Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neurobiol Aging ; 126: 113-122, 2023 06.
Article in English | MEDLINE | ID: mdl-36989547

ABSTRACT

Most patients with Alzheimer's disease (AD) develop neuropsychiatric symptoms (NPS) alongside cognitive decline, and apathy is one of the most common symptoms. Few preclinical studies have investigated the biological substrates underlying NPS in AD. In this study, we used a cross-sectional design to characterize apathy-like behaviors and assess memory in 5xFAD and wildtype control mice at 6, 12, and 16 months of age. Nest building, burrowing, and marble burying were used to test representative behaviors of apathy, and a composite score of apathy-like behavior was generated from these assays. Soluble Aß42 and plaques were quantified in the prefrontal cortex and hippocampus of the 5xFAD mice with the highest and lowest composite scores using ELISA and histology. Results suggest that 5xFAD mice develop significant apathy-like behaviors starting at 6 months of age that worsen with aging and are positively correlated with soluble Aß42 and plaques in the prefrontal cortex and hippocampus. Our findings highlight the utility of studying NPS in mouse models of AD to uncover important relationships with underlying neuropathology.


Subject(s)
Alzheimer Disease , Apathy , Mice , Animals , Alzheimer Disease/pathology , Amyloid beta-Peptides , Cross-Sectional Studies , Mice, Transgenic , Disease Models, Animal
2.
Curr Res Neurobiol ; 3: 100025, 2022.
Article in English | MEDLINE | ID: mdl-36518344

ABSTRACT

Conflicting evidence suggest that perturbations of GABAergic neurotransmission play crucial roles in disrupting cortical neuronal network oscillations, memory, and cognitive deficits in Alzheimer's disease (AD). However, the role and impact of sex differences on GABAergic transmission in AD are not well understood. Using an APP knock-in mouse model of AD, APPNLGF mice, we studied the effects of acute diazepam administration on memory and anxiety-like behavior to unveil sex-dependent dysregulation of GABAergic neurotransmission. We also examined sex differences in GABAA receptor subunit mRNA and protein expression and the role of epigenetic regulation in hippocampus of APPNLGF mice. We found that diazepam elicited dose-dependent suppression of locomotion in wildtype and APPNLGF mice. However, a low dose, which had no significant effect in both male and female wildtype as well as female APPNLGF mice, significantly suppressed locomotion in male APPNLGF mice. Furthermore, this low dose of diazepam was more efficacious at eliciting anxiolytic-like effects in male than female APPNLGF mice. The same low dose of diazepam disrupted recognition memory exclusively in male APPNLGF mice. Biochemical analyses revealed that hippocampal α1 and α5 GABAA receptor subunits mRNA and protein expression were significantly higher in male than female APPNLGF mice and were regulated by histone H3 tri-methylation (H3K4me3) but not histone H3 acetylation. The higher sensitivity of APPNLGF males to diazepam-induced behavioral effects may potentially be due to epigenetic-dependent upregulation of hippocampal α1 and α5 GABAA receptor subunits expression compared to female APPNLGF mice. These findings suggest that dysregulation of GABAergic neurotransmission plays a significant role in memory and affective behavior, particularly in male APPNLGF mice.

3.
Behav Brain Res ; 404: 113192, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33607163

ABSTRACT

Transgenic mouse models of Aß amyloidosis generated by knock-in of a humanized Aß sequence can offer some advantages over the transgenic models that overexpress amyloid precursor protein (APP). However, systematic comparison of memory, behavioral, and neuropathological phenotypes between these models has not been well documented. In this study, we compared memory and affective behavior in APPNLGF mice, an APP knock-in model, to two widely used mouse models of Alzheimer's disease, 5xFAD and APP/PS1 mice, at 10 months of age. We found that, despite similar deficits in working memory, object recognition, and social recognition memory, APPNLGF and 5xFAD mice but not APP/PS1 mice show compelling anxiety- and depressive-like behavior, and exhibited a marked impairment of social interaction. We quantified corticolimbic Aß plaques, which were lowest in APPNLGF, intermediate in APP/PS1, and highest in 5xFAD mice. Interestingly, analysis of plaque size revealed that plaques were largest in APP/PS1 mice, intermediate in 5xFAD mice, and smallest in APPNLGF mice. Finally, we observed a significantly higher percentage of the area occupied by plaques in both 5xFAD and APP/PS1 relative to APPNLGF mice. Overall, our findings suggest that the severity of Aß neuropathology is not directly correlated with memory and affective behavior impairments between these three transgenic mouse models. Additionally, APPNLGF may represent a valid mouse model for studying AD comorbid with anxiety and depression.


Subject(s)
Affect , Amyloid beta-Protein Precursor/genetics , Brain/pathology , Memory , Oligopeptides/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , Female , Locomotion , Male , Memory, Short-Term , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morris Water Maze Test , Oligopeptides/metabolism , Oligopeptides/physiology , Open Field Test , Social Interaction
4.
J Neurochem ; 158(4): 943-959, 2021 08.
Article in English | MEDLINE | ID: mdl-32813270

ABSTRACT

Signaling pathways mediated by corticotropin-releasing factor and its receptor 1 (CRF1) play a central role in stress responses. Dysfunction of the CRF system has been associated with neuropsychiatric disorders. However, dynamic changes in the CRF system during brain development and aging are not well investigated. In this study, we characterized CRF1, CRF, and corticotropin-releasing factor binding protein (CRFBP) expression in different brain regions in both male and female C57BL/6J mice from 1 to 18 months of age under basal conditions as well as after an acute 2-hr-restraint stress. We found that CRF and CRF1 levels tended to increase in the hippocampus and hypothalamus, and to decrease in the prefrontal cortex with aging, especially at 18 months of age, whereas CRFBP expression followed an opposite direction in these brain areas. We also observed area-specific sex differences in the expression of these three proteins. For example, CRF expression was lower in females than in males in all the brain regions examined except the prefrontal cortex. After acute stress, CRF and CRF1 were up-regulated at 1, 6, and 12 months of age, and down-regulated at 18 months of age. Females showed more robust changes compared to males of the same age. CRFBP expression either decreased or remained unchanged in most of the brain areas following acute stress. Our findings suggest that brain CRF1, CRF, and CRFBP expression changes dynamically across the lifespan and under stress condition in a sex- and regional-specific manner. Sex differences in the CRF system in response to stress may contribute to the etiology of stress-related neuropsychiatric disorders.


Subject(s)
Brain Chemistry/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Corticotropin-Releasing Hormone/biosynthesis , Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Receptors, Corticotropin-Releasing Hormone/genetics , Stress, Psychological/genetics , Stress, Psychological/metabolism , Animals , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Restraint, Physical , Sex Characteristics , Stress, Psychological/psychology
5.
Biol Psychiatry ; 85(12): 1036-1045, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30955840

ABSTRACT

BACKGROUND: The endocannabinoid and neurosteroid systems regulate emotions and stress responses. Activation of peroxisome proliferator-activated receptor (PPAR)-α by the endocannabinoid congener N-palmitoylethanolamine (PEA) regulates pathophysiological systems (e.g., inflammation, oxidative stress) and induces peripheral biosynthesis of allopregnanolone, a gamma-aminobutyric acidergic neurosteroid implicated in mood disorders. However, effects of PPAR-α on emotional behavior are poorly understood. METHODS: We studied the impact of PPAR-α activation on emotional behavior in a mouse model of posttraumatic stress disorder. Neurosteroid levels before and after PEA treatment were measured by gas chromatography-mass spectrometry in relevant brain regions of socially isolated versus group-housed mice exposed to the contextual fear conditioning test, elevated plus maze test, forced swim test, and tail suspension test. Neurosteroidogenic enzyme levels were quantified in hippocampus by Western blot. RESULTS: PEA administered in a model of conditioned contextual fear reconsolidation blockade facilitated fear extinction and fear extinction retention and induced marked antidepressive- and anxiolytic-like effects in socially isolated mice with reduced brain allopregnanolone levels. These effects were mimicked by the PPAR-α synthetic agonists, fenofibrate and GW7647, and were prevented by PPAR-α deletion, PPAR-α antagonists, and neurosteroid-enzyme inhibitors. Behavioral improvements correlated with PEA-induced upregulation of PPAR-α, neurosteroidogenic enzyme expression, and normalization of corticolimbic allopregnanolone levels. CONCLUSIONS: This evidence supports a previously unknown role for PPAR-α in behavior regulation and suggests new strategies for the treatment of neuropsychopathologies characterized by deficient neurosteroidogenesis, including posttraumatic stress disorder and major depressive disorder.


Subject(s)
Emotions/physiology , Ethanolamines/administration & dosage , PPAR alpha/physiology , Palmitic Acids/administration & dosage , Pregnanolone/biosynthesis , Stress Disorders, Post-Traumatic/physiopathology , Amides , Animals , Anxiety/physiopathology , Disease Models, Animal , Emotions/drug effects , Fear/drug effects , Fear/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , PPAR alpha/metabolism , Social Isolation
6.
Article in English | MEDLINE | ID: mdl-30586627

ABSTRACT

Post-traumatic stress disorder (PTSD) is a psychiatric condition characterized by drastic alterations in mood, emotions, social abilities and cognition. Notably, one aspect of PTSD, particularly in veterans, is its comorbidity with suicide. Elevated aggressiveness predicts high-risk to suicide in humans and despite the difficulty in reproducing a complex human suicidal behavior in rodents, aggressive behavior is a well reproducible behavioral trait of suicide. PTSD animal models are based on a peculiar phenotype, including exaggerated fear memory and impaired fear extinction associated with neurochemical dysregulations in the brain circuitry regulating emotion. The endocannabinoid and the neurosteroid systems regulate emotions and stress responses, and recent evidence shows these two systems are interrelated and critically compromised in neuropsychiatric disorders. For instance, levels of the neurosteroid, allopregnanolone, as well as those of the endocannabinoids, anandamide and its congener, palmitoylethanolamide are decreased in PTSD. Similarly, the endocannabinoid system and neurosteroid biosynthesis are altered in suicidal individuals. Selective serotonin reuptake inhibitors (SSRIs), the only FDA-approved treatments for PTSD, fail to help half of the treatment-seeking patients. This highlights the need for developing biomarker-based efficient therapies. One promising alternative to SSRIs points to stimulation of allopregnanolone biosynthesis as a treatment and a valid end-point to predict treatment response in PTSD patients. This review highlights running findings on the role of the endocannabinoid and neurosteroid systems in PTSD and suicidal behavior both in a preclinical and clinical perspective. A specific focus is given to predictive PTSD/suicide animal models. Ultimately, we discuss the idea that disruption of neurosteroid and endocannabinoid biosynthesis may offer a novel promising biomarker axis to develop new treatments for PTSD and, perhaps, suicidal behavior.


Subject(s)
Cannabinoids/therapeutic use , Disease Models, Animal , Neurotransmitter Agents/therapeutic use , Social Isolation , Stress Disorders, Post-Traumatic/drug therapy , Suicide Prevention , Animals , Humans
7.
Front Cell Neurosci ; 11: 208, 2017.
Article in English | MEDLINE | ID: mdl-28900387

ABSTRACT

Early trauma and stress exposure during a critical period of life may increase the risk of major depressive disorder (MDD) and post-traumatic stress disorder (PTSD) in adulthood. The first-choice treatment for MDD and PTSD are selective serotonin reuptake inhibitor (SSRI) antidepressants. Unfortunately, half of MDD and PTSD patients show resistance to the therapeutic effects of these drugs and more efficient treatments are essential. Both MDD and PTSD patients present reduced levels of allopregnanolone (Allo), a potent endogenous positive allosteric modulator of GABA action at GABAA receptors which are normalized by SSRIs in treatment responders. Thus, Allo analogs or drugs that stimulate its levels may offer an alternative in treating SSRIs-non-responders. We tested several drugs on the aggressive behavior of early and late adolescent socially-isolated (SI) mice, a model of PTSD. Isolation in early adolescence (PND 21) induced more severe aggression than mice isolated at PND 45. A single non-sedating administration of S-fluoxetine (S-FLX; 0.375-1.5 mg/kg), or of the Allo analogs ganaxolone (GNX; 10 mg/kg), BR351 (1-5 mg/kg), or BR297 (0.3125-2.5 mg/kg), or of the endocannabinoid, N-palmitoylethanolamine (PEA; 5-20 mg/kg) all decreased aggression more effectively in late than early adolescent SI mice. Importantly, the number of drug non-responders was higher in early than late SI mice for all the drugs tested. The non-responder rate was more elevated (12-64%) after S-FLX treatment, while 100% of mice responded to a single administration of PEA at the dose range of 15-20 mg/kg. Moreover, GNX, BR351, and BR297's antiaggressive effect persisted longer than S-FLX in both late and early SI mice. All drugs tested failed to alter locomotor activity of SI mice. Our results show that drugs that mimic Allo's action or that induce Allo biosynthesis may be valuable for the treatment of "SSRIs non-responder" patients.

8.
Br J Pharmacol ; 174(19): 3226-3241, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28456011

ABSTRACT

By rapidly modulating neuronal excitability, neurosteroids regulate physiological processes, such as responses to stress and development. Excessive stress affects their biosynthesis and causes an imbalance in cognition and emotions. The progesterone derivative, allopregnanolone (Allo) enhances extrasynaptic and postsynaptic inhibition by directly binding at GABAA receptors, and thus, positively and allosterically modulates the function of GABA. Allo levels are decreased in stress-induced psychiatric disorders, including depression and post-traumatic stress disorder (PTSD), and elevating Allo levels may be a valid therapeutic approach to counteract behavioural dysfunction. While benzodiazepines are inefficient, selective serotonin reuptake inhibitors (SSRIs) represent the first choice treatment for depression and PTSD. Their mechanisms to improve behaviour in preclinical studies include neurosteroidogenic effects at low non-serotonergic doses. Unfortunately, half of PTSD and depressed patients are resistant to current prescribed 'high' dosage of these drugs that engage serotonergic mechanisms. Unveiling novel biomarkers to develop more efficient treatment strategies is in high demand. Stress-induced down-regulation of neurosteroid biosynthesis and changes in GABAA receptor subunit expression offer a putative biomarker axis to develop new PTSD treatments. The advantage of stimulating Allo biosynthesis relies on the variety of neurosteroidogenic receptors to be targeted, including TSPO and endocannabinoid receptors. Furthermore, stress favours a GABAA receptor subunit composition with higher sensitivity for Allo. The use of synthetic analogues of Allo is a valuable alternative. Pregnenolone or drugs that stimulate its levels increase Allo but also sulphated steroids, including pregnanolone sulphate which, by inhibiting NMDA tonic neurotransmission, provides neuroprotection and cognitive benefits. In this review, we describe current knowledge on the effects of stress on neurosteroid biosynthesis and GABAA receptor neurotransmission and summarize available pharmacological strategies that by enhancing neurosteroidogenesis are relevant for the treatment of SSRI-resistant patients. Linked Articles This article is part of a themed section on Pharmacology of Cognition: a Panacea for Neuropsychiatric Disease? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.19/issuetoc.


Subject(s)
Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Steroids/metabolism , Stress, Psychological/metabolism , Animals , Antidepressive Agents/therapeutic use , Biomarkers/metabolism , Brain/metabolism , Cognitive Dysfunction/metabolism , Emotions , Humans , Mental Disorders/drug therapy , Mental Disorders/metabolism , Steroids/therapeutic use
9.
Neurosci Lett ; 649: 156-163, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28215878

ABSTRACT

This review highlights early efforts to translate pre-clinical and clinical findings regarding the role of neuroactive steroids in stress adaptation and PTSD into new therapeutics for PTSD. Numerous studies have demonstrated PTSD-related alterations in resting levels or the reactivity of neuroactive steroids and their targets. These studies also have demonstrated substantial variability in the dysfunction of specific neuroactive steroid systems among PTSD subpopulations. These variabilities have been related to the developmental timing of trauma, severity and type of trauma, genetic background, sex, reproductive state, lifestyle influences such as substance use and exercise, and the presence of comorbid conditions such as depression and chronic pain. Nevertheless, large naturalistic studies and a small placebo-controlled interventional study have revealed generally positive effects of glucocorticoid administration in preventing PTSD after trauma, possibly mediated by glucocorticoid receptor-mediated effects on other targets that impact PTSD risk, including other neuroactive steroid systems. In addition, clinical and preclinical studies show that administration of glucocorticoids, 17ß-estradiol, and GABAergic neuroactive steroids or agents that enhance their synthesis can facilitate extinction and extinction retention, depending on dose and timing of dose in relation to these complex PTSD-relevant recovery processes. This suggests that clinical trials designed to test neuroactive steroid therapeutics in PTSD may benefit from such considerations; typical continuous dosing regimens may not be optimal. In addition, validated and clinically accessible methods for identifying specific neuroactive steroid system abnormalities at the individual level are needed to optimize both clinical trial design and precision medicine based treatment targeting.


Subject(s)
Steroids/physiology , Steroids/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/drug therapy , Animals , Dehydroepiandrosterone/physiology , Dehydroepiandrosterone/therapeutic use , Estradiol/physiology , Estradiol/therapeutic use , Glucocorticoids/physiology , Glucocorticoids/therapeutic use , Humans , Pregnanolone/physiology , Pregnanolone/therapeutic use , Stress Disorders, Post-Traumatic/complications , Stress, Psychological/complications
10.
Psychopharmacology (Berl) ; 234(5): 749-760, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28013353

ABSTRACT

BACKGROUND: Allopregnanolone plays a role in the stress response and homeostasis. Alterations in the estrogen milieu during the perinatal period influence brain development in a manner that persists into adulthood. Accordingly, we showed that a single administration of estradiol benzoate (EB) on the day of birth decreases brain allopregnanolone concentrations in adult female rats. OBJECTIVE: We examined whether the persistent decrease in allopregnanolone concentrations, induced by neonatal EB treatment, might affect sensitivity to stress during adulthood. METHODS: Female rats were treated with 10 µg of EB or vehicle on the day of birth. During adulthood, the response to acute foot shock stress was assessed by measuring changes in brain allopregnanolone and corticosterone levels, as well as extracellular dopamine output in the medial prefrontal cortex (mPFC). RESULTS: Neonatal EB treatment enhanced stress-stimulated allopregnanolone levels in the hypothalamus, as well as extracellular dopamine output in the mPFC; this latest effect is reverted by subchronic progesterone treatment. By contrast, neonatal EB treatment did not alter stress-induced corticosterone levels, sensitivity to hypothalamic-pituitary-adrenal (HPA) axis negative feedback, or abundance of glucocorticoid and mineralocorticoid receptors. CONCLUSIONS: The persistent decrease in brain allopregnanolone concentrations, induced by neonatal EB treatment, enhances stress-stimulated allopregnanolone levels and extracellular dopamine output during adulthood. These effects are not associated to an impairment in HPA axis activity. Heightened sensitivity to stress is a risk factor for several neuropsychiatric disorders; these results suggest that exposure to estrogen during development may predispose individuals to such disorders.


Subject(s)
Brain/drug effects , Corticosterone/metabolism , Dopamine/metabolism , Estradiol/analogs & derivatives , Estrogens/pharmacology , Pregnanolone/metabolism , Stress, Psychological/metabolism , Animals , Animals, Newborn , Brain/metabolism , Electric Stimulation , Estradiol/pharmacology , Female , Hypothalamo-Hypophyseal System , Hypothalamus/drug effects , Pituitary-Adrenal System , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Progesterone/pharmacology , Progestins/pharmacology , Rats , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/metabolism
11.
Horm Behav ; 87: 35-46, 2017 01.
Article in English | MEDLINE | ID: mdl-27769760

ABSTRACT

Exposure of female rats to estradiol during the perinatal period has profound effects on GABAergic neurotransmission that are crucial to establish sexually dimorphic brain characteristics. We previously showed that neonatal ß-estradiol 3-benzoate (EB) treatment decreases brain concentrations of the neurosteroid allopregnanolone, a potent positive modulator of extrasynaptic GABAA receptors (GABAAR). We thus evaluated whether neonatal EB treatment affects GABAAR expression and function in the hippocampus of adult female rats. Neonatal EB administration increased the expression of extrasynaptic α4/δ subunit-containing GABAARs and the modulatory action of THIP on tonic currents mediated by these receptors. The same treatment decreased the expression of synaptic α1/α4/γ2 subunit-containing receptors, as well as phasic currents. These effects of neonatal EB treatment are not related to ambient allopregnanolone concentrations per se, given that vehicle-treated rats in diestrus, which have opposite neurosteroid levels than EB-treated rats, show similar changes in GABAARs. Rather, these changes may represent a compensatory mechanism to counteract the long-term reduction in allopregnanolone concentrations, induced by neonatal EB. Given that both α4/δ receptors and allopregnanolone are involved in memory consolidation, we evaluated whether neonatal EB treatment alters performance in the Morris water maze test during adulthood. Neonatal EB treatment decreased the latency and the cumulative search error to reach the platform, as well as thigmotaxis, suggesting improved learning, and also enhanced memory performance during the probe trial. These enduring changes in GABAAR plasticity may be relevant for the regulation of neuronal excitability in the hippocampus and for the etiology of psychiatric disorders that originate in development and show sex differences.


Subject(s)
Estradiol/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA-A/physiology , Spatial Learning/drug effects , Animals , Animals, Newborn , Estradiol/analogs & derivatives , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neurotransmitter Agents/pharmacology , Pregnanolone/pharmacology , Rats , Rats, Sprague-Dawley , Sex Characteristics , Synaptic Transmission/drug effects
12.
Psychopharmacology (Berl) ; 231(17): 3351-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24728651

ABSTRACT

RATIONALE: Allopregnanolone is a neurosteroid involved in depression, memory, social, and sexual behavior. We have previously demonstrated that treatment with a combination of ethinylestradiol (EE) and levonorgestrel (LNG), two compounds frequently used in hormonal contraception, decreased brain allopregnanolone concentrations. These changes may contribute to some of the emotional and sexual disorders observed in hormonal contraceptive users. OBJECTIVES: We thus examined whether the reduction in allopregnanolone concentrations induced by long-term EE/LNG administration was associated with altered emotional, learning, social, and sexual behaviors. METHODS: Rats were orally treated with a combination of EE (0.030 mg) and LNG (0.125 mg) once a day for 4 weeks and were subjected to behavioral tests 24 h after the last administration. RESULTS: EE/LNG treatment reduced immobility behavior in the forced swim test, without affecting sucrose preference and spatial learning and memory. In the resident-intruder test, EE/LNG-treated rats displayed a decrease in dominant behaviors associated with a reduction in social investigation. In the paced mating test, EE/LNG treated rats showed a reduction in proceptive behaviors, while the lordosis quotient was not affected. Progesterone, but not estradiol, administration to EE/LNG-treated rats increased sexual activity and cerebrocortical allopregnanolone concentrations. Prior administration of finasteride decreased allopregnanolone concentrations and abolished the increase in proceptivity induced by progesterone administration. CONCLUSIONS: The decrease in brain allopregnanolone concentrations induced by EE/LNG treatment is associated with a reduction in social behavior and sexual motivation in female rats. These results might be relevant to the side effects sometimes exhibited by women taking hormonal contraceptives.


Subject(s)
Contraceptives, Oral, Hormonal/pharmacology , Motivation/drug effects , Pregnanolone/antagonists & inhibitors , Sexual Behavior, Animal/drug effects , Social Behavior , Agonistic Behavior/drug effects , Anhedonia/drug effects , Animals , Cerebral Cortex/metabolism , Depression/chemically induced , Depression/psychology , Drug Combinations , Ethinyl Estradiol/pharmacology , Female , Levonorgestrel/pharmacology , Maze Learning/drug effects , Pregnanolone/metabolism , Rats , Rats, Sprague-Dawley , Swimming/psychology
13.
Alcohol Clin Exp Res ; 38(4): 948-58, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24428156

ABSTRACT

BACKGROUND: Ethanol (EtOH) administration increases brain allopregnanolone levels in rats, and this increase contributes to sensitivity to EtOH's behavioral effects. However, EtOH's effects on allopregnanolone may differ across species. We investigated the effects of acute EtOH administration on allopregnanolone, progesterone, and corticosterone levels in cerebral cortex and hippocampus of C57BL/6J and DBA/2J mice, 2 inbred strains with different alcohol sensitivity. METHODS: Naïve male C57BL/6J and DBA/2J mice received EtOH (1, 2, 3, or 4 g/kg, intraperitoneally [i.p.]) or saline and were euthanized 1 hour later. For the time-course study, mice received EtOH (2 g/kg, i.p.) and were euthanized 15, 30, 60, and 120 minutes later. Steroids were measured by radioimmunoassay. RESULTS: Acute EtOH administration did not alter cerebrocortical and hippocampal levels of allopregnanolone and progesterone in these strains at any of the doses and time points examined. Acute EtOH dose-dependently increased cerebrocortical corticosterone levels by 319, 347, and 459% in C57BL/6J mice at the doses of 2, 3, and 4 g/kg, and by 371, 507, 533, and 692% in DBA/2J mice at the doses of 1, 2, 3, and 4 g/kg, respectively. Similar changes were observed in the hippocampus. EtOH's effects on cerebrocortical corticosterone levels were also time dependent in both strains. Moreover, acute EtOH administration time-dependently increased plasma levels of progesterone and corticosterone. Finally, morphine administration increased cerebrocortical allopregnanolone levels in C57BL/6J (+77, +93, and +88% at 5, 10, and 30 mg/kg, respectively) and DBA/2J mice (+81% at 5 mg/kg), suggesting that the impairment in brain neurosteroidogenesis may be specific to EtOH. CONCLUSIONS: These results underline important species differences on EtOH-induced brain neurosteroidogenesis. Acute EtOH increases brain and plasma corticosterone levels but does not alter cerebrocortical and hippocampal concentrations of allopregnanolone and progesterone in naïve C57BL/6J and DBA/2J mice.


Subject(s)
Cerebral Cortex/metabolism , Ethanol/administration & dosage , Hippocampus/metabolism , Pregnanolone/blood , Animals , Cerebral Cortex/drug effects , Hippocampus/drug effects , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...