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










Publication year range
1.
bioRxiv ; 2024 May 11.
Article in English | MEDLINE | ID: mdl-38766105

ABSTRACT

Sleep problems are a prominent feature of mental health conditions including post-traumatic stress disorder (PTSD). Despite its potential importance, the role of sleep in the development of and/or recovery from trauma-related illnesses is not understood. Interestingly, there are reports that sleep deprivation immediately after a traumatic experience can reduce fear memories, an effect that could be utilized therapeutically in humans. While the mechanisms of this effect are not completely understood, one possible explanation for these findings is that immediate sleep deprivation interferes with consolidation of fear memories, rendering them weaker and more sensitive to intervention. Here, we allowed fear-conditioned mice to sleep immediately after fear conditioning during a time frame (18 hr) that includes and extends beyond periods typically associated with memory consolidation before subjecting them to 6 hr of sleep deprivation. Mice deprived of sleep with this delayed regimen showed dramatic reductions in fear during tests conducted immediately after sleep deprivation, as well as 24 hr later. This sleep deprivation regimen also increased levels of mRNA encoding brain-derived neurotrophic factor (BDNF), a molecule implicated in neuroplasticity, in the basolateral amygdala (BLA), a brain area implicated in fear and its extinction. These findings raise the possibility that the effects of our delayed sleep deprivation regimen are not due to disruption of memory consolidation, but instead are caused by BDNF-mediated neuroadaptations within the BLA that actively suppress expression of fear. Treatments that safely reduce expression of fear memories would have considerable therapeutic potential in the treatment of conditions triggered by trauma.

2.
bioRxiv ; 2023 Mar 24.
Article in English | MEDLINE | ID: mdl-36993188

ABSTRACT

Stress produces profound effects on behavior, including persistent alterations in sleep patterns. Here we examined the effects of two prototypical stress peptides, pituitary adenylate cyclase-activating polypeptide (PACAP) and corticotropin-releasing factor (CRF), on sleep architecture and other translationally-relevant endpoints. Male and female mice were implanted with subcutaneous transmitters enabling continuous measurement of electroencephalography (EEG) and electromyography (EMG), as well as body temperature and locomotor activity, without tethering that restricts free movement, body posture, or head orientation during sleep. At baseline, females spent more time awake (AW) and less time in slow wave sleep (SWS) than males. Mice then received intracerebral infusions of PACAP or CRF at doses producing equivalent increases in anxiety-like behavior. The effects of PACAP on sleep architecture were similar in both sexes and resembled those reported in male mice after chronic stress exposure. Compared to vehicle infusions, PACAP infusions decreased time in AW, increased time in SWS, and increased rapid eye movement sleep (REM) time and bouts on the day following treatment. In addition, PACAP effects on REM time remained detectable a week after treatment. PACAP infusions also reduced body temperature and locomotor activity. Under the same experimental conditions, CRF infusions had minimal effects on sleep architecture in either sex, causing only transient increases in SWS during the dark phase, with no effects on temperature or activity. These findings suggest that PACAP and CRF have fundamentally different effects on sleep-related metrics, and provide new insights into the mechanisms by which stress disrupts sleep.

3.
Biol Psychiatry ; 89(12): 1138-1149, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33715826

ABSTRACT

BACKGROUND: Stress is implicated in the pathophysiology of major depression and posttraumatic stress disorder. These conditions share core features, including motivational deficits, heighted anxiety, and sleep dysregulation. Chronic stress produces these same features in rodents, with some individuals being susceptible or resilient, as seen in humans. While stress-induced neuroadaptations within the nucleus accumbens are implicated in susceptibility-related dysregulation of motivational and emotional behaviors, their effects on sleep are unclear. METHODS: We used chemogenetics (DREADDs [designer receptors exclusively activated by designer drugs]) to examine the effects of selective alterations in activity of nucleus accumbens medium spiny neurons expressing dopamine D1 receptors (D1-MSNs) or dopamine D2 receptors (D2-MSNs) on sleep-related end points. Mice were implanted with wireless transmitters enabling continuous collection of data to quantify vigilance states over a 20-day test period. Parallel cohorts were examined in behavioral tests assessing stress susceptibility. RESULTS: D1- and D2-MSNs play dissociable roles in sleep regulation. Stimulation of inhibitory or excitatory DREADDs expressed in D1-MSNs exclusively affects rapid eye movement sleep, whereas targeting D2-MSNs affects slow wave sleep. The combined effects of D1-MSN inhibition and D2-MSN activation on sleep resemble those of chronic social defeat stress. Alterations in D1-MSN function also affect stress susceptibility in social behavior tests. Elevation of CREB (cAMP response element-binding protein) within D1-MSNs is sufficient to produce stress-like effects on rapid eye movement sleep. CONCLUSIONS: In addition to regulation of motivational and emotional behaviors, the nucleus accumbens also influences sleep, an end point with high translational relevance. These findings provide a neural basis for comorbidity in key features of stress-related illness.


Subject(s)
Nucleus Accumbens , Receptors, Dopamine D1 , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Sleep
4.
Behav Brain Res ; 396: 112884, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32871228

ABSTRACT

Accurate discrimination between safe and dangerous stimuli is essential for survival. Prior research has begun to uncover the neural structures that are necessary for learning this discrimination, but exploration of brain regions involved in this learning process has been mostly limited to males. Recent findings show sex differences in discrimination learning, with reduced fear expression to safe cues in females compared to males. Here, we used male and female Sprague Dawley rats to explore neural activation, as measured by Fos expression, in fear and safety learning related brain regions. Neural activation after fear discrimination (Discrimination) was compared between males and females, as well as with fear conditioned (Fear Only) and stimulus presented (Control) conditions. Correlations of discrimination ability and neural activation were also calculated. We uncovered a correlation between central amygdala (CeA) activation and discrimination abilities in males and females. Anterior medial bed nucleus of the stria terminalis (BNST) was the only region where sex differences in Fos counts were observed in the Discrimination condition, and the only region where neural activation significantly differed between Fear Only and Discrimination conditions. Together, these findings indicate the importance of fear expression circuitry in mediating discrimination responses and generate important questions for future investigation.


Subject(s)
Central Amygdaloid Nucleus/physiology , Conditioning, Classical/physiology , Discrimination Learning/physiology , Fear/physiology , Septal Nuclei/physiology , Sex Characteristics , Animals , Behavior, Animal/physiology , Central Amygdaloid Nucleus/metabolism , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Septal Nuclei/metabolism
5.
Neuroscience ; 379: 350-358, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29604383

ABSTRACT

Survival depends on adaptation to shifting environmental risks and opportunities. Regarding risks, the mechanisms which permit acquisition, recall, and flexible use of aversive associations is poorly understood. Drawing on the evidence that the orbital frontal cortex is critical to integrating outcome expectancies with flexible appetitive behavioral responses, we hypothesized that OFC would contribute to behavioral flexibility within an aversive learning domain. We introduce a fear conditioning procedure in which adult male rats were presented with shock-paired conditioned stimulus (CS+) or a safety cue (CS-). In a recall test, rats exhibit greater freezing to the CS+ than the CS-. Temporary inactivation of the ventrolateral OFC with muscimol prior to conditioning did not affect later discrimination, but inactivation after learning and prior to recall impaired discrimination between safety and danger cues. This result complements prior research in the appetitive domain and suggests that the OFC plays a general role in behavioral flexibility regardless of the valence of the CS.


Subject(s)
Conditioning, Psychological/physiology , Discrimination, Psychological/physiology , Fear/physiology , Inhibition, Psychological , Prefrontal Cortex/physiology , Animals , Anticipation, Psychological/drug effects , Anticipation, Psychological/physiology , Association , Auditory Perception/drug effects , Auditory Perception/physiology , Avoidance Learning/drug effects , Avoidance Learning/physiology , Conditioning, Psychological/drug effects , Discrimination, Psychological/drug effects , Electroshock , Fear/drug effects , Fear/psychology , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , GABA-A Receptor Agonists/pharmacology , Male , Muscimol/pharmacology , Prefrontal Cortex/drug effects , Rats, Sprague-Dawley , Visual Perception/drug effects , Visual Perception/physiology
6.
Learn Mem ; 25(1): 49-53, 2018 01.
Article in English | MEDLINE | ID: mdl-29246981

ABSTRACT

Distinguishing safety from danger is necessary for survival, but is aberrant in individuals with post-traumatic stress disorder (PTSD). While PTSD is more prevalent in women than men, research on sex differences in safety learning is limited. Here, female rats demonstrated greater fear discrimination than males in a CS+/CS- paradigm. To determine if this sex difference transferred to fear inhibition, rats were tested for conditioned inhibition in a summation test with the CS+ and CS- presented in compound; no sex difference emerged. The results suggest sex differences in the neural mechanisms of discrimination learning but not recall of a fear inhibitor.


Subject(s)
Conditioning, Psychological/physiology , Discrimination, Psychological/physiology , Fear/physiology , Inhibition, Psychological , Mental Recall/physiology , Sex Characteristics , Analysis of Variance , Animals , Cues , Electroshock , Female , Freezing Reaction, Cataleptic/physiology , Generalization, Psychological/physiology , Male , Psychological Tests , Rats, Sprague-Dawley
7.
Neurobiol Learn Mem ; 134 Pt B: 317-27, 2016 10.
Article in English | MEDLINE | ID: mdl-27523750

ABSTRACT

Veridical detection of safety versus danger is critical to survival. Learned signals for safety inhibit fear, and so when presented, reduce fear responses produced by danger signals. This phenomenon is termed conditioned inhibition of fear. Here, we report that CS+/CS- fear discrimination conditioning over 5 days in rats leads the CS- to become a conditioned inhibitor of fear, as measured by the classic tests of conditioned inhibition: summation and retardation of subsequent fear acquisition. We then show that NMDA-receptor antagonist AP5 injected to posterior insular cortex (IC) before training completely prevented the acquisition of a conditioned fear inhibitor, while intra-AP5 to anterior and medial IC had no effect. To determine if the IC contributes to the recall of learned fear inhibition, injections of the GABAA agonist muscimol were made to posterior IC before a summation test. This resulted in fear inhibition per se, which obscured inference to the effect of IC inactivation with recall of the safety cue. Control experiments sought to determine if the role of the IC in conditioned inhibition learning could be reduced to simpler fear discrimination function, but fear discrimination and recall were unaffected by AP5 or muscimol, respectively, in the posterior IC. These data implicate a role of posterior IC in the learning of conditioned fear inhibitors.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Conditioning, Classical/physiology , Fear/physiology , GABA-A Receptor Agonists/pharmacology , Inhibition, Psychological , Mental Recall/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Valine/analogs & derivatives , Animals , Conditioning, Classical/drug effects , Male , Mental Recall/drug effects , Muscimol/administration & dosage , Muscimol/pharmacology , Rats , Rats, Sprague-Dawley , Valine/administration & dosage , Valine/pharmacology
8.
Article in English | MEDLINE | ID: mdl-26344640

ABSTRACT

The capacity to discriminate between safety and danger is fundamental for survival, but is disrupted in individuals with posttraumatic stress disorder (PTSD). Acute stressors cause a release of serotonin (5-HT) in the forebrain, which is one mechanism for enhanced fear and anxiety; these effects are mediated by the 5-HT2Creceptor. Using a fear discrimination paradigm where a danger signal conditioned stimulus (CS+) co-terminates with a mild footshock and a safety signal (CS-) indicates the absence of shock, we demonstrate that danger/safety discrimination and fear inhibition develop over the course of 4 daily conditioning sessions. Systemic administration of the 5-HT2Creceptor antagonist SB 242084 (0.25 or 1.0mg/kg) prior to conditioning reduced behavioral freezing during conditioning, and improved learning and subsequent inhibition of fear by the safety signal. Discrimination was apparent in the first recall test, and discrimination during training was evident after 3days of conditioning versus 5days in the vehicle treated controls. These results suggest a novel therapeutic use for 5-HT2Creceptor antagonists to improve learning under stressful circumstances. Potential anatomical loci for 5-HT2Creceptor modulation of fear discrimination learning and cognitive performance enhancement are discussed. ETHICAL STATEMENT: John P. Christianson and Allison R. Foilb, the authors, verify that animal research was carried out in accordance with the National Institute of Health Guide for the Care and Use of Laboratory Animals (NIH Publications No. 80-23) and all procedures involving animals were reviewed and approved by the Boston College Animal Care and Use Committee. All efforts were made to limit the number of animals used and their suffering.


Subject(s)
Aminopyridines/pharmacology , Discrimination, Psychological/drug effects , Fear/drug effects , Indoles/pharmacology , Mental Recall/drug effects , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Discrimination, Psychological/physiology , Electroshock , Fear/physiology , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Inhibition, Psychological , Male , Mental Recall/physiology , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2C/metabolism
9.
Behav Brain Res ; 296: 249-253, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26386302

ABSTRACT

The ventral hippocampus (VH) is involved in the both the acquisition and recall of conditioned fear. Here, we tested the role of VH in acquisition and recall of a conditioned fear discrimination. Intra-VH vehicle or muscimol injections were made 1h prior to a CS+/CS- conditioning or prior to later recall. Vehicle treated rats exhibited discrimination with significantly greater freezing to the CS+ than to the CS- whereas muscimol treated rats did not freeze. Injections made before recall had no effect as both treatment groups displayed equal freezing in response to the CS+, and discrimination. While these results are consistent with several reports, the failure to influence fear discrimination upon recall appears to contrast with the hypothesized role of VH in recall of extinguished conditioned fear cues.


Subject(s)
Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Discrimination, Psychological/drug effects , Fear/drug effects , GABA-A Receptor Agonists/pharmacology , Hippocampus/physiology , Mental Recall/drug effects , Muscimol/pharmacology , Animals , Cues , GABA-A Receptor Agonists/administration & dosage , Male , Muscimol/administration & dosage , Rats , Rats, Sprague-Dawley
10.
Front Behav Neurosci ; 8: 348, 2014.
Article in English | MEDLINE | ID: mdl-25324750

ABSTRACT

Behaviorally controllable stressors confer protection from the neurochemical and behavioral consequences of future uncontrollable stressors, a phenomenon termed "behavioral immunization". Recent data implicate protein synthesis within the ventromedial prefrontal cortex (mPFC) as critical to behavioral immunization. Adult, male Sprague-Dawley rats were exposed to a series of controllable tailshocks and 1 week later to uncontrollable tailshocks, followed 24 h later by social exploration and shuttlebox escape tests. To test the involvement of N-methyl-D-aspartate receptors (NMDARs) and the extracellular signal-regulated kinase (ERK) cascade in behavioral immunization, either D-AP5 or the MEK inhibitor U0126 was injected to the prelimbic (PL) or infralimbic (IL) mPFC prior to controllable stress exposure. Phosphorylated ERK and P70S6K, regulators of transcription and translation, were quantified by Western blot or immunohistochemistry after controllable or uncontrollable tailshocks. Prior controllable stress prevented the social exploration and shuttlebox performance deficits caused by the later uncontrollable stressor, and this effect was blocked by injections of D-AP5 into mPFC. A significant increase in phosphorylated ERK1 and ERK2, but not P70S6K, occurred within the PL and IL in rats exposed to controllable stress, but not to uncontrollable stress. However, U0126 only prevented behavioral immunization when injected to the PL. We provide evidence that NMDAR and ERK dependent signaling within the PL region is required for behavioral immunization, a learned form of stressor resistance.

11.
Dev Neurosci ; 36(3-4): 261-8, 2014.
Article in English | MEDLINE | ID: mdl-24903860

ABSTRACT

Preadolescent animals display protracted hormonal stress responses mediated by the hypothalamic-pituitary-adrenal (HPA) axis compared to adults. Though the mechanisms that underlie this shift in stress reactivity are unknown, reduced glucocorticoid-dependent negative feedback on the HPA axis has been posited to contribute to this differential responsiveness. As the glucocorticoid receptors (GRs) are integral to this feedback response, we hypothesize that prior to puberty there will be fewer GRs in the neural-pituitary network that mediate negative feedback. To test this hypothesis we measured GR protein levels in the brains of preadolescent (28 days old), midadolescent (40 days old) and adult (77 days old) male rats via immunohistochemistry. Additionally, we assessed stress-induced plasma adrenocorticotropic hormone and corticosterone in prepubertal (30 days old) and adult (70 days old) male rats and examined GR protein levels via Western blot in the brain and pituitary. We found that despite substantial adolescent-related changes in hormonal responsiveness, no significant differences were found between these ages in GR protein levels in regions that are important in negative feedback, including the medial prefrontal cortex, paraventricular nucleus of the hypothalamus, hippocampal formation, and pituitary. These data indicate that the extended hormonal stress response exhibited by preadolescent animals is independent of significant pubertal changes in GR protein levels.


Subject(s)
Brain/growth & development , Brain/metabolism , Hormones/metabolism , Pituitary Gland/growth & development , Pituitary Gland/metabolism , Receptors, Glucocorticoid/metabolism , Stress, Psychological/metabolism , Adrenocorticotropic Hormone/metabolism , Aging/psychology , Animals , Corticosterone/metabolism , Hippocampus/growth & development , Hippocampus/metabolism , Male , Paraventricular Hypothalamic Nucleus/growth & development , Paraventricular Hypothalamic Nucleus/metabolism , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley
12.
Dev Neurobiol ; 72(5): 743-52, 2012 May.
Article in English | MEDLINE | ID: mdl-21990242

ABSTRACT

Pubertal development is marked by significant decreases in cellular proliferation and neurogenesis in the dentate gyrus of the hippocampal formation. Although it is unclear what mediates these developmental changes in the dentate gyrus, gonadal hormones have been implicated in modulating many neurobiological processes during puberty and various parameters of neurogenesis in adulthood. Thus, it is possible that the gradual and sustained increase in gonadal hormones experienced during puberty plays a role in these changes in neurogenesis. In this experiments, we first quantified cellular proliferation and neurogenesis using 5-bromo-2'-deoxyuridine (BrdU) and doublecortin (DCX) immunohistochemistry, respectively, in the dentate gyrus of prepubertal (30 d), midpubertal (45 d), and adult (90 d) male rats. We found the decline in BrdU and DCX cell numbers throughout these ages was coincident with increases in their plasma testosterone levels. We next tested whether exposure to the pubertal rise in gonadal hormones was necessary for this decrease in hippocampal neurogenesis to occur. Thus, we examined cellular proliferation and neurogenesis in intact 30 day (prepubertal) and 60-day-old (late-pubertal) rats, as well as 60-day-old rats that had previously been gonadectomized or sham-gonadectomized at 30 days of age. Although we again found the expected decline in BrdU and DCX cell numbers between 30 and 60 days of age in the intact groups, there were no differences among the 60-day-old animals, regardless of gonadal status. These data indicate that the pubertal-related decline in hippocampal cellular proliferation and neurogenesis is independent of the pubertal change in gonadal hormones.


Subject(s)
Cell Proliferation , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Neurogenesis/physiology , Sexual Maturation/physiology , Testosterone/metabolism , Animals , Doublecortin Protein , Male , Neurons/physiology , Rats , Rats, Sprague-Dawley , Stem Cells/physiology , Testis/growth & development , Testis/metabolism , Testosterone/blood
13.
J Endocrinol ; 210(3): 391-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21746793

ABSTRACT

Prepubertal rats display heightened hormonal stress reactivity compared with adults in that levels of ACTH and corticosterone take twice as long (i.e. 40-60 min) to return to baseline following an acute stressor. Despite this substantial change in stress responsiveness, and the critical nature of the adolescence period of development, the maturation of the hormonal stress response from the time of pubertal onset to adulthood has not been thoroughly investigated. To examine this, we measured ACTH, corticosterone, and testosterone in 30-, 40-, 50-, 60-, and 70-day-old (i.e. spanning pubertal and adolescent development) male rats before and after a 30 min session of restraint stress. We found that the adult-like ACTH stress response develops between 50 and 60 days of age, while the corticosterone response changes between 30 and 40 days of age. We also found that adrenal corticosterone concentrations paralleled the plasma corticosterone response following restraint, suggesting that stress-induced adrenal corticosterone synthesis decreases during adolescent development and may, at least in part, contribute to the differential stress response observed before and after puberty. Finally, stress leads to increases in testosterone secretion, but only after 50 days of age. Collectively, these results indicate that shifts in hormonal stress responses occur throughout adolescent maturation and that these responses show distinct developmental profiles.


Subject(s)
Neurosecretory Systems/physiology , Sexual Maturation/physiology , Stress, Physiological/physiology , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Age Factors , Animals , Corticosterone/blood , Corticosterone/metabolism , Male , Rats , Rats, Sprague-Dawley , Restraint, Physical/physiology , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...