Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Neurobiol Stress ; 31: 100651, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38933284

ABSTRACT

Stress is a major influence on mental health status; the ways that individuals respond to or copes with stressors determine whether they are negatively affected in the future. Stress responses are established by an interplay between genetics, environment, and life experiences. Psychosocial stress is particularly impactful during adolescence, a critical period for the development of mood disorders. In this study we compared two established, selectively-bred Sprague Dawley rat lines, the "internalizing" bred Low Responder (bLR) line versus the "externalizing" bred High Responder (bHR) line, to investigate how genetic temperament and adolescent environment impact future responses to social interactions and psychosocial stress, and how these determinants of stress response interact. Male bLR and bHR rats were exposed to social and environmental enrichment in adolescence prior to experiencing social defeat and were then assessed for social interaction and anxiety-like behavior. Adolescent enrichment caused rats to display more social interaction, as well as nominally less social avoidance, less submission during defeat, and resilience to the effects of social stress on corticosterone, in a manner that seemed more notable in bLRs. For bHRs, enrichment also caused greater aggression during a neutral social encounter and nominally during defeat, and decreased anxiety-like behavior. To explore the neurobiology underlying the development of social resilience in the anxious phenotype bLRs, RNA-seq was conducted on the hippocampus and nucleus accumbens, two brain regions that mediate stress regulation and social behavior. Gene sets previously associated with stress, social behavior, aggression and exploratory activity were enriched with differential expression in both regions, with a particularly large effect on gene sets that regulate social behaviors. Our findings provide further evidence that adolescent enrichment can serve as an inoculating experience against future stressors. The ability to induce social resilience in a usually anxious line of animals by manipulating their environment has translational implications, as it underscores the feasibility of intervention strategies targeted at genetically vulnerable adolescent populations.

2.
bioRxiv ; 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38645129

ABSTRACT

Stress is a major influence on mental health status; the ways that individuals respond to or copes with stressors determine whether they are negatively affected in the future. Stress responses are established by an interplay between genetics, environment, and life experiences. Psychosocial stress is particularly impactful during adolescence, a critical period for the development of mood disorders. In this study we compared two established, selectively-bred Sprague Dawley rat lines, the "internalizing" bred Low Responder (bLR) line versus the "externalizing" bred High Responder (bHR) line, to investigate how genetic temperament and adolescent environment impact future responses to social interactions and psychosocial stress, and how these determinants of stress response interact. Male bLR and bHR rats were exposed to social and environmental enrichment in adolescence prior to experiencing social defeat and were then assessed for social interaction and anxiety-like behavior. Adolescent enrichment caused rats to display more social interaction, as well as nominally less social avoidance, less submission during defeat, and resilience to the effects of social stress on corticosterone, in a manner that seemed more notable in bLRs. For bHRs, enrichment also caused greater aggression during a neutral social encounter and nominally during defeat, and decreased anxiety-like behavior. To explore the neurobiology underlying the development of social resilience in the anxious phenotype bLRs, RNA-seq was conducted on the hippocampus and nucleus accumbens, two brain regions that mediate stress regulation and social behavior. Gene sets previously associated with stress, social behavior, aggression and exploratory activity were enriched with differential expression in both regions, with a particularly large effect on gene sets that regulate social behaviors. Our findings provide further evidence that adolescent enrichment can serve as an inoculating experience against future stressors. The ability to induce social resilience in a usually anxious line of animals by manipulating their environment has translational implications, as it underscores the feasibility of intervention strategies targeted at genetically vulnerable adolescent populations.

