Sex differences in vasopressin neurons in the bed nucleus of the stria terminalis by in situ hybridization

Peptides ◽  
1989 ◽  
Vol 10 (3) ◽  
pp. 615-619 ◽  
Author(s):  
Margaret A. Miller ◽  
Linda Vician ◽  
Donald K. Clifton ◽  
Daniel M. Dorsa
2019 ◽  
Vol 21 (1) ◽  
pp. 115 ◽  
Author(s):  
Moeko Kanaya ◽  
Shimpei Higo ◽  
Hitoshi Ozawa

Estrogens play an essential role in multiple physiological functions in the brain, including reproductive neuroendocrine, learning and memory, and anxiety-related behaviors. To determine these estrogen functions, many studies have tried to characterize neurons expressing estrogen receptors known as ERα and ERβ. However, the characteristics of ERβ-expressing neurons in the rat brain still remain poorly understood compared to that of ERα-expressing neurons. The main aim of this study is to determine the neurochemical characteristics of ERβ-expressing neurons in the rat hypothalamus using RNAscope in situ hybridization (ISH) combined with immunofluorescence. Strong Esr2 signals were observed especially in the anteroventral periventricular nucleus (AVPV), bed nucleus of stria terminalis, hypothalamic paraventricular nucleus (PVN), supraoptic nucleus, and medial amygdala, as previously reported. RNAscope ISH with immunofluorescence revealed that more than half of kisspeptin neurons in female AVPV expressed Esr2, whereas few kisspeptin neurons were found to co-express Esr2 in the arcuate nucleus. In the PVN, we observed a high ratio of Esr2 co-expression in arginine-vasopressin neurons and a low ratio in oxytocin and corticotropin-releasing factor neurons. The detailed neurochemical characteristics of ERβ-expressing neurons identified in the current study can be very essential to understand the estrogen signaling via ERβ.


Endocrinology ◽  
1989 ◽  
Vol 125 (5) ◽  
pp. 2335-2340 ◽  
Author(s):  
MARGARET A. MILLER ◽  
JANICE H. URBAN ◽  
DANIEL M. DORSA

Endocrinology ◽  
2009 ◽  
Vol 150 (9) ◽  
pp. 4241-4247 ◽  
Author(s):  
Elaine K. Murray ◽  
Annie Hien ◽  
Geert J. de Vries ◽  
Nancy G. Forger

Abstract The principal nucleus of the bed nucleus of the stria terminalis (BNSTp) is larger in volume and contains more cells in male than female mice. These sex differences depend on testosterone and arise from a higher rate of cell death during early postnatal life in females. There is a delay of several days between the testosterone surge at birth and sexually dimorphic cell death in the BNSTp, suggesting that epigenetic mechanisms may be involved. We tested the hypothesis that chromatin remodeling plays a role in sexual differentiation of the BNSTp by manipulating the balance between histone acetylation and deacetylation using a histone deacetylase inhibitor. In the first experiment, a single injection of valproic acid (VPA) on the day of birth increased acetylation of histone H3 in the brain 24 h later. Next, males, females, and females treated neonatally with testosterone were administered VPA or saline on postnatal d 1 and 2 and killed at 21 d of age. VPA treatment did not influence volume or cell number of the BNSTp in control females but significantly reduced both parameters in males and testosterone-treated females. As a result, the sex differences were eliminated. VPA did not affect volume or cell number in the suprachiasmatic nucleus or the anterodorsal nucleus of the thalamus, which also did not differ between males and females. These findings suggest that a disruption in histone deacetylation may lead to long-term alterations in gene expression that block the masculinizing actions of testosterone in the BNSTp.


Sign in / Sign up

Export Citation Format

Share Document