Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neuroscience ; 468: 176-185, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34147563

ABSTRACT

Pharmacological and optogenetic studies have demonstrated that the basolateral amygdala (BLA) plays a pivotal role in regulating fear-conditioned changes in sleep, in particular, rapid eye movement sleep (REM). However, the linkage between BLA and REM regulation has been minimally examined. In this study, we optogenetically activated or inhibited BLA selectively during spontaneous REM, and determined the effects on REM amounts and on hippocampus regulated EEG-theta (θ) activity. Excitatory (CaMKIIα-hChR2 (E123A)-eYFP-WPRE) or inhibitory (CaMKIIα-eNpHR3.0-eYFP-WPRE) optogenetic constructs were stereotaxically delivered targeting glutamatergic cells in BLA (BLAGlu) of mice. Viral constructs without opsin (CaMKIIα-eYFP-WPRE) were used as controls. All mice were implanted with telemetry transmitters for monitoring electroencephalography (EEG), activity, and body temperature, and with optic cannulas for light delivery to the BLA. BLAGlu were optogenetically activated by blue light (473 nm), or inhibited by green light (532 nm), in 10 s epochs during REM, or non-REM (NREM), in undisturbed mice. Sleep amounts and EEG activity were analyzed. Projections from BLAGlu to neurons in hippocampus were immunohistochemically (IHC) examined. Brief optogenetic activation of BLAGlu during REM immediately reduced EEG theta activity (5-8 Hz, REM-θ), without affecting overall amount and propensity of sleep, while optogenetic inhibition increased REM-θ. Stimulation during NREM had no effect on EEG spectra or sleep. IHC results showed that glutamatergic and GABAergic cells in CA3 of the hippocampus received inputs from BLAGlu projection neurons. Activation of BLAGlu reduced, and inhibition increased, REM-θ without otherwise altering sleep. Optogenetic stimulation of BLAGlu may be useful for systematically manipulating sleep-related amygdalo-hippocampal interactions.


Subject(s)
Basolateral Nuclear Complex , Sleep, REM , Animals , Electroencephalography , Fear , Mice , Sleep
2.
Life (Basel) ; 12(1)2021 Dec 23.
Article in English | MEDLINE | ID: mdl-35054410

ABSTRACT

The basolateral amygdala (BLA) mediates the effects of stress and fear on rapid eye movement sleep (REM) and on REM-related theta (θ) oscillatory activity in the electroencephalograph (EEG), which is implicated in fear memory consolidation. We used optogenetics to assess the potential role of BLA glutamate neurons (BLAGlu) in regulating behavioral, stress and sleep indices of fear memory, and their relationship to altered θ. An excitatory optogenetic construct targeting glutamatergic cells (AAV-CaMKIIα-hChR2-eYFP) was injected into the BLA of mice. Telemetry was used for real-time monitoring of EEG, activity, and body temperature to determine sleep states and stress-induced hyperthermia (SIH). For 3 h following shock training (ST: 20 footshocks, 0.5 mA, 0.5 s, 1 min interval), BLA was optogenetically stimulated only during REM (REM + L) or NREM (NREM + L). Mice were then re-exposed to the fear context at 24 h, 48 h, and 1 week after ST and assessed for behavior, SIH, sleep and θ activity. Control mice were infected with a construct without ChR2 (eYFP) and studied under the same conditions. REM + L significantly reduced freezing and facilitated immediate recovery of REM tested at 24 h and 48 h post-ST during contextual re-exposures, whereas NREM + L had no significant effect. REM + L significantly reduced post-ST REM-θ, but attenuated REM-θ reductions at 24 h compared to those found in NREM + L and control mice. Fear-conditioned SIH persisted regardless of treatment. The results demonstrate that BLAGlu activity during post-ST REM mediates the integration of behavioral and sleep indices of fear memory by processes that are associated with θ oscillations within the amygdalo-hippocampal pathway. They also demonstrate that fear memories can remain stressful (as indicated by SIH) even when fear conditioned behavior (freezing) and changes in sleep are attenuated.

3.
Sleep ; 42(10)2019 10 09.
Article in English | MEDLINE | ID: mdl-31322681

ABSTRACT

STUDY OBJECTIVES: Sleep, in particular rapid eye movement (REM), has been linked to fear learning and extinction; however, their relationship is poorly understood. We determined how different delays of extinction training (ET) impact fear-conditioned behaviors, changes in sleep, and stress responses. METHODS: EEG activity, movement, and body temperature in mice were monitored via telemetry. Following contextual fear conditioning (shock training [ST]), separate groups of mice were reexposed to the context at 24-hour post-ST (24h ET-1) and at 48-hour post-ST (48h ET-1). Post-ET sleep amount and sleep-associated EEG (delta and theta) activity were compared to baseline and to post-ST sleep. Freezing, locomotion, grooming, and rearing were monitored to determine effects of ET on fear behaviors. Body temperature immediately after ET was monitored to assess stress-induced hyperthermia (SIH). RESULTS: 24h ET-1 and 48h ET-1 produced similar freezing and REM reductions, but dissimilar rearing activity and SIH. 24h ET-1 was followed by periods of suppressed REM-associated theta (REM-θ) activity, immediately after ET and during the subsequent dark period. Suppressed REM-θ was specific to sleep after 24h ET-1, and did not occur after ST, nor after 48h ET-1. CONCLUSIONS: ET-1 at 24 and 48 hours after ST was associated with similar freezing and REM amounts, but with differences in other overt behaviors, in REM-θ, and in SIH. Freezing was not predictive of changes in other fear-associated responses. This study demonstrated that consideration of time delay from fear acquisition to extinction is important when assessing the relationships between extinction and behavior, sleep, and stress responses.


Subject(s)
Extinction, Psychological/physiology , Fear/physiology , Freezing Reaction, Cataleptic/physiology , Locomotion/physiology , Sleep/physiology , Stress, Psychological/physiopathology , Animals , Electroencephalography/methods , Fear/psychology , Male , Mice , Mice, Inbred C57BL , Sleep, REM/physiology , Stress, Psychological/psychology , Time Factors
4.
J Neuroimmunol ; 319: 130-141, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29580714

ABSTRACT

Intranasal instillation of vesicular stomatitis virus (VSV) into mice given controllable stress (modeled by escapable foot shock, ES) resulted in enhanced pathogenicity and decreased survival relative to infected mice given uncontrollable stress (modeled by inescapable foot shock, IS) and non-shocked control mice. Survival likely reflected differential cytokine gene expression that may have been regulated by miR146a, a predicted stress-responsive upstream regulator. Controllability also enhanced the accumulation of brain T resident memory cells that persisted long after viral clearance. The unexpected facilitatory effect of ES on antiviral neuroimmune responses and pathogenicity may arise from differential immunoactivating and immunosuppressive effects of uncontrollable and controllable stress.


Subject(s)
Encephalitis, Viral/immunology , Helplessness, Learned , Stress, Psychological/immunology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Vesiculovirus
5.
Horm Behav ; 100: 20-28, 2018 04.
Article in English | MEDLINE | ID: mdl-29501756

ABSTRACT

The basolateral nucleus of the amygdala (BLA) plays a significant role in mediating individual differences in the effects of fear memory on sleep. Here, we assessed the effects of antagonizing corticotropin releasing factor receptor 1 (CRFR1) after shock training (ST) on fear-conditioned behaviors and sleep. Outbred Wistar rats were surgically implanted with electrodes for recording EEG and EMG and with bilateral guide cannulae directed at BLA. Data loggers were placed intraperitoneally to record core body temperature. The CRFR1 antagonist, antalarmin (ANT; 4.82 mM) was microinjected into BLA after shock training (ST: 20 footshocks, 0.8 mA, 0.5 s duration, 60 s interstimulus interval), and the effects on sleep, freezing and the stress response (stress-induced hyperthermia, SIH) were examined after ST and fearful context re-exposure alone at 7 days (CTX1) and 21 days (CTX2) post-ST. EEG and EMG recordings were scored for non-rapid eye movement sleep (NREM), rapid eye movement sleep (REM) and wakefulness. The rats were separated into 4 groups: Vehicle-vulnerable (Veh-Vul; n = 10), Veh-resilient (Veh-Res; n = 11), ANT-vulnerable (ANT-Vul; n = 8) and ANT-resilient (ANT-Res; n = 8) based on whether, compared to baseline, the rats showed a decrease or no change/increase in REM during the first 4 h following ST. Post-ST ANT microinjected into BLA attenuated the fear-conditioned reduction in REM in ANT-Vul rats on CTX1, but did not significantly alter REM in ANT-Res rats. However, compared to Veh treated rats, REM was reduced in ANT treated rats on CTX2. There were no group differences in freezing or SIH across conditions. Therefore, CRFR1 in BLA plays a role in mediating individual differences in sleep responses to stress and in the extinction of fear conditioned changes in sleep.


Subject(s)
Adaptation, Psychological/drug effects , Basolateral Nuclear Complex/drug effects , Body Temperature/drug effects , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Pyrimidines/pharmacology , Pyrroles/pharmacology , Sleep/drug effects , Animals , Basolateral Nuclear Complex/metabolism , Corticotropin-Releasing Hormone/antagonists & inhibitors , Electroencephalography , Fear/psychology , Male , Memory/drug effects , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/psychology , Rats , Rats, Wistar , Sleep/physiology , Sleep, REM/drug effects
6.
Neuroreport ; 29(2): 112-117, 2018 Jan 17.
Article in English | MEDLINE | ID: mdl-29135807

ABSTRACT

Controllability is an important factor in determining stress outcomes. Uncontrollable stress is associated with the development of psychopathology such as post-traumatic stress disorder, whereas controllable stress is associated with adaptive stress responses and positive outcomes. In this study, we investigated how controllability affects poststress neurobiology by assessing transcriptional levels of activity-dependent genes in medial prefrontal cortex (mPFC) and amygdala, regions important in mediating stress outcomes. Mice were subjected to either escapable shock (ES) or yoked inescapable shock (IS) as models of controllable and uncontrollable stress, respectively. Immediately (0 h) or at 2 h after shock training (20 trials; 0.5 mA, 5.0 s maximum duration; 1.0 min interstimulus interval), mice were killed, and we interrogated expression levels of the immediate-early genes, c-fos and Arc, and a delayed primary response gene, brain-derived neurotrophic factor, in mPFC, amygdala, and somatosensory cortex (a control region), using real-time reverse transcription quantitative PCR (RT qPCR). We found ES-associated up-regulation of brain-derived neurotrophic factor in amygdala as well as in mPFC. IS suppressed c-fos in mPFC (0 h) but induced more Arc in amygdala (2 h) in comparison with ES. Freezing, an index of fear memory, and serum level corticosterone, an index of the stress response, did not differ between mice trained with ES or IS. The data are discussed with respect to the potential functional involvements of the amygdala and mPFC in mediating differential outcomes of controllable and uncontrollable stress.


Subject(s)
Amygdala/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Stress, Psychological/metabolism , Animals , Corticosterone/blood , Electroshock , Freezing Reaction, Cataleptic , Gene Expression/physiology , Helplessness, Learned , Mice, Inbred BALB C , Random Allocation , Somatosensory Cortex/metabolism , Time Factors , Transcription, Genetic/physiology
7.
Sleep ; 40(4)2017 04 01.
Article in English | MEDLINE | ID: mdl-28199723

ABSTRACT

Study Objectives: Stressful events can directly produce significant alterations in subsequent sleep, in particular rapid eye movement sleep (REM); however, the neural mechanisms underlying the process are not fully known. Here, we investigated the role of the basolateral nuclei of the amygdala (BLA) in regulating the effects of stressful experience on sleep. Methods: We used optogenetics to briefly inhibit glutamatergic cells in BLA during the presentation of inescapable footshock (IS) and assessed effects on sleep, the acute stress response, and fear memory. c-Fos expression was also assessed in the amygdala and the medial prefrontal cortex (mPFC), both regions involved in coping with stress, and in brain stem regions implicated in the regulation of REM. Results: Compared to control mice, peri-shock inhibition of BLA attenuated an immediate reduction in REM after IS and produced a significant overall increase in REM. Moreover, upon exposure to the shock context alone, mice receiving peri-shock inhibition of BLA during training showed increased REM without altered freezing (an index of fear memory) or stress-induced hyperthermia (an index of acute stress response). Inhibition of BLA during REM under freely sleeping conditions enhanced REM only when body temperature was high, suggesting the effect was influenced by stress. Peri-shock inhibition of BLA also led to elevated c-Fos expression in the central nucleus of the amygdala and mPFC and differentially altered c-Fos activity in the selected brain stem regions. Conclusions: Glutamatergic cells in BLA can modulate the effects of stress on REM and can mediate effects of fear memory on sleep that can be independent of behavioral fear.


Subject(s)
Basolateral Nuclear Complex/physiology , Optogenetics , Sleep, REM/physiology , Stress, Psychological/physiopathology , Adaptation, Psychological/physiology , Animals , Basolateral Nuclear Complex/cytology , Electroshock , Fear/physiology , Freezing Reaction, Cataleptic , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Prefrontal Cortex/physiology , Proto-Oncogene Proteins c-fos/metabolism
8.
J Neuroimmunol ; 303: 31-42, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28041664

ABSTRACT

Rapid eye movement (REM) sleep is rapidly and persistently suppressed during vesicular stomatitis virus (VSV) encephalitis in C57Bl/6J (B6) mice. REM sleep suppression was associated with a complex global brain chemokine/cytokine response with bimodal kinetics although regionally distinct cytokine profiles were readily identified. Cytokine mRNA was translated either immediately or suppressed until the pathogen was cleared from the CNS. Innate signaling pathway (TLRs, RIG-I) activation occurred rapidly and sequentially prior to VSV neuroinvasion suggesting that antiviral states are quickly established in the CNS in advance of viral pathogen penetration. Il1ß suppressed REM sleep mimicking aspects of VSV-induced sleep alterations whereas some robustly induced chemokines may be protective of REM. Thus, multiple brain chemokines may mediate sleep across VSV encephalitis via differential somnogenic effects.


Subject(s)
Brain/immunology , Encephalitis, Viral/immunology , Inflammation Mediators/immunology , Sleep, REM/immunology , Transcriptional Activation/immunology , Vesicular stomatitis Indiana virus/immunology , Animals , Brain/metabolism , Brain/virology , Encephalitis, Viral/metabolism , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Sleep, REM/genetics , Transcriptional Activation/genetics
9.
Radiat Res ; 182(3): 292-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25029107

ABSTRACT

Previous ground-based experiments have shown that cranial irradiation with mission relevant (20 cGy) doses of 1 GeV/nucleon (56)Fe particles leads to a significant impairment in Attentional Set Shifting (ATSET) performance, a measure of executive function, in juvenile Wistar rats. However, the use of head only radiation exposure and the biological age of the rats used in that study may not be pertinent to determine the likelihood that ATSET will be impaired in Astronauts on deep space flights. In this study we have determined the impact that whole-body exposure to 10, 15 and 20 cGy of 1 GeV/nucleon (56)Fe particles had on the ability (at three months post exposure) of socially mature (retired breeder) Wistar rats to conduct the attentional set-shifting paradigm. The current study has established that whole-body exposures to 15 and 20 (but not 10) cGy of 1 GeV/nucleon (56)Fe particles results in the impairment of ATSET in both juvenile and socially mature rats. However, the exact nature of the impaired ATSET performance varied depending upon the age of the rats, whether whole-body versus cranial irradiation was used and the dose of 1 GeV/u (56)Fe received. Exposure of juvenile rats to 20 cGy of 1 GeV/nucleon (56)Fe particles led to a decreased ability to perform intra-dimensional shifting (IDS) irrespective of whether the rats received head only or whole-body exposures. Juvenile rats that received whole-body exposure also had a reduced ability to habituate to the assay and to complete intra-dimensional shifting reversal (IDR), whereas juvenile rats that received head only exposure had a reduced ability to complete compound discrimination reversal (CDR). Socially mature rats that received whole-body exposures to 10 cGy of 1 GeV/nucleon (56)Fe particles exhibited no obvious decline in set-shifting performance; however those exposed to 15 and 20 cGy had a reduced ability to perform simple discrimination (SD) and compound discrimination (CD). Exposure to 20 cGy of 1 GeV/nucleon (56)Fe particles also led to a decreased performance in IDR and to ∼25% of rats failing to habituate to the task. Most of these rats started to dig for the food reward but rapidly (within 15 s) gave up digging, suggesting that they had developed appropriate procedural memories about food retrieval, but had an inability to maintain attention on the task. Our preliminary data suggests that whole-body exposure to 20 cGy of 1 GeV/nucleon (56)Fe particles reduced the cholinergic (but not the GABAergic) readily releasable pool (RRP) in nerve terminals of the basal forebrain from socially-mature rats. This perturbation of the cholinergic RRP could directly lead to the loss of CDR and IDR performance, and indirectly [through the metabolic changes in the medial prefrontal cortex (mPFC)] to the loss of SD and CD performance. These findings provide the first evidence that attentional set-shifting performance in socially mature rats is impaired after whole-body exposure to mission relevant doses (15 and 20 cGy) of 1 GeV/nucleon (56)Fe particles, and importantly that a dose reduction down to 10 cGy prevents that impairment. The ability to conduct Discrimination tasks (SD and CD) and reversal learning (CDR) is reduced after exposure to 15 and 20 cGy of 1 GeV/nucleon (56)Fe particles, but at 20 cGy there is an additional decrement, ∼ 25% of rats are unable to maintain attention to task. These behavioral decrements are associated with a reduction in the cholinergic RRP within basal forebrain, which has been shown to play a major role in regulating the activity of the PFC.


Subject(s)
Attention/radiation effects , Cosmic Radiation , Executive Function/radiation effects , Animals , Dose-Response Relationship, Radiation , Male , Prosencephalon/radiation effects , Rats , Rats, Wistar , Synaptosomes/radiation effects , Whole-Body Irradiation
10.
Exp Brain Res ; 232(5): 1555-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24519098

ABSTRACT

Fear conditioning [inescapable shock training (ST)] and fearful context re-exposure (CR) alone can produce significant fear indicated by increased freezing and reductions in subsequent rapid eye movement (REM) sleep. Damage to or inactivation of the basolateral nucleus of the amygdala (BLA) prior to or after ST or prior to CR generally has been found to attenuate freezing in the shock training context. However, no one has examined the impact of BLA inactivation on fear-induced changes in sleep. Here, we used the GABAA agonist, muscimol (MUS), to inactivate BLA prior to ST, the period when fear is learned, and assessed sleep after ST and sleep and freezing after two CR sessions. Wistar rats (n = 14) were implanted with electrodes for recording sleep and with cannulae aimed bilaterally into BLA. After recovery, the animals were habituated to the injection procedure (handling) over 2 consecutive days and baseline sleep following handling was recorded. On experimental day 1, the rats were injected (0.5 µl) into BLA with either MUS (1.0 µM; n = 7) or vehicle (distilled water, n = 7) 30 min prior to ST (20 footshocks, 0.8 mA, 0.5-s duration, 60-s interstimulus interval). On experimental days 7 and 21, the animals experienced CR (CR1 and CR2, respectively) alone. Electroencephalogram and electromyogram were recorded for 8 h on each day, and the recording was scored for non-rapid eye movement sleep, REM sleep, and wakefulness. Freezing was examined during CR1 and CR2. MUS microinjections into BLA prior to ST blocked the post-training reduction in REM sleep seen in vehicle-treated rats. Furthermore, in MUS-treated rats, REM sleep after CR1 and CR2 was at baseline levels and freezing was significantly attenuated. Thus, BLA inactivation prior to ST blocks the effects of footshock stress on sleep and reduces fear memory, as indicated by the lack of freezing and changes in sleep after CR. These data indicate that BLA is an important regulator of stress-induced alterations in sleep and an important site for forming fear memories that can alter sleep.


