Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Bone Miner Res ; 35(6): 1107-1118, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31995643

RESUMO

Energy metabolism and bone homeostasis share several neuronal regulatory pathways. Within the ventral hypothalamus (VHT), the orexigenic neurons co-express Agouti-related peptide (AgRP) and neuropeptide Y (NPY) and the anorexigenic neurons co-express, α-melanocyte stimulating hormone derived from proopiomelanocortin (POMC), and cocaine and amphetamine-regulated transcript (CART). These neurons regulate both processes, yet their relative contribution is unknown. Previously, using genetically targeted activator protein (AP1) alterations as a tool, we showed in adult mice that AgRP or POMC neurons are capable of inducing whole-body energy catabolism and bone accrual, with different effects on bone resorption. Here, we investigated whether co-residing neurons exert similar regulatory effects. We show that AP1 antagonists targeted to NPY-producing or CART-producing neurons in adult mice stimulate energy expenditure, reduce body weight gain and adiposity and promote trabecular bone formation and mass, yet again via different effects on bone resorption, as measured by serum level of carboxy-terminal collagen type I crosslinks (CTX). In addition, AP1 antagonists promote neurite expansion, increasing neurite number, length, and surface area in primary hypothalamic neuronal cultures. Overall, our data demonstrate that the orexigenic NPY and anorexigenic CART neurons both have the capacity to stimulate energy burning state and increase bone mass. © 2020 American Society for Bone and Mineral Research.


Assuntos
Reabsorção Óssea , Proteínas do Tecido Nervoso , Neuropeptídeo Y , Fator de Transcrição AP-1/antagonistas & inibidores , Proteína Relacionada com Agouti/metabolismo , Animais , Osso Esponjoso/metabolismo , Metabolismo Energético , Hipotálamo/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo
2.
J Bone Miner Res ; 34(9): 1707-1720, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30998833

RESUMO

Energy metabolism and bone homeostasis share several regulatory pathways. The AP1 transcription factor ΔFosB and leptin both regulate energy metabolism and bone, yet whether their pathways intersect is not known. Transgenic mice overexpressing ΔFosB under the control of the Enolase 2 (ENO2) promoter exhibit high bone mass, high energy expenditure, low fat mass, and low circulating leptin levels. Because leptin is a regulator of bone and ΔFosB acts on leptin-responsive ventral hypothalamic (VHT) neurons to induce bone anabolism, we hypothesized that regulation of leptin may contribute to the central actions of ΔFosB in the VHT. To address this question, we used adeno-associated virus (AAV) expression of ΔFosB in the VHT of leptin-deficient ob/ob mice and genetic crossing of ENO2-ΔFosB with ob/ob mice. In both models, leptin deficiency prevented ΔFosB-triggered reduction in body weight, increase in energy expenditure, increase in glucose utilization, and reduction in pancreatic islet size. In contrast, leptin deficiency failed to prevent ΔFosB-triggered increase in bone mass. Unlike leptin deficiency, galanin deficiency blocked both the metabolic and the bone ΔFosB-induced effects. Overall, our data demonstrate that, while the catabolic energy metabolism effects of ΔFosB require intact leptin and galanin signaling, the bone mass-accruing effects of ΔFosB require galanin but are independent of leptin. © 2019 American Society for Bone and Mineral Research.


Assuntos
Osso e Ossos/anatomia & histologia , Metabolismo Energético , Galanina/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Animais , Peso Corporal , Deleção de Genes , Glucose/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Fosfopiruvato Hidratase/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
3.
J Bone Miner Res ; 27(8): 1649-58, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22461201

RESUMO

The regulation of bone and fat homeostasis and its relationship to energy expenditure has recently been the focus of increased attention because of its potential relevance to osteoporosis, obesity, and diabetes. Although central effectors within the hypothalamus have been shown to contribute to the regulation of both energy balance and bone homeostasis, little is known of the underlying mechanisms, including the possible involvement of transcriptional factors within the hypothalamus. Transgenic mice overexpressing ΔFosB, a splice variant of the AP-1 transcription factor FosB with mixed agonist-antagonistic properties, have increased energy expenditure and bone mass. Because these mice express ΔFosB in bone, fat, and hypothalamus, we sought to determine 1) whether overexpression of ΔFosB within the hypothalamus was sufficient to regulate energy expenditure and whether it would also regulate bone mass, and 2) whether these effects were the result of antagonism to AP-1. Our results show that stereotactic injection of an adeno-associated virus vector to restrict overexpression of ΔFosB to the ventral hypothalamus of wild-type mice induced a profound increase in both energy expenditure and bone formation and bone mass. This effect was phenocopied, at an even stronger level, by overexpression of a dominant-negative DNJunD, a pure AP-1 antagonist. Taken together, these results suggest that downregulation of AP-1 activity in the hypothalamus profoundly increases energy expenditure and bone formation, leading to both a decrease in adipose mass and an increase in bone mass. These findings may have physiological implications because ΔFosB is expressed and regulated in the hypothalamus.


Assuntos
Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Osteogênese/fisiologia , Fator de Transcrição AP-1/metabolismo , Animais , Peso Corporal/fisiologia , Dependovirus/metabolismo , Genes Dominantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Tamanho do Órgão , Ligação Proteica , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição AP-1/antagonistas & inibidores , Transcrição Gênica
4.
Endocrinology ; 150(1): 135-43, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18772235

RESUMO

Obesity and osteoporosis are major health issues affecting millions of individuals. Transgenic mice overexpressing DeltaFosB, an activator protein-1 transcription factor, under the control of the enolase 2 (ENO2) promoter exhibit both an increase in bone density and a decrease in adipose mass. Here we demonstrate that DeltaFosB overexpression increases fatty-acid oxidation and energy expenditure, leading to a decrease in adipocyte size and adipose mass. In addition, the ENO2-DeltaFosB mice exhibit increased insulin sensitivity and glucose tolerance. Targeted overexpression of DeltaFosB in adipocytes using the adipocyte protein 2 promoter failed to induce changes in fat or in bone, showing that the effect on metabolic activity is not due to cell-autonomous effects of DeltaFosB within adipocytes. Detailed analysis of the ENO2-DeltaFosB mice demonstrated that energy expenditure was increased in muscle, independent of locomotor activity. These findings provide evidence that signaling downstream of DeltaFosB is a potential target for not only osteoporosis but also obesity and diabetes.


Assuntos
Densidade Óssea/genética , Metabolismo Energético/efeitos dos fármacos , Insulina/farmacologia , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-fos/genética , Animais , Ingestão de Energia , Ácidos Graxos/metabolismo , Camundongos , Obesidade/genética , Tamanho do Órgão/genética , Osteocalcina/metabolismo , Fosfopiruvato Hidratase/genética , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-fos/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA