Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
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
2.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34654741

RESUMEN

Hypothalamic regulation of feeding and energy expenditure is a fundamental and evolutionarily conserved neurophysiological process critical for survival. Dysregulation of these processes, due to environmental or genetic causes, can lead to a variety of pathological conditions ranging from obesity to anorexia. Melanocortins and endogenous cannabinoids (eCBs) have been implicated in the regulation of feeding and energy homeostasis; however, the interaction between these signaling systems is poorly understood. Here, we show that the eCB 2-arachidonoylglycerol (2-AG) regulates the activity of melanocortin 4 receptor (MC4R) cells in the paraventricular nucleus of the hypothalamus (PVNMC4R) via inhibition of afferent GABAergic drive. Furthermore, the tonicity of eCBs signaling is inversely proportional to energy state, and mice with impaired 2-AG synthesis within MC4R neurons weigh less, are hypophagic, exhibit increased energy expenditure, and are resistant to diet-induced obesity. These mice also exhibit MC4R agonist insensitivity, suggesting that the energy state-dependent, 2-AG-mediated suppression of GABA input modulates PVNMC4R neuron activity to effectively respond to the MC4R natural ligands to regulate energy homeostasis. Furthermore, post-developmental disruption of PVN 2-AG synthesis results in hypophagia and death. These findings illustrate a functional interaction at the cellular level between two fundamental regulators of energy homeostasis, the melanocortin and eCB signaling pathways in the hypothalamic feeding circuitry.


Asunto(s)
Cannabinoides/metabolismo , Metabolismo Energético/fisiología , Homeostasis/fisiología , Receptor de Melanocortina Tipo 4/fisiología , Animales , Ácidos Araquidónicos/fisiología , Peso Corporal , Endocannabinoides/fisiología , Ayuno , Conducta Alimentaria/fisiología , Prueba de Tolerancia a la Glucosa , Glicéridos/fisiología , Resistencia a la Insulina , Ratones , Obesidad/genética , Receptor de Melanocortina Tipo 4/agonistas , Ácido gamma-Aminobutírico/metabolismo
3.
J Clin Endocrinol Metab ; 106(9): 2606-2616, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34036349

RESUMEN

CONTEXT: Pro-opiomelanocortin (POMC) and the melanocortin-4 receptor (MC4R) play a pivotal role in the leptin-melanocortin pathway. Mutations in these genes lead to monogenic types of obesity due to severe hyperphagia. In addition to dietary-induced obesity, a cardiac phenotype without hypertrophy has been identified in MC4R knockout mice. OBJECTIVE: We aimed to characterize cardiac morphology and function as well as tissue Na+ content in humans with mutations in POMC and MC4R genes. METHODS: A cohort of 42 patients (5 patients with bi-allelic POMC mutations, 6 heterozygous MC4R mutation carriers, 19 obese controls without known monogenic cause, and 12 normal weight controls) underwent cardiac magnetic resonance (CMR) imaging and 23Na-MRI. RESULTS: Monogenic obese patients with POMC or MC4R mutation respectively had a significantly lower left ventricular mass/body surface area (BSA) than nonmonogenic obese patients. Left ventricular end-diastolic volume/BSA was significantly lower in POMC- and MC4R-deficient patients than in nonmonogenic obese patients. Subcutaneous fat and skin Na+ content was significantly higher in POMC- and MC4R-deficient patients than in nonmonogenic obese patients. In these compartments, the water content was significantly higher in patients with POMC and MC4R mutation than in control groups. CONCLUSION: Patients with POMC or MC4R mutations carriers had a lack of transition to hypertrophy, significantly lower cardiac muscle mass/BSA, and stored more Na+ within the subcutaneous fat tissue than nonmonogenic obese patients. The results point towards the role of the melanocortin pathway for cardiac function and tissue Na+ storage and the importance of including cardiologic assessments into the diagnostic work-up of these patients.


Asunto(s)
Hipertrofia Ventricular Izquierda/etiología , Mutación , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/genética , Sodio/metabolismo , Función Ventricular Izquierda/fisiología , Adolescente , Agua Corporal/metabolismo , Femenino , Humanos , Hipertrofia Ventricular Izquierda/genética , Imagen por Resonancia Magnética , Masculino , Obesidad/complicaciones , Fenotipo , Proopiomelanocortina/fisiología , Receptor de Melanocortina Tipo 4/fisiología
4.
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
5.
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
6.
Obes Rev ; 21(5): e12992, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31889409

RESUMEN

In early childhood, individuals with Prader-Willi syndrome (PWS) experience excess weight gain and severe hyperphagia with food compulsivity, which often leads to early onset morbid obesity. Effective treatments for appetite suppression and weight control are currently unavailable for PWS. Our aim to further understand the pathogenesis of PWS led us to carry out a comprehensive search of the current and emerging therapies for managing hyperphagia and extreme weight gain in PWS. A literature search was performed using PubMed and the following keywords: "PWS" AND "therapy" OR "[drug name]"; reference lists, pharmaceutical websites, and the ClinicalTrials.gov registry were also reviewed. Articles presenting data from current standard treatments in PWS and also clinical trials of pharmacological agents in the pipeline were selected. Current standard treatments include dietary restriction/modifications, exercise, and growth hormone replacement, which appear to have limited efficacy for appetite and weight control in patients with PWS. The long-term safety and effectiveness of bariatric surgery in PWS remains unknown. However, many promising pharmacotherapies are in development and, if approved, will bring much needed choices into the PWS pharmacological armamentarium. With the progress that is currently being made in our understanding of PWS, an effective treatment may not be far off.


Asunto(s)
Hiperfagia/prevención & control , Obesidad Infantil/prevención & control , Síndrome de Prader-Willi/terapia , Acilación , Adolescente , Animales , Cirugía Bariátrica , Niño , Preescolar , Dietoterapia , Femenino , Ghrelina/sangre , Ghrelina/química , Hormona de Crecimiento Humana/deficiencia , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Hiperfagia/etiología , Lactante , Masculino , Oxitocina/uso terapéutico , Obesidad Infantil/etiología , Canales de Potasio/fisiología , Síndrome de Prader-Willi/complicaciones , Síndrome de Prader-Willi/fisiopatología , Receptor de Melanocortina Tipo 4/fisiología
7.
Nutrition ; 67-68: 110547, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31472367

RESUMEN

Obesity is a multifactorial, complex, and public health problem worldwide. Interaction between genes and environment as associated with diet may predispose an individual to obesity. In this sense, nutrigenetics appears to be a strategy that can improve understanding of the gene-diet interaction. The aim of this literature review was to summarize data from studies of genes involved in the regulation of energy intake (melanocortin 4 receptor [MC4R], fat mass and obesity-associated [FTO], ghrelin [GHRL], leptin [LEP], and cholecystokinin [CCK]) and diet interaction in obesity. The presence of polymorphisms in MC4R, FTO, leptin, and the respective receptor appear to be associated with higher energy and total lipid consumption. Polymorphisms in FTO, leptin, and leptin receptor are also related to increased intake of saturated fatty acids. Individuals with the MC4R, FTO, and ghrelin polymorphisms, who submitted themselves for weight loss intervention, appeared to achieve weight loss similar to individuals without polymorphisms in these genes. Additionally, protein seems to interact with these genes, which increases or decreases appetite, or to drive or lessen body weight recovery. Additionally, polymorphisms in these genes were found to be associated with inappropriate eating behaviors, such as increased consumption of sweets and snacks, consumption of large food portions, desire to eat, and eating associated with emotional issues. Preliminary data has supported the gene-diet interaction in determining weight loss and gain in individuals with polymorphisms in the genes involved in energy intake. Despite the advent of nutrigenetics in obesity, it is still too early to define the dietary management for weight loss based on the presence or absence of obesity polymorphisms.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/fisiología , Ingestión de Energía/genética , Leptina/fisiología , Obesidad/genética , Receptor de Melanocortina Tipo 4/fisiología , Receptores de Leptina/fisiología , Conducta Alimentaria/fisiología , Humanos , Nutrigenómica , Polimorfismo de Nucleótido Simple
8.
Curr Hypertens Rep ; 21(6): 46, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31028563

