Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
FASEB J ; 29(11): 4435-48, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26169937

RESUMO

The central melanocortin system is a key regulator of energy homeostasis. Recent studies indicate that tankyrases (TNKSs), which poly(ADP-ribosyl)ate target proteins and direct them toward proteasomal degradation, affect overall metabolism, but the exact molecular mechanisms remain unclear. We used zebrafish larvae as a model to study the mechanisms by which TNKS1b, the zebrafish ortholog of mammalian TNKS1, regulates glucose homeostasis and somatic growth. In situ hybridization revealed that TNKS1b mRNA is prominently expressed in the hypothalamus and pituitary of the embryonic and larval brain. In the pituitary, TNKS1b is coexpressed with pro-opiomelanocortin a (pomca) gene in corticotropes and melanotropes. Knockdown of TNKS1b reduced the linear growth of the larvae, stimulated insulin gene and glucose transporter 4 protein, and suppressed gluconeogenic phosphoenolpyruvate carboxykinase 1 gene. This result indicates rapid glucose utilization and reduction of gluconeogenesis in TNKS1b-deficient larvae. Knockdown of TNKS1b down-regulated pomca expression and diminished α-melanocyte-stimulating hormone in the pars intermedia. Furthermore, down-regulation of TNKS1b suppressed the expression of melanocortin receptor 3 and increased the expression of melanocortin receptor 4. The collective data suggest that TNKS1b modulates glucoregulatory mechanisms and the somatic growth of zebrafish larvae via the central melanocortin system.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipotálamo/embriologia , Hipófise/metabolismo , Pró-Opiomelanocortina/biossíntese , Tanquirases/biossíntese , Peixe-Zebra/embriologia , Animais , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Hipotálamo/citologia , Hipófise/citologia , Pró-Opiomelanocortina/genética , Receptor Tipo 3 de Melanocortina/biossíntese , Receptor Tipo 3 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/biossíntese , Receptor Tipo 4 de Melanocortina/genética , Tanquirases/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/genética
2.
Metab Brain Dis ; 30(5): 1129-37, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25936720

RESUMO

Substantial evidence demonstrated that maternal dietary nutrients can significantly determine the susceptibility to developing metabolic disorders in the offspring. Therefore, we aimed to investigate the later-life effects of maternal and postweaning diets interaction on epigenetic modification of the central nervous system in the offspring. We examined the effects of dams fed a high-fat, high-sucrose (FS) diet during pregnancy and lactation and weaned to FS diet continuously until 32 weeks of age. Then, DNA methylation and gene expressions of hypothalamic proopiomelanocortin (POMC) and melanocortin receptor 4 (MC4R) were determined in the offspring. Offspring of FS diet had heavier body weight, impaired glucose tolerance, decreased insulin sensitivity and higher serum leptin level at 32-week age (p < 0.05). The expression of POMC and MC4R genes were significantly increased in offspring exposed to FS diet during gestation, lactation and into 32-week age (p < 0.05). Consistently, hypomethylation of POMC promoter in the hypothalamus occurred in the FS diet offspring (p < 0.05), compared with the C group. However, no methylation was detected of MC4R promoter in both the two groups. Furthermore, POMC-specific methylation (%) was negatively associated with glucose response to a glucose load (r = -0.273, p = 0.039). Maternal and post-weaning high-fat diet predisposes the offspring for obesity, glucose intolerance and insulin resistance in later life. Our findings can advance our thinking around the DNA methylation status of the promoter of the POMC and MC4R genes between long-term high-fat, high-sucrose diet and glucose homeostasis in mouse.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Sacarose Alimentar/efeitos adversos , Glucose/metabolismo , Homeostase/fisiologia , Hipotálamo/metabolismo , Pró-Opiomelanocortina/biossíntese , Animais , Peso Corporal/fisiologia , Feminino , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal/sangue , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Receptor Tipo 4 de Melanocortina/biossíntese , Desmame
3.
Horm Behav ; 64(1): 95-102, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23707533

RESUMO

Neuropeptide Y (NPY) and nuclear factor-kappa B (NF-κB) are involved in regulating anorexia elicited by phenylpropanolamine (PPA), a sympathomimetic drug. This study explored whether NPY Y1 receptor (Y1R) is involved in this process, and a potential role for the proopiomelanocortin system was identified. Rats were given PPA once a day for 4days. Changes in the hypothalamic expression of the NPY, Y1R, NF-κB, and melanocortin receptor 4 (MC4R) levels were assessed and compared. The results indicated that food intake and NPY expression decreased, with the largest reductions observed on Day 2 (approximately 50% and 45%, respectively), whereas NF-κB, MC4R, and Y1R increased, achieving maximums on Day 2 (160%, 200%, and 280%, respectively). To determine the role of Y1R, rats were pretreated with Y1R antisense or a Y1R antagonist via intracerebroventricular injection 1h before the daily PPA dose. Y1R knockdown and inhibition reduced PPA anorexia and partially restored the normal expression of NPY, MC4R, and NF-κB. The data suggest that hypothalamic Y1R participates in the appetite-suppression from PPA by regulating MC4R and NF-κB. The results of this study increase our understanding of the molecular mechanisms in PPA-induced anorexia.


Assuntos
Depressores do Apetite/farmacologia , Comportamento Alimentar/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Fenilpropanolamina/farmacologia , Receptores de Melanocortina/efeitos dos fármacos , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Animais , Elementos Antissenso (Genética) , Western Blotting , Peso Corporal/fisiologia , Cateterismo , Ventrículos Cerebrais/fisiologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Colículos Inferiores , Injeções , Masculino , NF-kappa B/biossíntese , Ratos , Ratos Wistar , Receptor Tipo 4 de Melanocortina/biossíntese , Receptores de Melanocortina/biossíntese , Receptores de Neuropeptídeo Y/biossíntese , Receptores de Neuropeptídeo Y/genética
4.
J Neurosci ; 30(10): 3803-12, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20220015

RESUMO

Single-minded 1 (SIM1) mutations are one of the few known causes of nonsyndromic monogenic obesity in both humans and mice. Although the role of Sim1 in the formation of the hypothalamus has been described, its postdevelopmental, physiological functions have not been well established. Here we demonstrate that postnatal CNS deficiency of Sim1 is sufficient to cause hyperphagic obesity. We conditionally deleted Sim1 after birth using CaMKII-Cre (alpha-calcium/calmodulin-dependent protein kinase II-Cre) lines to recombine a floxed Sim1 allele. Conditional Sim1 heterozygotes phenocopied germ line Sim1 heterozygotes, displaying hyperphagic obesity and increased length. We also generated viable conditional Sim1 homozygotes, demonstrating that adult Sim1 expression is not essential for mouse or neuron survival and revealing a dosage-dependent effect of Sim1 on obesity. Using stereological cell counting, we showed that the phenotype of both germ line heterozygotes and conditional Sim1 homozygotes was not attributable to global hypocellularity of the paraventricular nucleus (PVN) of the hypothalamus. We also used retrograde tract tracing to demonstrate that the PVN of germ line heterozygous mice projects normally to the dorsal vagal complex and the median eminence. Finally, we showed that conditional Sim1 homozygotes and germ line Sim1 heterozygotes exhibit a remarkable decrease in hypothalamic oxytocin (Oxt) and PVN melanocortin 4 receptor (Mc4r) mRNA. These results demonstrate that the role of Sim1 in feeding regulation is not limited to formation of the PVN or its projections and that the hyperphagic obesity in Sim1-deficient mice may be attributable to changes in the leptin-melanocortin-oxytocin pathway.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Regulação da Expressão Gênica no Desenvolvimento , Hiperfagia/genética , Obesidade/genética , Ocitocina/antagonistas & inibidores , Receptor Tipo 4 de Melanocortina/antagonistas & inibidores , Proteínas Repressoras/deficiência , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Ingestão de Alimentos/genética , Feminino , Inativação Gênica , Hiperfagia/metabolismo , Hiperfagia/patologia , Hiperfagia/fisiopatologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Ocitocina/biossíntese , Ocitocina/genética , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Receptor Tipo 4 de Melanocortina/biossíntese , Receptor Tipo 4 de Melanocortina/genética , Proteínas Repressoras/genética , Reprodutibilidade dos Testes , Transdução de Sinais/genética
5.
Peptides ; 30(6): 1098-104, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19463742

RESUMO

The melanocortin system is crucial to regulation of energy homeostasis. The melanocortin receptor type 4 (MC4R) modulates insulin signaling via effects on c-Jun N-terminal kinase (JNK). The melanocortin agonist NDP-MSH dose-dependently inhibited JNK activity in HEK293 cells stably expressing the human MC4R; effects were reversed by melanocortin receptor antagonist. NDP-MSH time- and dose-dependently inhibited IRS-1(ser307) phosphorylation, effects also reversed by a specific melanocortin receptor antagonist. NDP-MSH augmented insulin-stimulated AKT phosphorylation in vitro. The melanocortin agonist melanotan II increased insulin-stimulated AKT phosphorylation in the rat hypothalamus in vivo. NDP-MSH increased insulin-stimulated glucose uptake in hypothalamic GT1-1 cells. The current study shows that the melanocortinergic system interacts with insulin signaling via novel effects on JNK activity.


Assuntos
Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/metabolismo , Transdução de Sinais , alfa-MSH/análogos & derivados , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Hipotálamo/metabolismo , Insulina/farmacologia , Proteínas Substratos do Receptor de Insulina/antagonistas & inibidores , Proteínas Substratos do Receptor de Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Peptídeos Cíclicos/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 4 de Melanocortina/biossíntese , Fatores de Tempo , alfa-MSH/farmacologia
6.
Neuropeptides ; 42(3): 293-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18359080

RESUMO

The aim of the present work was to study the potential involvement of melanocortin system in the anorectic mechanism of fluoxetine, a selective serotonin reuptake inhibitors, in obese Zucker rats. Male obese Zucker (fa/fa) rats were administered fluoxetine (10 mg/kg; i.p.) daily for two weeks. The control group was given 0.9% NaCl solution. RT-PCR for pro-opiomelanocortin (POMC), Agouti gene related peptide (AgRP) and melanocortin receptor 4 (MC4-R) in the hypothalamus, as well as regional immunostaining for alpha-melanocyte stimulating hormone (alpha-MSH) and MC4-R were carried out. Fluoxetine administration increased POMC expression and reduced MC4-R expression in the hypothalamus, without changes in AgRP mRNA levels. Moreover, an increase in the numbers of alpha-MSH positively immunostained neural cells in the hypothalamic arcuate nucleus (ARC), as well as a significant decrease in the numbers of neural cells positively immunostained for MC4-R in the paraventricular nucleus (PVN), without changes in lateral hypothalamic area (LHA), were observed. These results suggest the involvement of alpha-MSH in central fluoxetine anorectic action.


Assuntos
Depressores do Apetite , Fluoxetina/farmacologia , Hipotálamo/metabolismo , Obesidade/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , alfa-MSH/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Primers do DNA , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/metabolismo , RNA/biossíntese , RNA/isolamento & purificação , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Receptor Tipo 4 de Melanocortina/biossíntese , Receptor Tipo 4 de Melanocortina/efeitos dos fármacos , Receptor Tipo 4 de Melanocortina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , alfa-MSH/biossíntese
7.
J Vet Med Sci ; 66(11): 1323-7, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15585943

RESUMO

We investigated the effects of electrical foot shock stress on the melanocortin signaling cascade and the hypothalamus-pituitary-adrenal (HPA) system by observing levels of mRNA expression of corticotropin releasing factor (CRF), pro-opiomelanocortin (POMC), and melanocortin receptor subtype 4 (MC4R) in the rat amygdala and hypothalamus. When rats were exposed to electrical shock for 0.5 hr or 1 hr, plasma ACTH and corticosterone concentrations increased, indicating stress. The rats were then sacrificed to obtain RNA preparations from the brain tissue. In the amygdala, the expression of MC4R and POMC mRNA as well as CRF mRNA was significantly increased by electrical foot shock stress. In the hypothalamus, MC4R and POMC mRNA increased, but CRF mRNA remained unchanged. The duration of increased gene expression of MC4R and POMC in the amygdala was more sustained than in the hypothalamus. These results have provided the first evidence that exposure to stress increases expression of the MC4R system in the amygdala and hypothalamus.


Assuntos
Tonsila do Cerebelo/metabolismo , Hormônio Liberador da Corticotropina/biossíntese , Hipotálamo/metabolismo , Pró-Opiomelanocortina/biossíntese , Receptor Tipo 4 de Melanocortina/biossíntese , Estresse Fisiológico/metabolismo , Animais , Regulação da Expressão Gênica , Masculino , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
8.
Brain Res ; 992(1): 9-19, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-14604768

RESUMO

The melanocortinergic system plays an important role in promoting negative energy balance and preventing excessive fat deposition. This study has investigated the levels of mRNA expression of proopiomelanocortin (POMC), agouti-related protein (AgRP) and the melanocortin-4 receptor (MC4-R) in diet-induced obese (DIO) and diet-resistant (DR) mice. Thirty C57 mice were used in this study. Twenty-four mice were fed with a high-fat diet (HF: 40% of calories from fat, 20% from saturated fat) for 4 weeks and then classified as DIO and DR according to their body weight gain. Six mice were placed on a low-fat diet (LF: 10% of calories from fat, 1% from saturated fat) and were used as controls. After 22 weeks of feeding, visceral fat deposits were more than twice as heavy in the DIO mice as in the DR and LF mice, while the latter two groups had no significant difference. Using quantitative in situ hybridization techniques, this study found that the DIO mice had a significantly lower level of Arc POMC (-29%) and AgRP (-31%) mRNA expression than the DR and LF mice, respectively. The mice on high-fat diets had higher levels of AgRP mRNA expression in the bed nucleus of stria terminalis (BST), and ventral part of the lateral septal nucleus (LSV) than the LF mice. Furthermore, the DIO mice had a 40% higher level of MC4-R mRNA expression in the ventromedial hypothalamic nucleus (VMH) and posterodorsal part of the medial amygdaloid nucleus (MePD) than the LF mice. In conclusion, this study has demonstrated that differential expression of POMC, AgRP and MC4-R mRNA levels exists in DIO, DR and LF mice. These differences were shown to occur in the specific nuclei of the hypothalamus and other parts of the limbic system. These findings may assist in understanding the involvement of the melanocortinergic system in the regulation of body weight via the autonomic and limbic systems.


Assuntos
Dieta , Hipotálamo/metabolismo , Obesidade/metabolismo , Proteínas , Proteína Relacionada com Agouti , Animais , Peso Corporal , Dieta com Restrição de Gorduras , Ingestão de Energia , Expressão Gênica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pró-Opiomelanocortina/biossíntese , Biossíntese de Proteínas , RNA Mensageiro/análise , Receptor Tipo 4 de Melanocortina/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA