Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Neurol ; 330: 113327, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32387398

RESUMO

Numerous genes, and alterations in their expression, have been identified as risk factors for developing levodopa-induced dyskinesia (LID). However, our understanding of the complexities of molecular changes remains insufficient for development of clinical treatment. In the current study we used gene array, in situ hybridization, immunohistochemistry, and microdialysis to provide a unique compare and contrast assessment of the relationship of four candidate genes to LID, employing three genetically distinct rat strains (Sprague-Dawley (SD), Fischer-344 (F344) and Lewis-RT.1) showing differences in dyskinesia susceptibility and 'first-ever LID' versus 'chronic LID' expression in subjects displaying equal dyskinesia severity. In these studies, rat strains were easily distinguishable for their LID propensity with: 1) a majority of SD rats expressing LID (LID+) and a subset being resistant (LID-); 2) all F344 rats readily developing (LID+); and 3) all Lewis rats being LID-resistant (LID-). Following chronic levodopa, LID+ SD rats showed significant increases in candidate gene expression: Nr4a2/(Nurr1) > > Trh > Inhba = Fosb. However, SD rats with long-standing striatal dopamine (DA) depletion treated with first-ever versus chronic high-dose levodopa revealed that despite identical levels of LID severity: 1) Fosb and Nurr1 transcripts but not protein were elevated with acute LID expression; 2) FOSB/ΔFOSB and NURR1 proteins were elevated only with chronic LID; and 3) Trh transcript and protein were elevated only with chronic LID. Strikingly, despite similar levodopa-induced striatal DA release in both LID-expressing F344 and LID-resistant Lewis rats, Fosb, Trh, Inhba transcripts were significantly elevated in both strains; however, Nurr1 mRNA was significantly increased only in LID+ F344 rats. These findings suggest a need to reevaluate currently accepted genotype-to-phenotype relationships in the expression of LID, specifically that of Fosb, a transcription factor generally assumed to play a causal role, and Nurr1, a transcription factor that has received significant attention in PD research linked to its critical role in the survival and function of midbrain DA neurons but who's striatal expression, generally below levels of detection, has remained largely unexplored as a regulator of LID. Finally these studies introduce a novel 'model' (inbred F344 vs inbred Lewis) that may provide a powerful tool for investigating the role for 'dyskinesia-resistance' genes downstream of 'dyskinesia-susceptibility' genes in modulating LID expression, a concept that has received considerably less attention and offers a new ways of thinking about antidyskinetic therapies.


Assuntos
Antiparkinsonianos/toxicidade , Discinesia Induzida por Medicamentos/genética , Discinesia Induzida por Medicamentos/metabolismo , Levodopa/toxicidade , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Genótipo , Masculino , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo , Fenótipo , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew
2.
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
3.
Elife ; 72018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29761783

RESUMO

The adipokine leptin acts on the brain to regulate energy balance but specific functions in many brain areas remain poorly understood. Among these, the preoptic area (POA) is well known to regulate core body temperature by controlling brown fat thermogenesis, and we have previously shown that glutamatergic, long-form leptin receptor (Lepr)-expressing neurons in the POA are stimulated by warm ambient temperature and suppress energy expenditure and food intake. Here we further investigate the role of POA leptin signaling in body weight regulation and its relationship to body temperature regulation in mice. We show that POA Lepr signaling modulates energy expenditure in response to internal energy state, and thus contributes to body weight homeostasis. However, POA leptin signaling is not involved in ambient temperature-dependent metabolic adaptations. Our study reveals a novel cell population through which leptin regulates body weight.


Assuntos
Regulação da Temperatura Corporal , Metabolismo Energético , Homeostase , Leptina/metabolismo , Área Pré-Óptica/fisiologia , Transdução de Sinais , Animais , Peso Corporal , Camundongos
4.
J Clin Invest ; 128(3): 1125-1140, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29457782

RESUMO

Pro-opiomelanocortin (POMC) neurons function as key regulators of metabolism and physiology by releasing prohormone-derived neuropeptides with distinct biological activities. However, our understanding of early events in prohormone maturation in the ER remains incomplete. Highlighting the significance of this gap in knowledge, a single POMC cysteine-to-phenylalanine mutation at position 28 (POMC-C28F) is defective for ER processing and causes early onset obesity in a dominant-negative manner in humans through an unclear mechanism. Here, we report a pathologically important role of Sel1L-Hrd1, the protein complex of ER-associated degradation (ERAD), within POMC neurons. Mice with POMC neuron-specific Sel1L deficiency developed age-associated obesity due, at least in part, to the ER retention of POMC that led to hyperphagia. The Sel1L-Hrd1 complex targets a fraction of nascent POMC molecules for ubiquitination and proteasomal degradation, preventing accumulation of misfolded and aggregated POMC, thereby ensuring that another fraction of POMC can undergo normal posttranslational processing and trafficking for secretion. Moreover, we found that the disease-associated POMC-C28F mutant evades ERAD and becomes aggregated due to the presence of a highly reactive unpaired cysteine thiol at position 50. Thus, this study not only identifies ERAD as an important mechanism regulating POMC maturation within the ER, but also provides insights into the pathogenesis of monogenic obesity associated with defective prohormone folding.


Assuntos
Degradação Associada com o Retículo Endoplasmático , Retículo Endoplasmático/patologia , Hipotálamo/patologia , Obesidade/patologia , Pró-Opiomelanocortina/metabolismo , Animais , Axônios , Cisteína/química , Comportamento Alimentar , Feminino , Proteínas de Fluorescência Verde/metabolismo , Humanos , Inflamação , Peptídeos e Proteínas de Sinalização Intracelular , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neurônios/metabolismo , Fenilalanina/química , Pró-Opiomelanocortina/genética , Proteínas/metabolismo , Compostos de Sulfidrila , Ubiquitina/química , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
5.
Mol Cell Endocrinol ; 438: 77-88, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27614022

RESUMO

The last decade had witnessed a tremendous progress in our understanding of the causes of metabolic diseases including obesity. Among the contributing factors regulating energy balance are nutrient sensors such as sirtuins. Sirtuin1 (Sirt1), a NAD + - dependent deacetylase is affected by diet, environmental stress, and also plays a critical role in metabolic health by deacetylating proteins in many tissues, including liver, muscle, adipose tissue, heart, endothelium, and in the complexity of the hypothalamus. Because of its dependence on NAD+, Sirt1 also functions as a nutrient/redox sensor, and new novel data show a function of this enzyme in the maturation of hypothalamic peptide hormones controlling energy balance either through regulation of specific nuclear transcription factors or by regulating specific pro-hormone convertases (PCs) involved in the post-translational processing of pro-hormones. The post-translational processing mechanism of pro-hormones is critical in the pathogenesis of obesity as recently shown that metabolic and physiological triggers affect the biosynthesis and processing of many peptides hormones. Specific regulation of pro-hormone processing is likely another key step where final amounts of bioactive peptides can be tightly regulated. Different factors stimulate or inhibit pro-hormones biosynthesis in concert with an increase in the PCs involved in the maturation of bioactive hormones. Adding more complexity to the system, the new studies describe here suggest that Sirt1 could also regulate the fate of peptide hormone biosynthesis. The present review summarizes the recent progress in hypothalamic SIRT1 research with a particular emphasis on the tissue-specific control of neuropeptide hormone maturation. The series of studies done in mouse and rat models strongly advocate for the first time that a deacetylating enzyme could be a regulator in the maturation of peptide hormones and their processing enzymes. These discoveries are the culmination of the first in-depth understanding of the metabolic role of Sirt1 in the brain. It suggests that Sirt1 behaves differently in the brain than in organs such as the liver and pancreas, where the enzyme has been more commonly studied.


Assuntos
Hipotálamo/metabolismo , Hormônios Peptídicos/metabolismo , Pró-Proteína Convertases/metabolismo , Sirtuína 1/metabolismo , Animais , Humanos , Modelos Biológicos
6.
J Biol Chem ; 291(11): 5844-5859, 2016 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-26755731

RESUMO

Understanding the role of hypothalamic neuropeptides and hormones in energy balance is paramount in the search for approaches to mitigate the obese state. Increased hypothalamic-pituitary-adrenal axis activity leads to increased levels of glucocorticoids (GC) that are known to regulate body weight. The axis initiates the production and release of corticotropin-releasing hormone (CRH) from the paraventricular nucleus (PVN) of the hypothalamus. Levels of active CRH peptide are dependent on the processing of its precursor pro-CRH by the action of two members of the family of prohormone convertases 1 and 2 (PC1 and PC2). Here, we propose that the nutrient sensor sirtuin 1 (Sirt1) regulates the production of CRH post-translationally by affecting PC2. Data suggest that Sirt1 may alter the preproPC2 gene directly or via deacetylation of the transcription factor Forkhead box protein O1 (FoxO1). Data also suggest that Sirt1 may alter PC2 via a post-translational mechanism. Our results show that Sirt1 levels in the PVN increase in rats fed a high fat diet for 12 weeks. Furthermore, elevated Sirt1 increased PC2 levels, which in turn increased the production of active CRH and GC. Collectively, this study provides the first evidence supporting the hypothesis that PVN Sirt1 activates the hypothalamic-pituitary-adrenal axis and basal GC levels by enhancing the production of CRH through an increase in the biosynthesis of PC2, which is essential in the maturation of CRH from its prohormone, pro-CRH.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Pró-Proteína Convertase 2/metabolismo , Precursores de Proteínas/metabolismo , Sirtuína 1/metabolismo , Animais , Metabolismo Energético , Masculino , Obesidade/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Endocrinology ; 156(4): 1372-85, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25590244

RESUMO

Proopiomelanocortin (POMC)-derived peptides like α-melanocyte-stimulating hormone (MSH) substantially improve hepatic insulin sensitivity and regulate energy expenditure. Melanocortinergic agents are also powerful inducers of sexual arousal that are being investigated for a possible therapeutic role in erectile dysfunction. It is currently unclear whether reduced melanocortin (MC) activity may contribute to the sexual dysfunction accompanying obesity and type 2 diabetes. Male rodents with leptin and insulin resistance targeted to POMC neurons (leptin receptor [LepR]/insulin receptor [IR]POMC mice) exhibit obesity, hyperinsulinemia, hyperglycemia, and systemic insulin resistance. In this study, we demonstrate that LepR/IRPOMC males are also subfertile due to dramatic alterations in sexual behavior. Remarkably, these reproductive changes are accompanied by decreased α-MSH production not present when a single receptor type is deleted. Unexpectedly, behavioral sensitivity to α-MSH and MC receptor expression are also reduced in LepR/IRPOMC males, a potential adaptation of the MC system to altered α-MSH production. Together, these results suggest that concurrent insulin and leptin resistance in POMC neurons in individuals with obesity or type 2 diabetes can reduce endogenous α-MSH levels and impair sexual function.


Assuntos
Melanocortinas/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptor de Insulina/metabolismo , Receptores para Leptina/metabolismo , Disfunções Sexuais Fisiológicas/metabolismo , Agressão/fisiologia , Animais , Insulina/metabolismo , Resistência à Insulina/fisiologia , Leptina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Receptor de Insulina/genética , Receptores para Leptina/genética , Disfunções Sexuais Fisiológicas/genética
8.
Endocrinology ; 156(3): 961-74, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25549049

RESUMO

In the periphery, the nutrient-sensing enzyme Sirtuin 1 (silent mating type information regulation 2 homolog 1 [Sirt1]) reduces body weight in diet-induced obese (DIO) rodents. However, the role of hypothalamic Sirt1 in body weight and energy balance regulation is debated. The first studies to reveal that central Sirt1 regulates body weight came from experiments in our laboratory using Sprague-Dawley rats. Central inhibition of Sirt1 decreased body weight and food intake as a result of a forkhead box protein O1 (FoxO1)-mediated increase in the anorexigenic proopiomelanocortin (POMC) and decrease in the orexigenic Agouti-related peptide in the hypothalamic arcuate nucleus. Here, we demonstrate that central inhibition of Sirt1 in DIO decreased body weight and increased energy expenditure at higher levels as compared with the lean counterpart. Brain Sirt1 inhibition in DIO increased acetylated FoxO1, which in turn increased phosphorylated FoxO1 via improved insulin/phosphorylated AKT signaling. Elevated acetylated FoxO1 and phosphorylated FoxO1 increased POMC along with the α-melanocyte-stimulating hormone (α-MSH) maturation enzyme carboxypeptidase E, which resulted in more of the bioactive POMC product α-MSH released into the paraventricular nucleus. Increased in α-MSH led to augmented TRH levels and circulating T3 levels (triiodothyronine, thyroid hormone). These results indicate that inhibiting hypothalamic Sirt1 in DIO enhances the activity of the hypothalamic-pituitary-thyroid axis, which stimulates energy expenditure. Because we show that blocking central Sirt1 causes physiological changes that promote a negative energy balance in an obese individual, our results support brain Sirt1 as a significant target for weight loss therapeutics.


Assuntos
Peso Corporal/fisiologia , Carboxipeptidase H/metabolismo , Metabolismo Energético/fisiologia , Pró-Opiomelanocortina/metabolismo , Sirtuína 1/metabolismo , alfa-MSH/metabolismo , Animais , Carboxipeptidase H/genética , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/efeitos adversos , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação Enzimológica da Expressão Gênica , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Obesidade/induzido quimicamente , Obesidade/metabolismo , Pró-Opiomelanocortina/genética , Ratos , Ratos Sprague-Dawley , Sirtuína 1/genética , alfa-MSH/genética
9.
Mol Endocrinol ; 28(9): 1423-34, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24947673

RESUMO

In developed nations, the prevalence of obesity and its associated comorbidities continue to prevail despite the availability of numerous treatment strategies. Accumulating evidence suggests that multiple inputs from the periphery and within the brain act in concert to maintain energy metabolism at a constant rate. At the central level, the hypothalamus is the primary component of the nervous system that interprets adiposity or nutrient-related inputs; it delivers hormonal and behavioral responses with the ultimate purpose of regulating energy intake and energy consumption. At the molecular level, enzymes called nutrient energy sensors mediate metabolic responses of those tissues involved in energy balance ( 1 ). Two key energy/nutrient sensors, mammalian target of rapamycin and AMP-activated kinase, are involved in the control of food intake in the hypothalamus as well as in peripheral tissues ( 2 , 3 ). The third more recently discovered nutrient sensor, Sirtuin1 (Sirt1), a nicotinamide adenine dinucleotide-dependent deacetylase, functions to maintain whole-body energy homeostasis. Several studies have highlighted a role for both peripheral and central Sirt1 in regulating body metabolism, but its central role is still heavily debated. Owing to the opaqueness of central Sirt1's role in energy balance are its cell-specific functions. Because of its robust central expression, targeting cell-specific downstream mediators of Sirt1 signaling may help to combat obesity. However, when placed in the context of a physiologically relevant model, there is compelling evidence that central Sirt1 inhibition in itself is sufficient to promote negative energy balance in both the lean and diet-induced obese state.


Assuntos
Receptor Tipo 1 de Melanocortina/metabolismo , Sirtuína 1/metabolismo , Animais , Peso Corporal , Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Humanos , Hipotálamo/metabolismo , Modelos Biológicos , Neurônios/metabolismo , Obesidade/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Transdução de Sinais
10.
Endocrinology ; 155(7): 2423-35, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24773342

RESUMO

In the periphery, the nutrient-sensing enzyme Sirtuin 1 (silent mating type information regulation 2 homolog 1 [Sirt1]) reduces body weight in diet-induced obese (DIO) rodents. However, the role of Sirt1 in the brain, particularly the hypothalamus, in body weight and energy balance regulation is debated. Among the first studies to reveal that central Sirt1 regulates body weight came from experiments in our laboratory using Sprague Dawley rats. In that study, central inhibition of Sirt1 decreased body weight and food intake as a result of a Forkhead box protein O1 (FoxO1)-mediated increase in the anorexigenic proopiomelanocortin (POMC) and decrease in the orexigenic Agouti-related peptide in the hypothalamic arcuate nucleus. Here, we demonstrate that central inhibition of Sirt1 in DIO decreased body weight and increased energy expenditure at higher levels as compared with the lean counterpart. Brain Sirt1 inhibition in DIO increased acetylated FoxO1, which, in turn, increased phosphorylated FoxO1 via improved insulin/pAKT signaling. Elevated acetylated FoxO1 and phosphorylated FoxO1 increased POMC along with the α-MSH maturation enzyme carboxypeptidase E, which resulted in more of the bioactive POMC product α-MSH released into the paraventricular nucleus. Increased in α-MSH led to augmented TRH levels and circulating T3 levels (thyroid hormone). These results indicate that inhibiting hypothalamic Sirt1 in DIO enhances the activity of the hypothalamic-pituitary-thyroid axis, which stimulates energy expenditure. Because we show that blocking central Sirt1 causes physiological changes that promote a negative energy balance in an obese individual, our results support brain Sirt1 as a significant target for weight loss therapeutics.


Assuntos
Peso Corporal/fisiologia , Carboxipeptidase H/metabolismo , Metabolismo Energético/fisiologia , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Sirtuína 1/metabolismo , alfa-MSH/metabolismo , Acetilação , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Western Blotting , Carbazóis/farmacologia , Linhagem Celular Tumoral , Dieta Hiperlipídica/efeitos adversos , Ingestão de Alimentos/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Obesidade/etiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética
11.
Am J Physiol Endocrinol Metab ; 306(8): E904-15, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24518677

RESUMO

Hypothalamic proopiomelanocortin (POMC) neurons constitute a critical anorexigenic node in the central nervous system (CNS) for maintaining energy balance. These neurons directly affect energy expenditure and feeding behavior by releasing bioactive neuropeptides but are also subject to signals directly related to nutritional state such as the adipokine leptin. To further investigate the interaction of diet and leptin on hypothalamic POMC peptide levels, we exposed 8- to 10-wk-old male POMC-Discosoma red fluorescent protein (DsRed) transgenic reporter mice to either 24-48 h (acute) or 2 wk (chronic) food restriction, high-fat diet (HFD), or leptin treatment. Using semiquantitative immunofluorescence and radioimmunoassays, we discovered that acute fasting and chronic food restriction decreased the levels of adrenocorticotropic hormone (ACTH), α-melanocyte-stimulating hormone (α-MSH), and ß-endorphin in the hypothalamus, together with decreased DsRed fluorescence, compared with control ad libitum-fed mice. Furthermore, acute but not chronic HFD or leptin administration selectively increased α-MSH levels in POMC fibers and increased DsRed fluorescence in POMC cell bodies. HFD and leptin treatments comparably increased circulating leptin levels at both time points, suggesting that transcription of Pomc and synthesis of POMC peptide products are not modified in direct relation to the concentration of plasma leptin. Our findings indicate that negative energy balance persistently downregulated POMC peptide levels, and this phenomenon may be partially explained by decreased leptin levels, since these changes were blocked in fasted mice treated with leptin. In contrast, sustained elevation of plasma leptin by HFD or hormone supplementation did not significantly alter POMC peptide levels, indicating that enhanced leptin signaling does not chronically increase Pomc transcription and peptide synthesis.


Assuntos
Hipotálamo/metabolismo , Leptina/metabolismo , Estado Nutricional/fisiologia , Pró-Opiomelanocortina/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Dieta , Ingestão de Alimentos/efeitos dos fármacos , Leptina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pró-Opiomelanocortina/genética , Fatores de Tempo
12.
Peptides ; 47: 99-109, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23891651

RESUMO

The prohormone convertases, PC1/3 and PC2 are thought to be responsible for the activation of many prohormones through processing including the endogenous opioid peptides. We propose that maintenance of hormonal homeostasis can be achieved, in part, via alterations in levels of these enzymes that control the ratio of active hormone to prohormone. In order to test the hypothesis that exogenous opioids regulate the endogenous opioid system and the enzymes responsible for their biosynthesis, we studied the effect of short-term morphine or naltrexone treatment on pituitary PC1/3 and PC2 as well as on the level of pro-opiomelanocortin (POMC), the precursor gene for the biosynthesis of the endogenous opioid peptide, ß-endorphin. Using ribonuclease protection assays, we observed that morphine down-regulated and naltrexone up-regulated rat pituitary PC1/3 and PC2 mRNA. Immunofluorescence and Western blot analysis confirmed that the protein levels changed in parallel with the changes in mRNA levels and were accompanied by changes in the levels of phosphorylated cyclic-AMP response element binding protein. We propose that the alterations of the prohormone processing system may be a compensatory mechanism in response to an exogenous opioid ligand whereby the organism tries to restore its homeostatic hormonal milieu following exposure to the opioid, possibly by regulating the levels of multiple endogenous opioid peptides and other neuropeptides in concert.


Assuntos
Morfina/farmacologia , Entorpecentes/farmacologia , Pró-Opiomelanocortina/genética , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 2/genética , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Hipófise/efeitos dos fármacos , Hipófise/fisiologia , Pró-Opiomelanocortina/metabolismo , Pró-Proteína Convertase 1/antagonistas & inibidores , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 2/antagonistas & inibidores , Pró-Proteína Convertase 2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
13.
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
14.
Am J Physiol Endocrinol Metab ; 304(6): E640-50, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23321476

RESUMO

Protein posttranslational processing is a cellular mechanism fundamental to the generation of bioactive peptides, including the anorectic α-melanocyte-stimulating hormone (α-MSH) and thyrotropin-releasing hormone (TRH) peptides produced in the hypothalamic arcuate (ARC) and paraventricular (PVN) nuclei, respectively. Neuropeptide Y (NPY) promotes positive energy balance in part by suppressing α-MSH and TRH. The mechanism by which NPY regulates α-MSH output, however, is not well understood. Our results reveal that NPY inhibited the posttranslational processing of α-MSH's inactive precursor proopiomelanocortin (POMC) by decreasing the prohormone convertase-2 (PC2). We also found that early growth response protein-1 (Egr-1) and NPY-Y1 receptors mediated the NPY-induced decrease in PC2. NPY given intra-PVN also decreased PC2 in PVN samples, suggesting a reduction in PC2-mediated pro-TRH processing. In addition, NPY attenuated the α-MSH-induced increase in TRH production by two mechanisms. First, NPY decreased α-MSH-induced CREB phosphorylation, which normally enhances TRH transcription. Second, NPY decreased the amount of α-MSH in the PVN. Collectively, these results underscore the significance of the interaction between NPY and α-MSH in the central regulation of energy balance and indicate that posttranslational processing is a mechanism that plays a specific role in this interaction.


Assuntos
Regulação do Apetite , Núcleo Arqueado do Hipotálamo/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , alfa-MSH/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Infusões Intraventriculares , Masculino , Modelos Biológicos , Neuropeptídeo Y/administração & dosagem , Fosforilação , Pró-Opiomelanocortina/metabolismo , Pró-Proteína Convertase 2/metabolismo , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/metabolismo
15.
Mol Cell Endocrinol ; 365(1): 84-94, 2013 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-23000398

RESUMO

Thyrotropin-releasing hormone (TRH) in the paraventricular nucleus (PVN) of the hypothalamus is regulated by thyroid hormone (TH). cAMP response element binding protein (CREB) has also been postulated to regulate TRH expression but its interaction with TH signaling in vivo is not known. To evaluate the role of CREB in TRH regulation in vivo, we deleted CREB from PVN neurons to generate the CREB1(ΔSIM1) mouse. As previously shown, loss of CREB was compensated for by an up-regulation of CREM in euthyroid CREB1(ΔSIM1) mice but TSH, T4 and T3 levels were normal, even though TRH mRNA levels were elevated. Interestingly, TRH mRNA expression was also increased in the PVN of CREB1(ΔSIM1) mice in the hypothyroid state but became normal when made hyperthyroid. Importantly, CREM levels were similar in CREB1(ΔSIM1) mice regardless of thyroid status, demonstrating that the regulation of TRH by T3 in vivo likely occurs independently of the CREB/CREM family.


Assuntos
Modulador de Elemento de Resposta do AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Modulador de Elemento de Resposta do AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/fisiologia , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Hormônio Liberador de Tireotropina/genética , Tri-Iodotironina/metabolismo
16.
Endocrinology ; 154(1): 172-83, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23161869

RESUMO

In many mammals, body weight increases continuously throughout adulthood until late middle age. The hormone leptin is necessary for maintaining body weight, in that high levels of leptin promote negative energy balance. As animals age, however, their increase in body weight is accompanied by a steady rise in circulating leptin levels, indicating the progressive development of counterregulatory mechanisms to antagonize leptin's anorexigenic effects. Hypothalamic neurons coexpressing agouti-related peptide (AgRP) and neuropeptide Y are direct leptin targets. These neurons promote positive energy balance, and they inhibit anorexigenic proopiomelanocortin (POMC) neurons via direct neuropeptide action and release of γ-aminobutyric acid. We show here that AgRP and neuropeptide Y innvervation onto POMC neurons increases dramatically with age in male mice. This is associated with progressive increase of inhibitory postsynaptic currents and decrease of POMC firing rate with age. Neuronal activity is significantly attenuated in POMC neurons that receive a high density of AgRP puncta. These high-density AgRP inputs correlate with leptin levels in normal mice and are nearly absent in mice lacking leptin. The progression of increased AgRP innervation onto POMC somas is accelerated in hyperleptinemic, diet-induced obese mice. Together our study suggests that modulation of hypothalamic AgRP innervation constitutes one mechanism to counter the effects of the age-associated rise in leptin levels, thus sustaining body weight and fat mass at an elevated level in adulthood.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Gorduras na Dieta/efeitos adversos , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Eletrofisiologia , Imunofluorescência , Masculino , Camundongos , Neuropeptídeo Y/metabolismo , Radioimunoensaio
17.
Neurosci Lett ; 518(2): 86-91, 2012 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-22580206

RESUMO

The neuropeptide thyrotropin releasing hormone (TRH) is necessary for adequate cold-induced thermogenesis. TRH increases body temperature via both neuroendocrine and autonomic mechanisms. TRH neurons of the hypothalamic paraventricular nucleus (PVN) regulate thermogenesis through the activation of the hypothalamic-pituitary-thyroid axis during cold exposure. However, little is known about the role that TRH neurons play in mediating the sympathetic response to cold exposure. Here, we examined the response of TRH neurons of rats to cold exposure in hypothalamic regions including the PVN, the dorsomedial nucleus and the lateral hypothalamus along with areas of the ventral medulla including raphe obscurus, raphe pallidus (RPa) and parapyramidal regions. Our results using a double immunohistochemistry protocol to identify TRH and c-Fos (as a marker of cellular activity) followed by analysis of preproTRH gene expression demonstrate that only TRH neurons located in the PVN and the RPa are activated in animals exposed to short-term cold conditions.


Assuntos
Temperatura Baixa , Hipotálamo/fisiologia , Neurônios/metabolismo , Termogênese/fisiologia , Hormônio Liberador de Tireotropina/metabolismo , Animais , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Peptides ; 35(1): 42-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22421509

RESUMO

Prohormone convertases (PCs) 1 and 2 are the primary endoproteases involved in the post-translational processing of proThyrotropin Releasing Hormone (proTRH) to give rise to TRH and other proposed biologically active non-TRH peptides. Previous evidence suggests that PC1 is responsible for most proTRH cleavage events. Here, we used the PC1 and PC2 knockout (KO) mouse models to examine the effects of PC1 or PC2 loss on proTRH processing. The PC1KO mouse presented a decrease in five proTRH-derived peptides, whereas the PC2KO mouse showed only lesser reduction in three TRH (Gln-His-Pro), TRH-Gly (Gln-His-Pro-Gly), and the short forms preproTRH(178-184) (pFQ(7)) and preproTRH(186-199) (pSE(14)) of pFE(22) (preproTRH(178-199)). Also, PC1KO and not PC2KO showed a decrease in pEH(24) indicating that PC1 is more important in generating this peptide in the mouse, which differs from previous studies using rat proTRH. Furthermore, downstream effects on thyroid hormone levels were evident in PC1KO mice, but not PC2KO mice suggesting that PC1 plays the more critical role in producing bioactive hypophysiotropic TRH. Yet loss of PC1 did not abolish TRH entirely indicating a complementary action for both enzymes in the normal processing of proTRH. We also show that PC2 alone is responsible for catalyzing the conversion of pFE(22) to pFQ(7) and pSE(14), all peptides implicated in regulation of suckling-induced prolactin release. Collectively, results characterize the specific roles of PC1 and PC2 in proTRH processing in vivo.


Assuntos
Fragmentos de Peptídeos/biossíntese , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 2/genética , Precursores de Proteínas/biossíntese , Hormônio Liberador de Tireotropina/biossíntese , Sequência de Aminoácidos , Animais , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Núcleo Hipotalâmico Paraventricular/metabolismo , Pró-Proteína Convertase 1/deficiência , Pró-Proteína Convertase 2/deficiência , Homologia de Sequência de Aminoácidos , Tri-Iodotironina/biossíntese
19.
Am J Physiol Endocrinol Metab ; 300(6): E1002-11, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21406615

RESUMO

The adipose tissue-derived hormone leptin regulates energy balance through catabolic effects on central circuits, including proopiomelanocortin (POMC) neurons. Leptin activation of POMC neurons increases thermogenesis and locomotor activity. Protein tyrosine phosphatase 1B (PTP1B) is an important negative regulator of leptin signaling. POMC neuron-specific deletion of PTP1B in mice results in reduced high-fat diet-induced body weight and adiposity gain due to increased energy expenditure and greater leptin sensitivity. Mice lacking the leptin gene (ob/ob mice) are hypothermic and cold intolerant, whereas leptin delivery to ob/ob mice induces thermogenesis via increased sympathetic activity to brown adipose tissue (BAT). Here, we examined whether POMC PTP1B mediates the thermoregulatory response of CNS leptin signaling by evaluating food intake, body weight, core temperature (T(C)), and spontaneous physical activity (SPA) in response to either exogenous leptin or 4-day cold exposure (4°C) in male POMC-Ptp1b-deficient mice compared with wild-type controls. POMC-Ptp1b(-/-) mice were hypersensitive to leptin-induced food intake and body weight suppression compared with wild types, yet they displayed similar leptin-induced increases in T(C). Interestingly, POMC-Ptp1b(-/-) mice had increased BAT weight and elevated plasma triiodothyronine (T(3)) levels in response to a 4-day cold challenge, as well as reduced SPA 24 h after cold exposure, relative to controls. These data show that PTP1B in POMC neurons plays a role in short-term cold-induced reduction of SPA and may influence cold-induced thermogenesis via enhanced activation of the thyroid axis.


Assuntos
Temperatura Baixa , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Homeostase/genética , Homeostase/fisiologia , Neurônios/metabolismo , Pró-Opiomelanocortina/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/fisiologia , Animais , Grelina/sangue , Hipotálamo/metabolismo , Luz , Camundongos , Camundongos Knockout , Atividade Motora/fisiologia , Neurônios/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , RNA/biossíntese , RNA/genética , RNA/isolamento & purificação , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Grelina/biossíntese , Transdução de Sinais/fisiologia , Telemetria , Termogênese/fisiologia , Hormônios Tireóideos/sangue , Tireotropina/metabolismo
20.
PLoS One ; 5(11): e13861, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21085660

RESUMO

BACKGROUND: Dyskinesias associated with involuntary movements and painful muscle contractions are a common and severe complication of standard levodopa (L-DOPA, L-3,4-dihydroxyphenylalanine) therapy for Parkinson's disease. Pathologic neuroplasticity leading to hyper-responsive dopamine receptor signaling in the sensorimotor striatum is thought to underlie this currently untreatable condition. METHODOLOGY/PRINCIPAL FINDINGS: Quantitative real-time polymerase chain reaction (PCR) was employed to evaluate the molecular changes associated with L-DOPA-induced dyskinesias in Parkinson's disease. With this technique, we determined that thyrotropin releasing hormone (TRH) was greatly increased in the dopamine-depleted striatum of hemi-parkinsonian rats that developed abnormal movements in response to L-DOPA therapy, relative to the levels measured in the contralateral non-dopamine-depleted striatum, and in the striatum of non-dyskinetic control rats. ProTRH immunostaining suggested that TRH peptide levels were almost absent in the dopamine-depleted striatum of control rats that did not develop dyskinesias, but in the dyskinetic rats, proTRH immunostaining was dramatically up-regulated in the striatum, particularly in the sensorimotor striatum. This up-regulation of TRH peptide affected striatal medium spiny neurons of both the direct and indirect pathways, as well as neurons in striosomes. CONCLUSIONS/SIGNIFICANCE: TRH is not known to be a key striatal neuromodulator, but intrastriatal injection of TRH in experimental animals can induce abnormal movements, apparently through increasing dopamine release. Our finding of a dramatic and selective up-regulation of TRH expression in the sensorimotor striatum of dyskinetic rat models suggests a TRH-mediated regulatory mechanism that may underlie the pathologic neuroplasticity driving dopamine hyper-responsivity in Parkinson's disease.


Assuntos
Corpo Estriado/efeitos dos fármacos , Discinesia Induzida por Medicamentos/metabolismo , Levodopa/toxicidade , Doença de Parkinson Secundária/tratamento farmacológico , Hormônio Liberador de Tireotropina/metabolismo , Análise de Variância , Animais , Antiparkinsonianos/toxicidade , Comportamento Animal/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopamina/metabolismo , Discinesia Induzida por Medicamentos/etiologia , Discinesia Induzida por Medicamentos/genética , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Hormônio Liberador de Tireotropina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...