Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
1.
Obes Facts ; 16(5): 514-518, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37393902

RESUMEN

Leptin receptor (LEPR) deficiency is a rare genetic disorder that affects the body's ability to regulate appetite and weight. For patients and their families, the disorder seriously disrupts daily life; however, little is published about this impact. We here report the experiences of a 10.5-year-old girl with leptin receptor deficiency and her family. The diagnosis of this rare genetic obesity had a deep impact on the life of the child and her family. It led to a better understanding of the cause of the impaired appetite regulation and early-onset obesity with subsequently less judgement by others and improved cooperation of their social network and school on maintaining a healthy lifestyle for this girl. A strict eating regimen and lifestyle measures resulted in the first year after diagnosis in a significantly decreased body mass index (BMI), followed by BMI stabilization, still categorized as obesity class three. However, the troublesome challenge of how to manage the disruptive behaviour due to hyperphagia remained. Eventually, due to treatment with targeted pharmacotherapy, i.e., melanocortin-4 receptor agonists, her BMI continued to decrease due to resolving hyperphagia. The daily routine of the family and the atmosphere at home positively changed as they were no longer dominated by the food-focused behaviour of the child and the adherence to the strict eating regimen. This case report demonstrates the importance and impact of a rare genetic obesity disorder diagnosis in a family. Additionally, it highlights the value of genetic testing in patients with a high suspicion of a genetic obesity disorder as it can eventually lead to personalized treatment, such as guidance by specialized healthcare professionals and educated caregivers or targeted pharmacotherapy.


Asunto(s)
Errores Innatos del Metabolismo , Medicina de Precisión , Humanos , Niño , Femenino , Receptores de Leptina/genética , Obesidad/complicaciones , Obesidad/genética , Obesidad/tratamiento farmacológico , Hiperfagia/complicaciones , Hiperfagia/genética , Índice de Masa Corporal , Leptina/uso terapéutico , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/agonistas
2.
J Biomol Struct Dyn ; 41(24): 15411-15420, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37126536

RESUMEN

Obesity has emerged as a global issue, but with the complex structures of multiple related important targets and their agonists or antagonists determined, the mechanism of ligand-protein interaction may offer new chances for developing new generation agonists anti-obesity. Based on the molecule surface of the cryo-EM protein structure 7AUE, we tried to replace D-Ala3 with D-Met in setmelanotide as the linker site for fragment-growing with De novo evolution. The simulation results indicate that the derivatives could improve the binding abilities with the melanocortin 4 receptor and the selectivity over the melanocortin 1 receptor. The improved selectivity of the newly designed derivatives is mainly due to the shape difference of the molecular surface at the orthosteric peptide-binding pocket between melanocortin 4 receptor and melanocortin 1 receptor. The new extended fragments could not only enhance the binding affinities but also function as a gripper to seize the pore, making it easier to balance and stabilize the other component of the new derivatives. Although it is challenging to synthesize the compounds designed in silico, this study may perhaps serve as a trigger for additional anti-obesity research.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Receptor de Melanocortina Tipo 1 , Receptor de Melanocortina Tipo 4 , Humanos , Simulación del Acoplamiento Molecular , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/metabolismo , alfa-MSH/química , alfa-MSH/metabolismo , Obesidad
3.
Genet Med ; 25(7): 100857, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37092539

RESUMEN

PURPOSE: Recessive deficiency of proopiomelanocortin (POMC) causes childhood-onset severe obesity. Cases can now benefit from the melanocortin 4 receptor agonist setmelanotide. Furthermore, a phase 3 clinical trial is evaluating setmelanotide in heterozygotes for POMC. We performed a large-scale genetic analysis to assess the effect of heterozygous, pathogenic POMC variants on obesity. METHODS: A genetic analysis was performed in a family including 2 cousins with childhood-onset obesity. We analyzed the obesity status of heterozygotes for pathogenic POMC variants in the Human Gene Mutation Database. The association between heterozygous pathogenic POMC variants and obesity risk was assessed using 190,000 exome samples from UK Biobank. RESULTS: The 2 cousins carried a compound heterozygous pathogenic variant in POMC. Six siblings were heterozygotes; only 1 of them had obesity. In Human Gene Mutation Database, we identified 60 heterozygotes for pathogenic POMC variants, of whom 14 had obesity. In UK Biobank, heterozygous pathogenic POMC variants were not associated with obesity risk, but they modestly increased body mass index levels. CONCLUSION: Heterozygous pathogenic POMC variants do not contribute to monogenic obesity, but they slightly increase body mass index. Setmelanotide use in patients with obesity, which would only be based on the presence of a heterozygous POMC variant, can be questioned.


Asunto(s)
Obesidad Infantil , Proopiomelanocortina , Niño , Humanos , Índice de Masa Corporal , Heterocigoto , Mutación , Obesidad/genética , Obesidad Infantil/genética , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/agonistas , Fármacos Antiobesidad/uso terapéutico
4.
Neuropeptides ; 96: 102289, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36155088

RESUMEN

RATIONALE: Obesity is a major health problem worldwide. An understanding of the factors that drive feeding behaviors is key to the development of pharmaceuticals to decrease appetite and consumption. Proopiomelanocortin (POMC), the melanocortin peptide precursor, is essential in the regulation of body weight and ingestive behaviors. Deletion of POMC or impairment of melanocortin signaling in the brain results in hyperphagic obesity. Neurons in the hypothalamic arcuate nucleus produce POMC and project to many areas including the nucleus accumbens (NAcc), which is well established in the rewarding and reinforcing effects of both food and drugs of abuse. OBJECTIVE: These studies sought to determine the role of melanocortins in the NAcc on consumption of and motivation to obtain access to standard rodent chow. METHODS: Male, C57BL/6J mice were microinjected bilaterally into the NAcc (100 nl/side) with the melanocortin receptor 3/4 agonist melanotan-II (MT-II; 0.1, 0.3, and 1 nmol), and ingestive behaviors were examined in both home cage and operant food self-administration experiments. In addition, the ability of MT-II in the NAcc to produce aversive properties or affect metabolic rate were tested. RESULTS: MT-II injected into the NAcc significantly decreased consumption in both home cage and operant paradigms, and furthermore decreased appetitive responding to gain access to food. There was no development of conditioned taste avoidance or change in metabolic parameters following anorexic doses of MT-II. CONCLUSIONS: MT-II in the NAcc decreased both the motivation to eat and the amount of food consumed without inducing an aversive state or affecting metabolic rate, suggesting a role for melanocortin signaling in the NAcc that is selective for appetite and satiety without affecting metabolism or producing an aversive state.


Asunto(s)
Núcleo Accumbens , Péptidos Cíclicos , Proopiomelanocortina , Receptor de Melanocortina Tipo 4 , alfa-MSH , Animales , Masculino , Ratones , Melanocortinas/metabolismo , Ratones Endogámicos C57BL , Obesidad , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/agonistas , Péptidos Cíclicos/farmacología , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
5.
Life Sci ; 307: 120857, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-35931197

RESUMEN

AIMS: Melanocortin 4 receptor (MC4R) has a well-established role in regulating appetite, food intake and energy homeostasis. Setmelanotide is an MC4R agonist currently approved for weight loss in obese adults and children with mutations in components of the leptin-melanocortin pathway. This study aims to compare structural and functional aspects of the physiological MC4R agonist α-melanocyte-stimulating hormone (α-MSH) with setmelanotide. We also aim to show the binding affinity of setmelanotide to known MC4R human missense mutations associated with obesity. MAIN METHODS: AutoDock Vina was used in the structural analysis to calculate induced fit docking scores of ligand binding to MC4R wild type or the selected variants. HEK293-MC4R were utilized in the functional analysis of MC4R-actiavted pathways upon stimulating with α-MSH or setmelanotide. KEY FINDINGS: Our data shows that setmelanotide has a higher potency for cAMP formation and a weaker effect on ERK1/2 phosphorylation when compared to α-MSH indicating functional selectivity otherwise known as biased agonism. We also present structural data showing that setmelanotide has a higher binding affinity to MC4R compared to α-MSH. Lastly, we show that two loss-of-function and two gain-of-function MC4R variants change the conformation not only of the ligand binding pocket of the receptor but also of the peptide when bound to the receptor because the interaction network and the residues involved in the binding are altered. SIGNIFICANCE: Taken together, our study provides important insights into the diversity of MC4R signaling pathways which will facilitate the development of personalized anti-obesity drugs via refining MC4R agonists.


Asunto(s)
Fármacos Antiobesidad , Receptor de Melanocortina Tipo 4 , Adulto , Fármacos Antiobesidad/farmacología , Niño , Células HEK293 , Humanos , Leptina , Ligandos , Melanocortinas , Obesidad/metabolismo , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
6.
s.l; CONETEC; 9 jun. 2022.
No convencional en Español | BRISA/RedTESA | ID: biblio-1381050

RESUMEN

INTRODUCCIÓN: El sobrepeso y la obesidad se definen como una acumulación anormal o excesiva de grasa que puede ser perjudicial para la salud.1 En el caso de los adultos, la Organización Mundial de la Salud (OMS) define el sobrepeso a un Índice de Masa Corporal (IMC) igual o superior a 25, y a la obesidad a un IMC igual o superior a 30. En el caso de los niños de 5 a 19 años, el sobrepeso se define con un IMC para la edad con más de una desviación típica por encima de la mediana establecida en los patrones de crecimiento infantil de la OMS, y la obesidad es mayor que dos desviaciones típicas para dichos patrones. En algunos casos, la obesidad es causada por trastornos genéticos en la vía leptinamelanocortina, el sistema hipotalámico que controla el gasto de energía y la ingesta de alimentos. La hormona anorexígena leptina es secretada principalmente por el tejido adiposo y refleja las reservas de energía del cuerpo. La señalización de leptina hipotalámica conduce a la activación del receptor de melanocortina-4 (MC4R), lo que resulta en un mayor gasto de energía y saciedad. Cuando se altera esta señalización, los pacientes desarrollan hiperfagia y obesidad de aparición temprana. 1,2 En el estado de alimentación, la leptina estimula la producción de propiomelacortina (POMC), que es procesada por la pro proteína convertasa subtilisina y kexina tipo 1 (PCSK1) en péptidos de melanocortina (hormona estimulante de melanocitos α [α-MSH] y ß-MSH) que se unen y activan MC4R, por lo tanto reducir la ingesta de alimentos. Entre estos trastornos se destacan la deficiencia de POMC o PCSK1 y la deficiencia del receptor de leptina (LEPR) que se caracterizan por una obesidad severa que comienza a una edad temprana y se hereda con un patrón autosómico recesivo. TECNOLOGÍA: Setmelanotide (IMCIVREE™) es un agonista del receptor melanocortina-4 (MC4). Es un péptido cíclico de 8 aminoácidos análogo de la hormona estimuladora de melanocitos alfa ligando del receptor MC4 endógeno (α-MSH). Las hormonas reguladoras derivadas de la periferia (por ejemplo, la leptina) estimulan la expresión de α-MSH a través de POMC, que regula el hambre, la saciedad y el gasto de energía al unirse a los receptores MC4 en el hipotálamo. OBJETIVO: El objetivo del presente informe es evaluar rápidamente los parámetros de eficacia, seguridad, costos y recomendaciones disponibles acerca del empleo de setmelanotide para el tratamiento de la obesidad. MÉTODOS: Se realizó una búsqueda bibliográfica en las principales bases de datos tales como PUBMED, LILACS, BRISA, COCHRANE, SCIELO, EMBASE, TRIPDATABASE como así también en sociedades científicas, agencias reguladoras, financiadores de salud y agencias de evaluación de tecnologías sanitarias. Se priorizó la inclusión de revisiones sistemáticas, ensayos clínicos controlados aleatorizados, evaluación de tecnología sanitaria y guías de práctica clínica de alta calidad metodológica. RECOMENDACIONES: No se hallaron guías de práctica clínica actualizadas en Argentina y en el Mundo que recomienden la tecnología en la indicación evaluada. La guía de práctica clínica de la Asociación Estadounidense de Diabetes publicada en 2022 menciona la tecnología, aunque no la recomienda, entre las alternativas terapéuticas para el manejo y prevención de la obesidad y control de peso en pacientes con diabetes. CONCLUSIONES: La evidencia que sustenta la aprobación de comercialización de setmelanotide (IMCIVREE™) para el control crónico del peso en personas mayores de 6 años de edad con obesidad debida a la deficiencia de POMC, PCSK1 o LEPR por parte de las agencias regulatorias relevadas, se basa en un solo estudio de Fase III abierto y no controlado en pocos pacientes de países del hemisferio norte. Este estudio demostraría que setmelanotide, junto a medidas saludables, alcanzarían las metas de pérdida de peso propuestas en personas con obesidad severa al mediano plazo; sin embargo, esto no se traduciría en mejoras en estudios de laboratorio relevantes para el seguimiento y existe incertidumbre sobre su efecto en la calidad de vida reportada por estos pacientes. Las agencias regulatorias relevadas han autorizado recientemente la comercialización de setmelanotide en pacientes muy seleccionados mediante estudios genéticos, y bajo criterios de mantenimientos específicos. No se hallaron guías de práctica clínica actualizadas en Argentina y en el Mundo que recomienden la tecnología en la indicación evaluada. No se hallaron evaluaciones económicas publicadas, aunque el costo del fármaco es elevado.


Asunto(s)
Humanos , Receptor de Melanocortina Tipo 4/agonistas , Obesidad/tratamiento farmacológico , Argentina , Eficacia , Análisis Costo-Beneficio/economía
7.
Front Immunol ; 13: 1082036, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36703981

RESUMEN

Introduction: Alzheimer's disease (AD) is the most frequent cause of dementia and still lacks effective therapy. Clinical signs of AD include low levels of endogenous melanocortins (MCs) and previous studies have shown that treatment with MC analogs induces neuroprotection in the early stages of AD. Methods: We investigated the neuroprotective role of MCs in two transgenic mouse models of severe AD using 5 and 7 month-old (mo) 5XFAD mice and 9 and 12 mo 3xTg mice. These mice were subjected to a chronic stimulation of MC receptors (MCRs) with MC analogue Nle4-D-Phe7-α-melanocyte stimulating hormone (NDP-α-MSH, 340 µg/kg, i.p.). Mouse behavior and ex-vivo histological and biochemical analyses were performed after 50 days of treatment. Results: Our analysis demonstrated an improvement in cognitive abilities of AD mice at late stage of AD progression. We also showed that these protective effects are associated with decreased levels of hyperphosphorylated Tau but not with Aß burden, that was unaffected in the hippocampus and in the cortex of AD mice. In addition, an age-dependent NDP effect on glial reactivity was observed only in 3xTg mice whereas a global downregulation of p38 mitogen-activated protein kinase was selectively observed in 7 mo 5XFAD and 14 mo 3xTg mice. Conclusion: Our results suggest that MCR stimulation by NDP-α-MSH could represent a promising therapeutic strategy in managing cognitive decline also at late stage of AD, whereas the effects on neuroinflammation may be restricted to specific stages of AD progression.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Receptor de Melanocortina Tipo 4 , Animales , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Cognición , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Ratones Transgénicos , Receptor de Melanocortina Tipo 4/agonistas
8.
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
10.
Front Endocrinol (Lausanne) ; 12: 690387, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421821

RESUMEN

The molecular sensors underlying nutrient-stimulated GLP-1 secretion are currently being investigated. Peripheral administration of melanocortin-4 receptor (MC4R) agonists have been reported to increase GLP-1 plasma concentrations in mice and humans but it is unknown whether this effect results from a direct effect on the GLP-1 secreting L-cells in the intestine, from other effects in the intestine or from extra-intestinal effects. We investigated L-cell expression of MC4R in mouse and human L-cells by reanalyzing publicly available RNA sequencing databases (mouse and human) and by RT-qPCR (mouse), and assessed whether administration of MC4R agonists to a physiologically relevant gut model, isolated perfused mouse and rat small intestine, would stimulate GLP-1 secretion or potentiate glucose-stimulated secretion. L-cell MC4R expression was low in mouse duodenum and hardly detectable in the ileum and MC4R expression was hardly detectable in human L-cells. In isolated perfused mouse and rat intestine, neither intra-luminal nor intra-arterial administration of NDP-alpha-MSH, a potent MC4R agonist, had any effect on GLP-1 secretion (P ≥0.98, n = 5-6) from the upper or lower-half of the small intestine in mice or in the lower half in rats. Furthermore, HS014-an often used MC4R antagonist, which we found to be a partial agonist-did not affect the glucose-induced GLP-1 response in the rat, P = 0.62, n = 6). Studies on transfected COS7-cells confirmed bioactivity of the used compounds and that concentrations employed were well within in the effective range. Our combined data therefore suggest that MC4R-activated GLP-1 secretion in rodents either exclusively occurs in the colon or involves extra-intestinal signaling.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Intestino Delgado/metabolismo , Células L/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Células COS , Chlorocebus aethiops , Bases de Datos Factuales , Humanos , Intestino Delgado/efectos de los fármacos , Células L/efectos de los fármacos , Masculino , Ratones , Ratas , Ratas Wistar , Receptor de Melanocortina Tipo 4/agonistas , Transducción de Señal/efectos de los fármacos , alfa-MSH/farmacología
11.
Science ; 372(6544): 808-814, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33858992

RESUMEN

Obesity is a global epidemic that causes morbidity and impaired quality of life. The melanocortin receptor 4 (MC4R) is at the crux of appetite, energy homeostasis, and body-weight control in the central nervous system and is a prime target for anti-obesity drugs. Here, we present the cryo-electron microscopy (cryo-EM) structure of the human MC4R-Gs signaling complex bound to the agonist setmelanotide, a cyclic peptide recently approved for the treatment of obesity. The work reveals the mechanism of MC4R activation, highlighting a molecular switch that initiates satiation signaling. In addition, our findings indicate that calcium (Ca2+) is required for agonist, but not antagonist, efficacy. These results fill a gap in the understanding of MC4R activation and could guide the design of future weight-management drugs.


Asunto(s)
Fármacos Antiobesidad/química , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/química , Saciedad , alfa-MSH/análogos & derivados , Fármacos Antiobesidad/farmacología , Apetito , Sitios de Unión , Calcio/química , Calcio/fisiología , Microscopía por Crioelectrón , Diseño de Fármacos , Células HEK293 , Humanos , Ligandos , Mutación , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Conformación Proteica en Hélice alfa , Dominios Proteicos , Receptor de Melanocortina Tipo 4/genética , Transducción de Señal , alfa-MSH/química , alfa-MSH/farmacología
12.
Gastroenterology ; 161(2): 536-547.e2, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33848536

RESUMEN

OBJECTIVE: Hypothalamic melanocortin 4 receptors (MC4R) are a key regulator of energy homeostasis. Brain-penetrant MC4R agonists have failed, as concentrations required to suppress food intake also increase blood pressure. However, peripherally located MC4R may also mediate metabolic benefits of MC4R activation. Mc4r transcript is enriched in mouse enteroendocrine L cells and peripheral administration of the endogenous MC4R agonist, α-melanocyte stimulating hormone (α-MSH), triggers the release of the anorectic hormones Glucagon-like peptide-1 (GLP-1) and peptide tyrosine tyrosine (PYY) in mice. This study aimed to determine whether pathways linking MC4R and L-cell secretion exist in humans. DESIGN: GLP-1 and PYY levels were assessed in body mass index-matched individuals with or without loss-of-function MC4R mutations following an oral glucose tolerance test. Immunohistochemistry was performed on human intestinal sections to characterize the mucosal MC4R system. Static incubations with MC4R agonists were carried out on human intestinal epithelia, GLP-1 and PYY contents of secretion supernatants were assayed. RESULTS: Fasting PYY levels and oral glucose-induced GLP-1 secretion were reduced in humans carrying a total loss-of-function MC4R mutation. MC4R was localized to L cells and regulates GLP-1 and PYY secretion from ex vivo human intestine. α-MSH immunoreactivity in the human intestinal epithelia was predominantly localized to L cells. Glucose-sensitive mucosal pro-opiomelanocortin cells provide a local source of α-MSH that is essential for glucose-induced GLP-1 secretion in small intestine. CONCLUSION: Our findings describe a previously unidentified signaling nexus in the human gastrointestinal tract involving α-MSH release and MC4R activation on L cells in an autocrine and paracrine fashion. Outcomes from this study have direct implications for targeting mucosal MC4R to treat human metabolic disorders.


Asunto(s)
Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Mucosa Intestinal/metabolismo , Péptido YY/metabolismo , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , alfa-MSH/metabolismo , Comunicación Autocrina , Glucemia/metabolismo , Estudios de Casos y Controles , Células Enteroendocrinas/efectos de los fármacos , Glucosa/administración & dosificación , Prueba de Tolerancia a la Glucosa , Humanos , Mucosa Intestinal/efectos de los fármacos , Mutación con Pérdida de Función , Comunicación Paracrina , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/genética , Vías Secretoras , Transducción de Señal , Factores de Tiempo , alfa-MSH/farmacología
13.
Nat Commun ; 12(1): 2057, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824339

RESUMEN

Lipocalin 2 (LCN2) was recently identified as an endogenous ligand of the type 4 melanocortin receptor (MC4R), a critical regulator of appetite. However, it remains unknown if this molecule influences appetite during cancer cachexia, a devastating clinical entity characterized by decreased nutrition and progressive wasting. We demonstrate that LCN2 is robustly upregulated in murine models of pancreatic cancer, its expression is associated with reduced food consumption, and Lcn2 deletion is protective from cachexia-anorexia. Consistent with LCN2's proposed MC4R-dependent role in cancer-induced anorexia, pharmacologic MC4R antagonism mitigates cachexia-anorexia, while restoration of Lcn2 expression in the bone marrow is sufficient in restoring the anorexia feature of cachexia. Finally, we observe that LCN2 levels correlate with fat and lean mass wasting and is associated with increased mortality in patients with pancreatic cancer. Taken together, these findings implicate LCN2 as a pathologic mediator of appetite suppression during pancreatic cancer cachexia.


Asunto(s)
Apetito , Caquexia/complicaciones , Lipocalina 2/metabolismo , Neoplasias Pancreáticas/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anorexia/sangre , Anorexia/complicaciones , Barrera Hematoencefálica/patología , Médula Ósea/patología , Caquexia/sangre , Línea Celular Tumoral , Modelos Animales de Enfermedad , Conducta Alimentaria , Femenino , Eliminación de Gen , Humanos , Lipocalina 2/sangre , Masculino , Ratones Noqueados , Persona de Mediana Edad , Modelos Biológicos , Músculos/patología , Neutrófilos/patología , Tamaño de los Órganos , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/genética , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/metabolismo , Regulación hacia Arriba
14.
Mol Metab ; 48: 101206, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33684608

RESUMEN

BACKGROUND: Over the past 20 years, insights from human and mouse genetics have illuminated the central role of the brain leptin-melanocortin pathway in controlling mammalian food intake, with genetic disruption resulting in extreme obesity, and more subtle polymorphic variations influencing the population distribution of body weight. At the end of 2020, the U.S. Food and Drug Administration (FDA) approved setmelanotide, a melanocortin 4 receptor agonist, for use in individuals with severe obesity due to either pro-opiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) deficiency. SCOPE OF REVIEW: Herein, we chart the melanocortin pathway's history, explore its pharmacology, genetics, and physiology, and describe how a neuropeptidergic circuit became an important druggable obesity target. MAJOR CONCLUSIONS: Unravelling the genetics of the subset of severe obesity has revealed the importance of the melanocortin pathway in appetitive control; coupling this with studying the molecular pharmacology of compounds that bind melanocortin receptors has brought a new obesity drug to the market. This process provides a drug discovery template for complex disorders, which for setmelanotide took 25 years to transform from a single gene into an approved drug.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Homeostasis/efectos de los fármacos , Melanocortinas/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Receptor de Melanocortina Tipo 4/agonistas , Transducción de Señal/efectos de los fármacos , alfa-MSH/análogos & derivados , Animales , Fármacos Antiobesidad/farmacología , Aprobación de Drogas/historia , Descubrimiento de Drogas/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Ratones , Obesidad/epidemiología , Receptor de Melanocortina Tipo 4/metabolismo , Estados Unidos/epidemiología , alfa-MSH/farmacología , alfa-MSH/uso terapéutico
15.
Lancet Diabetes Endocrinol ; 8(12): 960-970, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33137293

RESUMEN

BACKGROUND: The melanocortin 4 receptor (MC4R), a component of the leptin-melanocortin pathway, plays a part in bodyweight regulation. Severe early-onset obesity can be caused by biallelic variants in genes that affect the MC4R pathway. We report the results from trials of the MC4R agonist setmelanotide in individuals with severe obesity due to either pro-opiomelanocortin (POMC) deficiency obesity or leptin receptor (LEPR) deficiency obesity. METHODS: These single-arm, open-label, multicentre, phase 3 trials were done in ten hospitals across Canada, the USA, Belgium, France, Germany, the Netherlands, and the UK. Participants aged 6 years or older with POMC or LEPR deficiency obesity received open-label setmelanotide for 12 weeks. Participants with at least 5 kg weight loss (or ≥5% if weighing <100 kg at baseline) entered an 8-week placebo-controlled withdrawal sequence (including 4 weeks each of blinded setmelanotide and placebo treatment) followed by 32 additional weeks of open-label treatment. The primary endpoint, which was assessed in participants who received at least one dose of study medication and had a baseline assessment (full analysis set), was the proportion of participants with at least 10% weight loss compared with baseline at approximately 1 year. A key secondary endpoint was mean percentage change in the most hunger score of the 11-point Likert-type scale at approximately 1 year on the therapeutic dose, which was assessed in a subset of participants aged 12 years or older in the full analysis set who demonstrated at least 5 kg weight loss (or ≥5% in paediatric participants if baseline bodyweight was <100 kg) over the 12-week open-label treatment phase and subsequently proceeded into the placebo-controlled withdrawal sequence, regardless of later disposition. These studies are registered with ClinicalTrials.gov, NCT02896192 and NCT03287960. FINDINGS: Between Feb 14, 2017, and Sept 7, 2018, ten participants were enrolled in the POMC trial and 11 participants were enrolled in the LEPR trial, and included in the full analysis and safety sets. Eight (80%) participants in the POMC trial and five (45%) participants in the LEPR trial achieved at least 10% weight loss at approximately 1 year. The mean percentage change in the most hunger score was -27·1% (n=7; 90% CI -40·6 to -15·0; p=0·0005) in the POMC trial and -43·7% (n=7; -54·8 to -29·1; p<0·0001) in the LEPR trial. The most common adverse events were injection site reaction and hyperpigmentation, which were reported in all ten participants in the POMC trial; nausea was reported in five participants and vomiting in three participants. In the LEPR trial, the most commonly reported treatment-related adverse events were injection site reaction in all 11 participants, skin disorders in five participants, and nausea in four participants. No serious treatment-related adverse events occurred in both trials. INTERPRETATION: Our results support setmelanotide for the treatment of obesity and hyperphagia caused by POMC or LEPR deficiency. FUNDING: Rhythm Pharmaceuticals.


Asunto(s)
Insuficiencia Suprarrenal/complicaciones , Fármacos Antiobesidad/uso terapéutico , Obesidad/tratamiento farmacológico , Proopiomelanocortina/deficiencia , Receptor de Melanocortina Tipo 4/agonistas , Receptores de Leptina/deficiencia , alfa-MSH/análogos & derivados , Adolescente , Adulto , Niño , Método Doble Ciego , Femenino , Estudios de Seguimiento , Humanos , Masculino , Obesidad/complicaciones , Obesidad/etiología , Obesidad/patología , Pronóstico , Adulto Joven , alfa-MSH/uso terapéutico
16.
Curr Biol ; 30(23): 4579-4593.e7, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32976803

RESUMEN

Locomotion requires energy, yet animals need to increase locomotion in order to find and consume food in energy-deprived states. While such energy homeostatic coordination suggests brain origin, whether the central melanocortin 4 receptor (Mc4r) system directly modulates locomotion through motor circuits is unknown. Here, we report that hypothalamic Pomc neurons in zebrafish and mice have long-range projections into spinal cord regions harboring Mc4r-expressing V2a interneurons, crucial components of the premotor networks. Furthermore, in zebrafish, Mc4r activation decreases the excitability of spinal V2a neurons as well as swimming and foraging, while systemic or V2a neuron-specific blockage of Mc4r promotes locomotion. In contrast, in mice, electrophysiological recordings revealed that two-thirds of V2a neurons in lamina X are excited by the Mc4r agonist α-MSH, and acute inhibition of Mc4r signaling reduces locomotor activity. In addition, we found other Mc4r neurons in spinal lamina X that are inhibited by α-MSH, which is in line with previous studies in rodents where Mc4r agonists reduced locomotor activity. Collectively, our studies identify spinal V2a interneurons as evolutionary conserved second-order neurons of the central Mc4r system, providing a direct anatomical and functional link between energy homeostasis and locomotor control systems. The net effects of this modulatory system on locomotor activity can vary between different vertebrate species and, possibly, even within one species. We discuss the biological sense of this phenomenon in light of the ambiguity of locomotion on energy balance and the different living conditions of the different species.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Interneuronas/metabolismo , Locomoción/fisiología , Proopiomelanocortina/metabolismo , Médula Espinal/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Núcleo Arqueado del Hipotálamo/citología , Evolución Biológica , Fenómenos Electrofisiológicos/efectos de los fármacos , Ratones , Modelos Animales , Red Nerviosa/fisiología , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Pez Cebra , Proteínas de Pez Cebra/agonistas , Proteínas de Pez Cebra/genética
17.
Neurotox Res ; 38(2): 421-433, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32378055

RESUMEN

Excessive alcohol intake affects hippocampal function and neuronal communication through oxidative stress and mitochondrial impairment. Previous studies have suggested that the melanocortin system (MCS) plays an essential role in alcohol consumption and addiction. The MCS is a hypothalamic region involved in regulating inflammatory processes in the brain, and its pharmacological activation through the melanocortin-4 receptor (MC4R) reduces both alcohol consumption and the neuroinflammatory responses in the brain. However, the cellular mechanisms involved in the beneficial actions of MCS against ethanol toxicity are not entirely understood. The objective of this study was to investigate the protective role of the MC4R pharmacological activator RO27-3225 on oxidative damage and mitochondrial impairment present in hippocampal neuronal cultures acutely exposed to ethanol (50, 75 mM, 24 h). Pre-treatment with RO27-3225 (250 nM, 1 h) prevented reactive oxygen species (ROS) increase, dysregulation of cytosolic calcium homeostasis, and mitochondrial potential loss induced by ethanol. Improvement of mitochondrial failure produced by RO27-3225 was accompanied by a significant increase in ATP production in ethanol-treated neurons. More importantly, RO27-3225 promoted the activation of the antioxidant pathway Nrf-2, demonstrated by an increase in the expression and nuclear translocation of Nrf-2, and upregulation of mRNA levels of NAD(P)H quinone oxidoreductase 1 (NQO1), an antioxidant enzyme which expression is activated by this pathway. These results suggest that the stimulation of MC4R prevents oxidative damage and mitochondrial stress induced by ethanol through the activation of the Nrf-2 pathway in cultured hippocampal neurons. These results are novel and demonstrate the critical function of MC4R in promoting antioxidant defense and reducing mitochondrial damage produced by ethanol in the brain.


Asunto(s)
Calcio/metabolismo , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/fisiología , Péptidos/farmacología , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Antioxidantes , Células Cultivadas , Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Hipocampo/citología , Inflamación/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/genética , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptor de Melanocortina Tipo 4/agonistas
18.
Mol Metab ; 35: 100956, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32244183

RESUMEN

OBJECTIVE: Histaminergic neurons of the tuberomammillary nucleus (TMN) are wake-promoting and contribute to the regulation of energy homeostasis. Evidence indicates that melanocortin 4 receptors (MC4R) are expressed within the TMN. However, whether the melanocortin system influences the activity and function of TMN neurons expressing histidine decarboxylase (HDC), the enzyme required for histamine synthesis, remains undefined. METHODS: We utilized Hdc-Cre mice in combination with whole-cell patch-clamp electrophysiology and in vivo chemogenetic techniques to determine whether HDC neurons receive metabolically relevant information via the melanocortin system. RESULTS: We found that subsets of HDC-expressing neurons were excited by melanotan II (MTII), a non-selective melanocortin receptor agonist. Use of melanocortin receptor selective agonists (THIQ, [D-Trp8]-γ-MSH) and inhibitors of synaptic transmission (TTX, CNQX, AP5) indicated that the effect was mediated specifically by MC4Rs and involved a glutamatergic dependent presynaptic mechanism. MTII enhanced evoked excitatory post-synaptic currents (EPSCs) originating from electrical stimulation of the perifornical lateral hypothalamic area (PeFLH), supportive of melanocortin effects on the glutamatergic PeFLH projection to the TMN. Finally, in vivo chemogenetic inhibition of HDC neurons strikingly enhanced the anorexigenic effects of intracerebroventricular administration of MTII, suggesting that MC4R activation of histaminergic neurons may restrain the anorexigenic effects of melanocortin system activation. CONCLUSIONS: These experiments identify a functional interaction between the melanocortin and histaminergic systems and suggest that HDC neurons act naturally to restrain the anorexigenic effect of melanocortin system activation. These findings may have implications for the control of arousal and metabolic homeostasis, especially in the context of obesity, in which both processes are subjected to alterations.


Asunto(s)
Histamina/metabolismo , Histidina Descarboxilasa/metabolismo , Área Hipotalámica Lateral/citología , Área Hipotalámica Lateral/metabolismo , Melanocortinas/metabolismo , Neuronas/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Histidina Descarboxilasa/genética , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Péptidos Cíclicos/farmacología , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/genética , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
19.
PLoS One ; 15(2): e0229617, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32109250

RESUMEN

An increasing prevalence of overweight and obesity in people living with HIV has been associated with initiation of antiretroviral therapy with integrase strand transfer inhibitors (INSTIs). An off-target inhibition of the endogenous ligand binding to the human melanocortin 4 receptor (MC4R) has been suggested as a potential mechanism for clinical body weight gain following initiation of dolutegravir, an INSTI. In this study, we interrogated several INSTIs for their capacity for antagonism or agonism of MC4R in an in vitro cell-based assays including at concentrations far exceeding plasma concentrations reached at the recommended dosages. Our results indicate that while INSTIs do exhibit the capacity to antagonize MC4R, this occurs at concentrations well above predicted clinical exposure and is thus an implausible explanation for INSTI-associated weight gain.


Asunto(s)
Inhibidores de Integrasa VIH/efectos adversos , Receptor de Melanocortina Tipo 4/efectos de los fármacos , Aumento de Peso/efectos de los fármacos , Peso Corporal , Infecciones por VIH/tratamiento farmacológico , Inhibidores de Integrasa VIH/farmacología , Inhibidores de Integrasa VIH/uso terapéutico , Humanos , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/antagonistas & inhibidores
20.
Ann Pharmacother ; 54(7): 684-690, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31893927

RESUMEN

Objective: To review data regarding bremelanotide, a recently approved therapy for hypoactive sexual desire disorder (HSDD). Data Sources: Literature search of Medline, SCOPUS, and EMBASE was performed using the search terms bremelanotide, bremelanotide injection, Vyleesi, and melanocortin 4 receptor agonist between January 1, 1996, and December 15, 2019. Reference lists from included articles were also reviewed for pertinent citations. Study Selection/Data Extraction: We included phase 2 and 3 trials of bremelanotide. There were 2 reports of phase 3 trials and 2 reports of phase 2 trials. Additional information from supplementary analyses was also referenced. Data Synthesis: Bremelanotide demonstrates significant improvement in desire and a significant decrease in distress related to lack of desire. The most common adverse effects include nausea (39.9%), facial flushing (20.4%), and headache (11%). Relevance to Patient Care and Clinical Practice: Bremelanotide is the second Food and Drug Administration-approved medication for the treatment of HSDD. Bremelanotide's place in therapy is unknown, as the HSDD guidelines were last updated in 2017. Although the trials met statistical significance for change in sexual desire elements and distress related to sexual desire, the clinical benefit may only be modest. Conclusion: Bremelanotide is a subcutaneous injection that can be administered as needed approximately 45 minutes prior to sexual activity. Bremelanotide is safe and has limited drug-drug interactions, including no clinically significant interactions with ethanol. Prescribing guidelines recommend no more than 1 dose in 24 hours and no more than 8 doses per month. Individuals should discontinue use after 8 weeks without benefit.


Asunto(s)
Libido/efectos de los fármacos , Péptidos Cíclicos/uso terapéutico , Disfunciones Sexuales Psicológicas/tratamiento farmacológico , alfa-MSH/uso terapéutico , Ensayos Clínicos como Asunto , Interacciones Farmacológicas , Femenino , Humanos , Inyecciones Subcutáneas , Náusea/inducido químicamente , Péptidos Cíclicos/administración & dosificación , Péptidos Cíclicos/efectos adversos , Péptidos Cíclicos/farmacocinética , Receptor de Melanocortina Tipo 3/agonistas , Receptor de Melanocortina Tipo 4/agonistas , Resultado del Tratamiento , alfa-MSH/administración & dosificación , alfa-MSH/efectos adversos , alfa-MSH/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...