Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 156(3): 396-7, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24485448

RESUMO

Vogt et al. demonstrate that, in mice, maternal high-fat feeding during lactation is sufficient to program the offspring for impaired energy and glucose homeostasis throughout their lifetime. They reveal that the resulting abnormal insulin signaling in the offspring interferes with the formation of hypothalamic neural circuits that contribute to metabolic status.


Assuntos
Dieta Hiperlipídica , Hiperglicemia/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Lactação , Obesidade/metabolismo , Animais , Feminino , Masculino , Gravidez
2.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34654741

RESUMO

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.


Assuntos
Canabinoides/metabolismo , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Receptor Tipo 4 de Melanocortina/fisiologia , Animais , Ácidos Araquidônicos/fisiologia , Peso Corporal , Endocanabinoides/fisiologia , Jejum , Comportamento Alimentar/fisiologia , Teste de Tolerância a Glucose , Glicerídeos/fisiologia , Resistência à Insulina , Camundongos , Obesidade/genética , Receptor Tipo 4 de Melanocortina/agonistas , Ácido gama-Aminobutírico/metabolismo
3.
J Biol Chem ; 288(24): 17675-88, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23640886

RESUMO

It was shown previously that abnormal prohormone processing or inactive proconverting enzymes that are responsible for this processing cause profound obesity. Our laboratory demonstrated earlier that in the diet-induced obesity (DIO) state, the appetite-suppressing neuropeptide α-melanocyte-stimulating hormone (α-MSH) is reduced, yet the mRNA of its precursor protein proopiomelanocortin (POMC) remained unaltered. It was also shown that the DIO condition promotes the development of endoplasmic reticulum (ER) stress and leptin resistance. In the current study, using an in vivo model combined with in vitro experiments, we demonstrate that obesity-induced ER stress obstructs the post-translational processing of POMC by decreasing proconverting enzyme 2, which catalyzes the conversion of adrenocorticotropin to α-MSH, thereby decreasing α-MSH peptide production. This novel mechanism of ER stress affecting POMC processing in DIO highlights the importance of ER stress in regulating central energy balance in obesity.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Estresse do Retículo Endoplasmático , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Processamento de Proteína Pós-Traducional , Hormônio Adrenocorticotrópico/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/patologia , Linhagem Celular , Dieta Hiperlipídica/efeitos adversos , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica , Leptina/fisiologia , Masculino , Camundongos , Obesidade/etiologia , Obesidade/patologia , Pró-Opiomelanocortina/genética , Pró-Proteína Convertase 2/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , alfa-MSH/metabolismo , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
4.
Elife ; 112022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35323110

RESUMO

The ascending prevalence of obesity in recent decades is commonly associated with soaring morbidity and mortality rates, resulting in increased health-care costs and decreased quality of life. A systemic state of stress characterized by low-grade inflammation and pathological formation of reactive oxygen species (ROS) usually manifests in obesity. The transcription factor nuclear factor erythroid-derived 2-like 2 (NRF2) is the master regulator of the redox homeostasis and plays a critical role in the resolution of inflammation. Here, we show that the natural isothiocyanate and potent NRF2 activator sulforaphane reverses diet-induced obesity through a predominantly, but not exclusively, NRF2-dependent mechanism that requires a functional leptin receptor signaling and hyperleptinemia. Sulforaphane does not reduce the body weight or food intake of lean mice but induces an anorectic response when coadministered with exogenous leptin. Leptin-deficient Lepob/ob mice and leptin receptor mutant Leprdb/db mice display resistance to the weight-reducing effect of sulforaphane, supporting the conclusion that the antiobesity effect of sulforaphane requires functional leptin receptor signaling. Furthermore, our results suggest the skeletal muscle as the most notable site of action of sulforaphane whose peripheral NRF2 action signals to alleviate leptin resistance. Transcriptional profiling of six major metabolically relevant tissues highlights that sulforaphane suppresses fatty acid synthesis while promoting ribosome biogenesis, reducing ROS accumulation, and resolving inflammation, therefore representing a unique transcriptional program that leads to protection from obesity. Our findings argue for clinical evaluation of sulforaphane for weight loss and obesity-associated metabolic disorders.


Assuntos
Leptina , Receptores para Leptina , Animais , Inflamação/metabolismo , Isotiocianatos/farmacologia , Leptina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética , Obesidade/metabolismo , Qualidade de Vida , Espécies Reativas de Oxigênio , Sulfóxidos
5.
Nat Metab ; 4(1): 44-59, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35039672

RESUMO

The adipose tissue-derived hormone leptin can drive decreases in food intake while increasing energy expenditure. In diet-induced obesity, circulating leptin levels rise proportionally to adiposity. Despite this hyperleptinemia, rodents and humans with obesity maintain increased adiposity and are resistant to leptin's actions. Here we show that inhibitors of the cytosolic enzyme histone deacetylase 6 (HDAC6) act as potent leptin sensitizers and anti-obesity agents in diet-induced obese mice. Specifically, HDAC6 inhibitors, such as tubastatin A, reduce food intake, fat mass, hepatic steatosis and improve systemic glucose homeostasis in an HDAC6-dependent manner. Mechanistically, peripheral, but not central, inhibition of HDAC6 confers central leptin sensitivity. Additionally, the anti-obesity effect of tubastatin A is attenuated in animals with a defective central leptin-melanocortin circuitry, including db/db and MC4R knockout mice. Our results suggest the existence of an HDAC6-regulated adipokine that serves as a leptin-sensitizing agent and reveals HDAC6 as a potential target for the treatment of obesity.


Assuntos
Desacetilase 6 de Histona/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Leptina/metabolismo , Obesidade/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Peso Corporal , Dieta Hiperlipídica , Relação Dose-Resposta a Droga , Metabolismo Energético/efeitos dos fármacos , Ativação Enzimática , Regulação da Expressão Gênica/efeitos dos fármacos , Desacetilase 6 de Histona/genética , Desacetilase 6 de Histona/metabolismo , Inibidores de Histona Desacetilases/química , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Obesos , Modelos Biológicos , Obesidade/tratamento farmacológico , Obesidade/etiologia , Transdução de Sinais/efeitos dos fármacos
6.
Diabetes ; 70(9): 2081-2091, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34183373

RESUMO

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.


Assuntos
Metabolismo Energético/fisiologia , Obesidade/metabolismo , Proteoglicanas/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Fármacos Antiobesidade/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/genética , Proteoglicanas/genética , Receptor Tipo 4 de Melanocortina/genética , Transdução de Sinais/fisiologia , Termogênese/fisiologia , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
7.
Am J Physiol Endocrinol Metab ; 299(6): E976-89, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20858755

RESUMO

The hypothalamic-pituitary-thyroid (HPT) axis is a major contributor in maintaining energy expenditure and body weight, and the adipocyte hormone leptin regulates this axis by increasing TRH levels in the fed state. Leptin stimulates TRH directly in the hypothalamic paraventricular nucleus (PVN; direct pathway) and indirectly by regulating proopiomelnocortin neurons in the hypothalamic arcuate nucleus (ARC; indirect pathway). Whereas the indirect pathway is fully functional in lean animals, it is inactive during diet-induced obesity (DIO) because of the establishment of leptin resistance. Despite this, the HPT axis activity in obese humans and rodents remains within the normal levels or slightly higher. Therefore, in this study, we aimed to determine the mechanism(s) by which the HPT axis is still active despite leptin resistance. With a combination of using the Sprague-Dawley rat physiological model and the Zuker rat that bears a mutation in the leptin receptor, we were able to demonstrate that under DIO conditions the HPT axis is regulated at the central level, but only through the direct pathway of leptin action on TRH neurons. Deiodinase enzymes, which are present in many tissues and responsible for converting thyroid hormones, were not statistically different between lean and DIO animals. These data suggest that the increase in T(4/3) seen in obese animals is due mostly to central leptin action. We also found that T(3) feedback inhibition on the prepro-TRH gene is controlled partially by leptin-induced pSTAT3 signaling via the TRH promoter. This interactive relationship between T(3) and pSTAT3 signaling appears essential to maintain the HPT axis at normal levels in conditions such as obesity.


