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










Publication year range
1.
Biol Psychiatry ; 84(8): 611-623, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29887035

ABSTRACT

BACKGROUND: The ability to effectively cope with stress is a critical determinant of disease susceptibility. The lateral habenula (LHb) and the endocannabinoid (ECB) system have independently been shown to be involved in the selection of stress coping strategies, yet the role of ECB signaling in the LHb remains unknown. METHODS: Using a battery of complementary techniques in rats, we examined the localization of type-1 cannabinoid receptors (CB1Rs) and assessed the behavioral and neuroendocrine effects of intra-LHb CB1R manipulations. We further tested the extent to which the ECB system in the LHb is impacted following chronic unpredictable stress or social defeat stress, and whether manipulation of LHb CB1Rs can bias coping strategies in rats with a history of chronic stress. RESULTS: Electron microscopy studies revealed CB1R expression on presynaptic axon terminals, postsynaptic membranes, mitochondria, and glial processes in the rat LHb. In vivo microdialysis experiments indicated that acute stress increased the amount of 2-arachidonoylglycerol in the LHb, while intra-LHb CB1R blockade increased basal corticosterone, augmented proactive coping strategies, and reduced anxiety-like behavior. Basal LHb 2-arachidonoylglycerol content was similarly elevated in rats that were subjected to chronic unpredictable stress or social defeat stress and positively correlated with adrenal weight. Finally, intra-LHb CB1R blockade increased proactive behaviors in response to a novel conspecific, increasing approach behaviors irrespective of stress history and decreasing the latency to be attacked during an agonistic encounter. CONCLUSIONS: Alterations in LHb ECB signaling may be relevant for development of stress-related pathologies in which LHb dysfunction and stress-coping impairments are hallmark symptoms.


Subject(s)
Adaptation, Psychological , Habenula/metabolism , Maze Learning , Spatial Memory , Stress, Psychological/physiopathology , Animals , Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Corticosterone/blood , Depression/metabolism , Disease Models, Animal , Endocannabinoids/pharmacology , Female , Glycerides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism
2.
Addict Biol ; 23(5): 1117-1129, 2018 09.
Article in English | MEDLINE | ID: mdl-28940879

ABSTRACT

Repeated cycles of alcohol [ethanol (EtOH)] intoxication and withdrawal dysregulate excitatory glutamatergic systems in the brain and induce neuroadaptations in the medial prefrontal cortex (mPFC) that contribute to cognitive dysfunction. The mPFC is composed of subdivisions that are functionally distinct, with dorsal regions facilitating drug-cue associations and ventral regions modulating new learning in the absence of drug. A key modulator of glutamatergic activity is the holoenzyme calcium/calmodulin-dependent protein kinase II (CaMKII) that phosphorylates ionotropic glutamate receptors. Here, we examined the hypothesis that abstinence from chronic intermittent EtOH (CIE) exposure dysregulates CaMKII activity in the mPFC to impair cognitive flexibility. We used an operant model of strategy set shifting in male Long-Evans rats demonstrating reduced susceptibility to trial omissions during performance in a visual cue-guided task versus albino strains. Relative to naïve controls, rats experiencing approximately 10 days of abstinence from CIE vapor exposure demonstrated impaired performance during a procedural shift from visual cue to spatial location discrimination. Phosphorylation of CaMKII subtype α was upregulated in the dorsal, but not ventral mPFC of CIE-exposed rats, and was positively correlated with perseverative-like responding during the set shift. The findings suggest that abstinence from CIE exposure induces an undercurrent of kinase activity (e.g. CaMKII), which may promote aberrant glutamatergic responses in select regions of the mPFC. Given the role of the mPFC in modulating executive control of behavior, we propose that increased CaMKII subtype α activity reflects a dysregulated 'top-down' circuit that interferes with adaptive behavioral performance under changing environmental demands.


Subject(s)
Alcoholism/complications , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cognitive Dysfunction/etiology , Ethanol/pharmacology , Prefrontal Cortex/metabolism , Alcoholism/genetics , Alcoholism/physiopathology , Animals , Behavior, Animal/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Central Nervous System Depressants/pharmacology , Cognition/drug effects , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Male , Phosphorylation , Rats , Rats, Long-Evans
3.
Biol Psychiatry ; 83(3): 203-213, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29066224

ABSTRACT

BACKGROUND: The neuropeptide oxytocin (OT) is a key regulator of social and emotional behaviors. The effects of OT are context dependent, and it has been proposed that OT increases the salience of both positive and negative social cues. Here we tested whether the bed nucleus of the stria terminalis (BNST) mediates anxiogenic effects of OT. METHODS: First, we studied the effects of systemic administration of an OT receptor (OTR) antagonist L-368,899 on social behavior in male and female California mice exposed to social defeat. We examined the effect of L-368,899 on G protein activation and used early growth response factor 1 immunohistochemistry to identify potential sites of OTR action. Finally, we examined the effects of L-368,899 infused in the BNST on behavior. RESULTS: A single dose of systemic L-368,899 increased social approach in stressed female mice and decreased social approach in male mice naïve to defeat. L-368,899 prevented OT activation of G proteins and did not activate G proteins in the absence of OT. Intranasal OT, which reduces social approach in female mice but not male mice, increased early growth response factor 1 immunoreactivity in the nucleus accumbens core and anteromedial BNST in female mice but not in male mice. Stressed female mice that received an infusion of L-368,899 into the anteromedial BNST but not the nucleus accumbens core increased social approach and decreased social vigilance responses. CONCLUSIONS: Our results suggest that OTR activation in anteromedial BNST induces a vigilance response in which individuals avoid, yet attend to, unfamiliar social contexts. Our results suggest that OTR antagonists may have unappreciated therapeutic potential for stress-induced psychiatric disorders.


Subject(s)
Behavior, Animal , Camphanes/pharmacology , Oxytocin/pharmacology , Piperazines/pharmacology , Receptors, Oxytocin , Septal Nuclei , Sex Characteristics , Social Behavior , Stress, Psychological , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Camphanes/administration & dosage , Disease Models, Animal , Female , Male , Mice , Nucleus Accumbens/drug effects , Oxytocin/administration & dosage , Piperazines/administration & dosage , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
4.
J Neurosci ; 37(7): 1853-1861, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28202787

ABSTRACT

Persons with alcoholism who are abstinent exhibit persistent impairments in the capacity for response inhibition, and this form of impulsivity is significantly associated with heightened relapse risk. Brain-imaging studies implicate aberrant prefrontal cortical function in this behavioral pathology, although the underlying mechanisms are not understood. Here we present evidence that deficient activation of glycine and serine release in the ventral medial prefrontal cortex (vmPFC) contributes to increased motor impulsivity during protracted abstinence from long-term alcohol exposure. Levels of 12 neurotransmitters were monitored in the rat vmPFC during the performance of a challenging variant of the five-choice serial reaction time task (5-CSRTT) in which alcohol-exposed rats exhibit excessive premature responding. Following long-term ethanol exposure, rats showed blunted task-related recruitment of vmPFC glycine and serine release, and the loss of an inverse relationship between levels of these neurotransmitters and premature responding normally evident in alcohol-naive subjects. Intra-vmPFC administration of the glycine transport inhibitor ALX5407 prevented excessive premature responding by alcohol-exposed rats, and this was reliant on NMDA glycine site availability. Alcohol-exposed rats and controls did not differ in their premature responding and glycine and serine levels in vmPFC during the performance of the standard 5-CSRTT. Collectively, these findings provide novel insight into cortical neurochemical mechanisms contributing to increased impulsivity following long-term alcohol exposure and highlight the NMDA receptor coagonist site as a potential therapeutic target for increased impulsivity that may contribute to relapse risk.SIGNIFICANCE STATEMENT Persons with alcoholism demonstrate increased motor impulsivity during abstinence; however, the neuronal mechanisms underlying these behavioral effects remain unknown. Here, we took advantage of an animal model that shows deficiencies in inhibitory control following prolonged alcohol exposure to investigate the neurotransmitters that are potentially responsible for dysregulated motor impulsivity following long-term alcohol exposure. We found that increased motor impulsivity is associated with reduced recruitment of glycine and serine neurotransmitters in the ventromedial prefrontal cortex (vmPFC) cortex in rats following long-term alcohol exposure. Administration of glycine transport inhibitor ALX5407 in the vmPFC alleviated deficits in impulse control.


Subject(s)
Alcohol Abstinence , Alcohol Drinking/physiopathology , Glycine/metabolism , Impulsive Behavior/physiology , Signal Transduction/physiology , Animals , Central Nervous System Depressants/adverse effects , Choice Behavior/drug effects , Disease Models, Animal , Ethanol/adverse effects , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Male , Neurotransmitter Agents/metabolism , Photic Stimulation , Quinolones/pharmacology , Rats , Rats, Wistar , Reaction Time/drug effects , Sarcosine/analogs & derivatives , Sarcosine/pharmacology , Serine/metabolism , Serine/pharmacology , Signal Transduction/drug effects
5.
Neuropharmacology ; 110(Pt A): 59-68, 2016 11.
Article in English | MEDLINE | ID: mdl-27452721

ABSTRACT

Vasopressin V1a receptors (V1aR) are thought to contribute to the pathophysiology of psychiatric disorders such as anxiety and depression, sparking interest in V1aR as a therapeutic target. Although the global effects of V1aR have been documented, less is known about the specific neural circuits mediating these effects. Moreover, few studies have examined context-specific V1aR function in both males and females. By using the California mouse, we first studied the effects of sex and social defeat stress on V1aR binding in the forebrain. In females but not males, V1aR binding in the bed nucleus of the stria terminalis (BNST) was negatively correlated to social interaction behavior. In females stress also increased V1aR binding in the nucleus accumbens (NAc). Infusions of V1aR antagonist in to the medioventral BNST (BNSTmv) had anxiogenic effects only in animals naïve to defeat. For males, inhibition of V1aR in BNSTmv had anxiogenic effects in social and nonsocial contexts, but for females, anxiogenic effects were limited to social contexts. In stressed females, inhibition of V1aR in the NAc shell had no effect on social interaction behavior, but had an anxiogenic effect in an open field test. These data suggest that V1aR in BNSTmv have anxiolytic and prosocial effects in males, and that in females, prosocial and anxiolytic effects of V1aR appear to be mediated independently by receptors in the BNSTmv and NAc shell, respectively. These findings suggest that males have more overlap in neural circuits modulating anxiety in social and nonsocial contexts than females.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/metabolism , Anxiety/metabolism , Interpersonal Relations , Receptors, Vasopressin/metabolism , Septal Nuclei/metabolism , Sex Characteristics , Animals , Antidiuretic Hormone Receptor Antagonists/administration & dosage , Antidiuretic Hormone Receptor Antagonists/toxicity , Anxiety/chemically induced , Anxiety/psychology , Female , Infusions, Intraventricular , Male , Mice , Protein Binding/physiology , Random Allocation , Receptors, Vasopressin/agonists , Septal Nuclei/drug effects , Vasopressins/administration & dosage , Vasopressins/metabolism
6.
Biol Psychiatry ; 80(5): 406-14, 2016 09 01.
Article in English | MEDLINE | ID: mdl-26620251

ABSTRACT

BACKGROUND: Oxytocin (OT) is considered to be a stress-buffering hormone, dampening the physiologic effects of stress. However, OT can also be anxiogenic. We examined acute and long-lasting effects of social defeat on OT neurons in male and female California mice. METHODS: We used immunohistochemistry for OT and c-fos cells to examine OT neuron activity immediately after defeat (n = 6-9) and 2 weeks (n = 6-9) and 10 weeks (n = 4-5) later. We quantified Oxt messenger RNA with quantitative polymerase chain reaction (n = 5-9). Intranasal OT was administered to naïve and stressed mice tested in social interaction and resident-intruder tests (n = 8-14). RESULTS: Acute exposure to a third episode of defeat increased OT/c-fos colocalizations in the paraventricular nucleus of both sexes. In the medioventral bed nucleus of the stria terminalis, defeat increased Oxt messenger RNA, total OT neurons, and OT/c-fos colocalizations in female mice but not male mice. Intranasal OT failed to reverse stress-induced social withdrawal in female mice and reduced social interaction behavior in female mice naïve to defeat. In contrast, intranasal OT increased social interaction in stressed male mice and reduced freezing in the resident-intruder test. CONCLUSIONS: Social defeat induces long-lasting increases in OT production and OT/c-fos cells in the medioventral bed nucleus of the stria terminalis of female mice but not male mice. Intranasal OT largely reversed the effects of stress on behavior in male mice, but effects were mixed in female mice. These results suggest that changes in OT-sensitive networks contribute to sex differences in behavioral responses to stress.


Subject(s)
Neurons/drug effects , Oxytocin/administration & dosage , Oxytocin/metabolism , Sex Characteristics , Stress, Psychological/drug therapy , Stress, Psychological/pathology , Administration, Intranasal , Animals , Disease Models, Animal , Female , Male , Mice , Neurons/metabolism , Oxytocin/genetics , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Social Behavior , Time Factors
7.
Eur J Neurosci ; 41(4): 434-41, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25615538

ABSTRACT

Behavioral flexibility is a component of executive functioning that allows individuals to adapt to changing environmental conditions. Independent lines of research indicate that the mu opioid receptor (MOR) is an important mediator of behavioral flexibility and responses to psychosocial stress. The current study bridges these two lines of research and tests the extent to which social defeat and MOR affect behavioral flexibility and whether sex moderates these effects in California mice (Peromyscus californicus). Males and females assigned to social defeat or control conditions were tested in a Barnes maze. In males, defeat impaired behavioral flexibility but not acquisition. Female performance was unaffected by defeat. MOR binding in defeated and control mice in the orbitofrontal cortex (OFC), striatum and hippocampus was examined via autoradiography. Stressed males had reduced MOR binding in the OFC whereas females were unaffected. The MOR antagonist beta-funaltrexamine (1 mg/kg) impaired performance in males naïve to defeat during the reversal phase but had no effect on females. Finally, we examined the effects of the MOR agonist morphine (2.5 and 5 mg/kg) on stressed mice. As expected, morphine improved behavioral flexibility in stressed males. The stress-induced deficits in behavioral flexibility in males are consistent with a proactive coping strategy, including previous observations that stressed male California mice exhibit strong social approach and aggression. Our pharmacological data suggest that a down-regulation of MOR signaling in males may contribute to sex differences in behavioral flexibility following stress. This is discussed in the framework of coping strategies for individuals with mood disorders.


Subject(s)
Aggression , Maze Learning , Receptors, Opioid, mu/metabolism , Stress, Psychological/metabolism , Animals , Female , Frontal Lobe/metabolism , Hippocampus/metabolism , Male , Peromyscus , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Sex Factors , Stress, Psychological/physiopathology
8.
Front Neuroendocrinol ; 35(4): 447-58, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24685383

ABSTRACT

Estradiol can modulate neural activity and behavior via both genomic and nongenomic mechanisms. Environmental cues have a major impact on the relative importance of these signaling pathways with significant consequences for behavior. First we consider how photoperiod modulates nongenomic estrogen signaling on behavior. Intriguingly, short days permit rapid effects of estrogens on aggression in both rodents and song sparrows. This highlights the importance of considering photoperiod as a variable in laboratory research. Next we review evidence for rapid effects of estradiol on ecologically-relevant behaviors including aggression, copulation, communication, and learning. We also address the impact of endocrine disruptors on estrogen signaling, such as those found in corncob bedding used in rodent research. Finally, we examine the biochemical mechanisms that may mediate rapid estrogen action on behavior in males and females. A common theme across these topics is that the effects of estrogens on social behaviors vary across different environmental conditions.


Subject(s)
Behavior/drug effects , Brain/drug effects , Environment , Estradiol/pharmacology , Estrogens/pharmacology , Signal Transduction/drug effects , Animals , Brain/metabolism , Humans
9.
Physiol Behav ; 128: 86-91, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24518867

ABSTRACT

Photoperiodic regulation of aggression has been well established in several vertebrate species, with rodents demonstrating increased aggression in short day photoperiods as compared to long day photoperiods. Previous work suggests that estrogens regulate aggression via rapid nongenomic pathways in short days and act more slowly in long days, most likely via genomic pathways. The current study therefore examines the role of melatonin in mediating aggression and estrogen-dependent gene transcription. In Experiment 1, male California mice were housed under long day photoperiods and were treated with either 0.3 µg/g of melatonin, 40 mg/kg of the melatonin receptor antagonist luzindole, or vehicle for 10 days. We found that melatonin administration significantly increased aggression as compared to mice receiving vehicle, but this phenotype was not completely ameliorated by luzindole. In Experiment 2, male California mice were injected with either 1mg/kg of the aromatase inhibitor letrozole or vehicle, and oxytocin receptor (OTR), estrogen receptor alpha (ERα), and c-fos gene expression was examined in the bed nucleus of the stria terminalis (BNST) and medial preoptic area (MPOA). In the BNST, but not MPOA, OTR mRNA was significantly downregulated following letrozole administration, indicating that OTR is an estrogen-dependent gene in the BNST. In contrast, ERα was not estrogen dependent in either brain region. In the MPOA, OTR mRNA was inhibited by melatonin, and luzindole suppressed this effect. C-fos and ERα did not differ between treatments in any brain region examined. These results suggest that it is unlikely that melatonin facilitates aggression via broad spectrum regulation of estrogen-dependent gene expression. Instead, melatonin may act via regulation of other transcription factors such as extracellular signal regulated kinase.


Subject(s)
Aggression/drug effects , Melatonin/pharmacology , Receptors, Melatonin/antagonists & inhibitors , Aggression/physiology , Aggression/psychology , Animals , Estrogen Receptor alpha/biosynthesis , Estrogens/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Letrozole , Male , Melatonin/physiology , Nitriles/pharmacology , Peromyscus/metabolism , Peromyscus/physiology , Peromyscus/psychology , Proto-Oncogene Proteins c-fos/biosynthesis , Receptors, Melatonin/physiology , Receptors, Oxytocin/biosynthesis , Triazoles/pharmacology , Tryptamines/pharmacology
10.
Horm Behav ; 64(3): 557-65, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23763907

ABSTRACT

In several vertebrate species, the effects of estrogens on male aggressive behavior can be modulated by environmental cues. In song sparrows and rodents, estrogens modulate aggression in the nonbreeding season or winter-like short days, respectively. The behavioral effects of estrogens are rapid, which generally is considered indicative of nongenomic processes. The current study further examined the hypothesis that estradiol acts nongenomically under short days by utilizing a protein synthesis inhibitor, cycloheximide (CX). Mice were housed in either short or long day photoperiods, and treated with an aromatase inhibitor. One hour before resident-intruder testing mice were injected with either CX or saline vehicle, and 30 min later were treated orally with either cyclodextrin conjugated estradiol or vehicle. Under short days, mice treated with estradiol showed a rapid decrease in aggressive behavior, independent of CX administration. CX alone had no effect on aggression. These results show that protein synthesis is not required for the rapid effects of estradiol on aggression, strongly suggesting that these effects are mediated by nongenomic processes. We also showed that estradiol suppressed c-fos immunoreactivity in the caudal bed nucleus of the stria terminalis under short days. No effects of estradiol on behavior or c-fos expression were observed in mice housed under long days. Previously we had also demonstrated that cage bedding influenced the directional effects of estrogens on aggression. Here, we show that the phenomenon of rapid action of estradiol on aggression under short days is a robust result that generalizes to different bedding conditions.


Subject(s)
Aggression/drug effects , Estradiol/pharmacology , Photoperiod , Animals , Arginine Vasopressin/metabolism , Circadian Rhythm/physiology , Cycloheximide/pharmacology , Female , Male , Peromyscus , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Seasons
11.
Horm Behav ; 63(3): 543-50, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23384773

ABSTRACT

There is compelling evidence for important sex differences in behavioral and hormonal responses to psychosocial stress. Here we examined the effects of gonadal hormones on behavioral responses to social defeat stress in monogamous California mice (Peromyscus californicus). Three episodes of social defeat induced social withdrawal in intact females but not males. Gonadectomy blocked corticosterone responses to defeat in females and sensitized male corticosterone responses. However, gonadectomy had no effects on social interaction behavior, suggesting that social withdrawal is not dependent on gonadal hormones in the adult California mouse. In contrast, defeat reduced exploratory behavior in the open field test for intact but not castrated males. We also examined the effects of social defeat on social interaction behavior when California mice were raised on corncob bedding, which has estrogenic properties. In this dataset of over 300 mice, we observed that social defeat did not induce social withdrawal when females were raised on corncob bedding. This finding suggests that the use of corncob in rodent studies could mask important sex differences in the effects of stress on brain and behavior. Although gonadal hormones do not affect social withdrawal behavior in adults, our data suggest that hormones may act earlier in development to induce a more resilient social phenotype.


Subject(s)
Behavior, Animal/physiology , Corticosterone/physiology , Gonadal Hormones/physiology , Phytoestrogens/pharmacology , Sex Characteristics , Animals , Castration , Corticosterone/metabolism , Female , Housing, Animal , Male , Neuropsychological Tests , Peromyscus , Phenotype , Stress, Psychological/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...