; d) da definição dos principais genes; e e) da abordagem das principais perspectivas de intervenção.


Obesity is defined as excess body fat accumulated in the adipose tissue when the individual reaches BMI values equal to or greater than 30 kg/m2. It is one of the main risk factors for several non-communicable diseases (NCDs), such as Type 2 Diabetes mellitus (T2D), cardiovascular diseases, high blood pressure, stroke and even cancer. Although obesity is directly related to excessive calorie intake in relation to daily energy expenditure, its etiology may be associated with low levels of physical activity, neuroendocrine changes, and genetic factors. Considering the genetic component, it can be classified as syndromic and be associated with chromosomal or numerical changes, or as non-syndromic and being related mainly to single nucleotide polymorphisms (SNPs) in alleles that act as monogenic inheritance, or with an interaction of several genes (multifactorial polygenic). Although there are many different etiologies, obesity is usually treated using the same approach, disregarding the physiology that triggered it. Thus, the aim of this study was to address non-syndromic genetic obesity through a) a brief description of a historical perspective on its understanding; b) the exposure of the main molecular mechanisms involved in weight control, c) the compilation of the key genes and related SNPs, d) the definition of the key genes and e) the approach of the main intervention representations.


Asunto(s)
Humanos , Masculino , Femenino , Peso Corporal/genética , Epigenómica , Genes/genética , Obesidad/genética , Índice de Masa Corporal , Expresión Génica/genética , Polimorfismo de Nucleótido Simple/genética , Receptor de Melanocortina Tipo 4/genética , Melanocortinas/genética , Receptores de Leptina/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Hipotálamo/fisiopatología , Obesidad/fisiopatología
7.
Am J Physiol Endocrinol Metab ; 322(5): E436-E445, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35344393

RESUMEN

The melanocortin 4 receptor (MC4R) plays an important role in the regulation of appetite and energy expenditure in humans and rodents. Impairment of MC4R signaling causes severe obesity. MC4R mainly couples to the G-protein Gs. Ligand binding to MC4R activates adenylyl cyclase resulting in increased intracellular cAMP levels. cAMP acts as a secondary messenger, regulating various cellular processes. MC4R can also couple with Gq and other signaling pathways. Therefore, the contribution of MC4R/Gs signaling to energy metabolism and appetite remains unclear. To study the effect of Gs signaling activation in MC4R cells on whole body energy metabolism and appetite, we generated a novel mouse strain that expresses a Gs-coupled designer receptors exclusively activated by designer drugs [Gs-DREADD (GsD)] selectively in MC4R-expressing cells (GsD-MC4R mice). Chemogenetic activation of the GsD by a designer drug [deschloroclozapine (DCZ); 0.01∼0.1 mg/kg body wt] in MC4R-expressing cells significantly increased oxygen consumption and locomotor activity. In addition, GsD activation significantly reduced the respiratory exchange ratio, promoting fatty acid oxidation, but did not affect core (rectal) temperature. A low dose of DCZ (0.01 mg/kg body wt) did not suppress food intake, but a high dose of DCZ (0.1 mg/kg body wt) suppressed food intake in MC4R-GsD mice, although either DCZ dose (0.01 or 0.1 mg/kg body wt) did not affect food intake in the control mice. In conclusion, the current study demonstrated that the stimulation of Gs signaling in MC4R-expressing cells increases energy expenditure and locomotor activity and suppresses appetite.NEW & NOTEWORTHY We report that Gs signaling in melanocortin 4 receptor (MC4R)-expressing cells regulates energy expenditure, appetite, and locomotor activity. These findings shed light on the mechanism underlying the regulation of energy metabolism and locomotor activity by MC4R/cAMP signaling.


Asunto(s)
Proteínas de Unión al GTP , Obesidad , Receptor de Melanocortina Tipo 4 , Animales , Ingestión de Alimentos , Metabolismo Energético , Proteínas de Unión al GTP/metabolismo , Locomoción , Ratones , Obesidad/metabolismo , Receptor de Melanocortina Tipo 4/genética
8.
Mol Metab ; 55: 101401, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34823066

RESUMEN

OBJECTIVE: The paraventricular nucleus of hypothalamus (PVN), an integrative center in the brain, orchestrates a wide range of physiological and behavioral responses. While the PVN melanocortin 4 receptor (MC4R) signaling (PVNMC4R+) is involved in feeding regulation, the neuroanatomical organization of PVNMC4R+ connectivity and its role in other physiological regulations are incompletely understood. Here we aimed to better characterize the input-output organization of PVNMC4R+ neurons and test their physiological functions beyond feeding. METHODS: Using a combination of viral tools, we mapped PVNMC4R+ circuits and tested the effects of chemogenetic activation of PVNMC4R+ neurons on thermoregulation, cardiovascular control, and other behavioral responses beyond feeding. RESULTS: We found that PVNMC4R+ neurons innervate many different brain regions that are known to be important not only for feeding but also for neuroendocrine and autonomic control of thermoregulation and cardiovascular function, including but not limited to the preoptic area, median eminence, parabrachial nucleus, pre-locus coeruleus, nucleus of solitary tract, ventrolateral medulla, and thoracic spinal cord. Contrary to these broad efferent projections, PVNMC4R+ neurons receive monosynaptic inputs mainly from other hypothalamic nuclei (preoptic area, arcuate and dorsomedial hypothalamic nuclei, supraoptic nucleus, and premammillary nucleus), the circumventricular organs (subfornical organ and vascular organ of lamina terminalis), the bed nucleus of stria terminalis, and the parabrachial nucleus. Consistent with their broad efferent projections, chemogenetic activation of PVNMC4R+ neurons not only suppressed feeding but also led to an apparent increase in heart rate, blood pressure, and brown adipose tissue temperature. These physiological changes accompanied acute transient hyperactivity followed by hypoactivity and resting-like behavior. CONCLUSIONS: Our results elucidate the neuroanatomical organization of PVNMC4R+ circuits and shed new light on the roles of PVNMC4R+ pathways in autonomic control of thermoregulation, cardiovascular function, and biphasic behavioral activation.


Asunto(s)
Núcleo Hipotalámico Paraventricular/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Regulación de la Temperatura Corporal/fisiología , Encéfalo/metabolismo , Núcleo Hipotalámico Dorsomedial/metabolismo , Técnicas de Sustitución del Gen/métodos , Hipotálamo/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Neuronas/metabolismo , Receptor de Melanocortina Tipo 4/fisiología , Médula Espinal/metabolismo
9.
Mol Metab ; 53: 101317, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34400348

RESUMEN

OBJECTIVE: Homo- or heterodimerization of G protein-coupled receptors (GPCRs) generally alters the normal functioning of these receptors and mediates their responses to a variety of physiological stimuli in vivo. It is well known that melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R) are key regulators of appetite and energy homeostasis in the central nervous system (CNS). However, the GPCR partners of MC3R and MC4R are not well understood. Our objective is to analyze single cell RNA-seq datasets of the hypothalamus to explore and identify novel GPCR partners of MC3R and MC4R and examine the pharmacological effect on the downstream signal transduction and membrane translocation of melanocortin receptors. METHODS: We conducted an integrative analysis of multiple single cell RNA-seq datasets to reveal the expression pattern and correlation of GPCR families in the mouse hypothalamus. The emerging GPCRs with important metabolic functions were selected for cloning and co-immunoprecipitation validation. The positive GPCR partners were then tested for the pharmacological activation, competitive binding assay and surface translocation ELISA experiments. RESULTS: Based on the expression pattern of GPCRs and their function enrichment results, we narrowed down the range of potential GPCR interaction with MC3R and MC4R for further confirmation. Co-immunoprecipitation assay verified 23 and 32 novel GPCR partners that interacted with MC3R and MC4R in vitro. The presence of these GPCR partners exhibited different effects in the physiological regulation and signal transduction of MC3R and MC4R. CONCLUSIONS: This work represented the first large-scale screen for the functional GPCR complex of central melanocortin receptors and defined a composite metabolic regulatory GPCR network of the hypothalamic nucleuses.


Asunto(s)
Melanocortinas/metabolismo , Receptor de Melanocortina Tipo 3/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Hipotálamo/metabolismo , Ratones , Receptor de Melanocortina Tipo 3/genética , Receptor de Melanocortina Tipo 4/genética , Transducción de Señal
10.
Handb Clin Neurol ; 181: 301-310, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34238466

RESUMEN

Neural circuits in the hypothalamus play a key role in the regulation of human energy homeostasis. A critical circuit involves leptin-responsive neurons in the hypothalamic arcuate nucleus (the infundibular nucleus in humans) expressing the appetite-suppressing neuropeptide proopiomelanocortin (POMC) and the appetite-stimulating Agouti-related peptide. In the fed state, the POMC-derived melanocortin peptide α-melanocyte-stimulating hormone stimulates melanocortin-4 receptors (MC4Rs) expressed on second-order neurons in the paraventricular nucleus of the hypothalamus (PVN). Agonism of MC4R leads to reduced food intake and increased energy expenditure. Disruption of this hypothalamic circuit by inherited mutations in the genes encoding leptin, the leptin receptor, POMC, and MC4R can lead to severe obesity in humans. The characterization of these and closely related genetic obesity syndromes has informed our understanding of the neural pathways by which leptin regulates energy balance, neuroendocrine function, and the autonomic nervous system. A broader understanding of these neural and molecular mechanisms has paved the way for effective mechanism-based therapies for patients whose severe obesity is driven by disruption of these pathways.


Asunto(s)
Obesidad , Proopiomelanocortina , Metabolismo Energético/genética , Humanos , Hipotálamo/metabolismo , Leptina/genética , Leptina/metabolismo , Obesidad/genética , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/genética , Receptores de Leptina/genética , Síndrome
11.
Diabetes ; 70(9): 2081-2091, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34183373

RESUMEN

Work in recent decades has established that metabolic hormones released by endocrine cells and diverse other cell types serve to regulate nutrient intake and energy homeostasis. Tsukushi (TSK) is a leucine-rich repeat-containing protein secreted primarily by the liver that exerts an inhibitory effect on brown fat sympathetic innervation and thermogenesis. Despite this, physiological regulation of TSK and the mechanisms underlying its effects on energy balance remain poorly understood. Here we show that hepatic expression and plasma concentrations of TSK are induced by feeding and regulated by melanocortin-4 receptor (MC4R) signaling. We generated TSK and MC4R-double-knockout mice to elucidate the nature of cross talk between TSK and the central regulatory circuit of energy balance. Remarkably, TSK inactivation restores energy balance, ameliorates hyperphagia, and improves metabolic health in MC4R-deficient mice. TSK ablation enhances thermogenic gene expression in brown fat, dampens obesity-association inflammation in the liver and adipose tissue, and protects MC4R-null mice from diet-induced nonalcoholic steatohepatitis. At the cellular level, TSK deficiency augments feeding-induced c-Fos expression in the paraventricular nucleus of the hypothalamus. These results illustrate physiological cross talk between TSK and the central regulatory circuit in maintaining energy balance and metabolic homeostasis.


Asunto(s)
Metabolismo Energético/fisiología , Obesidad/metabolismo , Proteoglicanos/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Fármacos Antiobesidad/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/genética , Proteoglicanos/genética , Receptor de Melanocortina Tipo 4/genética , Transducción de Señal/fisiología , Termogénesis/fisiología , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
12.
Nat Commun ; 12(1): 3525, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112797

RESUMEN

Contrasting to the established role of the hypothalamic agouti-related protein (AgRP) neurons in feeding regulation, the neural circuit and signaling mechanisms by which they control energy expenditure remains unclear. Here, we report that energy expenditure is regulated by a subgroup of AgRP neurons that send non-collateral projections to neurons within the dorsal lateral part of dorsal raphe nucleus (dlDRN) expressing the melanocortin 4 receptor (MC4R), which in turn innervate nearby serotonergic (5-HT) neurons. Genetic manipulations reveal a bi-directional control of energy expenditure by this circuit without affecting food intake. Fiber photometry and electrophysiological results indicate that the thermo-sensing MC4RdlDRN neurons integrate pre-synaptic AgRP signaling, thereby modulating the post-synaptic serotonergic pathway. Specifically, the MC4RdlDRN signaling elicits profound, bi-directional, regulation of body weight mainly through sympathetic outflow that reprograms mitochondrial bioenergetics within brown and beige fat while feeding remains intact. Together, we suggest that this AgRP neural circuit plays a unique role in persistent control of energy expenditure and body weight, hinting next-generation therapeutic approaches for obesity and metabolic disorders.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Conducción Nerviosa/fisiología , Neuronas Serotoninérgicas/fisiología , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Peso Corporal , Cromatografía Liquida , Ingestión de Alimentos/fisiología , Metabolismo Energético/genética , Masculino , Ratones , Conducción Nerviosa/efectos de los fármacos , Conducción Nerviosa/efectos de la radiación , Obesidad/metabolismo , Optogenética , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/efectos de la radiación , Serotonina/metabolismo , Serotonina/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Espectrometría de Masas en Tándem , Temperatura
13.
J Exp Med ; 218(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33978701

RESUMEN

Atypical antipsychotics such as risperidone cause drug-induced metabolic syndrome. However, the underlying mechanisms remain largely unknown. Here, we report a new mouse model that reliably reproduces risperidone-induced weight gain, adiposity, and glucose intolerance. We found that risperidone treatment acutely altered energy balance in C57BL/6 mice and that hyperphagia accounted for most of the weight gain. Transcriptomic analyses in the hypothalamus of risperidone-fed mice revealed that risperidone treatment reduced the expression of Mc4r. Furthermore, Mc4r in Sim1 neurons was necessary for risperidone-induced hyperphagia and weight gain. Moreover, we found that the same pathway underlies the obesogenic effect of olanzapine-another commonly prescribed antipsychotic drug. Remarkably, whole-cell patch-clamp recording demonstrated that risperidone acutely inhibited the activity of hypothalamic Mc4r neurons via the opening of a postsynaptic potassium conductance. Finally, we showed that treatment with setmelanotide, an MC4R-specific agonist, mitigated hyperphagia and obesity in both risperidone- and olanzapine-fed mice.


Asunto(s)
Antipsicóticos/farmacología , Receptor de Melanocortina Tipo 4/metabolismo , Risperidona/farmacología , Aumento de Peso/efectos de los fármacos , Animales , Femenino , Hiperfagia/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Obesidad/metabolismo , Olanzapina/farmacología , Potasio/metabolismo , Potenciales Sinápticos/efectos de los fármacos , Transcriptoma/efectos de los fármacos , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
14.
Pharmacol Res Perspect ; 9(3): e00777, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34014033

RESUMEN

The melanocortin-4 receptor (MC4R) antagonistic peptide TCMCB07 was developed for the treatment of cachexia. The objectives of this study were to examine pharmacokinetics and safety of TCMCB07 administered subcutaneously to healthy dogs. Dogs were treated with high- (2.25 mg kg-1 ) (n = 5) and low-dose TCMCB07 (0.75 mg kg-1 ) (n = 5) once daily for 28 days with a 14-day washout period between groups. Histamine levels, complete blood count, chemistry panel, blood pressure, 24-hour Holter recording, and pharmacokinetic parameters were monitored in the high-dose group. Physical examination changes were limited to weight gain and darkening of the coat color. There was no elevation of plasma histamine within 24 hours of injection but there was a significant elevation of plasma histamine across time. An approximately doubled eosinophil count and an approximately 25% increase, and then 25% decrease back to pre-treatment plasma phosphorous were also found, although both remained within the reference interval. Serial blood pressure and 24-hour Holter monitors revealed no clinically relevant changes. A difference was found in the AUC between dosing groups and a significant effect of dose, time, and interaction was noted for Vd . Low-dose TCMCB07 had a Cmax of 2.1 ug ml-1 at day 28, compared to high-dose TCMCB07 which had a Cmax 3.6 ug ml-1 at day 28. Once-daily subcutaneous administration of TCMCB07 was well-tolerated for up to 28 days in dogs when administered at doses one and three times (0.75 mg kg-1 and 2.25 mg kg-1 ) the predicted therapeutic dose and pharmacokinetic parameters are described. SIGNIFICANCE STATEMENT: Melanocortin-4 receptor (MC4R) antagonistic peptide TCMCB07 is safe at both low and high doses in dogs. Therapy was tolerated well as determined by physical examination, clinical pathology, and cardiovascular parameters; darkening of the coat was noted with treatment and resolved with discontinuation. Pharmacokinetics are described and further study in the naturally occurring canine model is warranted.


Asunto(s)
Péptidos Cíclicos/farmacocinética , Receptor de Melanocortina Tipo 4/antagonistas & inhibidores , Animales , Arritmias Cardíacas/inducido químicamente , Presión Arterial/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Perros , Femenino , Histamina/sangre , Inyecciones Subcutáneas , Masculino , Péptidos Cíclicos/efectos adversos , Fósforo/sangre
15.
Endocrinology ; 162(6)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33834205

RESUMEN

The paraventricular nucleus of the hypothalamus (PVH) is a heterogeneous collection of neurons that play important roles in modulating feeding and energy expenditure. Abnormal development or ablation of the PVH results in hyperphagic obesity and defects in energy expenditure whereas selective activation of defined PVH neuronal populations can suppress feeding and may promote energy expenditure. Here, we characterize the contribution of calcitonin receptor-expressing PVH neurons (CalcRPVH) to energy balance control. We used Cre-dependent viral tools delivered stereotaxically to the PVH of CalcR2Acre mice to activate, silence, and trace CalcRPVH neurons and determine their contribution to body weight regulation. Immunohistochemistry of fluorescently-labeled CalcRPVH neurons demonstrates that CalcRPVH neurons are largely distinct from several PVH neuronal populations involved in energy homeostasis; these neurons project to regions of the hindbrain that are implicated in energy balance control, including the nucleus of the solitary tract and the parabrachial nucleus. Acute activation of CalcRPVH neurons suppresses feeding without appreciably augmenting energy expenditure, whereas their silencing leads to obesity that may be due in part due to loss of PVH melanocortin-4 receptor signaling. These data show that CalcRPVH neurons are an essential component of energy balance neurocircuitry and their function is important for body weight maintenance. A thorough understanding of the mechanisms by which CalcRPVH neurons modulate energy balance might identify novel therapeutic targets for the treatment and prevention of obesity.


Asunto(s)
Metabolismo Energético/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Receptores de Calcitonina/fisiología , Animales , Ingestión de Alimentos/fisiología , Metabolismo Energético/genética , Conducta Alimentaria/fisiología , Homeostasis/fisiología , Hipotálamo/metabolismo , Hipotálamo/fisiología , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/metabolismo , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Receptor de Melanocortina Tipo 4/fisiología , Receptores de Calcitonina/genética , Receptores de Calcitonina/metabolismo
16.
Sci China Life Sci ; 64(3): 419-433, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32803714

RESUMEN

Cenpj is a centrosomal protein located at the centrosomes and the base of cilia, it plays essential roles in regulating neurogenesis and cerebral cortex development. Although centrosomal and cilium dysfunction are one of the causes of obesity, insulin resistance, and type 2 diabetes, the role that Cenpj plays in the regulation of body weight remains unclear. Here, we deleted Cenpj by crossing Cenpjflox/flox mice with Nkx2.1-Cre mice. Loss of the centrosomal protein Cenpj in Nkx2.1-expressing cells causes morbid obesity in mice at approximately 4 months of age with expended brain ventricles but no change of brain size. We found that hypothalamic cells exhibited reduced proliferation and increased apoptosis upon Cenpj depletion at the embryonic stages, resulting in a dramatic decrease in the number of Proopiomelanocortin (POMC) neurons and electrophysiological dysfunction of NPY neurons in the arcuate nucleus (ARC) in adults. Furthermore, depletion of Cenpj also reduced the neuronal projection from the ARC to the paraventricular nucleus (PVN), with decreased melanocortin-4 receptors (MC4R) expression in PVN neurons. The study defines the roles that Cenpj plays in regulating hypothalamus development and body weight, providing a foundation for further understanding of the pathological mechanisms of related diseases.


Asunto(s)
Técnicas de Silenciamiento del Gen , Hipotálamo/fisiopatología , Proteínas Asociadas a Microtúbulos/genética , Obesidad Mórbida/fisiopatología , Animales , Apoptosis , Línea Celular , Proliferación Celular , Hipotálamo/embriología , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Obesidad Mórbida/genética , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Factor Nuclear Tiroideo 1/genética
17.
Diabetologia ; 64(1): 181-194, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33052459

RESUMEN

AIMS/HYPOTHESIS: Melanocortin 4 receptor (MC4R) mutation is the most common cause of known monogenic obesity in humans. Unexpectedly, humans and rodents with MC4R deficiency do not develop hyperglycaemia despite chronic obesity and insulin resistance. To explain the underlying mechanisms for this phenotype, we determined the role of MC4R in glucose homeostasis in the presence and absence of obesity in mice. METHODS: We used global and hypothalamus-specific MC4R-deficient mice to investigate the brain regions that contribute to glucose homeostasis via MC4R. We performed oral, intraperitoneal and intravenous glucose tolerance tests in MC4R-deficient mice that were either obese or weight-matched to their littermate controls to define the role of MC4R in glucose regulation independently of changes in body weight. To identify the integrative pathways through which MC4R regulates glucose homeostasis, we measured renal and adrenal sympathetic nerve activity. We also evaluated glucose homeostasis in adrenaline (epinephrine)-deficient mice to investigate the role of adrenaline in mediating the effects of MC4R in glucose homeostasis. We employed a graded [13C6]glucose infusion procedure to quantify renal glucose reabsorption in MC4R-deficient mice. Finally, we measured the levels of renal glucose transporters in hypothalamus-specific MC4R-deficient mice and adrenaline-deficient mice using western blotting to ascertain the molecular mechanisms underlying MC4R control of glucose homeostasis. RESULTS: We found that obese and weight-matched MC4R-deficient mice exhibited improved glucose tolerance due to elevated glucosuria, not enhanced beta cell function. Moreover, MC4R deficiency selectively in the paraventricular nucleus of the hypothalamus (PVH) is responsible for reducing the renal threshold for glucose as measured by graded [13C6]glucose infusion technique. The MC4R deficiency suppressed renal sympathetic nerve activity by 50% in addition to decreasing circulating adrenaline and renal GLUT2 levels in mice, which contributed to the elevated glucosuria. We further report that adrenaline-deficient mice recapitulated the increased excretion of glucose in urine observed in the MC4R-deficient mice. Restoration of circulating adrenaline in both the MC4R- and adrenaline-deficient mice reversed their phenotype of improved glucose tolerance and elevated glucosuria, demonstrating the role of adrenaline in mediating the effects of MC4R on glucose reabsorption. CONCLUSIONS/INTERPRETATION: These findings define a previously unrecognised function of hypothalamic MC4R in glucose reabsorption mediated by adrenaline and renal GLUT2. Taken together, our findings indicate that elevated glucosuria due to low sympathetic tone explains why MC4R deficiency does not cause hyperglycaemia despite inducing obesity and insulin resistance. Graphical abstract.


Asunto(s)
Hexosas/metabolismo , Homeostasis/fisiología , Receptor de Melanocortina Tipo 4/fisiología , Bases de Schiff/metabolismo , Animales , Glucemia/metabolismo , Cruzamientos Genéticos , Epinefrina/deficiencia , Epinefrina/fisiología , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 2/fisiología , Glucosuria/fisiopatología , Hipotálamo/química , Insulina/sangre , Resistencia a la Insulina/fisiología , Riñón/inervación , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/fisiopatología , Receptor de Melanocortina Tipo 4/deficiencia , Sistema Nervioso Simpático/fisiopatología
18.
Nutrients ; 12(11)2020 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-33202557

RESUMEN

The dysfunction of melanocortin signaling has been associated with obesity, given the important role in the regulation of energy homeostasis, food intake, satiety and body weight. In the hypothalamus, the melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R) contribute to the stability of these processes, but MC3R and MC4R are also localized in the mesolimbic dopamine system, the region that responds to the reinforcing properties of highly palatable food (HPF) and where these two receptors seem to affect food reward and motivation. Loss of function of the MC4R, resulting from genetic mutations, leads to overeating in humans, but to date, a clear understanding of the underlying mechanisms and behaviors that promote overconsumption of caloric foods remains unknown. Moreover, the MC4R demonstrated to be a crucial modulator of the stress response, factor that is known to be strictly related to binge eating behavior. In this review, we will explore the preclinical and clinical studies, and the controversies regarding the involvement of melanocortin system in altered eating patterns, especially binge eating behavior, food reward and motivation.


Asunto(s)
Bulimia/genética , Ingestión de Alimentos/genética , Conducta Alimentaria , Hiperfagia/genética , Obesidad/genética , Receptor de Melanocortina Tipo 4/genética , Índice de Masa Corporal , Ingestión de Alimentos/psicología , Humanos , Hipotálamo/metabolismo , Motivación , Mutación , Obesidad/psicología , Receptor de Melanocortina Tipo 3/genética , Receptor de Melanocortina Tipo 3/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Recompensa
19.
Int J Mol Sci ; 21(19)2020 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-32987823

RESUMEN

The melanocortin receptor 4 (MC4R) signaling system consists of MC4R, MC4R ligands [melanocyte-stimulating hormone (MSH), adrenocorticotropin (ACTH), agouti-related protein (AgRP)], and melanocortin-2 receptor accessory protein 2 (MRAP2), and it has been proposed to play important roles in feeding and growth in vertebrates. However, the expression and functionality of this system have not been fully characterized in teleosts. Here, we cloned tilapia MC4R, MRAP2b, AgRPs (AgRP, AgRP2), and POMCs (POMCa1, POMCb) genes and characterized the interaction of tilapia MC4R with MRAP2b, AgRP, α-MSH, and ACTH in vitro. The results indicate the following. (1) Tilapia MC4R, MRAP2b, AgRPs, and POMCs share high amino acid identity with their mammalian counterparts. (2) Tilapia MRAP2b could interact with MC4R expressed in CHO cells, as demonstrated by Co-IP assay, and thus decrease MC4R constitutive activity and enhance its sensitivity to ACTH1-40. (3) As in mammals, AgRP can function as an inverse agonist and antagonist of MC4R, either in the presence or absence of MRAP2b. These data, together with the co-expression of MC4R, MRAP2b, AgRPs, and POMCs in tilapia hypothalamus, suggest that as in mammals, ACTH/α-MSH, AgRP, and MRAP2 can interact with MC4R to control energy balance and thus play conserved roles in the feeding and growth of teleosts.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cíclidos/metabolismo , Hipotálamo/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal , Animales
20.
Toxicon ; 187: 57-64, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32882257

RESUMEN

There is a lack of information available on the anorexic action of fusarenon-x (FX), which is a sesquiterpenoid mycotoxin. In this study, we investigated the changes in the hypothalamus and small intestine related to appetite after oral FX exposure. The time-course change of food intake after oral FX exposure (0.5, 1.0, and 2.5 mg/kg bw) in B6C3F1 mice showed that 2.5 mg/kg bw of FX significantly suppressed food intake during 3-6 h compared to the control. Furthermore, the total food intake for 24 h was lower in the group exposed to FX than in the control. The FX exposure (2.5 mg/kg bw for 3 h) significantly increased mRNA levels of anorexic hormones (pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcription (CART)) without changing the mRNA levels of orexigenic hormones. In addition, FX exposure indicated significantly higher mRNA levels of possible downstream targets of anorexic POMC neurons, such as the melanocortin 4 receptor (MC4R), brain-derived neurotrophic factor (BDNF) and tyrosine kinase receptor B (TrkB), in the hypothalamus compared to the control. FX exposure also significantly increased the mRNA level of inflammatory cytokines (tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß)) and activated nuclear factor-kappa B (NF-κB), which is a regulatory factor for POMC in the hypothalamus. In the intestine, FX exposure did not affect the mRNA level of anorexic peptide YY but significantly elevated that of anorexic cholecystokinin (CCK) and regulatory factors for CCK (calcium-sensing receptor (CaSR), the transient receptor potential ankyrin-1 channel (TRPA1), and transient receptor potential cation channel subfamily M member 5 (TRPM5)). These results suggest that FX sequentially induces inflammatory cytokine expression, NF-κB activation, and POMC expression in the hypothalamus. FX also induces CCK expression in the intestine possibly via induction of CaSR, TRPM5, and TRPA1 expression. These changes will eventually lead to the anorexic action of FX.


Asunto(s)
Hipotálamo/efectos de los fármacos , Intestinos/efectos de los fármacos , Tricotecenos/toxicidad , Animales , Anorexia , Masculino , Ratones , FN-kappa B/metabolismo , Proopiomelanocortina , Receptor de Melanocortina Tipo 4
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA