Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 16(7)2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38611051

ABSTRACT

Tamoxifen, a selective estrogen receptor modulator (SERM), is commonly used as an adjuvant drug therapy for estrogen-receptor-positive breast cancers. Though effective at reducing the rate of cancer recurrence, patients often report unwanted cognitive and affective side effects. Despite this, the impacts of chronic tamoxifen exposure on the brain are poorly understood, and rodent models of tamoxifen exposure do not replicate the chronic oral administration seen in patients. We, therefore, used long-term ad lib consumption of medicated food pellets to model chronic tamoxifen exposure in a clinically relevant way. Adult female Long-Evans Hooded rats consumed tamoxifen-medicated food pellets for approximately 12 weeks, while control animals received standard chow. At the conclusion of the experiment, blood and brain samples were collected for analyses. Blood tamoxifen levels were measured using a novel ultra-performance liquid chromatography-tandem mass spectrometry assay, which found that this administration paradigm produced serum levels of tamoxifen similar to those in human patients. In the brain, brain-derived neurotrophic factor (BDNF) was visualized in the hippocampus using immunohistochemistry. Chronic oral tamoxifen treatment resulted in a decrease in BDNF expression across several regions of the hippocampus. These findings provide a novel method of modeling and measuring chronic oral tamoxifen exposure and suggest a putative mechanism by which tamoxifen may cause cognitive and behavioral changes reported by patients.

2.
J Neuroendocrinol ; 35(7): e13278, 2023 07.
Article in English | MEDLINE | ID: mdl-37127859

ABSTRACT

Sleep disruptions are a common occurrence during the peripartum period. While physical and environmental factors associated with pregnancy and newborn care account for some sleep disruptions, there is evidence that peripartum fluctuations in estrogens may independently impact sleep. However, the impact of these large fluctuations in estrogens on peripartum sleep is unclear because it is difficult to tease apart the effects of estrogens on sleep from effects associated with the growth and development of the fetus or parental care. We therefore used a hormone-simulated pseudopregnancy (HSP) in female Syrian hamsters to test the hypothesis that pregnancy-like increases in estradiol decrease sleep in the absence of other factors. Adult female Syrian hamsters were ovariectomized and given daily hormone injections that simulate estradiol levels during early pregnancy, late pregnancy, and the postpartum period. Home cage video recordings were captured at seven timepoints and videos were analyzed for actigraphy. During "late pregnancy," total sleep time and sleep efficiency were decreased in hormone-treated animals during the white light period compared to pretest levels. Likewise, during "late pregnancy," locomotion was increased in the white light period for hormone-treated animals compared to pretest levels. These changes continued into the "postpartum period" for animals who continued to receive estradiol treatment, but not for animals who were withdrawn from estradiol. At the conclusion of the experiment, animals were euthanized and cFos expression was quantified in the ventral lateral preoptic area (VLPO) and lateral hypothalamus (LH). Animals who continued to receive high levels of estradiol during the "postpartum" period had significantly more cFos in the VLPO and LH than animals who were withdrawn from hormones or vehicle controls. Together, these data suggest that increased levels of estradiol during pregnancy are associated with sleep suppression, which may be mediated by increased activation of hypothalamic nuclei.


Subject(s)
Estradiol , Pseudopregnancy , Cricetinae , Animals , Pregnancy , Female , Estradiol/pharmacology , Mesocricetus , Estrogens/pharmacology , Sleep
3.
Horm Behav ; 149: 105312, 2023 03.
Article in English | MEDLINE | ID: mdl-36645923

ABSTRACT

In placental mammals, estradiol levels are chronically elevated during pregnancy, but quickly drop to prepartum levels following birth. This may produce an "estrogen withdrawal" state that has been linked to changes in affective states in humans and rodents during the postpartum period. The neural mechanisms underlying these affective changes, however, are understudied. We used a hormone-simulated pseudopregnancy (HSP), a model of postpartum estrogen withdrawal, in adult female C57BL/6 mice to test the impact of postpartum estradiol withdrawal on several behavioral measures of anxiety and motivation. We found that estradiol withdrawal following HSP increased anxiety-like behavior in the elevated plus maze, but not in the open field or marble burying tests. Although hormone treatment during HSP consistently increased sucrose consumption, sucrose preference was generally not impacted by hormone treatment or subsequent estradiol withdrawal. In the social motivation test, estradiol withdrawal decreased the amount of time spent in proximity to a social stimulus animal. These behavioral changes were accompanied by changes in the expression of ∆FosB, a transcription factor correlated with stable long-term plasticity, in the nucleus accumbens (NAc). Specifically, estrogen-withdrawn females had higher ∆FosB expression in the nucleus accumbens core, but ∆FosB expression did not vary across hormone conditions in the nucleus accumbens shell. Using transgenic reporter mice, we found that this increase in ∆FosB occurred in both D1- and D2-expressing cells in the NAc core. Together, these results suggest that postpartum estrogen withdrawal impacts anxiety and motivation and increases ∆FosB in the NAc core.


Subject(s)
Estradiol , Nucleus Accumbens , Animals , Female , Mice , Pregnancy , Estradiol/pharmacology , Estrogens/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Nucleus Accumbens/metabolism , Placenta/metabolism , Receptors, Dopamine D1/metabolism , Sucrose
4.
Neurosci Biobehav Rev ; 132: 679-690, 2022 01.
Article in English | MEDLINE | ID: mdl-34808191

ABSTRACT

Major life transitions often co-occur with significant fluctuations in hormones that modulate the central nervous system. These hormones enact neuroplastic mechanisms that prepare an organism to respond to novel environmental conditions and/or previously unencountered cognitive, emotional, and/or behavioral demands. In this review, we will explore several examples of how hormones mediate neuroplastic changes in order to produce adaptive responses, particularly during transitions in life stages. First, we will explore hormonal influences on social recognition in both males and females as they transition to sexual maturity. Next, we will probe the role of hormones in mediating the transitions to motherhood and fatherhood, respectively. Finally, we will survey the long-term impact of reproductive experience on neuroplasticity in females, including potential protective effects and risk factors associated with reproductive experience in mid-life and beyond. Ultimately, a more complete understanding of how hormones influence neuroplasticity throughout the lifespan, beyond development, is necessary for understanding how individuals respond to life changes in adaptive ways.


Subject(s)
Hormones , Reproduction , Female , Humans , Male , Neuronal Plasticity/physiology , Reproduction/physiology
5.
Biol Psychiatry ; 89(9): 929-938, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33487439

ABSTRACT

BACKGROUND: Estrogen increases dramatically during pregnancy but quickly drops below prepregnancy levels at birth and remains suppressed during the postpartum period. Clinical and rodent work suggests that this postpartum drop in estrogen results in an estrogen withdrawal state that is related to changes in affect, mood, and behavior. How estrogen withdrawal affects oxytocin (OT) neurocircuitry has not been examined. METHODS: We used a hormone-simulated pseudopregnancy followed by estrogen withdrawal in Syrian hamsters, a first for this species. Ovariectomized females were given daily injections to approximate hormone levels during gestation and then withdrawn from estrogen to simulate postpartum estrogen withdrawal. These hamsters were tested for behavioral assays of anxiety and anhedonia during estrogen withdrawal. Neuroplasticity in OT-producing neurons in the paraventricular nucleus of the hypothalamus and its efferent targets was measured. RESULTS: Estrogen-withdrawn females had increased anxiety-like behaviors in the elevated plus maze and open field tests but did not differ from control females in sucrose preference. Furthermore, estrogen-withdrawn females had more OT-immunoreactive cells and OT messenger RNA in the paraventricular nucleus of the hypothalamus and an increase in OT receptor density in the dorsal raphe nucleus. Finally, blocking OT receptors in the dorsal raphe nucleus during estrogen withdrawal prevented the high-anxiety behavioral phenotype in estrogen-withdrawn females. CONCLUSIONS: Estrogen withdrawal induces OT neuroplasticity in the paraventricular nucleus of the hypothalamus and dorsal raphe nucleus to increase anxiety-like behavior during the postpartum period. More broadly, these experiments suggest Syrian hamsters as a novel organism in which to model the effects of postpartum estrogen withdrawal on the brain and anxiety-like behavior.


Subject(s)
Dorsal Raphe Nucleus , Oxytocin , Anxiety , Estrogens , Female , Humans , Hypothalamus , Paraventricular Hypothalamic Nucleus , Postpartum Period , Pregnancy
6.
Front Behav Neurosci ; 13: 227, 2019.
Article in English | MEDLINE | ID: mdl-31636548

ABSTRACT

Low levels of desire and arousal are the primary sexual dysfunctions in women, necessitating neurobiological studies of sexual motivation in female animal models. As the mesocorticolimbic system is a primary neural circuit underlying sexual motivation, the goal of this study was to test the hypothesis that medial prefrontal cortex (mPFC) glutamate mediates sexual behavior activation of the nucleus accumbens. Glutamatergic neurons in the mPFC were activated by sex behavior, and these sex-activated cells shown to project to the nucleus accumbens. During sexual interactions with the male, glutamate transients recorded in the nucleus accumbens of female hamsters were specifically associated with the receipt of intromissions from the male. Further, inhibition of the mPFC during sex significantly decreased nucleus accumbens activation. Glutamatergic medial prefrontal cortical input to the nucleus accumbens mediates the activity in the nucleus accumbens during female sexual behavior. These results offer novel insights into the neurobiology of the motivational control of female sexual behavior and provide attractive avenues for pursuing target-specific and clinically-relevant therapies for sexual dysfunction in women.

7.
Behav Neurosci ; 133(4): 378-384, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30869949

ABSTRACT

Motivated behaviors share the common feature of activating the mesolimbic dopamine system. Repeated experience with motivated behaviors can cause long-lasting structural changes in the nucleus accumbens (NAc). The molecular mechanisms underlying this experience-dependent plasticity in the NAc have been well described following experience with drugs of abuse. In particular, the transcription factor Delta FosB (ΔFosB) is a key regulator of drug-related neuroplasticity. Fewer studies have examined the molecular mechanisms underlying experience-dependent plasticity in the NAc following naturally motivated behaviors, but previous research has demonstrated that sexual experience increases the accumulation of ΔFosB in the NAc of female hamsters and male rats. Sex behavior is unique among motivated behaviors in that the expression of the behavior varies drastically between males and females of the same species. Despite this, a quantitative comparison of ΔFosB following sex experience in males and females of the same species has never been conducted. We therefore used Western blotting to test the hypothesis that sex experience increases ΔFosB in both male and female Syrian hamsters following repeated sexual experience. We found that sex experience significantly increases ΔFosB protein in male and female Syrian hamsters. Further, ΔFosB protein levels did not differ between males and females following sex experience. Interestingly, repeated sex experience only led to increased copulatory efficiency in female hamsters; male copulatory efficiency did not improve with repeated experience. Together, these data demonstrate that ΔFosB is increased following sexual reward in both males and females but may be uncoupled from behavioral plasticity in males. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Subject(s)
Motivation/physiology , Proto-Oncogene Proteins c-fos/metabolism , Sexual Behavior, Animal/physiology , Animals , Brain/physiology , Copulation/physiology , Cricetinae , Female , Male , Mesocricetus/metabolism , Neuronal Plasticity/physiology , Nucleus Accumbens/physiology , Proto-Oncogene Proteins c-fos/physiology , Reward , Sex Factors
8.
Neuropharmacology ; 156: 107451, 2019 09 15.
Article in English | MEDLINE | ID: mdl-30502376

ABSTRACT

Although many people think of aggression as a negative or undesirable emotion, it is a normal part of many species' repertoire of social behaviors. Purposeful and controlled aggression can be adaptive in that it warns other individuals of perceived breaches in social contracts with the goal of dispersing conflict before it escalates into violence. Aggression becomes maladaptive, however, when it escalates inappropriately or impulsively into violence. Despite ample data demonstrating that impulsive aggression and violence occurs in both men and women, aggression has historically been considered a uniquely masculine trait. As a result, the vast majority of studies attempting to model social aggression in animals, particularly those aimed at understanding the neural underpinnings of aggression, have been conducted in male rodents. In this review, we summarize the state of the literature on the neurobiology of social aggression in female rodents, including social context, hormonal regulation and neural sites of aggression regulation. Our goal is to put historical research in the context of new research, emphasizing studies using ecologically valid methods and modern sophisticated techniques. This article is part of the Special Issue entitled 'Current status of the neurobiology of aggression and impulsivity'.


Subject(s)
Aggression/physiology , Brain/physiology , Animals , Female , Male , Mice , Models, Neurological , Neurons , Rats , Sex Characteristics
9.
J Undergrad Neurosci Educ ; 15(1): A67-A71, 2016.
Article in English | MEDLINE | ID: mdl-27980473

ABSTRACT

The traditional approach to teaching neuroscience often involves presenting a topic like one might present a "murder mystery"; evidence is presented serially until the final answer is revealed. Although this approach mirrors the scientific discovery process, it is not always effective at engaging students, particularly those who are less familiar with the scientific concepts being presented as evidence. By the time the answer arrives, students may be too overwhelmed to absorb it. One way to combat this is to reverse the order of presentation. By starting with the final condition and working backwards through the underlying neuroscientific concepts, students have a relatable framework in which to couch the scientific detail necessary to understand neural phenomena. It was with this approach in mind that the course, "Fundamental Neuroscience: Understanding Ourselves" was designed. Taught for the past seven years at the University of Minnesota, the course uses the best-selling book The Brain That Changes Itself by Norman Doidge in lieu of a traditional textbook. Each chapter focuses on a case study of a particular neuropsychological problem or, in some cases, the work of a particular neuroscientist. This material is then used as a launching point to delve deeper into the neurobiological mechanisms underlying the particular disorder. In our experience, the result is that students from a wide variety of academic backgrounds are able to engage with the material throughout the entire lesson and apply their new knowledge broadly across the discipline of Neuroscience. This article aims to provide an in-depth presentation of the course, including potential challenges of working with a pop-science text. Further, we extend our discussion to a newly-developed companion course using non-traditional texts and how these courses fit into a Neuroscience minor.

10.
Biol Psychiatry ; 79(8): 685-92, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26342498

ABSTRACT

BACKGROUND: Escalated aggression is a behavioral sign of numerous psychiatric disorders characterized by a loss of control. The neurobiology underlying escalated aggression is unknown and is particularly understudied in females. Research in our laboratory demonstrated that repeated aggressive experience in female hamsters resulted in an escalated response to future aggressive encounters and an increase in dendritic spine density on nucleus accumbens (NAc) neurons. We hypothesized that the activation of group I metabotropic glutamate receptor signaling though the fragile X mental retardation protein (FMRP) pathway may underlie synaptic plasticity associated with aggression escalation. METHODS: Female hamsters were given five daily aggression tests with or without prior treatment with the metabotropic glutamate receptor 5 (mGluR5) antagonist 2-methyl-6-(phenylethynyl)-pyridine. Following aggression testing, messenger RNA expression and protein levels were measured in the nucleus accumbens for postsynaptic density protein 95 (PSD-95) and SAP90/PSD-95-associated protein 3, as well as the levels of phosphorylated FMRP. RESULTS: Experience-dependent escalation of aggression in female hamsters depends on activation of mGluR5 receptors. Furthermore, aggressive experience decreases phosphorylation of FMRP in the NAc, which is coupled to a long-term increase in the expression of the synaptic scaffolding proteins PSD-95 and SAP90/PSD-95-associated protein 3. Finally, the experience-dependent increase in PSD-95 is prevented by antagonism of the mGluR5 receptor. CONCLUSIONS: Activation of the FMRP pathway by group I metabotropic glutamate receptors is involved in regulating synaptic plasticity following aggressive experience. The NAc is a novel target for preclinical studies of the treatment of escalated aggression, with the added benefit that emerging therapeutic approaches are likely to be effective in treating pathologic aggression in both female and male subjects.


Subject(s)
Aggression/physiology , Nucleus Accumbens/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Aggression/drug effects , Animals , Blotting, Western , Caudate Nucleus/drug effects , Caudate Nucleus/metabolism , Cricetinae , Excitatory Amino Acid Antagonists/pharmacology , Female , Fragile X Mental Retardation Protein/metabolism , Nerve Tissue Proteins/metabolism , Nucleus Accumbens/drug effects , Ovariectomy , Phosphorylation , Psychological Tests , Putamen/drug effects , Putamen/metabolism , Pyridines/pharmacology , RNA, Messenger/metabolism , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors
11.
Neurosci Lett ; 550: 41-5, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23850605