3.
Neurobiol Stress ; 17: 100437, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35242893

ABSTRACT

Evaluating and coping with stressful social events as they unfold is a critical strategy in overcoming them without long-lasting detrimental effects. Individuals display a wide range of responses to stress, which can manifest in a variety of outcomes for the brain as well as subsequent behavior. Chronic Social Defeat Stress (CSDS) in mice has been widely used to model individual variation following a social stressor. Following a course of repeated intermittent psychological and physical stress, mice diverge into separate populations of social reactivity: resilient (socially interactive) and susceptible (socially avoidant) animals. A rich body of work reveals distinct neurobiological and behavioral consequences of this experience that map onto the resilient and susceptible groups. However, the range of factors that emerge over the course of defeat have not been fully described. Therefore, in the current study, we focused on characterizing behavioral, physiological, and neuroendocrine profiles of mice in three separate phases: before, during, and following CSDS. We found that following CSDS, traditional read-outs of anxiety-like and depression-like behaviors do not map on to the resilient and susceptible groups. By contrast, behavioral coping strategies used during the initial social stress encounter better predict which mice will eventually become resilient or susceptible. In particular, mice that will emerge as susceptible display greater escape behavior on Day 1 of social defeat than those that will emerge as resilient, indicating early differences in coping mechanisms used between the two groups. We further show that the social avoidance phenotype in susceptible mice is specific to the aggressor strain and does not generalize to conspecifics or other strains, indicating that there may be features of threat discrimination that are specific to the susceptible mice. Our findings suggest that there are costs and benefits to both the resilient and susceptible outcomes, reflected in their ability to cope and adapt to the social stressor.

4.
Transl Psychiatry ; 12(1): 105, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35292624

ABSTRACT

Microglia play critical roles in healthy brain development and function, as well as the neuropathology underlying a range of brain diseases. Despite evidence for a role of microglia in affective regulation and mood disorders, little is known regarding how variation in microglia status relates to individual differences in emotionality. Using a selective breeding model, we have generated rat lines with unique temperamental phenotypes that reflect broad emotional traits: bred low responder rats (bLRs) are novelty-averse and model a passive coping style, whereas bred high responder rats (bHRs) are highly exploratory and model an active coping style. To identify a functional role of microglia in these phenotypes, we administered minocycline, an antibiotic with potent microglia inhibiting properties and observed shifts in forced swim, sucrose preference, and social interaction behaviors in bLRs. Using detailed anatomical analyses, we compared hippocampal microglia profiles of bHRs and bLRs and found that although the lines had similar numbers of microglia, selective breeding was associated with a shift in the morphological features of these cells. Specifically, microglia from bLRs were characterized by a hyper-ramified morphology, with longer processes and more complicated branching patterns than microglia from bHRs. This morphology is thought to reflect an early stage of microglia activation and suggests that bLR microglia are in a reactive state even when animals are not overtly challenged. Taken together, our results provide novel evidence linking variation in inborn temperament with differences in the baseline status of microglia and implicate a role for microglia in shaping enduring emotional characteristics.


Subject(s)
Microglia , Temperament , Animals , Emotions/physiology , Problem Solving , Rats , Selective Breeding
5.
Bio Protoc ; 10(15): e3699, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-33659365

ABSTRACT

Stress is crucial to the survival of an organism, but excessive stress can lead to psychological disorders including depression, anxiety, substance abuse, and suicidality. The prevailing notion is that chronic stress promotes adverse outcomes on brain and body health, whereas acute stressors are generally benign. Notably, acute events such mass shootings or natural disasters are now emerging as significant sources of cognitive and emotional problems including post-traumatic stress disorder (PTSD). These events are characterized by the simultaneous occurrence of physical, emotional, and social stresses, which last minutes to hours. Hence, there is a need to model such multiple concurrent acute stresses (MAS) to uncover the mechanisms by which they lead to profound adverse outcomes. The MAS paradigm described here involves simultaneously exposing a rodent to several different stressors including restraint, crowding, and jostling alongside peers in a brightly lit and very noisy environment. Moreover, the MAS paradigm can be used once or imposed repeatedly to emulate complex, repeated modern life stresses, advancing our mechanistic understanding of consequent mental and cognitive impairments.

6.
Neuropsychopharmacology ; 45(3): 515-523, 2020 02.
Article in English | MEDLINE | ID: mdl-31698409

ABSTRACT

In humans, early-life adversity is associated with impairments in learning and memory that may emerge later in life. In rodent models, early-life adversity directly impacts hippocampal neuron structure and connectivity with progressive deficits in long-term potentiation and spatial memory function. Previous work has demonstrated that augmented release and actions of the stress-activated neuropeptide, CRH, contribute to the deleterious effects of early-life adversity on hippocampal dendritic arborization, synapse number and memory-function. Early-life adversity increases hippocampal CRH expression, and blocking hippocampal CRH receptor type-1 (CRHR1) immediately following early-life adversity prevented the consequent memory and LTP defects. Here, we tested if blocking CRHR1 in young adults ameliorates early-life adversity-provoked memory deficits later in life. A weeklong course of a CRHR1 antagonist in 2-month-old male rats prevented early-life adversity-induced deficits in object recognition memory that emerged by 12 months of age. Surprisingly, whereas the intervention did not mitigate early-life adversity-induced spatial memory losses at 4 and 8 months, it restored hippocampus-dependent location memory in 12-month-old rats that experienced early-life adversity. Neither early-life adversity nor CRHR1 blockade in the adult influenced anxiety- or depression-related behaviors. Altogether, these findings suggest that cognitive deficits attributable to adversity during early-life-sensitive periods are at least partially amenable to interventions later in life.


Subject(s)
Aging/psychology , Memory Disorders/prevention & control , Memory Disorders/psychology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological/drug therapy , Stress, Psychological/psychology , Age Factors , Aging/drug effects , Aging/physiology , Animals , Animals, Newborn , Female , Injections, Intraventricular , Male , Memory Disorders/etiology , Pregnancy , Pyrimidines/administration & dosage , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/physiology , Stress, Psychological/complications
7.
Brain Res ; 1715: 106-114, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30880118

ABSTRACT

In previous studies, early-life fibroblast growth factor-2 (FGF2) administration conferred resilience to developing anxiety-like behavior in vulnerable animals in adulthood. To follow up on this work, we administered FGF2 the day after birth to animals that differ in emotional behavior and further explored its long-term effects on affective behavior and circuitry. Selectively-bred "high responder" rats (bHRs) exhibit low levels of anxiety-like and depression-like behavior, whereas selectively-bred "low responders" (bLRs) display high levels of anxiety-like and depression-like behavior. We found that early-life administration of FGF2 decreased negative affect in bLRs during the early post-natal period, as indexed by 40 kHz ultrasonic vocalizations (USVs) in response to a brief maternal separation on PND11. FGF2 also increased positive affect during the juvenile period, as measured by 50 kHz USVs in response to heterospecific hand play ("tickling") after weaning. In general, we found that bHRs produced more 50 kHz USVs than bLRs. In adulthood, we measured opioid ligand and receptor expression in brain regions implicated in USV production and affect regulation by mRNA in situ hybridization. Within multiple affective brain regions, bHRs had greater expression of the mu opioid receptor than bLRs. FGF2 increased mu opioid expression in bLRs. The bLRs had more kappa and less delta receptor expression than bHRs, and FGF2 increased prodynorphin in bLRs. Our results provide support for further investigations into the role of growth factors and endogenous opioids in the treatment of disorders characterized by altered affect, such as anxiety and depression.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Receptors, Opioid, mu/metabolism , Vocalization, Animal/physiology , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Emotions , Exploratory Behavior/drug effects , Fibroblast Growth Factor 2/metabolism , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Ultrasonic Waves , Vocalization, Animal/drug effects
8.
Horm Behav ; 86: 64-70, 2016 11.
Article in English | MEDLINE | ID: mdl-27693608

ABSTRACT

We aimed to determine the short-term effects of early-life stress in the form of maternal separation (MS) on anxiety-like behavior in male rat pups. In order to assess anxiety, we measured 40kHz separation-induced ultrasonic vocalizations (USV) on postnatal day (PND) 11. We further aimed to evaluate the potential involvement of two neurochemical systems known to regulate social and anxiety-like behaviors throughout life: oxytocin (OT) and fibroblast growth factor 2 (FGF2). For these purposes, we tested the effects of neonatal administration (on PND1) of an acute dose of FGF2 on USV and its potential interaction with MS. In addition, we validated the anxiolytic effects of OT and measured oxytocin receptor (OTR) gene expression, binding and epigenetic regulation via histone acetylation. Our results show that MS potentiated USV while acute administration of OT and FGF2 attenuated them. Further, we found that both FGF2 and MS increased OTR gene expression and the association of acH3K14 with the OTR promoter in the bed nucleus of the stria terminalis (BNST). Comparable changes, though not as pronounced, were also found for the central amygdala (CeA). Our findings suggest that FGF2 may exert its anxiolytic effects in male MS rats by a compensatory increase in the acetylation of the OTR promoter to overcome reduced OT levels in the BNST.


Subject(s)
Anxiety/genetics , Behavior, Animal/drug effects , Fibroblast Growth Factor 2/pharmacology , Growth and Development/drug effects , Maternal Deprivation , Receptors, Oxytocin/genetics , Septal Nuclei/drug effects , Animals , Animals, Newborn , Anxiety/metabolism , Anxiety/physiopathology , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/growth & development , Central Amygdaloid Nucleus/metabolism , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Growth and Development/genetics , Male , Oxytocin/metabolism , Oxytocin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Oxytocin/metabolism , Septal Nuclei/growth & development , Septal Nuclei/metabolism
9.
Hippocampus ; 26(12): 1618-1632, 2016 12.
Article in English | MEDLINE | ID: mdl-27657911

ABSTRACT

Memory and related cognitive functions are progressively impaired in a subgroup of individuals experiencing childhood adversity and stress. However, it is not possible to identify vulnerable individuals early, a crucial step for intervention. In this study, high-resolution magnetic resonance imaging (MRI) and intra-hippocampal diffusion tensor imaging (DTI) were employed to examine for structural signatures of cognitive adolescent vulnerabilities in a rodent model of early-life adversity. These methods were complemented by neuroanatomical and functional assessments of hippocampal network integrity during adolescence, adulthood and middle-age. The high-resolution MRI identified selective loss of dorsal hippocampal volume, and intra-hippocampal DTI uncovered disruption of dendritic structure, consistent with disrupted local connectivity, already during late adolescence in adversity-experiencing rats. Memory deteriorated over time, and stunting of hippocampal dendritic trees was apparent on neuroanatomical analyses. Thus, disrupted hippocampal neuronal structure and connectivity, associated with cognitive impairments, are detectable via non-invasive imaging modalities in rats experiencing early-life adversity. These high-resolution imaging approaches may constitute promising tools for prediction and assessment of at-risk individuals in the clinic. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hippocampus/diagnostic imaging , Memory Disorders/diagnostic imaging , Memory Disorders/etiology , Stress, Psychological/complications , Stress, Psychological/diagnostic imaging , Animals , Cohort Studies , Corticosterone/blood , Crowding , Diffusion Tensor Imaging , Environment , Female , Hippocampus/growth & development , Hippocampus/pathology , Housing, Animal , Light , Magnetic Resonance Imaging , Male , Memory Disorders/blood , Memory Disorders/pathology , Models, Animal , Noise , Organ Size , Pyramidal Cells/pathology , Radioimmunoassay , Random Allocation , Rats, Sprague-Dawley , Stress, Psychological/blood , Stress, Psychological/pathology
10.
Neurobiol Stress ; 2: 10-19, 2015.
Article in English | MEDLINE | ID: mdl-25884016

ABSTRACT

Epilepsy is more prevalent in populations with high measures of stress, but the neurobiological mechanisms are unclear. Stress is a common precipitant of seizures in individuals with epilepsy, and may provoke seizures by several mechanisms including changes in neurotransmitter and hormone levels within the brain. Importantly, stress during sensitive periods early in life contributes to 'brain programming', influencing neuronal function and brain networks. However, it is unclear if early-life stress influences limbic excitability and promotes epilepsy. Here we used an established, naturalistic model of chronic early-life stress (CES), and employed chronic cortical and limbic video-EEGs combined with molecular and cellular techniques to probe the contributions of stress to age-specific epilepsies and network hyperexcitability and identify the underlying mechanisms. In control male rats, EEGs obtained throughout development were normal and no seizures were observed. EEGs demonstrated epileptic spikes and spike series in the majority of rats experiencing CES, and 57% of CES rats developed seizures: Behavioral events resembling the human age-specific epilepsy infantile spasms occurred in 11/23 (48%), accompanied by EEG spikes and/or electrodecrements, and two additional rats (9%) developed limbic seizures that involved the amygdala. Probing for stress-dependent, endogenous convulsant molecules within amygdala, we examined the expression of the pro-convulsant neuropeptide corticotropin-releasing hormone (CRH), and found a significant increase of amygdalar--but not cortical--CRH expression in adolescent CES rats. In conclusion, CES of limited duration has long-lasting effects on brain excitability and may promote age-specific seizures and epilepsy. Whereas the mechanisms involved require further study, these findings provide important insights into environmental contributions to early-life seizures.

11.
Dev Psychobiol ; 56(8): 1675-88, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24910169

ABSTRACT

A close association between early-life experience and cognitive and emotional outcomes is found in humans. In experimental models, early-life experience can directly influence a number of brain functions long-term. Specifically, and often in concert with genetic background, experience regulates structural and functional maturation of brain circuits and alters individual neuronal function via large-scale changes in gene expression. Because adverse experience during sensitive developmental periods is often associated with neuropsychiatric disease, there is an impetus to create realistic models of distinct early-life experiences. These can then be used to study causality between early-life experiential factors and cognitive and emotional outcomes, and to probe the underlying mechanisms. Although chronic early-life stress has been linked to the emergence of emotional and cognitive disorders later in life, most commonly used rodent models of involve daily maternal separation and hence intermittent early-life stress. We describe here a naturalistic and robust chronic early-life stress model that potently influences cognitive and emotional outcomes. Mice and rats undergoing this stress develop structural and functional deficits in a number of limbic-cortical circuits. Whereas overt pathological memory impairments appear during adulthood, emotional and cognitive vulnerabilities emerge already during adolescence. This naturalistic paradigm, widely adopted around the world, significantly enriches the repertoire of experimental tools available for the study of normal brain maturation and of cognitive and stress-related disorders including depression, autism, post-traumatic stress disorder, and dementia.


Subject(s)
Animals, Newborn , Brain , Disease Models, Animal , Stress, Psychological , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Animals, Newborn/physiology , Brain/growth & development , Brain/metabolism , Brain/physiopathology , Mice , Rats , Stress, Psychological/complications , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
12.
Trends Neurosci ; 35(5): 315-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22386641

ABSTRACT

Learning and memory processes carried out within the hippocampus are influenced by stress in a complex manner, and the mechanisms by which stress modulates the physiology of the hippocampus are not fully understood. This review addresses how the production and release of the neuropeptide corticotropin-releasing hormone (CRH) within the hippocampus during stress influences neuronal structure and hippocampal function. CRH functions in the contexts of acute and chronic stresses taking place during development, adulthood and aging. Current challenges are to uncover how the dynamic actions of CRH integrate with the well-established roles of adrenal-derived steroid stress hormones to shape the cognitive functions of the hippocampus in response to stress.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Dendritic Spines/pathology , Hippocampus/physiology , Stress, Psychological/metabolism , Animals , Humans , Stress, Psychological/physiopathology
13.
J Neurosci ; 30(39): 13005-15, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20881118

ABSTRACT

Chronic stress impairs learning and memory in humans and rodents and disrupts long-term potentiation (LTP) in animal models. These effects are associated with structural changes in hippocampal neurons, including reduced dendritic arborization. Unlike the generally reversible effects of chronic stress on adult rat hippocampus, we have previously found that the effects of early-life stress endure and worsen during adulthood, yet the mechanisms for these clinically important sequelae are poorly understood. Stress promotes secretion of the neuropeptide corticotropin-releasing hormone (CRH) from hippocampal interneurons, activating receptors (CRF(1)) located on pyramidal cell dendrites. Additionally, chronic CRF(1) occupancy negatively affects dendritic arborization in mouse organotypic slice cultures, similar to the pattern observed in middle-aged, early-stressed (CES) rats. Here we found that CRH expression is augmented in hippocampus of middle-aged CES rats, and then tested whether the morphological defects and poor memory performance in these animals involve excessive activation of CRF(1) receptors. Central or peripheral administration of a CRF(1) blocker following the stress period improved memory performance of CES rats in novel-object recognition tests and in the Morris water maze. Consonant with these effects, the antagonist also prevented dendritic atrophy and LTP attenuation in CA1 Schaffer collateral synapses. Together, these data suggest that persistently elevated hippocampal CRH-CRF(1) interaction contributes importantly to the structural and cognitive impairments associated with early-life stress. Reducing CRF(1) occupancy post hoc normalized hippocampal function during middle age, thus offering potential mechanism-based therapeutic interventions for children affected by chronic stress.


Subject(s)
Cognition Disorders/metabolism , Corticotropin-Releasing Hormone/metabolism , Hippocampus/metabolism , Neurons/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/metabolism , Animals , Animals, Newborn , Chronic Disease , Cognition Disorders/physiopathology , Disease Models, Animal , Female , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Transgenic , Neurons/pathology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/physiology , Stress, Psychological/physiopathology
14.
Eur J Neurosci ; 32(3): 469-82, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20704594

ABSTRACT

In Syrian hamsters, reproductive behavior relies on the perception of chemical signals released from conspecifics. The medial amygdala (MEA) processes sexual odors through functionally distinct, but interconnected, sub-regions; the anterior MEA (MEAa) appears to function as a chemosensory filter to distinguish between opposite-sex and same-sex odors, whereas the posterodorsal MEA (MEApd) is critical for generating attraction specifically to opposite-sex odors. To identify how these sub-regions interact during odor processing, we measured odor-induced Fos expression, an indirect marker of neuronal activation, in the absence of either MEAa or MEApd processing. In Experiment 1, electrolytic lesions of the MEAa decreased Fos expression throughout the posterior MEA in male hamsters exposed to either female or male odors, whereas MEApd lesions had no effect on Fos expression within the MEAa. These results indicate that the MEAa normally enhances processing of sexual odors within the MEApd and that this interaction is primarily unidirectional. Furthermore, lesions of the MEAa, but not the MEApd, decreased Fos expression within several connected forebrain nuclei, suggesting that the MEAa provides the primary excitatory output of the MEA during sexual odor processing. In Experiment 2, we observed a similar pattern of decreased Fos expression, using fiber-sparing, NMDA lesions of the MEAa, suggesting that the decreases in Fos expression were not attributable exclusively to damage to passing fibers. Taken together, these results provide the first direct test of how the different sub-regions within the MEA interact during odor processing, and highlight the role of the MEAa in transmitting sexual odor information to the posterior MEA, as well as to related forebrain nuclei.


Subject(s)
Amygdala/physiology , Odorants , Olfactory Pathways/physiology , Olfactory Perception/physiology , Prosencephalon/physiology , Sexual Behavior, Animal/physiology , Animals , Cricetinae , Female , Immunohistochemistry , Male , Mesocricetus , Proto-Oncogene Proteins c-fos/metabolism
15.
Physiol Behav ; 94(4): 545-51, 2008 Jul 05.
Article in English | MEDLINE | ID: mdl-18485425

ABSTRACT

Rodent reproductive behavior relies heavily on odor processing, and evidence suggests that many odor-guided sexual behaviors are shaped by prior experience. We sought to determine if exposure to male odors during development is required for the adult expression of proceptive sexual behavior toward male odors in female Syrian hamsters. Exposure to male odors was restricted in naïve subjects by removing all male siblings from the litter at three to five days of age. Control litters were also culled, but included equal numbers of male and female pups. As adults, naïve females displayed investigatory preferences toward male odors in a Y-maze that were comparable to control females; this preference was observed whether contact with the odor stimuli was prevented of allowed. In contrast, naïve females vaginal scent-marked equally toward male and female volatile odors, suggesting an inability to target behavior toward sexually relevant odors. However, naïve females marked preferentially toward male odors when allowed to contact the odor stimuli. These results provide evidence for the experience-dependent development of vaginal marking behavior toward volatile components of sexual odors. Furthermore, they suggest that distinct mechanisms regulate the development of odor preferences and vaginal marking behavior in this species.


Subject(s)
Animal Communication , Choice Behavior/physiology , Discrimination Learning/physiology , Sexual Behavior, Animal/physiology , Smell/physiology , Age Factors , Animals , Cricetinae , Critical Period, Psychological , Estrous Cycle , Female , Male , Mesocricetus , Pheromones/metabolism , Recognition, Psychology/physiology , Scent Glands/physiology , Vagina/metabolism
16.
Physiol Behav ; 91(1): 134-41, 2007 May 16.
Article in English | MEDLINE | ID: mdl-17374544

ABSTRACT

Proceptive and receptive behaviors of female rodents, such as golden hamsters, are often regulated by changes in circulating levels of ovarian hormones. However, less is known about how ovarian hormones might regulate female hamster's attraction and preference for volatile odor from males. To evaluate this, we assessed female preference by recording investigation and proximity to male and female volatile odorants in a Y-maze across all days of the estrous cycle (Experiments 1 and 2) or following ovariectomy (Experiment 3). In Experiment 1, female subjects were tested four times, once on each day of their estrous cycle. Females showed a preference for male odors on diestrus day 1 and to a lesser degree on proestrus, but showed no preference on the day of behavioral estrus. Irrespective of cycle day, preference was apparent in the first few days of testing and disappeared by the fourth day, suggesting that repeated testing attenuated female preference. To avoid this problem, in Experiment 2 each animal was tested only on one day of the 4-day estrous cycle. Female preference for male volatile odors over those from females was observed on each day of their estrous cycle, including estrus. Moreover, following gonadectomy (Experiment 3) female hamsters still preferred male volatile odors to those of females. Taken together, this suggests that circulating levels of gonadal hormones do not influence preference for male volatile odors in female hamsters.


Subject(s)
Gonadal Steroid Hormones/blood , Gonadal Steroid Hormones/physiology , Odorants , Sexual Behavior, Animal/drug effects , Animals , Cricetinae , Estrous Cycle/physiology , Exploratory Behavior/drug effects , Female , Male , Mesocricetus , Ovariectomy
17.
Eur J Neurosci ; 24(12): 3541-52, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17229102

ABSTRACT

In rodent species, such as the Syrian hamster, the expression of sexual preference requires neural integration of social chemosensory signals and steroid hormone cues. Although anatomical data suggest that separate pathways within the nervous system process these two signals, the functional significance of this separation is not well understood. Specifically, within the medial amygdala, the anterior region (MEa) receives input from the olfactory bulbs and other chemosensory areas, whereas the posterodorsal region (MEpd) contains a dense population of steroid receptors and receives less substantial chemosensory input. Consequently, the MEa may subserve a primarily discriminative function, whereas the MEpd may mediate the permissive effects of sex steroids on sexual preference. To test these hypotheses, we measured preference and attraction to female and male odors in males with lesions of either the MEa or MEpd. In Experiment 1, lesions of either region eliminated opposite-sex odor preferences. Importantly, MEpd-lesioned males displayed decreased attraction toward female odors, suggesting decreased sexual motivation. In contrast, MEa-lesioned males displayed high levels of investigation of both male and female odors, suggesting an inability to categorize the relevance of the odor stimuli. In Experiment 2, we verified that both MEa- and MEpd-lesioned males could discriminate between female and male odors, thereby eliminating the possibility that the observed lack of preference reflected a sensory deficit. Taken together, these results suggest that both the MEa and MEpd are critical for the expression of opposite-sex odor preference, although they appear to mediate distinct aspects of this behavior.


Subject(s)
Amygdala/physiology , Odorants , Sex Characteristics , Sexual Behavior, Animal/physiology , Steroids/blood , Amygdala/anatomy & histology , Analysis of Variance , Animals , Behavior, Animal , Cricetinae , Discrimination, Psychological/physiology , Functional Laterality , Male , Mesocricetus , Radioimmunoassay/methods , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...