RESUMEN

PURPOSE OF REVIEW: To highlight the role of the brain melanocortin 4 receptor (MC4R) for sympathetic nervous system (SNS) activation in hypertension. RECENT FINDINGS: Hypertension is the most significant risk factor for developing cardiovascular disease. Although excess weight gain is associated with at least two thirds of primary hypertension cases, the pathophysiological mechanisms involved remain the subject of intense investigation. Multiple studies demonstrate an important role for increased sympathetic nervous system (SNS) activity in development and maintenance of hypertension, and that the brain MC4R modulates SNS activity to thermogenic, cardiovascular, and kidney tissues. These studies also support the concept that MC4R activation is critical for obesity-induced hypertension as well as other forms of hypertension associated with increased SNS activity. MC4R is a potential target for antiobesity therapy, although there are challenges in using MC4R agonists to induce weight loss without evoking increases in SNS activity.


Asunto(s)
Hipertensión/fisiopatología , Obesidad/fisiopatología , Receptor de Melanocortina Tipo 4/fisiología , Sistema Nervioso Simpático/fisiopatología , Apetito/fisiología , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Metabolismo Energético/fisiología , Humanos , Hipertensión/etiología , Obesidad/complicaciones , Receptor de Melanocortina Tipo 4/agonistas , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología
9.
Diabetes Metab Res Rev ; 35(5): e3149, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30851142

RESUMEN

AIMS: Amylin, a pancreatic hormone cosecreted with insulin, exerts important anorexic and weight-loss effects. Melanocortin 4 receptor (MC4R) signalling plays a critical role in energy homeostasis; however, its role on amylin-dependent regulation of food intake and adaptive thermogenesis of interscapular brown adipose tissue (IBAT) are unclear. In this study, we examined the effects of amylin on food intake and thermogenesis on IBAT via the MC4R pathway in mice. MATERIALS AND METHODS: Acute food consumption and thermogenesis in IBAT were measured in male wild-type (WT) and MC4R-deficient mice following intraperitoneal injection of amylin and SHU9119, an MC3R/4R antagonist, to determine the role of the central melanocortin system on the hypothalamus and IBAT. RESULTS: Amylin (50 µg/kg) suppressed feeding and stimulated thermogenesis on IBAT via activation of the MC4R system in mice. Pharmacological blockade of MC4R using SHU9119 (50 µg/kg) attenuated amylin-induced inhibition of feeding and stimulation of thermogenesis in IBAT. No changes were observed when SHU9119 was injected alone. Moreover, amylin significantly increased MC4R expression and c-Fos neuronal signals in the arcuate nucleus and significantly increased acetyl-CoA carboxylase (ACC) phosphorylation in the hypothalamus and IBAT and uncoupling protein-1 (UCP1) expression in the IBAT of WT mice via the MC4R pathway. CONCLUSION: The melanocortin system was involved in amylin-induced suppression of food intake and activation of thermogenesis in both the hypothalamus and IBAT via modulation of ACC phosphorylation and UCP1 expression.


Asunto(s)
Ingestión de Alimentos , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Receptor de Melanocortina Tipo 4/fisiología , Termogénesis/efectos de los fármacos , Termogénesis/genética , Acetil-CoA Carboxilasa/genética , Acetil-CoA Carboxilasa/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/genética , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Melanocortina Tipo 4/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
10.
PLoS One ; 14(1): e0210389, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30629642

RESUMEN

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by impaired social interactions, difficulty with communication, and repetitive behavior patterns. In humans affected by ASD, there is a male pre-disposition towards the condition with a male to female ratio of 4:1. In part due to the complex etiology of ASD including genetic and environmental interplay, there are currently no available medical therapies to improve the social deficits of ASD. Studies in rodent models and humans have shown promising therapeutic effects of oxytocin in modulating social adaptation. One pharmacological approach to stimulating oxytocinergic activity is the melanocortin receptor 4 agonist Melanotan-II (MT-II). Notably the effects of oxytocin on environmental rodent autism models has not been investigated to date. We used a maternal immune activation (MIA) mouse model of autism to assess the therapeutic potential of MT-II on autism-like features in adult male mice. The male MIA mice exhibited autism-like features including impaired social behavioral metrics, diminished vocal communication, and increased repetitive behaviors. Continuous administration of MT-II to male MIA mice over a seven-day course resulted in rescue of social behavioral metrics. Normal background C57 male mice treated with MT-II showed no significant alteration in social behavioral metrics. Additionally, there was no change in anxiety-like or repetitive behaviors following MT-II treatment of normal C57 mice, though there was significant weight loss following subacute treatment. These data demonstrate MT-II as an effective agent for improving autism-like behavioral deficits in the adult male MIA mouse model of autism.


Asunto(s)
Trastorno Autístico/tratamiento farmacológico , Péptidos Cíclicos/uso terapéutico , alfa-MSH/análogos & derivados , Animales , Conducta Animal/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor de Melanocortina Tipo 4/metabolismo , Receptor de Melanocortina Tipo 4/fisiología , Conducta Social , alfa-MSH/uso terapéutico
11.
Acta Physiol (Oxf) ; 225(4): e13222, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30466186

RESUMEN

AIM: We previously demonstrated that central nervous system (CNS) melanocortin 4 receptors (MC4R) play a key role in regulating blood pressure (BP) in some conditions associated with increased SNS activity, including obesity. In this study, we examined whether activation of CNS MC4R contributes to chronic intermittent hypoxia (CIH)-induced hypertension and ventilatory responses to hypercapnia. METHODS: Rats were instrumented with an intracerebroventricular (ICV) cannula in the lateral cerebral ventricle for continuous infusion of MC4R antagonist (SHU-9119) and telemetry probes for measuring mean arterial pressure (MAP) and heart rate (HR). Untreated and SHU-9119-treated rats as well as obese and lean MC4R-deficient rats were exposed to CIH for 7-18 consecutive days. RESULTS: Chronic intermittent hypoxia reduced cumulative food intake by 18 ± 5 g while MAP and HR increased by 10 ± 3 mm Hg and 9 ± 5 bpm in untreated rats. SHU-9119 increased food intake (from 15 ± 1 to 46 ± 3 g) and prevented CIH-induced reduction in food intake. CIH-induced hypertension was not attenuated by MC4R antagonism (average increase of 10 ± 1 vs 9 ± 1 mm Hg for untreated and SHU-9119 treated rats). In obese MC4R-deficient rats, CIH for 7 days raised BP by 11 ± 4 mm Hg. However, when MC4R-deficient rats were food restricted to prevent obesity, CIH-induced hypertension was attenuated by 32%. We also found that MC4R deficiency was associated with impaired ventilatory responses to hypercapnia independently of obesity. CONCLUSION: These results show that obesity and the CNS melanocortin system interact in complex ways to elevate BP during CIH and that MC4R may be important in the ventilatory responses to hypercapnia.