ABSTRACT

The nucleus accumbens (NAc) is an important brain region for motivation, reinforcement, and reward. Afferents to the NAc can be divided into two anatomically segregated neurochemical phenotypes: dopaminergic inputs, primarily from the midbrain ventral tegmental area (VTA) and glutamatergic inputs from several cortical and sub-cortical structures. A population of glutamatergic neurons exists within the VTA and evidence from rats and mice suggests that these VTA axons may co-release dopamine and glutamate into the NAc. Our laboratory has used sexual experience in Syrian hamsters as a model of experience-dependent plasticity within the NAc. Given that both dopamine and glutamate are involved in this plasticity, it is important to determine whether these neurotransmitters are co-expressed within the mesolimbic pathway of hamsters. We therefore used immunofluorescent staining to investigate the possible co-localization of tyrosine hydroxylase (TH), a dopaminergic marker, and vesicular glutamate transporter 2 (VGLUT2), a glutamatergic marker, within the mesolimbic pathway. PCR analyses identified VGLUT2 gene expression in the VTA. No co-localization of TH and VGLUT2 protein was detected in NAc fibers, nor was there a difference in immunolabeling between males and females. Further studies are needed to resolve this absence of anatomical co-localization of TH and VGLUT2 in hamster striatal afferents with reports of functional co-release in other rodents.


Subject(s)
Neurons/metabolism , Nucleus Accumbens/metabolism , Tyrosine 3-Monooxygenase/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Animals , Cricetinae , Female , Male , Mesocricetus , Synapses/metabolism
12.
Horm Behav ; 61(2): 204-11, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22210198

ABSTRACT

In many species, including Syrian hamsters, the generation of male reproductive behavior depends critically on the perception of female odor cues from conspecifics in the environment. The behavioral response to these odors is mediated by a network of steroid-sensitive ventral forebrain nuclei including the medial amygdala (MA), posterior bed nucleus of the stria terminalis (BNST) and medial preoptic area (MPOA). Previous studies have demonstrated that each of these three nuclei is required for appropriate sexual behavior and that MA preferentially sends female odor information directly to BNST and MPOA. It is unknown, however, how the functional connections between MA and BNST and/or MPOA are organized to generate different aspects of reproductive behavior. Therefore, the following experiments used the asymmetrical pathway lesion technique to test the role of the functional connections between MA and BNST and/or MPOA in odor preference and copulatory behaviors. Lesions that functionally disconnected MA from MPOA eliminated copulatory behavior but did not affect odor preference. In contrast, lesions that functionally disconnected MA from BNST eliminated preference for volatile female odors but did not affect preference for directly contacted odors or copulatory behavior. These results therefore demonstrate a double dissociation in the functional connections required for attraction to volatile sexual odors and copulation and, more broadly, suggest that appetitive and consummatory reproductive behaviors are mediated by distinct neural pathways.


Subject(s)
Amygdala/physiology , Olfactory Perception/physiology , Preoptic Area/physiology , Septal Nuclei/physiology , Sexual Behavior, Animal/physiology , Animals , Appetitive Behavior/physiology , Copulation/physiology , Cricetinae , Female , Male , Mesocricetus , Neural Pathways/physiology , Odorants
13.
Physiol Behav ; 105(4): 1021-7, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22138441

ABSTRACT

Vaginal marking is a stereotyped scent marking behavior in female Syrian hamsters used to attract male hamsters for mating. Although the modulation of vaginal marking by hormones and odors is well understood, the motor control of this proceptive reproductive behavior remains unknown. Therefore, we used X-ray videography to visualize individual bone movements during vaginal marking. Kinematic analyses revealed several consistent motor patterns of vaginal marking. Despite exhibiting a diversity of trial-to-trial non-marking behaviors (e.g. locomotor stepping), we found that lowering and raising the pelvis consistently corresponded with coordinated flexion and extension cycles of the hip, knee, and tail, suggesting that these movements are fundamental to vaginal marking behavior. Surprisingly, we observed only small changes in the angles of the pelvic and sacral regions, suggesting previous reports of pelvic rotation during vaginal marking may need to be reconsidered. From these kinematic data, we inferred that vaginal marking is primarily due to the actions of hip and knee extensor muscles of the trailing leg working against gravity to support the weight of the animal as it controls the descent of the pelvis to the ground. The cutaneous trunci muscle likely mediates the characteristic flexion of the tail. Interestingly, this tail movement occurred on the same time scale as the joint kinematics suggesting possible synergistic recruitment of these muscle groups. These data therefore provide new targets for future studies examining the peripheral control of female reproductive behaviors.


Subject(s)
Animal Communication , Biomechanical Phenomena/physiology , Bone and Bones/physiology , Pheromones/physiology , Vagina/metabolism , X-Rays , Animals , Cricetinae , Female , Mesocricetus , Sexual Behavior, Animal/physiology
14.
Horm Behav ; 59(4): 536-48, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21316366

ABSTRACT

In many rodent species, including Syrian hamsters, the expression of appropriate social behavior depends critically on the perception and identification of conspecific odors. The behavioral response to these odors is mediated by a network of steroid-sensitive ventral forebrain nuclei including the medial amygdala (Me), posterior bed nucleus of the stria terminalis (BNST), and medial preoptic area (MPOA). Although it is well-known that Me, BNST, and MPOA are densely interconnected and each uniquely modulates odor-guided social behaviors, the degree to which conspecific odor information and steroid hormone cues are directly relayed between these nuclei is unknown. To answer this question, we injected the retrograde tracer, cholera toxin B (CTB), into the BNST or MPOA of male subjects and identified whether retrogradely-labeled cells in Me and BNST 1) expressed immediate early genes (IEGs) following exposure to male and/or female odors or 2) expressed androgen receptor (AR). Although few retrogradely-labeled cells co-localized with IEGs, a higher percentage of BNST- and MPOA-projecting cells in the posterior Me (MeP) expressed IEGs in response to female odors than to male odors. The percentage of retrogradely-labeled cells that expressed IEGs did not, however, differ between and female and male odor-exposed groups in the anterior Me (MeA), posterointermediate BNST (BNSTpi), or posteromedial BNST (BNSTpm). Many retrogradely-labeled cells co-localized with AR, and a higher percentage of retrogradely-labeled MeP and BNSTpm cells expressed AR than retrogradely-labeled MeA and BNSTpi cells, respectively. Together, these data demonstrate that Me, BNST, and MPOA interact as a functional circuit to process sex-specific odor cues and hormone information in male Syrian hamsters.


Subject(s)
Amygdala/physiology , Neurons/physiology , Olfactory Perception/physiology , Preoptic Area/physiology , Septal Nuclei/physiology , Analysis of Variance , Animals , Cricetinae , Cues , Female , Immunohistochemistry , Male , Mesocricetus , Neural Pathways/physiology , Neuronal Tract-Tracers , Odorants , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Androgen/metabolism
15.
Eur J Neurosci ; 32(3): 483-93, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20597978

ABSTRACT

In Syrian hamsters (Mesocricetus auratus), the expression of reproductive behavior requires the perception of social odors. The behavioral response to these odors is mediated by a network of ventral forebrain nuclei, including the posterior bed nucleus of the stria terminalis (pBNST). Previous studies have tested the role of the pBNST in reproductive behavior, but the use of large, fiber-damaging lesions in these studies make it difficult to attribute post-lesion deficits to the pBNST specifically. Thus, the current study used discrete, excitotoxic lesions of the pBNST to test the role of the pBNST in opposite-sex odor preference and copulatory behavior in both sexually-naive and sexually-experienced males. Lesions of the pBNST decreased sexually-naive males' investigation of volatile female odors, resulting in an elimination of opposite-sex odor preference. This elimination of preference was not due to a sensory deficit, as males with pBNST lesions were able to discriminate between odors. When, however, subjects were given sexual experience prior to pBNST lesions, their preference for volatile opposite-sex odors remained intact post-lesion. Similarly, when sexually-naive or sexually-experienced subjects were allowed to contact the social odors during the preference test, lesions of the pBNST decreased males' investigation of female odors but did not eliminate preference for opposite-sex odors, regardless of sexual experience. Finally, lesions of the pBNST delayed the copulatory sequence in sexually-naive, but not sexually-experienced, males such that they took longer to mount, intromit, ejaculate and display long intromissions. Together, these results demonstrate that the pBNST plays a unique and critical role in both appetitive and consummatory aspects of male reproductive behaviors.


Subject(s)
Odorants , Olfactory Perception/physiology , Septal Nuclei/physiology , Sexual Behavior, Animal/physiology , Smell/physiology , Analysis of Variance , Animals , Cricetinae , Female , Immunohistochemistry , Male , Mesocricetus , Olfactory Pathways/physiology , Radioimmunoassay , Sensory Thresholds/physiology , Volatilization
SELECTION OF CITATIONS
SEARCH DETAIL
...