Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Horm Behav ; 125: 104815, 2020 09.
Article in English | MEDLINE | ID: mdl-32640197

ABSTRACT

Decision-making is a complex process essential to daily adaptation in many species. Risk is an inherent aspect of decision-making and it is influenced by gonadal hormones. Testosterone and 17ß-estradiol may modulate decision making and impact the mesocorticolimbic dopamine pathway. Here, we explored sex differences, the effect of gonadal hormones and the dopamine agonist amphetamine on risk-based decision making. Intact or gonadectomised (GDX) male and female rats underwent to a probabilistic discounting task. High and low doses of testosterone propionate (1.0 or 0.2 mg) and 17ß-estradiol benzoate (0.3 µg) were administered to assess acute effects on risk-based decision making. After 3-days of washout period, intact and GDX rats received high or low (0.5 or 0.125 mg/kg) doses of amphetamine and re-tested in the probabilistic discounting task. Under baseline conditions, males made more risky choices during probability discounting compared to female rats, particularly in the lower probability blocks, but GDX did not influence risky choice. The high, but not the low dose, of testosterone modestly reduced risky decision making in GDX male rats. Conversely, 17ß-estradiol had no significant effect on risky choice regardless of GDX status in either sex. Lastly, a higher dose of amphetamine increased risky decision making in both intact males and females, but had no effect in GDX rats. These findings demonstrated sex differences in risk-based decision making, with males showing a stronger bias toward larger, uncertain rewards. GDX status influenced the effects of amphetamine, suggesting different dopaminergic regulation in risk-based choices among males and females.


Subject(s)
Amphetamine/pharmacology , Cognition , Decision Making , Sex Characteristics , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Castration , Cognition/drug effects , Cognition/physiology , Decision Making/drug effects , Decision Making/physiology , Delay Discounting/drug effects , Delay Discounting/physiology , Dopamine/pharmacology , Dopamine Agonists/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Male , Rats , Rats, Long-Evans , Reward , Risk Reduction Behavior , Testosterone/pharmacology
2.
Endocrinology ; 160(9): 2128-2136, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31219567

ABSTRACT

Androgens (testosterone and DHT) increase adult hippocampal neurogenesis by increasing survival of new neurons in male rats and mice via an androgen receptor pathway, but it is not known whether androgens regulate neurogenesis in female rats and whether the effect is age-dependent. We investigated the effects of DHT, a potent androgen, on neurogenesis in young adult and middle-aged male and female rats. Rats were gonadectomized and injected with the DNA synthesis marker bromodeoxyuridine (BrdU). The following day, rats began receiving daily injections of oil or DHT for 30 days. We evaluated cell proliferation (Ki67) and survival of new neurons (BrdU and BrdU/NeuN) in the hippocampus of male and female rats by using immunohistochemistry. As expected, DHT increased the number of BrdU+ cells in young males but surprisingly not in middle-aged males or in young and middle-aged females. In middle age, DHT increased the proportion of BrdU/NeuN cells, an effect driven by females. Androgen receptor expression also increased with aging in both female and male rats, which may contribute to a lack of DHT neurogenic effect in middle age. Our results indicate that DHT regulates adult hippocampal neurogenesis in a sex- and age-dependent manner.


Subject(s)
Dihydrotestosterone/pharmacology , Hippocampus/drug effects , Neurogenesis/drug effects , Age Factors , Animals , Female , Hippocampus/physiology , Male , Rats , Rats, Sprague-Dawley , Sex Factors
3.
Horm Behav ; 87: 69-79, 2017 01.
Article in English | MEDLINE | ID: mdl-27984032

ABSTRACT

The hippocampus and dorsal striatum are important structures involved in place and response learning strategies respectively. Both sex and estrous cycle phase differences in learning strategy preference exist following cue competition paradigms. Furthermore, significant effects of sex and learning strategy on hippocampal neural plasticity have been reported. However, associations between learning strategy and immediate early gene (IEG) expression in the hippocampus and dorsal striatum are not completely understood. In the current study we investigated the effects of sex and estrous cycle phase on strategy choice and IEG expression in the hippocampus and dorsal striatum of rats following cue competition training in the Morris water maze. We found that proestrous rats were more likely to choose a place strategy than non-proestrous or male rats. Although male cue strategy users travelled greater distances than the other groups on the first day of training, there were no other sex or strategy differences in the ability to reach a hidden or a visible platform. Female place strategy users exhibited greater zif268 expression and male place strategy users exhibited greater cFos expression compared to all other groups in CA3. Furthermore, cue strategy users had greater expression of cFos in the dorsal striatum than place strategy users. Shorter distances to reach a visible platform were associated with less activation of cFos in CA3 and CA1 of male place strategy users. Our findings indicate multiple differences in brain activation with sex and strategy use, despite limited behavioral differences between the sexes on this cue competition paradigm.


Subject(s)
Competitive Behavior/physiology , Estrous Cycle/physiology , Genes, Immediate-Early/genetics , Hippocampus/metabolism , Maze Learning/physiology , Animals , Choice Behavior/physiology , Cues , Dentate Gyrus/metabolism , Female , Male , Rats , Rats, Sprague-Dawley , Sex Characteristics , Sexual Behavior/physiology , Transcriptional Activation
4.
Neuropharmacology ; 107: 278-293, 2016 08.
Article in English | MEDLINE | ID: mdl-27018449

ABSTRACT

Depression is more prevalent in women than in men, and women are at a heightened risk for depression during the postpartum and perimenopause. There is also evidence to suggest that the ovarian hormone milieu may dictate antidepressant efficacy. Thus, it is important to investigate the role of ovarian hormones in the pathogenesis of depression and in the mechanisms that may underlie antidepressant efficacy. In the present study, we used 10-month-old female Sprague-Dawley rats to examine the effects of long-term ovarian hormone deprivation on the development of depressive-like endophenotypes after chronic stress, and on antidepressant efficacy. Four months following ovariectomy (OVX) or sham surgery, all rats were subjected to 6 weeks of chronic unpredictable stress (CUS). During the last 3 weeks of CUS, rats received daily injections of fluoxetine (5 mg/kg) or vehicle. All rats were assessed on measures of anxiety- and depressive-like behavior, hypothalamic-pituitary-adrenal (HPA) negative feedback inhibition, and on markers of neurogenesis and microglia in the dentate gyrus. Our findings demonstrate that long-term ovarian hormone deprivation increased anxiety and depressive-like behavior, as seen by increased immobility in the forced swim test and latency to feed in the novelty suppressed feeding test, and decreased sucrose preference. Further, long-term OVX resulted in impaired HPA negative feedback inhibition, as seen in the dexamethasone suppression test. Fluoxetine treatment showed limited behavioral and neuroendocrine efficacy, however it reduced microglial (Iba-1) expression, and increased cell proliferation, neurogenesis (via cell survival), and the expression of the polysialylated neuronal cell adhesion molecule (PSA-NCAM) in the dentate gyrus, although these effects varied by region (dorsal, ventral) and ovarian status. Taken together, our findings demonstrate that ovarian hormones may impart resilience against the behavioral and neuroendocrine consequences of chronic unpredictable stress, and may modulate the effects of fluoxetine on cell proliferation, neurogenesis, and PSA-NCAM in the middle-aged female.


Subject(s)
Corticosterone/blood , Fluoxetine/therapeutic use , Ovariectomy/adverse effects , Ovary/metabolism , Stress, Psychological/blood , Stress, Psychological/drug therapy , Age Factors , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Disease Models, Animal , Estradiol/blood , Female , Fluoxetine/pharmacology , Ovariectomy/trends , Rats , Rats, Sprague-Dawley , Stress, Psychological/psychology
5.
Front Neuroendocrinol ; 41: 129-52, 2016 04.
Article in English | MEDLINE | ID: mdl-26988999

ABSTRACT

Neurogenesis within the adult hippocampus is modulated by endogenous and exogenous factors. Here, we review the role of sex hormones in the regulation of adult hippocampal neurogenesis in males and females. The review is framed around the potential functional implications of sex hormone regulation of adult hippocampal neurogenesis, with a focus on cognitive function and mood regulation, which may be related to sex differences in incidence and severity of dementia and depression. We present findings from preclinical studies of endogenous fluctuations in sex hormones relating to reproductive function and ageing, and from studies of exogenous hormone manipulations. In addition, we discuss the modulating roles of sex, age, and reproductive history on the relationship between sex hormones and neurogenesis. Because sex hormones have diverse targets in the central nervous system, we overview potential mechanisms through which sex hormones may influence hippocampal neurogenesis. Lastly, we advocate for a more systematic consideration of sex and sex hormones in studying the functional implications of adult hippocampal neurogenesis.


Subject(s)
Gonadal Steroid Hormones/physiology , Hippocampus/physiology , Neurogenesis/physiology , Adult , Animals , Female , Hippocampus/metabolism , Humans , Male
6.
Horm Behav ; 79: 58-69, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26774465

ABSTRACT

Hypogonadal men are more likely to develop depression, while testosterone supplementation shows antidepressant-like effects in hypogonadal men and facilitates antidepressant efficacy. Depression is associated with hypothalamic-pituitary-adrenal (HPA) axis hyperactivity and testosterone exerts suppressive effects on the HPA axis. The hippocampus also plays a role in the feedback regulation of the HPA axis, and depressed patients show reduced hippocampal neuroplasticity. We assessed the antidepressant-like effects of testosterone with, or without, imipramine on behavioral and neural endophenotypes of depression in a chronic unpredictable stress (CUS) model of depression. A 21-day CUS protocol was used on gonadectomized male Sprague-Dawley rats treated with vehicle, 1mg of testosterone propionate, 10mg/kg of imipramine, or testosterone and imipramine in tandem. Testosterone treatment reduced novelty-induced hypophagia following CUS exposure, but not under non-stress conditions, representing state-dependent effects. Further, testosterone increased the latency to immobility in the forced swim test (FST), reduced basal corticosterone, and reduced adrenal mass in CUS-exposed rats. Testosterone also facilitated the effects of imipramine by reducing the latency to immobility in the FST and increasing sucrose preference. Testosterone treatment had no significant effect on neurogenesis, though the combination of testosterone and imipramine increased PSA-NCAM expression in the ventral dentate gyrus. These findings demonstrate the antidepressant- and anxiolytic-like effects of testosterone within a CUS model of depression, and provide insight into the mechanism of action, which appears to be independent of enhanced hippocampal neurogenesis.


Subject(s)
Antidepressive Agents/pharmacology , Imipramine/pharmacology , Neuronal Plasticity/drug effects , Stress, Psychological/psychology , Testosterone/pharmacology , Animals , Chronic Disease , Corticosterone/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Depression/pathology , Depression/prevention & control , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Male , Neurogenesis/drug effects , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/drug therapy , Stress, Psychological/pathology
7.
Neuropsychopharmacology ; 41(6): 1670-80, 2016 May.
Article in English | MEDLINE | ID: mdl-26530284

ABSTRACT

Antidepressant drugs are too often ineffective, the exact mechanism of efficacy is still ambiguous, and there has been a paucity of novel targets for pharmacotherapy. In an attempt to understand the pathogenesis of depression and subsequently develop more efficacious antidepressant drugs, multiple theories have been proposed, including the modulation of neurotransmission, the upregulation of neurogenesis and neurotrophic factors, normalizing hypothalamic-pituitary-adrenal reactivity, and the reduction of neuroinflammation; all of which have supporting lines of evidence. Therefore, an ideal molecular target for novel pharmaceutical intervention would function at the confluence of these theories. The polysialylated form of the neural cell adhesion molecule (PSA-NCAM) functions broadly, serving to mediate synaptic plasticity, neurogenesis, neurotrophic factor signaling, and inflammatory signaling throughout the brain; all of which are associated with the pathophysiology and treatment of depression. Moreover, the expression of PSA-NCAM is reduced by depression, and conversely enhanced by antidepressant treatment, particularly within the hippocampus. Here we demonstrate that selectively cleaving the polysialic acid moiety, using the bacteriophage-derived enzyme endoneuraminidase N, completely inhibits the antidepressant efficacy of the selective-serotonin reuptake inhibitor fluoxetine (FLX) in a chronic unpredictable stress model of depression. We also observe a corresponding attenuation of FLX-induced hippocampal neuroplasticity, including decreased hippocampal neurogenesis, synaptic density, and neural activation. These data indicate that PSA-NCAM-mediated neuroplasticity is necessary for antidepressant action; therefore PSA-NCAM represents an interesting, and novel, target for pharmacotherapy.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Fluoxetine/pharmacology , Neural Cell Adhesion Molecule L1/physiology , Sialic Acids/physiology , Animals , Corticosterone/blood , Depression/drug therapy , Disease Models, Animal , Glycoside Hydrolases/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neural Cell Adhesion Molecule L1/deficiency , Neural Cell Adhesion Molecule L1/metabolism , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Sialic Acids/deficiency , Sialic Acids/metabolism , Testosterone/blood , Treatment Outcome
8.
Hippocampus ; 24(3): 280-92, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24132958

ABSTRACT

Cannabinoid exposure during adolescence has adverse effects on neuroplasticity, emotional behavior, cognition, and reward sensitivity in adult rats. We investigated whether escalating doses of the cannabinoid receptor 1 (CB1 R) agonist, HU-210, in adolescence would affect adult hippocampal neurogenesis and behavioral processes putatively modulated by hippocampal neurogenesis, in adult male and female Sprague-Dawley rats. Escalating doses of HU-210 (25, 50, and 100 µg/kg), or vehicle were administered from postnatal day (PND) 35 to 46. Animals were left undisturbed until PND 70, when they were treated with 5-bromo-2-deoxyuridine (BrdU; 200 mg/kg) and perfused 21 days later to examine density of BrdU-ir and BrdU/NeuN cells in the dentate gyrus. In another cohort, hypothalamic-pituitary-adrenal (HPA) axis reactivity to an acute restraint stress (30 min; PND 75) and behavioral sensitization to d-amphetamine sulfate (1-2 mg/kg; PND 105-134) were assessed in adulthood. Adolescent HU-210 administration suppressed the density of BrdU-ir cells in the dentate gyrus in adult male, but not adult female rats. Adolescent HU-210 administration also induced significantly higher peak corticosterone levels and reminiscent of the changes in neurogenesis, this effect was more pronounced in adult males than females. However, adolescent cannabinoid treatment resulted in significantly higher stereotypy scores in adult female, but not male, rats. Thus, adolescent CB1 R activation suppressed hippocampal neurogenesis and increased stress responsivity in adult males, but not females, and enhanced amphetamine sensitization in adult female, but not male, rats. Taken together, increased CB1 R activation during adolescence results in sex-dependent, long-term, changes to hippocampal structure and function, an effect that may shed light on differing vulnerabilities to developing disorders following adolescent cannabinoid exposure, based on sex.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Dentate Gyrus/drug effects , Dextroamphetamine/pharmacology , Dronabinol/analogs & derivatives , Neurogenesis/physiology , Receptor, Cannabinoid, CB1/drug effects , Sex Characteristics , Sexual Maturation/physiology , Stereotyped Behavior/drug effects , Stress, Physiological/physiology , Stress, Psychological/physiopathology , Animals , Cannabinoid Receptor Agonists/administration & dosage , Corticosterone/blood , DNA Replication/drug effects , Dentate Gyrus/growth & development , Dentate Gyrus/physiology , Dose-Response Relationship, Drug , Dronabinol/administration & dosage , Dronabinol/pharmacology , Estrus , Female , Hypothalamo-Hypophyseal System/physiopathology , Injections, Intraperitoneal , Male , Pituitary-Adrenal System/physiopathology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/physiology , Restraint, Physical/adverse effects , Sexual Maturation/drug effects , Single-Blind Method , Stereotyped Behavior/physiology
9.
Neural Plast ; 2013: 805497, 2013.
Article in English | MEDLINE | ID: mdl-23691371

ABSTRACT

Depression is a devastating and prevalent disease, with profound effects on neural structure and function; however the etiology and neuropathology of depression remain poorly understood. Though antidepressant drugs exist, they are not ideal, as only a segment of patients are effectively treated, therapeutic onset is delayed, and the exact mechanism of these drugs remains to be elucidated. Several theories of depression do exist, including modulation of monoaminergic neurotransmission, alterations in neurotrophic factors, and the upregulation of adult hippocampal neurogenesis, and are briefly mentioned in the review. However none of these theories sufficiently explains the pathology and treatment of depression unto itself. Recently, neural plasticity theories of depression have postulated that multiple aspects of brain plasticity, beyond neurogenesis, may bridge the prevailing theories. The term "neural plasticity" encompasses an array of mechanisms, from the birth, survival, migration, and integration of new neurons to neurite outgrowth, synaptogenesis, and the modulation of mature synapses. This review critically assesses the role of adult hippocampal neurogenesis and the cell adhesion molecule, PSA-NCAM (which is known to be involved in many facets of neural plasticity), in depression and antidepressant treatment.


Subject(s)
Depressive Disorder/physiopathology , Hippocampus/physiopathology , Neural Cell Adhesion Molecule L1/physiology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Sialic Acids/physiology , Animals , Depressive Disorder/metabolism , Hippocampus/metabolism , Humans , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism
10.
Psychoneuroendocrinology ; 36(9): 1327-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21481538

ABSTRACT

The incidence of depression is 2-3× higher in women particularly during the reproductive years, an occurrence that has been associated with levels of sex hormones. The age-related decline of testosterone levels in men corresponds with the increased acquisition of depressive symptoms, and hormone replacement therapy can be efficacious in treating depression in hypogonadal men. Although it is not possible to model depression in rodents, it is possible to model some of the symptoms of depression including a dysregulated stress response and altered neuroplasticity. Among animal models of depression, chronic mild unpredictable stress (CMS) is a common paradigm used to induce depressive-like behaviours in rodents, disrupt the hypothalamic-pituitary adrenal axis and decrease hippocampal neuroplasticity. The purpose of this study was to assess the effect of hypogonadism, produced by gonadectomy, on the acquisition of depressive-like behaviours and changes in hippocampal neuroplasticity in adult male Sprague-Dawley rats. A 21-day unpredictable CMS protocol was used on gonadectomised (GDX) and sham-operated males which produced an attenuation of weight gain in the GDX males receiving CMS treatment (GDX-CMS). Behavioural analysis was carried out to assess anxiety- and depressive-like behaviours. The combination of GDX and CMS produced greater passive behaviours within the forced swim test than CMS exposure alone. Similarly, hippocampal cell proliferation, neurogenesis and the expression of the neuroplastic protein polysialated neural cell adhesion molecule (PSA-NCAM) were all significantly reduced in the GDX-CMS group compared to all other treatment groups. These findings indicate that testicular hormones confer resiliency to chronic stress in males therefore reducing the likelihood of developing putative physiological, behavioural or neurological depressive-like phenotypes.


Subject(s)
Depressive Disorder/etiology , Hypogonadism/complications , Mental Disorders/etiology , Animals , Corticosterone/blood , Disease Progression , Disease Susceptibility , Exercise Test , Hypogonadism/blood , Male , Orchiectomy , Phenotype , Physical Conditioning, Animal/physiology , Rats , Rats, Sprague-Dawley , Stress, Psychological/blood , Stress, Psychological/complications , Stress, Psychological/physiopathology , Swimming/physiology
11.
Dev Neurobiol ; 68(14): 1580-90, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18844212

ABSTRACT

Newborn cells of the adult dentate gyrus in the hippocampus are characterized by their abundant expression of polysialic acid (PSA), a carbohydrate attached to the neural cell adhesion molecule (NCAM). PSA+ newborn cells of the dentate gyrus form clusters with proliferating neural progenitor cells, migrate away from these clusters, and terminally differentiate. To identify the roles of PSA in the development of adult progenitors of the dentate gyrus, we injected endoneuraminidase N (endoN) into the hippocampus of adult rats to specifically cleave PSA from NCAM. Two days later, we administered the mitotic marker, 5-bromo-2'-deoxyuridine (BrdU). Three days after BrdU injection, BrdU+ cells were found inside and outside the clusters of newborn cells. In endoN-treated animals, the total number of BrdU+ cells was not changed but significantly more BrdU+ cells were present within clusters, suggesting that PSA normally facilitates the migration of progenitors away from the clusters. Seven days post-BrdU injection, endoN-treated animals had significantly more BrdU+ cells which were also positive for the mature neuronal nuclear marker NeuN compared with controls, indicating that the loss of PSA from progenitor cells increases neuronal differentiation. This report is the first demonstration that PSA is involved in controlling the spatio-temporal neuronal maturation of adult hippocampal progenitors in the normal brain. In vitro, the removal of PSA from adult-derived neural progenitors significantly enhanced neuronal differentiation, strengthening our in vivo findings and indicating that PSA removal on isolated progenitor cells, apart from a complex in vivo environment, induces neuronal maturation.


Subject(s)
Adult Stem Cells/drug effects , Cell Differentiation/drug effects , Cell Movement/physiology , Hippocampus/cytology , Neurons/physiology , Sialic Acids/metabolism , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Count , Cell Movement/drug effects , Doublecortin Domain Proteins , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factors/pharmacology , Glycoside Hydrolases/pharmacology , Ki-67 Antigen/metabolism , Membrane Transport Proteins/metabolism , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Monocarboxylic Acid Transporters , Neurons/drug effects , Neuropeptides/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...