Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Neurobiol Stress ; 20: 100470, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36039150

ABSTRACT

Research over the past few decades has established a role for the endocannabinoid system in contributing to the neural and endocrine responses to stress exposure. The two endocannabinoid ligands, anandamide (AEA) and 2-arachidonoyl glycerol (2-AG), both play roles in regulating the stress response and both exhibit dynamic changes in response to stress exposure. Most of this previous research, however, was conducted in male rodents. Given that, especially in rodents, the stress response is influenced by sex, an understanding of how these dynamic responses of endocannabinoids in response to stress is influenced by sex could provide insight into sex differences of the acute stress response. We exposed adult, Sprague Dawley rats to different commonly utilized acute stress modalities, specifically restraint, swim and foot shock stress. Thirty minutes following stress onset, we excised the amygdala, hippocampus and medial prefrontal cortex, corticolimbic brain regions involved in the stress response, to measure endocannabinoid levels. When AEA levels were altered in response to restraint and swim stress, they were reduced, whereas exposure to foot shock stress led to an increase in the amygdala. 2-AG levels, when they were altered by stress exposure were only increased, specifically in males in the amygdala following swim stress, and in the hippocampus and medial prefrontal cortex overall following foot shock stress. This increase in 2-AG levels following stress only in males was the only sex difference found in stress-induced changes in endocannabinoid levels. There were no consistent sex differences observed. Collectively, these data contribute to our further understanding of the interactions between stress and endocannabinoid function.

2.
Neuropharmacology ; 146: 154-162, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30496752

ABSTRACT

Early-life stress modulates the development of cortico-limbic circuits and increases vulnerability to adult psychopathology. Given the important stress-buffering role of endocannabinoid (eCB) signaling, we performed a comprehensive investigation of the developmental trajectory of the eCB system and the impact of exposure to early life stress induced by repeated maternal separation (MS; 3 h/day) from postnatal day 2 (PND2) to PND12. Tissue levels of the eCB molecules anandamide (AEA) and 2-arachidonoylglycerol (2-AG) were measured after MS exposures, as well under basal conditions at juvenile (PND14), adolescent (PND40) and adult (PND70) timepoints in the prefrontal cortex (PFC), amygdala and hippocampus. We also examined the effects of MS on CB1 receptor binding in these three brain regions at PND40 and PND70. AEA content was found to increase from PND2 into adulthood in a linear manner across all brain regions, while 2-AG was found to exhibit a transient spike during the juvenile period (PND12-14) within the amygdala and PFC, but increased in a linear manner across development in the hippocampus. Exposure to MS resulted in bidirectional changes in AEA and 2-AG tissue levels within the amygdala and hippocampus and produced a sustained reduction in eCB function in the hippocampus at adulthood. CB1 receptor densities across all brain regions were generally found to be downregulated later in life following exposure to MS. Collectively, these data demonstrate that early life stress can alter the normative ontogeny of the eCB system, resulting in a sustained deficit in eCB function, particularly within the hippocampus, in adulthood.


Subject(s)
Endocannabinoids/metabolism , Maternal Deprivation , Stress, Psychological/metabolism , Amygdala/metabolism , Animals , Arachidonic Acids/metabolism , Female , Glycerides/metabolism , Hippocampus/metabolism , Humans , Male , Polyunsaturated Alkamides/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism
3.
Proc Natl Acad Sci U S A ; 115(29): 7605-7610, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29967158

ABSTRACT

Endocannabinoid signaling regulates feeding and metabolic processes and has been linked to obesity development. Several hormonal signals, such as glucocorticoids and ghrelin, regulate feeding and metabolism by engaging the endocannabinoid system. Similarly, studies have suggested that leptin interacts with the endocannabinoid system, yet the mechanism and functional relevance of this interaction remain elusive. Therefore, we explored the interaction between leptin and endocannabinoid signaling with a focus on fatty acid amide hydrolase (FAAH), the primary degradative enzyme for the endocannabinoid N-arachidonoylethanolamine (anandamide; AEA). Mice deficient in leptin exhibited elevated hypothalamic AEA levels and reductions in FAAH activity while leptin administration to WT mice reduced AEA content and increased FAAH activity. Following high fat diet exposure, mice developed resistance to the effects of leptin administration on hypothalamic AEA content and FAAH activity. At a functional level, pharmacological inhibition of FAAH was sufficient to prevent leptin-mediated effects on body weight and food intake. Using a novel knock-in mouse model recapitulating a common human polymorphism (FAAH C385A; rs324420), which reduces FAAH activity, we investigated whether human genetic variance in FAAH affects leptin sensitivity. While WT (CC) mice were sensitive to leptin-induced reductions in food intake and body weight gain, low-expressing FAAH (AA) mice were unresponsive. These data demonstrate that FAAH activity is required for leptin's hypophagic effects and, at a translational level, suggest that a genetic variant in the FAAH gene contributes to differences in leptin sensitivity in human populations.


Subject(s)
Amidohydrolases/metabolism , Arachidonic Acids/metabolism , Eating , Endocannabinoids/metabolism , Energy Metabolism/drug effects , Hypothalamus/metabolism , Leptin/pharmacology , Polyunsaturated Alkamides/metabolism , Amidohydrolases/genetics , Animals , Body Weight/drug effects , Body Weight/genetics , Dietary Fats/pharmacology , Eating/drug effects , Eating/genetics , Gene Knock-In Techniques , Leptin/deficiency , Male , Mice , Mice, Knockout , Polymorphism, Genetic
4.
Psychoneuroendocrinology ; 66: 151-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26821211

ABSTRACT

Sustained exposure to stress or corticosteroids is known to cause changes in brain endocannabinoid (eCB) signaling, such that tissue contents of the eCBs N-arachidonylethanolamine (AEA) are generally reduced while 2-arachidonoylglycerol (2-AG) levels increase. These changes in eCB signaling are important for many of the aspects of chronic stress, such as anxiety, reward sensitivity and stress adaptation, yet the mechanisms mediating these changes are not fully understood. We have recently found that the stress-related neuropeptide corticotropin-releasing hormone (CRH), acting through the CRH type 1 receptor (CRHR1), can reduce AEA content by increasing its hydrolysis by the enzyme fatty acid amide hydrolase (FAAH) as well as increase 2-AG contents. As extra-hypothalamic CRH is upregulated by chronic corticosteroid or stress exposure, we hypothesized that increased CRH signaling through CRHR1 contributes to the effects of chronic corticosteroid exposure on the eCB system within the amygdala and prefrontal cortex. Male rats were exposed to 7 days of systemic corticosterone capsules, with or without concurrent exposure to a CRHR1 antagonist, after which we examined eCB content. Consistent with previous studies in the amygdala, sustained corticosterone exposure increases CRH mRNA in the prefrontal cortex. As was shown previously, FAAH activity was increased and AEA contents were reduced within the amygdala and prefrontal cortex following chronic corticosterone exposure. Chronic corticosterone exposure also elevated 2-AG content in the prefrontal cortex but not the amygdala. These corticosteroid-driven changes were all blocked by systemic CRHR1 antagonism. Consistent with these data indicating sustained increases in CRH signaling can mediate the effects of chronic elevations in corticosteroids, CRH overexpressing mice also exhibited increased FAAH-mediated AEA hydrolysis in the amygdala and prefrontal cortex compared to wild type. CRH overexpression increased 2-AG content in the amygdala, but not the prefrontal cortex. These data indicate that chronic elevations in CRH signaling, as is seen following exposure to chronic elevations in corticosterone or stress, drive persistent changes in eCB function. As reductions in AEA signaling mediate the effects of CRH and chronic stress on anxiety, these data provide a mechanism linking these processes.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Endocannabinoids/physiology , Glucocorticoids/pharmacology , Limbic System/drug effects , Prefrontal Cortex/drug effects , Amygdala/drug effects , Amygdala/metabolism , Animals , Endocannabinoids/metabolism , Limbic System/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Restraint, Physical/psychology , Signal Transduction/drug effects , Stress, Psychological/metabolism
5.
Int Rev Neurobiol ; 125: 49-84, 2015.
Article in English | MEDLINE | ID: mdl-26638764

ABSTRACT

Adolescence is a period characterized by many distinct physical, behavioral, and neural changes during the transition from child- to adulthood. In particular, adolescent neural changes often confer greater plasticity and flexibility, yet with this comes the potential for heightened vulnerability to external perturbations such as stress exposure or recreational drug use. There is substantial evidence to suggest that factors such as adolescent stress exposure have longer lasting and sometimes more deleterious effects on an organism than stress exposure during adulthood. Moreover, the adolescent neuroendocrine response to stress exposure is different from that of adults, suggesting that further maturation of the adolescent hypothalamic-pituitary-adrenal (HPA) axis is required. The endocannabinoid (eCB) system is a potential candidate underlying these age-dependent differences given that it is an important regulator of the adult HPA axis and neuronal development. Therefore, this review will focus on (1) the functionality of the adolescent HPA axis, (2) eCB regulation of the adult HPA axis, (3) dynamic changes in eCB signaling during the adolescent period, (4) the effects of adolescent stress exposure on the eCB system, and (5) modulation of HPA axis activity and emotional behavior by adolescent cannabinoid treatment. Collectively, the emerging picture suggests that the eCB system mediates interactions between HPA axis stress responsivity, emotionality, and maturational stage. These findings may be particularly relevant to our understanding of the development of affective disorders and the risks of adolescent cannabis consumption on emotional health and stress responsivity.


Subject(s)
Adolescent Behavior , Brain/growth & development , Endocannabinoids/physiology , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Signal Transduction , Stress, Psychological/physiopathology , Adolescent , Brain/physiopathology , Humans , Signal Transduction/physiology
6.
Neuropharmacology ; 99: 89-97, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26192544

ABSTRACT

The endocannabinoid (eCB) system is known to regulate neural, endocrine and behavioral responses to stress in adults; however there is little knowledge regarding how this system governs the development and maturation of these responses. Previous work has reported dynamic and time-specific changes in CB1 receptor expression, N-arachidonylethanolamine (AEA) content and fatty acid amide hydrolase (FAAH) activity within corticolimbic structures throughout the peri-adolescent period. To examine whether fluctuations in adolescent eCB activity contribute to the development of adult stress responsivity and emotionality, we treated male Sprague-Dawley rats daily with the CB1R antagonist, AM-251 (5 mg/kg), or vehicle between post-natal days (PND) 35-45. Following this treatment, emotional behavior, HPA axis stress reactivity and habituation to repeated restraint stress, as well as corticolimbic eCB content were examined in adulthood (PND 75). Behaviorally, AM-251-treated males exhibited more active stress-coping behavior in the forced swim test, greater risk assessment behavior in the elevated plus maze and no significant differences in general motor activity. Peri-adolescent AM-251 treatment modified corticosterone habituation to repeated restraint exposure compared to vehicle. Peri-adolescent CB1R antagonism induced moderate changes in adult corticolimbic eCB signaling, with a significant decrease in amygdalar AEA, an increase in hypothalamic AEA and an increase in prefrontal cortical CB1R expression. Together, these data indicate that peri-adolescent endocannabinoid signaling contributes to the maturation of adult neurobehavioral responses to stress.


Subject(s)
Brain/growth & development , Cannabinoid Receptor Antagonists/toxicity , Endocannabinoids/metabolism , Piperidines/toxicity , Pyrazoles/toxicity , Receptor, Cannabinoid, CB1/metabolism , Stress, Psychological/metabolism , Adaptation, Psychological/drug effects , Adaptation, Psychological/physiology , Adrenocorticotropic Hormone/metabolism , Animals , Brain/drug effects , Brain/metabolism , Corticosterone/metabolism , Disease Models, Animal , Emotions/drug effects , Emotions/physiology , Male , Motor Activity/drug effects , Motor Activity/physiology , Random Allocation , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Restraint, Physical , Risk-Taking
7.
J Neurosci ; 35(9): 3879-92, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25740517

ABSTRACT

Corticotropin-releasing hormone (CRH) is a central integrator in the brain of endocrine and behavioral stress responses, whereas activation of the endocannabinoid CB1 receptor suppresses these responses. Although these systems regulate overlapping functions, few studies have investigated whether these systems interact. Here we demonstrate a novel mechanism of CRH-induced anxiety that relies on modulation of endocannabinoids. Specifically, we found that CRH, through activation of the CRH receptor type 1 (CRHR1), evokes a rapid induction of the enzyme fatty acid amide hydrolase (FAAH), which causes a reduction in the endocannabinoid anandamide (AEA), within the amygdala. Similarly, the ability of acute stress to modulate amygdala FAAH and AEA in both rats and mice is also mediated through CRHR1 activation. This interaction occurs specifically in amygdala pyramidal neurons and represents a novel mechanism of endocannabinoid-CRH interactions in regulating amygdala output. Functionally, we found that CRH signaling in the amygdala promotes an anxious phenotype that is prevented by FAAH inhibition. Together, this work suggests that rapid reductions in amygdala AEA signaling following stress may prime the amygdala and facilitate the generation of downstream stress-linked behaviors. Given that endocannabinoid signaling is thought to exert "tonic" regulation on stress and anxiety responses, these data suggest that CRH signaling coordinates a disruption of tonic AEA activity to promote a state of anxiety, which in turn may represent an endogenous mechanism by which stress enhances anxiety. These data suggest that FAAH inhibitors may represent a novel class of anxiolytics that specifically target stress-induced anxiety.


Subject(s)
Amygdala/metabolism , Amygdala/physiopathology , Anxiety/metabolism , Anxiety/physiopathology , Arachidonic Acids/metabolism , Corticotropin-Releasing Hormone/metabolism , Endocannabinoids/metabolism , Polyunsaturated Alkamides/metabolism , Adrenocorticotropic Hormone/metabolism , Amidohydrolases/metabolism , Animals , Hydrolysis , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/physiology , Stress, Psychological/metabolism , Stress, Psychological/psychology
8.
Behav Brain Res ; 271: 106-10, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24907533

ABSTRACT

The medial prefrontal cortex (mPFC) is known to regulate higher order processes like cognitive flexibility. Accumulating behavioral evidence suggests that endocannabinoid (eCB) signaling regulates neuronal architecture within the PFC, as well as certain forms of cognitive flexibility; however, all of these studies have been performed in male rodents and it is currently unknown whether the eCB system performs a similar role in females. To this extent, dendritic morphology of layer II/III neurons in the infra- and prelimbic regions of the mPFC was analyzed and cognitive ability and flexibility in a fixed-platform Morris water maze task was assessed in adult female CB1 receptor knockout (CB1KO) mice. Similar to data generated in male mice, female mice exhibited no difference in acquisition relative to wildtype (WT); however, during reversal learning, CB1KO females spent more time in the original training quadrant and took significantly longer to learn the location of the new platform relative to WT. Within the mPFC, female mice had reduced length and complexity of layer II/III neurons within the prelimbic, but not infralimbic region of the PFC. Taken together, these findings indicate that the role of eCB signaling in cognitive flexibility is independent of sex and disrupted CB1 receptor signaling results in compromised structure and function of the PFC, at least within the prelimbic division.


Subject(s)
Cognition , Neurons/metabolism , Prefrontal Cortex/cytology , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/metabolism , Animals , Behavior, Animal , Female , Male , Maze Learning , Memory , Mice , Mice, Inbred C57BL , Mice, Knockout , Reversal Learning , Sex Factors , Stress, Psychological/metabolism
9.
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
10.
Synapse ; 67(1): 4-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22987804

ABSTRACT

Fatty acid amide hydrolase (FAAH) regulates tissue concentrations of N-acylethanolamines (NAEs), including the endocannabinoid, N-arachidonylethanolamide (anandamide, AEA). FAAH activity and NAEs are widely distributed throughout the brain and FAAH activity regulates an array of processes including emotion, cognition, inflammation, and feeding. However, there is relatively little research describing how this system develops throughout adolescence, particularly within limbic circuits regulating stress and reward processing. Thus, this study characterized temporal changes in NAE content (AEA, oleoylethanolamine [OEA], and palmitoylethanolamide [PEA]) and FAAH activity across the peri-adolescent period, in four corticolimbic structures (amygdala, hippocampus, prefrontal cortex, and hypothalamus). Brain tissue of male Sprague-Dawley rats was collected on postnatal days (PND) 25, 35, 45, and 70, representing pre-adolescence, early- to mid-adolescence, late adolescence, and adulthood, respectively. Tissue was analyzed for AEA, OEA, and PEA content as well as FAAH activity at each time point. AEA, OEA, and PEA exhibited a similar temporal pattern in all four brain regions. NAE concentrations were lowest at PND 25 and highest at PND 35. NAE concentrations decreased between PNDs 35 and 45 and increased between PNDs 45 and 70. FAAH activity mirrored the pattern of NAE content in which it decreased between PNDs 25 and 35, increased between PNDs 35 and 45, and decreased between PNDs 45 and 70. These age-dependent patterns of NAE content and FAAH activity demonstrate temporal specificity to the development of this system and could contribute to alterations in stress sensitivity, emotionality, and executive function which also fluctuate during this developmental period.


Subject(s)
Ethanolamines/metabolism , Limbic System/metabolism , Age Factors , Amidohydrolases/metabolism , Animals , Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Limbic System/growth & development , Male , Polyunsaturated Alkamides/metabolism , Rats , Rats, Sprague-Dawley
11.
J Neurosci ; 31(29): 10506-15, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21775596

ABSTRACT

The mechanisms subserving the ability of glucocorticoid signaling within the medial prefrontal cortex (mPFC) to terminate stress-induced activation of the hypothalamic-pituitary-adrenal (HPA) axis are not well understood. We report that antagonism of the cannabinoid CB(1) receptor locally within the mPFC prolonged corticosterone secretion following cessation of stress in rats. Mice lacking the CB(1) receptor exhibited a similar prolonged response to stress. Exposure of rats to stress produced an elevation in the endocannabinoid 2-arachidonoylglycerol within the mPFC that was reversed by pretreatment with the glucocorticoid receptor antagonist RU-486 (20 mg/kg). Electron microscopic and electrophysiological data demonstrated the presence of CB(1) receptors in inhibitory-type terminals impinging upon principal neurons within layer V of the prelimbic region of the mPFC. Bath application of corticosterone (100 nm) to prefrontal cortical slices suppressed GABA release onto principal neurons in layer V of the prelimbic region, when examined 1 h later, which was prevented by application of a CB(1) receptor antagonist. Collectively, these data demonstrate that the ability of stress-induced glucocorticoid signaling within mPFC to terminate HPA axis activity is mediated by a local recruitment of endocannabinoid signaling. Endocannabinoid activation of CB(1) receptors decreases GABA release within the mPFC, likely increasing the outflow of the principal neurons of the prelimbic region to contribute to termination of the stress response. These data support a model in which endocannabinoid signaling links glucocorticoid receptor engagement to activation of corticolimbic relays that inhibit corticosterone secretion.


Subject(s)
Arachidonic Acids/metabolism , Glycerides/metabolism , Signal Transduction/physiology , Stress, Psychological/metabolism , Stress, Psychological/pathology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Corticosterone/pharmacology , Disease Models, Animal , Electric Stimulation/methods , Endocannabinoids , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Hormone Antagonists/pharmacology , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Microscopy, Electron, Transmission , Mifepristone/pharmacology , Patch-Clamp Techniques/methods , Piperidines/pharmacology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/deficiency , Signal Transduction/drug effects , Stress, Psychological/drug therapy , gamma-Aminobutyric Acid/metabolism
12.
Proc Natl Acad Sci U S A ; 107(20): 9406-11, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20439721

ABSTRACT

Secretion of glucocorticoid hormones during stress produces an array of physiological changes that are adaptive and beneficial in the short term. In the face of repeated stress exposure, however, habituation of the glucocorticoid response is essential as prolonged glucocorticoid secretion can produce deleterious effects on metabolic, immune, cardiovascular, and neurobiological function. Endocannabinoid signaling responds to and regulates the activity of the hypothalamic-pituitary-adrenal (HPA) axis that governs the secretion of glucocorticoids; however, the role this system plays in adaptation of the neuroendocrine response to repeated stress is not well characterized. Herein, we demonstrate a divergent regulation of the two endocannabinoid ligands, N-arachidonylethanolamine (anandamide; AEA) and 2-arachidonoylglycerol (2-AG), following repeated stress such that AEA content is persistently decreased throughout the corticolimbic stress circuit, whereas 2-AG is exclusively elevated within the amygdala in a stress-dependent manner. Pharmacological studies demonstrate that this divergent regulation of AEA and 2-AG contribute to distinct forms of HPA axis habituation. Inhibition of AEA hydrolysis prevented the development of basal hypersecretion of corticosterone following repeated stress. In contrast, systemic or intra-amygdalar administration of a CB(1) receptor antagonist before the final stress exposure prevented the repeated stress-induced decline in corticosterone responses. The present findings demonstrate an important role for endocannabinoid signaling in the process of stress HPA habituation, and suggest that AEA and 2-AG modulate different components of the adrenocortical response to repeated stressor exposure.


Subject(s)
Adaptation, Physiological/physiology , Cannabinoid Receptor Modulators/metabolism , Signal Transduction/physiology , Stress, Physiological/physiology , Adaptation, Physiological/drug effects , Analysis of Variance , Animals , Arachidonic Acids/antagonists & inhibitors , Arachidonic Acids/pharmacology , Benzamides/pharmacology , Carbamates/pharmacology , Corticosterone/blood , Endocannabinoids , Male , Piperidines/pharmacology , Polyunsaturated Alkamides/antagonists & inhibitors , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Signal Transduction/drug effects
13.
Psychoneuroendocrinology ; 35(8): 1265-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20207489

ABSTRACT

Sex differences have been identified in many of the behavioral and physiological effects of cannabinoids. While estrogen has been linked to some of these variations, the effects of estrogen on cannabinoid receptor binding have not been characterized within regions of the brain specifically implicated in stress responsivity and emotional behavior. To examine sex differences, and the role of estradiol, in regulation of the cannabinoid receptor, we compared the binding site density of the cannabinoid receptor within the amygdala, hippocampus and hypothalamus in males, cycling females, ovariectomized (OVX) females and estradiol-treated OVX females (OVX+E). Our data reveal that males and OVX females have higher amounts of hypothalamic and lower amounts of amygdalar cannabinoid receptor binding relative to both cycling females and OVX+E females. Within the hippocampus, ovariectomy resulted in an upregulation of cannabinoid receptor binding. These data provide a putative biochemical mechanism mediating the observed behavioral and physiological sex differences in the effects of cannabinoids, particularly with respect to stress and emotional behavior.


Subject(s)
Limbic System/drug effects , Receptors, Cannabinoid/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Cannabinoid Receptor Agonists , Estradiol/pharmacology , Estrous Cycle/drug effects , Estrous Cycle/metabolism , Estrous Cycle/physiology , Female , Hippocampus/drug effects , Hippocampus/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Limbic System/metabolism , Male , Ovariectomy , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley
14.
Hippocampus ; 20(4): 513-23, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19489006

ABSTRACT

Voluntary exercise and endogenous cannabinoid activity have independently been shown to regulate hippocampal plasticity. The aim of the current study was to determine whether the endocannabinoid system is regulated by voluntary exercise and if these changes contribute to exercise-induced enhancement of cell proliferation. In Experiment 1, 8 days of free access to a running wheel increased the agonist binding site density of the cannabinoid CB(1) receptor; CB(1) receptor-mediated GTPgammaS binding; and the tissue content of the endocannabinoid anandamide in the hippocampus but not in the prefrontal cortex. In Experiment 2, the CB(1) receptor antagonist AM251 (1 mg kg(-1)) was administered daily to animals given free access to a running wheel for 8 days, after which cell proliferation in the hippocampus was examined through immunohistochemical analysis of the cell cycle protein Ki-67. Voluntary exercise increased proliferation of progenitor cells, as evidenced by the increase in the number of Ki-67 positive cells in the granule cell layer of the dentate gyrus (DG) in the hippocampus. However, this effect was abrogated by concurrent treatment with AM251, indicating that the increase in endocannabinoid signaling in the hippocampus is required for the exercise-induced increase in cell proliferation. These data demonstrate that the endocannabinoid system in the hippocampus is sensitive to environmental change and suggest that it is a mediator of experience-induced plasticity.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Cell Proliferation , Hippocampus/metabolism , Physical Conditioning, Animal/physiology , Receptor, Cannabinoid, CB1/metabolism , Analysis of Variance , Animals , Cell Count , Cell Cycle/physiology , Immunohistochemistry , Neurogenesis , Neurons/metabolism , Prefrontal Cortex/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Signal Transduction , Stem Cells
15.
Eur J Pharmacol ; 602(1): 54-7, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-19049807

ABSTRACT

An organism's ability to adapt successfully to stress reflects an equilibrium that requires not only an appropriate response, but also an ability to control that response. The hypothalamic-pituitary-adrenal (HPA) axis contributes to these homeostatic actions. Previous research implicates involvement of the serotonergic 5-HT(2A) receptors of the hypothalamic paraventricular nucleus (PVN) in HPA axis activation. However, the sensitivity of these receptors to activate the PVN under conditions of chronically elevated glucocorticoids is not known. To this extent, we investigated the effects of chronic corticosterone administration on c-fos expression induced by the serotonergic 5-HT(2A/2C) receptor agonist DOI within the PVN. Under resting conditions, DOI evokes a robust activation of the PVN; however, following chronic treatment with corticosterone, this response is abolished. These results indicate that chronically elevated glucocorticoid levels desensitize serotonergic 5-HT(2A) receptors within the PVN, a phenomenon which may contribute to HPA axis suppression following protracted glucocorticoid hypersecretion.


Subject(s)
Glucocorticoids/pharmacology , Neurons/drug effects , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Animals , Glucocorticoids/administration & dosage , Immunohistochemistry , Male , Rats , Rats, Long-Evans , Time Factors
16.
J Comp Psychol ; 120(3): 217-28, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16893259

ABSTRACT

The acoustic frequency ranges in birdsongs provide important absolute pitch cues for the recognition of conspecifics. Black-capped chickadees (Poecile atricapillus), mountain chickadees (Poecile gambeli), and zebra finches (Taeniopygia guttata) were trained to sort tones contiguous in frequency into 8 ranges on the basis of associations between response to the tones in each range and reward. All 3 species acquired accurate frequency-range discriminations, but zebra finches acquired the discrimination in fewer trials and to a higher standard than black-capped or mountain chickadees, which did not differ appreciably in the discrimination. Chickadees' relatively poorer accuracy was traced to poorer discrimination of tones in the higher frequency ranges. During transfer tests, the discrimination generalized to novel tones when the training tones were included, but not when they were omitted.


Subject(s)
Auditory Perception , Finches , Pitch Discrimination , Songbirds , Vocalization, Animal , Animal Communication , Animals , Conditioning, Operant , Female , Generalization, Stimulus , Male , Seasons , Sound Spectrography , Species Specificity , Transfer, Psychology
17.
J Comp Psychol ; 119(4): 371-80, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16366770

ABSTRACT

Acoustic communication in black-capped chickadees (Poecile atricapillus) has been studied intensively, the "chick-a-dee" call being among the most well described. This call consists of 4 note types; chickadees perceive these notes as open-ended categories and do so in a continuous manner, with As more similar to Bs and Bs more similar to Cs. Acoustic features contributing to the note-type differentiation are unknown. Recent analyses suggested that certain acoustic features may play a role in note-type classification. Here, the authors tested black-capped chickadees in an operant-conditioning paradigm to determine which features were controlling note-type perception. The results suggest that the note pitch and the frequency modulation in the initial portion of the note control the perception of note types.


Subject(s)
Echolocation , Visual Perception , Vocalization, Animal , Animals , Birds , Discrimination, Psychological , Female , Male
18.
Article in English | MEDLINE | ID: mdl-14499306

ABSTRACT

Melatonin has been demonstrated to increase activity in the forced swim test (FST), a putative model of antidepressant efficacy, indicating that it may possess antidepressant-like qualities. It has been suggested that corticosterone can interfere with the efficacy of antidepressants, an effect that has previously been demonstrated in the FST. This experiment examined the effects of melatonin and corticosterone, independently and in combination, on the behaviours of both male and female rats in the FST. Corticosterone, melatonin, combined vehicles or a combined melatonin/corticosterone regimen were administered for 20 days, after which the animals were observed in the FST. As seen in previous research, melatonin elicited an antidepressant-like effect in the FST by reducing immobile behaviour (P<.01) and increasing active behaviour (P<.01). Corticosterone was found to reduce activity (P<.01) and increase immobility (P<.01), as well as attenuate the anti-immobility effects of melatonin (P=.03). These findings suggest that while melatonin may possess antidepressant-like qualities, high levels of corticosterone seem capable of attenuating these effects.


Subject(s)
Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Corticosterone/pharmacology , Depression/drug therapy , Melatonin/therapeutic use , Sex Characteristics , Animals , Antidepressive Agents/antagonists & inhibitors , Depression/psychology , Drug Combinations , Drug Interactions , Female , Immobilization/physiology , Male , Melatonin/antagonists & inhibitors , Rats , Rats, Long-Evans , Swimming/physiology
19.
Physiol Behav ; 77(2-3): 217-25, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12419397

ABSTRACT

The present study investigated sex differences and the effect of a high level of estradiol in the female meadow vole on performance in the forced swim test (FST) and the Morris water maze in meadow voles. Female meadow voles were ovariectomized (OVX) and administered either vehicle (sesame oil) or estradiol for 2 days prior to performing the FST. Four days following the FST, all animals were run in the Morris water maze. Results indicated that estradiol-injected female meadow voles showed more 'depressive-like' behaviors in the FST (greater time spent immobile and less time spent swimming) than vehicle-treated female or male meadow voles. In addition, estradiol-treated females had impaired performance (greater latencies and distance swam to reach the hidden platform) than both vehicle-treated female and male meadow voles, consistent with previous data. Despite the fact that estradiol administration increased 'depressive-like' behaviors in the FST and impaired performance in the Morris water maze, there was no correlation between the two behaviors indicating that 'depressive-like' behaviors did not account for the differences seen in spatial performance in the Morris water maze. To our knowledge, this is the first demonstration in rodents indicating that estradiol-mediated changes in behavior in the FST is not indicative of subsequent performance in the Morris water maze.


Subject(s)
Arvicolinae/physiology , Depression/psychology , Estradiol/pharmacology , Maze Learning/drug effects , Animals , Diving/psychology , Female , Motor Activity/drug effects , Ovariectomy , Swimming/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...