Abstract

Central estrogen signaling coordinates energy expenditure, reproduction, and in concert with peripheral estrogen impacts skeletal homeostasis in females. Here, we ablate estrogen receptor alpha (ERα) in the medial basal hypothalamus and find a robust bone phenotype only in female mice that results in exceptionally strong trabecular and cortical bones, whose density surpasses other reported mouse models. Stereotaxic guided deletion of ERα in the arcuate nucleus increases bone mass in intact and ovariectomized females, confirming the central role of estrogen signaling in this sex-dependent bone phenotype. Loss of ERα in kisspeptin (Kiss1)-expressing cells is sufficient to recapitulate the bone phenotype, identifying Kiss1 neurons as a critical node in this powerful neuroskeletal circuit. We propose that this newly-identified female brain-to-bone pathway exists as a homeostatic regulator diverting calcium and energy stores from bone building when energetic demands are high. Our work reveals a previously unknown target for treatment of age-related bone disease.

Highlights

  • Central estrogen signaling coordinates energy expenditure, reproduction, and in concert with peripheral estrogen impacts skeletal homeostasis in females

  • Our investigation to understand the complex role of estrogen signaling in the medial basal hypothalamus (MBH) establishes that ERα-expressing Kiss[1] arcuate nucleus (ARC) neurons are central to restraining a powerful brain–bone axis in female mice

  • This assertion stems from the sex-dependent, high bone mass phenotype that emerged from three independent, intersectional strategies that target central ERα signaling

Read more

Summary

Introduction

Central estrogen signaling coordinates energy expenditure, reproduction, and in concert with peripheral estrogen impacts skeletal homeostasis in females. Stereotaxic guided deletion of ERα in the arcuate nucleus increases bone mass in intact and ovariectomized females, confirming the central role of estrogen signaling in this sex-dependent bone phenotype. Loss of ERα in kisspeptin (Kiss1)-expressing cells is sufficient to recapitulate the bone phenotype, identifying Kiss[1] neurons as a critical node in this powerful neuroskeletal circuit. Trabecular and cortical bone volume are modestly elevated after deleting ERα using the non-inducible and developmentally promiscuous POMC-Cre[7,8] In both of these mouse models (Esr1Nestin-Cre and Esr1POMC-Cre), the elevated bone mass in females vanishes following ovariectomy, underscoring the essential role of gonadal sex-steroids in generating and/or maintaining these bone phenotypes. We go on to define Kiss[1] neurons as the critical estrogen-responsive subpopulation in the ARC for promoting this remarkable, robust bone mass in female mice

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call