Subject(s)
Basolateral Nuclear Complex/physiopathology , Fear/physiology , Memory/physiology , Sleep/physiology , Stress Disorders, Post-Traumatic/pathology , Analysis of Variance , Animals , Basolateral Nuclear Complex/drug effects , Conditioning, Classical/physiology , Disease Models, Animal , Electroencephalography , Electromyography , GABA-A Receptor Agonists/pharmacology , Memory/drug effects , Microinjections , Muscimol/pharmacology , Rats , Rats, Wistar , Sleep/drug effects
11.
Brain Behav Immun ; 35: 125-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24055862

ABSTRACT

Intranasal application of vesicular stomatitis virus (VSV) produces a well-characterized model of viral encephalitis in mice. Within one day post-infection (PI), VSV travels to the olfactory bulb and, over the course of 7 days, it infects regions and tracts extending into the brainstem followed by clearance and recovery in most mice by PI day 14 (PI 14). Infectious diseases are commonly accompanied by excessive sleepiness; thus, sleep is considered a component of the acute phase response to infection. In this project, we studied the relationship between sleep and VSV infection using C57BL/6 (B6) and BALB/c mice. Mice were implanted with transmitters for recording EEG, activity and temperature by telemetry. After uninterrupted baseline recordings were collected for 2 days, each animal was infected intranasally with a single low dose of VSV (5×10(4) PFU). Sleep was recorded for 15 consecutive days and analyzed on PI 0, 1, 3, 5, 7, 10, and 14. Compared to baseline, amounts of non-rapid eye movement sleep (NREM) were increased in B6 mice during the dark period of PI 1-5, whereas rapid eye movement sleep (REM) was significantly reduced during the light periods of PI 0-14. In contrast, BALB/c mice showed significantly fewer changes in NREM and REM. These data demonstrate sleep architecture is differentially altered in these mouse strains and suggests that, in B6 mice, VSV can alter sleep before virus progresses into brain regions that control sleep.


Subject(s)
Behavior, Animal/physiology , Encephalitis, Viral/psychology , Rhabdoviridae Infections/psychology , Sleep/physiology , Vesicular stomatitis Indiana virus , Animals , Electroencephalography , Encephalitis, Viral/physiopathology , Encephalitis, Viral/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rhabdoviridae Infections/physiopathology , Rhabdoviridae Infections/virology , Sleep, REM/physiology
12.
Sleep ; 36(4): 471-80, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23564994

ABSTRACT

STUDY OBJECTIVE: To determine whether corticotropin-releasing factor (CRF) in the basolateral amygdala (BLA) modulated sleep and fear-conditioned alterations in sleep. DESIGN: After 2 days of habituation to recording procedures, baseline sleep recordings were obtained. The animals were then habituated to the handling procedure necessary for microinjections over 2 consecutive days. In experiment 1, rats received microinjections of 0.5 µL antalarmin (1.61 or 4.82 mM), a CRF receptor 1 antagonist, or distilled water once a week for 3 wk. In experiment 2, rats received a microinjection of either antalarmin or vehicle prior to inescapable shock training (ST; 20 shocks; 0.8 mA, 0.5 sec; 1 min interstimulus interval). The animals were placed back in the context 7 days later for 30 min without shock (CR; context re-exposure). Sleep was recorded for 8 h after each manipulation. SETTING: NA. SUBJECTS: Outbred Wistar rats. INTERVENTIONS: The rats were surgically implanted with electrodes for recording the electroencephalogram and electromyogram for determining arousal state and with bilateral guide cannulae directed at BLA. MEASUREMENTS AND RESULTS: Antalarmin microinjected into BLA did not significantly alter sleep under undisturbed conditions. However, antalarmin microinjected bilaterally into BLA prior to ST blocked reductions in rapid eye movement sleep that ST normally produces. Further, the single microinjection prior to ST blocked the reduction in rapid eye movement typically seen after subsequent CR. Behavioral freezing, an indicator of fear memory, was not altered. CONCLUSIONS: CRF in BLA is involved in regulating stress-induced alterations in sleep and it plays a role in modulating how stressful memories influence sleep.


Subject(s)
Amygdala/metabolism , Conditioning, Psychological/physiology , Corticotropin-Releasing Hormone/metabolism , Fear/physiology , Sleep/physiology , Animals , Behavior, Animal , Electroencephalography/methods , Electromyography/methods , Electroshock/methods , Habituation, Psychophysiologic/physiology , Memory/physiology , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Rats , Rats, Wistar , Sleep, REM/physiology
13.
Sleep ; 36(3): 421-30, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23449731

ABSTRACT

STUDY OBJECTIVES: Controllable stress, modeled by escapable shock (ES), can produce significant alterations in post-stress sleep, including increased rapid eye movement (REM) sleep. Recent work has demonstrated that post-stress sleep may be influenced by stressor predictability, modeled by predictive auditory cues. In this study, we trained mice with ES, either signaled (SES) or unsignaled (UES) by auditory cues, and investigated the effects of predictability on escape learning and sleep associated with ES. DESIGN: Adult male BALB/cJ mice were implanted for recording electroencephalography and activity via telemetry. After the mice recovered from surgery, baseline sleep recordings were obtained. The mice were then randomly assigned to SES and UES conditions. Both groups had control over the duration of footshocks (0.5 mA; 5.0 sec maximum duration) by moving to the non-occupied chamber in a shuttlebox. SES mice were presented tones (90 dB, 2 kHz, 10 sec maximum duration) that started 5.0 sec prior to and co-terminated with footshocks. UES mice were presented identical tones that were not synchronized to shock presentation. ES training continued for 2 consecutive days (EST1 and EST2) with 20 footshock presentations (1 min inter-stimulus intervals). Seven days after EST2, the animals were re-exposed to the training chamber (context) alone for 30 min. MEASUREMENTS AND RESULTS: Escape latency was used to determine successful or unsuccessful escape learning. Sleep was scored for 20 h for baseline and on each treatment day. Freezing in the training context was scored as a behavioral index of fear. Nine of 14 SES mice successfully learned escape (SESl), and 5 failed to learn escape (SESf). Compared with baseline, SESl mice, but not SESf mice, showed significantly increased post-shock REM. All UES mice learned escape and showed enhanced post-shock REM. Freezing and sleep did not differ among groups on the context re-exposure day. CONCLUSIONS: The results indicate that information available in a stressful situation can affect an animal's ability to learn an appropriate response and post-stress sleep. CITATION: Machida M; Yang L; Wellman LL; Sanford LD. Effects of stressor predictability on escape learning and sleep in mice. SLEEP 2013;36(3):421-430.


Subject(s)
Avoidance Learning/physiology , Escape Reaction/physiology , Sleep Wake Disorders/etiology , Stress, Psychological/complications , Stress, Psychological/physiopathology , Acoustic Stimulation/methods , Animals , Behavior, Animal/physiology , Conditioning, Psychological/physiology , Cues , Electroencephalography/methods , Electroshock/methods , Male , Mice , Mice, Inbred BALB C , Sleep Wake Disorders/physiopathology , Sleep, REM , Telemetry/methods
14.
Biol Pharm Bull ; 34(1): 146-9, 2011.
Article in English | MEDLINE | ID: mdl-21212533

ABSTRACT

Glycogen synthase kinase-3ß (GSK-3ß) is a serine/threonine kinase that phosphorylate protein substrates involved in Alzheimer's disease (AD), such as microtubule-associated protein tau and amyloid precursor protein (APP). GSK-3ß consists of two splice variants; the major short form (GSK-3ß1) distributes in many organs and the minor long form (GSK-3ß2), whose structural difference is the insert of only 13 amino acid residues to the C-terminal side of the catalytic site of GSK-3ß1, is present in central nervous system. However, the physiological significances of the two variants are unclear. Here we examined whether the phosphorylation activities of two variants to tau and APP are different in cells. We found that GSK-3ß2 has lower phosphorylation activity to tau at AD-relevant epitope (Ser396) than GSK-3ß1 in cells, whereas the two variants exhibit equivalent levels of phosphorylation activities to APP. Recombinant GSK-3ß2 has also lower phosphorylation activity to tau than GSK-3ß1 in vitro, although the phosphorylation activities of the two variants to a synthetic peptide substrate pGS-2 are comparable. Furthermore, the deletion of the C-terminal tail (CT) of GSK-3ß2 resulted in considerable reduction of tau phosphorylation activity as compared with GSK-3ß1, suggesting that the lower phosphorylation activity of GSK-3ß2 to tau is attributed to weak interaction with tau through its unique higher-order structure of CT constructed by the 13 amino acids insertion. Such information may provide a clue for understanding of the physiological significance of the two splice variants of GSK-3ß and a new insight into the regulation of tau phosphorylation in central nervous system.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , tau Proteins/metabolism , HEK293 Cells , Humans , Phosphorylation , Protein Isoforms , Protein Structure, Tertiary
15.
Radiat Res ; 174(5): 618-23, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20726706

ABSTRACT

Exposure to galactic cosmic radiation (GCR) is considered to be a potential health risk in long-term space travel, and it represents a significant risk to the central nervous system (CNS). The most harmful component of GCR is the HZE [high-mass, highly charged (Z), high-energy] particles, e.g. (56)Fe. In ground-based experiments, exposure to HZE-particle radiation induces pronounced deficits in hippocampus-dependent learning and memory in rodents. The mechanisms underlying these impairments are mostly unknown, but some studies suggest that HZE-particle exposure perturbs the regulation of long-term potentiation (LTP) at the CA1 synapse in the hippocampus. In this study, we irradiated rats with 60 cGy of 1 GeV (56)Fe-particle radiation and established its impact on hippocampal glutamatergic neurotransmissions at 3 and 6 months after exposure. Exposure to 60 cGy (56)Fe-particle radiation significantly (P < 0.05) reduced hyperosmotic sucrose evoked [(3)H]-glutamate release from hippocampal synaptosomes, a measure of the readily releasable vesicular pool (RRP). This HZE-particle-induced reduction in the glutamatergic RRP persisted for at least 6 months after exposure. At 90 days postirradiation, there was a significant reduction in the expression of the NR1, NR2A and NR2B subunits of the glutamatergic NMDA receptor. The level of the NR2A protein remained suppressed at 180 days postirradiation, but the level of NR2B and NR1 proteins returned to or exceeded normal levels, respectively. Overall, this study shows that hippocampal glutamatergic transmission is sensitive to relative low doses of (56)Fe particles. Whether the observed HZE-particle-induced change in glutamate transmission, which plays a critical role in learning and memory, is the cause of HZE-particle-induced neurocognitive impairment requires further investigation.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/radiation effects , Iron/chemistry , Synaptosomes/metabolism , Synaptosomes/radiation effects , Animals , Dose-Response Relationship, Radiation , Hippocampus/metabolism , Male , Molecular Weight , Protein Subunits/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Vesicles/metabolism , Synaptic Vesicles/radiation effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...