Asunto(s)
Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Obesidad/fisiopatología , Receptor de Melanocortina Tipo 4/fisiología , Sistema Nervioso Simpático/fisiopatología , Animales , Barorreflejo , Glucemia , Presión Sanguínea , Peso Corporal , Ingestión de Alimentos , Frecuencia Cardíaca , Hematócrito , Hipercapnia/complicaciones , Hipoxia/complicaciones , Insulina/sangre , Leptina/sangre , Masculino , Obesidad/complicaciones , Ventilación Pulmonar , Ratas Sprague-Dawley , Ratas Transgénicas
12.
J Neuroendocrinol ; 31(1): e12670, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30561082

RESUMEN

Energy stores in fat tissue are determined in part by the activity of hypothalamic neurones expressing the melanocortin-4 receptor (MC4R). Even a partial reduction in MC4R expression levels in mice, rats or humans produces hyperphagia and morbid obesity. Thus, it is of great interest to understand the molecular basis of neuromodulation by the MC4R. The MC4R is a G protein-coupled receptor that signals efficiently through GαS , and this signalling pathway is essential for normal MC4R function in vivo. However, previous data from hypothalamic slice preparations indicated that activation of the MC4R depolarised neurones via G protein-independent regulation of the ion channel Kir7.1. In the present study, we show that deletion of Kcnj13 (ie, the gene encoding Kir7.1) specifically from MC4R neurones produced resistance to melanocortin peptide-induced depolarisation of MC4R paraventricular nucleus neurones in brain slices, resistance to the sustained anorexic effect of exogenously administered melanocortin peptides, late onset obesity, increased linear growth and glucose intolerance. Some MC4R-mediated phenotypes appeared intact, including Agouti-related peptide-induced stimulation of food intake and MC4R-mediated induction of peptide YY release from intestinal L cells. Thus, a subset of the consequences of MC4R signalling in vivo appears to be dependent on expression of the Kir7.1 channel in MC4R cells.


Asunto(s)
Hipotálamo/fisiopatología , Neuronas/fisiología , Obesidad/fisiopatología , Canales de Potasio de Rectificación Interna/fisiología , Receptor de Melanocortina Tipo 4/fisiología , Animales , Conducta Alimentaria/fisiología , Femenino , Masculino , Potenciales de la Membrana , Ratones Endogámicos C57BL , Ratones Noqueados , Canales de Potasio de Rectificación Interna/genética
13.
J Clin Invest ; 128(7): 3160-3170, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29911992

RESUMEN

It is critical for survival to assign positive or negative valence to salient stimuli in a correct manner. Accordingly, harmful stimuli and internal states characterized by perturbed homeostasis are accompanied by discomfort, unease, and aversion. Aversive signaling causes extensive suffering during chronic diseases, including inflammatory conditions, cancer, and depression. Here, we investigated the role of melanocortin 4 receptors (MC4Rs) in aversive processing using genetically modified mice and a behavioral test in which mice avoid an environment that they have learned to associate with aversive stimuli. In normal mice, robust aversions were induced by systemic inflammation, nausea, pain, and κ opioid receptor-induced dysphoria. In sharp contrast, mice lacking MC4Rs displayed preference or indifference toward the aversive stimuli. The unusual flip from aversion to reward in mice lacking MC4Rs was dopamine dependent and associated with a change from decreased to increased activity of the dopamine system. The responses to aversive stimuli were normalized when MC4Rs were reexpressed on dopamine D1 receptor-expressing cells or in the striatum of mice otherwise lacking MC4Rs. Furthermore, activation of arcuate nucleus proopiomelanocortin neurons projecting to the ventral striatum increased the activity of striatal neurons in an MC4R-dependent manner and elicited aversion. Our findings demonstrate that melanocortin signaling through striatal MC4Rs is critical for assigning negative motivational valence to harmful stimuli.


Asunto(s)
Cuerpo Estriado/fisiología , Motivación/fisiología , Receptor de Melanocortina Tipo 4/fisiología , Animales , Reacción de Prevención/fisiología , Conducta Animal/fisiología , Benzazepinas/administración & dosificación , Cuerpo Estriado/efectos de los fármacos , Dopamina/fisiología , Antagonistas de Dopamina/administración & dosificación , Femenino , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proopiomelanocortina/fisiología , Receptor de Melanocortina Tipo 4/deficiencia , Receptor de Melanocortina Tipo 4/genética , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/fisiología , Recompensa
14.
J Biol Chem ; 293(28): 10993-11005, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29794140

RESUMEN

Central melanocortin 4 receptors (MC4Rs) stimulate energy expenditure and inhibit food intake. MC4Rs activate the G protein Gsα, but whether Gsα mediates all MC4R actions has not been established. Individuals with Albright hereditary osteodystrophy (AHO), who have heterozygous Gsα-inactivating mutations, only develop obesity when the Gsα mutation is present on the maternal allele because of tissue-specific genomic imprinting. Furthermore, evidence in mice implicates Gsα imprinting within the central nervous system (CNS) in this disorder. In this study, we examined the effects of Gsα in MC4R-expressing cells on metabolic regulation. Mice with homozygous Gsα deficiency in MC4R-expressing cells (MC4RGsKO) developed significant obesity with increased food intake and decreased energy expenditure, along with impaired insulin sensitivity and cold-induced thermogenesis. Moreover, the ability of the MC4R agonist melanotan-II (MTII) to stimulate energy expenditure and to inhibit food intake was impaired in MC4RGsKO mice. MTII failed to stimulate the secretion of the anorexigenic hormone peptide YY (PYY) from enteroendocrine L cells, a physiological response mediated by MC4R-Gsα signaling, even though baseline PYY levels were elevated in these mice. In Gsα heterozygotes, mild obesity and reduced energy expenditure were present only in mice with a Gsα deletion on the maternal allele in MC4R-expressing cells, whereas food intake was unaffected. These results demonstrate that Gsα signaling in MC4R-expressing cells is required for controlling energy balance, thermogenesis, and peripheral glucose metabolism. They further indicate that Gsα imprinting in MC4R-expressing cells contributes to obesity in Gsα knockout mice and probably in individuals with Albright hereditary osteodystrophy as well.


Asunto(s)
Metabolismo Energético , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Glucosa/metabolismo , Obesidad/etiología , Receptor de Melanocortina Tipo 4/fisiología , Termogénesis , Animales , Ingestión de Alimentos , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Impresión Genómica , Homocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
Pharmacol Res ; 129: 10-19, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29329999

RESUMEN

It is well known that melanocortin-4 receptors (MC4Rs) and central melanocortin pathways regulate food intake, energy expenditure, and glucose homeostasis. Importantly, MC4R deficiency is the most common monogenic cause of human obesity. Interestingly, MC4Rs expressed by distinct central nuclei are responsible for the different physiological function of MC4R stimulation. In addition, MC4Rs activate multiple intracellular and/or synaptic signaling molecules for the regulation of neuronal circuits. Therefore, MC4Rs and the downstream signal molecules are plausible targets for development of novel therapeutics against obesity and obesity-related metabolic disorders. In this review, we discuss recent findings on the neuronal circuits and signaling molecules that are responsible for MC4R control energy balance and autonomic function. Further, we review status of MC4R agonists as novel therapeutics for obesity syndrome. We believe that comprehensive understanding of signaling molecules involved in MC4R control of neuronal circuits will help to design MC4R agonists as safe and effective anti-obesity drugs.


Asunto(s)
Neuronas/fisiología , Obesidad/tratamiento farmacológico , Receptor de Melanocortina Tipo 4/fisiología , Animales , Humanos , Ligandos , Obesidad/fisiopatología , Receptor de Melanocortina Tipo 4/agonistas , Transmisión Sináptica , Síndrome
16.
Metabolism ; 70: 152-159, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28403939

RESUMEN

BACKGROUND: Brain estrogen receptor-α (ERα) is essential for estrogenic regulation of energy homeostasis and reproduction. We previously showed that ERα expressed by pro-opiomelanocortin (POMC) neurons mediates estrogen's effects on food intake, body weight, negative regulation of hypothalamic-pituitary-gonadal axis (HPG axis) and fertility. RESULTS AND CONCLUSIONS: We report here that global deletion of a key downstream receptor for POMC peptide, the melanocortin 4 receptor (MC4R), did not affect normal negative feedback regulation of estrogen on the HPG axis, estrous cyclicity and female fertility. Furthermore, loss of the MC4R did not influence estrogenic regulation on food intake and body weight. These results indicate that the MC4R is not required for estrogen's effects on metabolic and reproductive functions.


Asunto(s)
Estrógenos , Homeostasis , Receptor de Melanocortina Tipo 4/fisiología , Reproducción , Animales , Peso Corporal , Ingestión de Alimentos , Metabolismo Energético , Retroalimentación Fisiológica , Femenino , Ratones , Ratones Endogámicos C57BL , Embarazo , Proopiomelanocortina
17.
Neuroscience ; 346: 102-112, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28093215

RESUMEN

The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that is expressed in several brain nuclei playing a crucial role in the regulation of energy balance controlling the homeostasis of the organism. It displays both agonist-evoked and constitutive activity, and moreover, it can couple to different G proteins. Most of the research on MC4R has been focused on agonist-induced activity, while the molecular and cellular basis of MC4R constitutive activity remains scarcely studied. We have previously shown that neuronal N-type voltage-gated calcium channels (CaV2.2) are inhibited by MC4R agonist-dependent activation, while the CaV subtypes that carry L- and P/Q-type current are not. Here, we tested the hypothesis that MC4R constitutive activity can affect CaV, with focus on the channel subtypes that can control transcriptional activity coupled to depolarization (L-type, CaV1.2/1.3) and neurotransmitter release (N- and P/Q-type, CaV2.2 and CaV2.1). We found that MC4R constitutive activity inhibits specifically CaV1.2/1.3 and CaV2.1 subtypes of CaV. We also explored the signaling pathways mediating this inhibition, and thus propose that agonist-dependent and basal MC4R activation modes signal differentially through Gs and Gi/o pathways to impact on different CaV subtypes. In addition, we found that chronic incubation with MC4R endogenous inverse agonist, agouti and agouti-related peptide (AgRP), occludes CaV inhibition in a cell line and in amygdaloid complex cultured neurons as well. Thus, we define new mechanisms of control of the main mediators of depolarization-induced calcium entry into neurons by a GPCR that displays constitutive activity.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Neuronas/fisiología , Receptor de Melanocortina Tipo 4/fisiología , Proteína Relacionada con Agouti/administración & dosificación , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología , Animales , Femenino , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor de Melanocortina Tipo 4/agonistas , Transducción de Señal
18.
Reproduction ; 153(3): 267-276, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28003536

RESUMEN

Deletion of the melanocortin-4-receptor (Mc4r) gene in mice causes hyperphagia, followed by hyperinsulinemia, obesity and progressive infertility. Evidence shows that the number of developed corpora lutea is reduced in obese MC4R-knockout (MC4R KO) female mice, but the mechanism is unclear. The effect of hyperphagia and obesity by MC4R KO on pulsatile luteinizing hormone (LH) secretion and ovulation remains unknown. In MC4R KO mice and wild-type littermates (WT LM) during the diestrus period throughout different ages, we examined and monitored their metabolic status, pulsatile LH profiles, follicular morphology and the number of corpora lutea. MC4R KO mice were hyperphagic, obese, hyperglycemic, hyperinsulinemic and demonstrated insulin resistance and hepatic steatosis. Irregular estrous cycles and significant changes in the LH secretion profiles were observed in sexually matured 16- to 28-week MC4R KO mice, without any difference in testosterone levels. In addition, MC4R KO mice at 16 weeks of age had significantly fewer corpora lutea than same age WT LM mice. The ovary examinations of MC4R KO mice at 28 weeks of age showed predominantly antral and preovulatory follicles with no corpora lutea. These findings were consistent with the decrease in total, pulsatile, mass and basal LH releases in MC4R KO mice. The characteristics of hormone profiles in obese MC4R KO mice indicate that MC4R plays an important role in regulating LH release, ovulation and reproductive ability probably via hyperphagia-induced obesity. Further study of correlation between metabolic and reproductive regulatory hormones is warranted to dissect the pathological mechanism underlying obesity-induced infertility.Free Chinese abstract: A Chinese translation of this abstract is freely available at http://www.reproduction-online.org/content/153/3/267/suppl/DC1.


Asunto(s)
Cuerpo Lúteo/fisiopatología , Hígado Graso/patología , Hormona Luteinizante/metabolismo , Sistemas Neurosecretores , Receptor de Melanocortina Tipo 4/fisiología , Reproducción/fisiología , Animales , Hígado Graso/etiología , Hígado Graso/metabolismo , Femenino , Hiperglucemia/etiología , Hiperglucemia/patología , Hiperinsulinismo/etiología , Hiperinsulinismo/patología , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovario/fisiopatología , Ovulación/fisiología
19.
Nat Neurosci ; 19(2): 206-19, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26814590

RESUMEN

The melanocortin system provides a conceptual blueprint for the central control of energetic state. Defined by four principal molecular components--two antagonistically acting ligands and two cognate receptors--this phylogenetically conserved system serves as a prototype for hierarchical energy balance regulation. Over the last decade the application of conditional genetic techniques has facilitated the neuroanatomical dissection of the melanocortinergic network and identified the specific neural substrates and circuits that underscore the regulation of feeding behavior, energy expenditure, glucose homeostasis and autonomic outflow. In this regard, the melanocortin-4 receptor is a critical coordinator of mammalian energy homeostasis and body weight. Drawing on recent advances in neuroscience and genetic technologies, we consider the structure and function of the melanocortin-4 receptor circuitry and its role in energy homeostasis.


Asunto(s)
Metabolismo Energético/genética , Metabolismo Energético/fisiología , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/fisiología , Animales , Peso Corporal/fisiología , Ingestión de Energía/genética , Ingestión de Energía/fisiología , Conducta Alimentaria/fisiología , Homeostasis/genética , Homeostasis/fisiología , Humanos
20.
J Clin Invest ; 126(1): 40-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26595811

RESUMEN

Activation of brain melanocortin 4 receptors (MC4Rs) leads to reduced food intake, increased energy expenditure, increased insulin sensitivity, and reduced linear growth. MC4R effects on energy expenditure and glucose metabolism are primarily mediated by the G protein G(s)α in brain regions outside of the paraventricular nucleus of the hypothalamus (PVN). However, the G protein(s) that is involved in MC4R-mediated suppression of food intake and linear growth, which are believed to be regulated primarily though action in the PVN, is unknown. Here, we show that PVN-specific loss of G(q)α and G11α, which stimulate PLC, leads to severe hyperphagic obesity, increased linear growth, and inactivation of the hypothalamic-pituitary-adrenal axis, without affecting energy expenditure or glucose metabolism. Moreover, we demonstrate that the ability of an MC4R agonist delivered to PVN to inhibit food intake is lost in mice lacking G(q/11)α in the PVN but not in animals deficient for G(s)α. The blood pressure response to the same MC4R agonist was only lost in animals lacking G(s)α specifically in the PVN. Together, our results exemplify how different physiological effects of GPCRs may be mediated by different G proteins and identify a pathway for appetite regulation that could be selectively targeted by G(q/11)α-biased MC4R agonists as a potential treatment for obesity.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Receptor de Melanocortina Tipo 4/fisiología , Animales , Colesterol/metabolismo , Femenino , Sistema Hipotálamo-Hipofisario/fisiología , Resistencia a la Insulina , Melanocortinas/farmacología , Ratones , Ratones Noqueados , Obesidad/etiología , Sistema Hipófiso-Suprarrenal/fisiología , Receptor de Melanocortina Tipo 4/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...