Assuntos
Hipotálamo/metabolismo , Leptina/metabolismo , Obesidade/metabolismo , Obesidade/fisiopatologia , Glândula Tireoide/metabolismo , Glândula Tireoide/fisiopatologia , Análise de Variância , Animais , Western Blotting , Temperatura Corporal , Dieta , Metabolismo Energético , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Hipotálamo/fisiopatologia , Imuno-Histoquímica , Modelos Lineares , Masculino , Neurônios/metabolismo , Neurônios/fisiologia , Obesidade/etiologia , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Receptores para Leptina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Hormônio Liberador de Tireotropina/metabolismo , Tiroxina/sangue , Tri-Iodotironina/sangue
8.
Trends Endocrinol Metab ; 30(3): 163-176, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30691778

RESUMO

Overweight and obesity pose significant health problems globally, and are causatively linked to metabolic dysregulation. The hypothalamus integrates neural, nutritional, and hormonal cues to regulate homeostasis, including circadian rhythm, body temperature, thirst, food intake, energy expenditure, and glucose metabolism. Hypothalamic neuropeptides play a fundamental role in these processes. Studies during the past two decades suggest a role of central endoplasmic reticulum (ER) stress in the pathophysiology of obesity. This review covers recent findings on the role of ER stress and neuropeptide processing in the central regulation of energy homeostasis, with special emphasis on proopiomelanocortin (POMC)-encoding neurons. In addition, the role of neuroinflammation in the context of obesity is briefly discussed.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Hipotálamo/metabolismo , Animais , Metabolismo Energético/fisiologia , Humanos , Resposta a Proteínas não Dobradas/fisiologia
9.
J Endocrinol ; 241(3): R81-R96, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30959481

RESUMO

Leptin resistance refers to states in which leptin fails to promote its anticipated effects, frequently coexisting with hyperleptinaemia. Leptin resistance is closely associated with obesity and also observed in physiological situations such as pregnancy and in seasonal animals. Leptin resensitisation refers to the reversion of leptin-resistant states and is associated with improvement in endocrine and metabolic disturbances commonly observed in obesity and a sustained decrease of plasma leptin levels, possibly below a critical threshold level. In obesity, leptin resensitisation can be achieved with treatments that reduce body adiposity and leptinaemia, or with some pharmacological compounds, while physiological leptin resistance reverts spontaneously. The restoration of leptin sensitivity could be a useful strategy to treat obesity, maintain weight loss and/or reduce the recidivism rate for weight regain after dieting. This review provides an update and discussion about reversion of leptin-resistant states and modulation of the molecular mechanisms involved in each situation.


Assuntos
Leptina/sangue , Obesidade/sangue , Transdução de Sinais , Adiposidade , Animais , Pressão Sanguínea , Peso Corporal , Dieta , Ingestão de Alimentos , Ingestão de Energia , Feminino , Fertilidade , Humanos , Hiperglicemia/sangue , Masculino , Camundongos , Fosforilação , Fotoperíodo , Gravidez , Prenhez , Termogênese , Redução de Peso
10.
Endocrinology ; 160(4): 863-879, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30802281

RESUMO

Leptin signals to regulate food intake and energy expenditure under conditions of normative energy homeostasis. The central expression and function of leptin receptor B (LepRb) have been extensively studied during the past two decades; however, the mechanisms by which LepRb signaling dysregulation contributes to the pathophysiology of obesity remains unclear. The paraventricular nucleus of the hypothalamus (PVN) plays a crucial role in regulating energy balance as well as the neuroendocrine axes. The role of LepRb expression in the PVN in regard to the regulation of physiological function of leptin has been controversial. The single-minded homolog 1 gene (Sim1) is densely expressed in the PVN and in parts of the amygdala, making Sim1-Cre mice a useful model for examining molecular mechanisms regulating PVN function. In this study, we characterized the physiological role of LepRb in Sim1-expressing neurons using LepRb-floxed × Sim1-Cre mice. Sim1-specific LepRb-deficient mice were surprisingly hypophagic on regular chow but gained more weight upon exposure to a high-fat diet than did their control littermates. We show that Sim1-specific deletion of a single LepRb gene copy caused decreased surface and core body temperatures as well as decreased energy expenditure in ambient room temperatures in both female and male mice. Furthermore, cold-induced adaptive (nonshivering) thermogenesis is disrupted in homozygous knockout mice. A defective thermoregulatory response was associated with defective cold-induced upregulation of uncoupling protein 1 in brown adipose tissue and reduced serum T4. Our study provides novel functional evidence supporting LepRb signaling in Sim1 neurons in the regulation of body weight, core body temperature, and cold-induced adaptive thermogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Hipotálamo/metabolismo , Neurônios/metabolismo , Receptores para Leptina/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/fisiologia , Termogênese/fisiologia , Tecido Adiposo Marrom/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Calorimetria Indireta , Metabolismo Energético/fisiologia , Leptina/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora/fisiologia , Receptores para Leptina/genética , Proteínas Repressoras/genética , Proteína Desacopladora 1/metabolismo
11.
J Neuroendocrinol ; 31(1): e12670, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30561082

RESUMO

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.


Assuntos
Hipotálamo/fisiopatologia , Neurônios/fisiologia , Obesidade/fisiopatologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Receptor Tipo 4 de Melanocortina/fisiologia , Animais , Comportamento Alimentar/fisiologia , Feminino , Masculino , Potenciais da Membrana , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canais de Potássio Corretores do Fluxo de Internalização/genética
12.
Sci Adv ; 4(8): eaat0866, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30140740

RESUMO

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


Assuntos
Metabolismo Energético , Comportamento Alimentar , Homeostase , Potenciais Pós-Sinápticos Inibidores/fisiologia , Neurônios/fisiologia , Receptor Tipo 3 de Melanocortina/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
13.
Elife ; 62017 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-28829041

RESUMO

Haploinsufficiency of the melanocortin-4 receptor, the most common monogenetic obesity syndrome in humans, is associated with a reduction in autonomic tone, bradycardia, and incidence of obesity-associated hypertension. Thus, it has been assumed that melanocortin obesity syndrome may be protective with respect to obesity-associated cardiovascular disease. We show here that absence of the melanocortin-4 receptor (MC4R) in mice causes dilated cardiomyopathy, characterized by reduced contractility and increased left ventricular diameter. This cardiomyopathy is independent of obesity as weight matched diet induced obese mice do not display systolic dysfunction. Mc4r cardiomyopathy is characterized by ultrastructural changes in mitochondrial morphology and cardiomyocyte disorganization. Remarkably, testing of myocardial tissue from Mc4r-/- mice exhibited increased ADP stimulated respiratory capacity. However, this increase in respiration correlates with increased reactive oxygen species production - a canonical mediator of tissue damage. Together this study identifies MC4R deletion as a novel and potentially clinically important cause of heart failure.


Assuntos
Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Receptor Tipo 4 de Melanocortina/deficiência , Difosfato de Adenosina/metabolismo , Animais , Respiração Celular/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/ultraestrutura , Miocárdio/patologia , Miócitos Cardíacos/patologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/toxicidade
14.
J Mol Endocrinol ; 56(4): T157-74, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26939593

RESUMO

The melanocortin peptides derived from pro-opiomelanocortin (POMC) were originally understood in terms of the biological actions of α-melanocyte-stimulating hormone (α-MSH) on pigmentation and adrenocorticotrophic hormone on adrenocortical glucocorticoid production. However, the discovery of POMC mRNA and melanocortin peptides in the CNS generated activities directed at understanding the direct biological actions of melanocortins in the brain. Ultimately, discovery of unique melanocortin receptors expressed in the CNS, the melanocortin-3 (MC3R) and melanocortin-4 (MC4R) receptors, led to the development of pharmacological tools and genetic models leading to the demonstration that the central melanocortin system plays a critical role in the regulation of energy homeostasis. Indeed, mutations in MC4R are now known to be the most common cause of early onset syndromic obesity, accounting for 2-5% of all cases. This review discusses the history of these discoveries, as well as the latest work attempting to understand the molecular and cellular basis of regulation of feeding and energy homeostasis by the predominant melanocortin peptide in the CNS, α-MSH.


Assuntos
Metabolismo Energético , Comportamento Alimentar , Homeostase , alfa-MSH/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Clonagem Molecular , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Optogenética/métodos , Pró-Opiomelanocortina/metabolismo , Isoformas de Proteínas , Receptores de Melanocortina/genética , Receptores de Melanocortina/metabolismo , Transdução de Sinais , alfa-MSH/farmacologia
15.
Cell Metab ; 20(1): 73-84, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24836559

RESUMO

Bromodomain-containing protein 7 (BRD7) is a member of the bromodomain-containing protein family that is known to play a role as tumor suppressors. Here, we show that BRD7 is a component of the unfolded protein response (UPR) signaling through its ability to regulate X-box binding protein 1 (XBP1) nuclear translocation. BRD7 interacts with the regulatory subunits of phosphatidylinositol 3-kinase (PI3K) and increases the nuclear translocation of both p85α and p85ß and the spliced form of XBP1 (XBP1s). Deficiency of BRD7 blocks the nuclear translocation of XBP1s. Furthermore, our in vivo studies have shown that BRD7 protein levels are reduced in the liver of obese mice, and reinstating BRD7 levels in the liver restores XBP1s nuclear translocation, improves glucose homeostasis, and ultimately reduces the blood glucose levels in the obese and diabetic mouse models.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Glucose/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/química , Estresse do Retículo Endoplasmático , Células HEK293 , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Obesos , Fosfatidilinositol 3-Quinase/química , Fosfatidilinositol 3-Quinase/deficiência , Subunidades Proteicas/química , Subunidades Proteicas/deficiência , Subunidades Proteicas/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição/química , Proteína 1 de Ligação a X-Box
16.
PLoS One ; 4(12): e8322, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20020036

RESUMO

Sirt1 is an evolutionarily conserved NAD(+) dependent deacetylase involved in a wide range of processes including cellular differentiation, apoptosis, as well as metabolism, and aging. In this study, we investigated the role of hypothalamic Sirt1 in energy balance. Pharmacological inhibition or siRNA mediated knock down of hypothalamic Sirt1 showed to decrease food intake and body weight gain. Central administration of a specific melanocortin antagonist, SHU9119, reversed the anorectic effect of hypothalamic Sirt1 inhibition, suggesting that Sirt1 regulates food intake through the central melanocortin signaling. We also showed that fasting increases hypothalamic Sirt1 expression and decreases FoxO1 (Forkhead transcription factor) acetylation suggesting that Sirt1 regulates the central melanocortin system in a FoxO1 dependent manner. In addition, hypothalamic Sirt1 showed to regulate S6K signaling such that inhibition of the fasting induced Sirt1 activity results in up-regulation of the S6K pathway. Thus, this is the first study providing a novel role for the hypothalamic Sirt1 in the regulation of food intake and body weight. Given the role of Sirt1 in several peripheral tissues and hypothalamus, potential therapies centered on Sirt1 regulation might provide promising therapies in the treatment of metabolic diseases including obesity.


Assuntos
Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Sirtuína 1/metabolismo , Acetilação , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/fisiologia , Linhagem Celular Tumoral , Metabolismo Energético/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Hipotálamo/enzimologia , Melanocortinas/metabolismo , Camundongos , Modelos Animais , Modelos Biológicos , Proteínas do Tecido Nervoso/metabolismo , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Melanocortina/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais
17.
J Biol Chem ; 283(46): 31438-48, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18779326

RESUMO

The biogenesis of rat thyrotropin releasing hormone (TRH) involves the processing of its precursor (proTRH) into five biologically active TRH peptides and several non-TRH peptides where two of them had been attributed potential biological functions. This process implicates 1) proper folding of proTRH in the endoplasmic reticulum after its biosynthesis and exit to the Golgi apparatus and beyond, 2) initial processing of proTRH in the trans Golgi network and, 3) sorting of proTRH-derived peptides to the regulated secretory pathway. Previous studies have focused on elucidating the processing and sorting determinants of proTRH. However, the role of protein folding in the sorting of proTRH remains unexplored. Here we have investigated the role in the secretion of proTRH of a sequence comprising 22 amino acid residues, located at the N-terminal region of proTRH, residues 31-52. Complete deletion of these 22 amino acids dramatically compromised the biosynthesis of proTRH, manifested as a severe reduction in the steady state level of proTRH in the endoplasmic reticulum. This effect was largely reproduced by the deletion of only three amino acid residues, 40PGL42, within the proTRH31-52 sequence. The decreased steady state level of the mutant DeltaPGL was due to enhanced endoplasmic reticulum-associated protein degradation. However, the remnant of DeltaPGL that escaped degradation was properly processed and sorted to secretory granules. Thus, these results suggest that the N-terminal domain within the prohormone sequence does not act as "sorting signal" in late secretion; instead, it seems to play a key role determining the proper folding pathway of the precursor and, thus, its stability.


Assuntos
Precursores de Proteínas/metabolismo , Via Secretória , Hormônio Liberador de Tireotropina/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Deleção de Genes , Camundongos , Dados de Sequência Molecular , Mutação/genética , Precursores de Proteínas/química , Precursores de Proteínas/genética , Ratos , Hormônio Liberador de Tireotropina/química , Hormônio Liberador de Tireotropina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA