Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
J Bone Miner Res ; 35(6): 1107-1118, 2020 06.
Article in English | MEDLINE | ID: mdl-31995643

ABSTRACT

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.


Subject(s)
Bone Resorption , Nerve Tissue Proteins , Neuropeptide Y , Transcription Factor AP-1/antagonists & inhibitors , Agouti-Related Protein/metabolism , Animals , Cancellous Bone/metabolism , Energy Metabolism , Hypothalamus/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism
2.
J Bone Miner Res ; 34(9): 1707-1720, 2019 09.
Article in English | MEDLINE | ID: mdl-30998833

ABSTRACT

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.


Subject(s)
Bone and Bones/anatomy & histology , Energy Metabolism , Galanin/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Proto-Oncogene Proteins c-fos/genetics , Animals , Body Weight , Gene Deletion , Glucose/metabolism , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Phosphopyruvate Hydratase/metabolism , Proto-Oncogene Proteins c-fos/metabolism
3.
Aging (Albany NY) ; 9(2): 353-369, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28121620

ABSTRACT

The ventral hypothalamus (VHT) integrates several physiological cues to maintain glucose homeostasis and energy balance. Aging is associated with increased glucose intolerance but the underlying mechanisms responsible for age-related metabolic decline, including neuronal signaling in the VHT, remain elusive. We have shown that mice with VHT-targeted overexpression of ∆FosB, a splice variant of the AP1 transcription factor FosB, exhibit increased energy expenditure, leading to decreased adiposity. Here, we show that VHT-targeted overexpression of ∆FosB also improves glucose tolerance, increases insulin sensitivity in target organs and thereby suppresses insulin secretion. These effects are also observed by the overexpression of dominant negative JunD, demonstrating that they occur via AP1 antagonism within the VHT. Furthermore, the improved glucose tolerance and insulin sensitivity persisted in aged animals overexpressing ∆FosB in the VHT. These beneficial effects on glucose metabolism were abolished by peripheral sympathectomy and α-adrenergic, but not ß-adrenergic, blockade. Taken together, our results show that antagonizing AP1 transcription activity in the VHT leads to a marked improvement in whole body glucose homeostasis via activation of the SNS, conferring protection against age-related impairment in glucose metabolism. These findings may open novel avenues for therapeutic intervention in diabetes and age-related glucose intolerance.


Subject(s)
Aging/metabolism , Blood Glucose/metabolism , Hypothalamus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Sympathetic Nervous System/metabolism , Adiposity/physiology , Aging/genetics , Animals , Energy Metabolism/physiology , Glucose Intolerance/metabolism , Homeostasis/physiology , Insulin Resistance/physiology , Mice , Proto-Oncogene Proteins c-fos/genetics
4.
J Bone Miner Res ; 27(8): 1649-58, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22461201

ABSTRACT

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.


Subject(s)
Energy Metabolism/physiology , Hypothalamus/metabolism , Osteogenesis/physiology , Transcription Factor AP-1/metabolism , Animals , Body Weight/physiology , Dependovirus/metabolism , Genes, Dominant , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Organ Size , Protein Binding , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL