Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 53
1.
PLoS One ; 16(12): e0260577, 2021.
Article En | MEDLINE | ID: mdl-34898621

Females are more affected by psychiatric illnesses including eating disorders, depression, and post-traumatic stress disorder than males. However, the neural mechanisms mediating these sex differences are poorly understood. Animal models can be useful in exploring such neural mechanisms. Conditioned taste aversion (CTA) is a behavioral task that assesses how animals process the competition between associated reinforcing and aversive stimuli in subsequent task performance, a process critical to healthy behavior in many domains. The purpose of the present study was to identify sex differences in this behavior and associated neural responses. We hypothesized that females would value the rewarding stimulus (Boost®) relative to the aversive stimulus (LiCl) more than males in performing CTA. We evaluated behavior (Boost® intake, LiCl-induced behaviors, ultrasonic vocalizations (USVs), CTA performance) and Fos activation in relevant brain regions after the acute stimuli [acute Boost® (AB), acute LiCl (AL)] and the context-only task control (COT), Boost® only task (BOT) and Boost®-LiCl task (BLT). Acutely, females drank more Boost® than males but showed similar aversive behaviors after LiCl. Females and males performed CTA similarly. Both sexes produced 55 kHz USVs anticipating BOT and inhibited these calls in the BLT. However, more females emitted both 22 kHz and 55 kHz USVs in the BLT than males: the latter correlated with less CTA. Estrous cycle stage also influenced 55 kHz USVs. Fos responses were similar in males and females after AB or AL. Females engaged the gustatory cortex and ventral tegmental area (VTA) more than males during the BOT and males engaged the amygdala more than females in both the BOT and BLT. Network analysis of correlated Fos responses across brain regions identified two unique networks characterizing the BOT and BLT, in both of which the VTA played a central role. In situ hybridization with RNAscope identified a population of D1-receptor expressing cells in the CeA that responded to Boost® and D2 receptor-expressing cells that responded to LiCl. The present study suggests that males and females differentially process the affective valence of a stimulus to produce the same goal-directed behavior.


Conditioning, Psychological , Proto-Oncogene Proteins c-fos/metabolism , Acoustic Stimulation , Amygdala/drug effects , Amygdala/metabolism , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/radiation effects , Female , Lithium Chloride/pharmacology , Male , Rats , Sex Characteristics , Ultrasonics
2.
Proc Natl Acad Sci U S A ; 116(17): 8576-8581, 2019 04 23.
Article En | MEDLINE | ID: mdl-30877252

The neural circuits underlying memory change over prolonged periods after learning, in a process known as systems consolidation. Postlearning spontaneous reactivation of memory-related neural ensembles is thought to mediate this process, although a causal link has not been established. Here we test this hypothesis in mice by using optogenetics to selectively reactivate neural ensembles representing a contextual fear memory (sometimes referred to as engram neurons). High-frequency stimulation of these ensembles in the retrosplenial cortex 1 day after learning produced a recent memory with features normally observed in consolidated remote memories, including higher engagement of neocortical areas during retrieval, contextual generalization, and decreased hippocampal dependence. Moreover, this effect was only present if memory ensembles were reactivated during sleep or light anesthesia. These results provide direct support for postlearning memory ensemble reactivation as a mechanism of systems consolidation, and show that this process can be accelerated by ensemble reactivation in an unconscious state.


Cerebral Cortex/physiology , Conditioning, Psychological/physiology , Fear/physiology , Memory Consolidation/physiology , Optogenetics/methods , Animals , Cerebral Cortex/radiation effects , Conditioning, Psychological/radiation effects , Fear/radiation effects , Female , Male , Memory Consolidation/radiation effects , Mice , Mice, Transgenic
3.
Environ Sci Pollut Res Int ; 25(11): 10894-10903, 2018 Apr.
Article En | MEDLINE | ID: mdl-29397508

Exposure of pregnant women to radiofrequency (RF) devices raises questions on their possible health consequences for their progeny. We examined the hazard threshold of gestational RF on the progeny's glial homeostasis, sensory-motor gating, emotionality, and novelty seeking and tested whether maternal immune activation would increase RF toxicity. Pregnant dams were daily restrained with loop antennas adjoining the abdomen (fetus body specific absorption rates (SAR): 0, 0.7, or 2.6 W/kg) and received three lipopolysaccharide (LPS) intra-peritoneal injections (0 or 80 µg/kg). Scores in the prepulse startle inhibition, fear conditioning, open field, and elevated plus maze were assessed at adolescence and adulthood. Glial fibrillary acidic protein (GFAP) and interleukines-1ß (ILs) were quantified. LPS induced a SAR-dependent reduction of the prepulse startle inhibition in adults. Activity in the open field was reduced at 2.6 W/kg at adolescence. GFAP and ILs, emotional memory, and anxiety-related behaviors were not modified. These data support the hypothesis that maternal immune activation increased the developmental RF exposure-induced long-term neurobiological impairments. These data support the fact that fetuses who receive combined environmental exposures with RF need special attention for protection.


Cell Phone , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/epidemiology , Radiation Exposure/adverse effects , Radio Waves/adverse effects , Animals , Cerebrum/radiation effects , Conditioning, Psychological/radiation effects , Exploratory Behavior/radiation effects , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Maze Learning/radiation effects , Mice , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Rats, Sprague-Dawley , Reflex, Startle/radiation effects
4.
Radiat Res ; 189(4): 345-353, 2018 04.
Article En | MEDLINE | ID: mdl-29351056

Clinical management of primary and secondary central nervous system (CNS) malignancies frequently includes radiotherapy to forestall tumor growth and recurrence after surgical resection. While cranial radiotherapy remains beneficial, adult and pediatric brain tumor survivors suffer from a wide range of debilitating and progressive cognitive deficits. Although this has been recognized as a significant problem for decades, there remains no clinical recourse for the unintended neurocognitive sequelae associated with these types of cancer treatments. In previous work, multiple mechanisms have been identified that contribute to radiation-induced cognitive dysfunction, including the inhibition of neurogenesis caused by the depletion of radiosensitive populations of stem and progenitor cells in the hippocampus. To explore the potential neuroprotective properties of a pro-neurogenic compound NSI-189, Long-Evans rats were subjected to a clinically relevant fractionated irradiation protocol followed by four weeks of NSI-189 administered daily by oral gavage. Animals were then subjected to five different behavioral tasks followed by an analysis of neurogenesis, hippocampal volume and neuroinflammation. Irradiated cohorts manifested significant behavioral decrements on all four spontaneous exploration tasks. Importantly, NSI-189 treatment resulted in significantly improved performance in four of these tasks: novel place recognition, novel object recognition, object in place and temporal order. In addition, there was a trend of improved performance in the contextual phase of the fear conditioning task. Importantly, enhanced cognition in the NSI-189-treated cohort was found to persist one month after the cessation of drug treatment. These neurocognitive benefits of NSI-189 coincided with a significant increase in neurogenesis and a significant decrease in the numbers of activated microglia compared to the irradiated cohort that was given vehicle alone. The foregoing changes were not accompanied by major changes in hippocampal volume. These data demonstrate that oral administration of a pro-neurogenic compound exhibiting anti-inflammatory indications could impart long-term neurocognitive benefits in the irradiated brain.


Aminopyridines/administration & dosage , Aminopyridines/pharmacology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Piperazines/administration & dosage , Piperazines/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/etiology , Administration, Oral , Animals , Cognition/drug effects , Cognition/radiation effects , Cognitive Dysfunction/pathology , Cognitive Dysfunction/physiopathology , Conditioning, Psychological/drug effects , Conditioning, Psychological/radiation effects , Cranial Irradiation/adverse effects , Fear/psychology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Hippocampus/radiation effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Organ Size/drug effects , Organ Size/radiation effects , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Rats , Recognition, Psychology/drug effects , Recognition, Psychology/radiation effects
5.
Radiat Res ; 188(5): 532-551, 2017 11.
Article En | MEDLINE | ID: mdl-28945526

Astronauts traveling to Mars will be exposed to chronic low doses of galactic cosmic space radiation, which contains highly charged, high-energy (HZE) particles. 56Fe-HZE-particle exposure decreases hippocampal dentate gyrus (DG) neurogenesis and disrupts hippocampal function in young adult rodents, raising the possibility of impaired astronaut cognition and risk of mission failure. However, far less is known about how exposure to other HZE particles, such as 28Si, influences hippocampal neurogenesis and function. To compare the influence of 28Si exposure on indices of neurogenesis and hippocampal function with previous studies on 56Fe exposure, 9-week-old C57BL/6J and Nestin-GFP mice (NGFP; made and maintained for 10 or more generations on a C57BL/6J background) received whole-body 28Si-particle-radiation exposure (0, 0.2 and 1 Gy, 300 MeV/n, LET 67 KeV/µ, dose rate 1 Gy/min). For neurogenesis assessment, the NGFP mice were injected with the mitotic marker BrdU at 22 h postirradiation and brains were examined for indices of hippocampal proliferation and neurogenesis, including Ki67+, BrdU+, BrdU+NeuN+ and DCX+ cell numbers at short- and long-term time points (24 h and 3 months postirradiation, respectively). In the short-term group, stereology revealed fewer Ki67+, BrdU+ and DCX+ cells in 1-Gy-irradiated group relative to nonirradiated control mice, fewer Ki67+ and DCX+ cells in 0.2 Gy group relative to control group and fewer BrdU+ and DCX+ cells in 1 Gy group relative to 0.2 Gy group. In contrast to the clearly observed radiation-induced, dose-dependent reductions in the short-term group across all markers, only a few neurogenesis indices were changed in the long-term irradiated groups. Notably, there were fewer surviving BrdU+ cells in the 1 Gy group relative to 0- and 0.2-Gy-irradiated mice in the long-term group. When the short- and long-term groups were analyzed by sex, exposure to radiation had a similar effect on neurogenesis indices in male and female mice, although only male mice showed fewer surviving BrdU+ cells in the long-term group. Fluorescent immunolabeling and confocal phenotypic analysis revealed that most surviving BrdU+ cells in the long-term group expressed the neuronal marker NeuN, definitively confirming that exposure to 1 Gy 28Si radiation decreased the number of surviving adult-generated neurons in male mice relative to both 0- and 0.2-Gy-irradiated mice. For hippocampal function assessment, 9-week-old male C57BL/6J mice received whole-body 28Si-particle exposure and were then assessed long-term for performance on contextual and cued fear conditioning. In the context test the animals that received 0.2 Gy froze less relative to control animals, suggesting decreased hippocampal-dependent function. However, in the cued fear conditioning test, animals that received 1 Gy froze more during the pretone portion of the test, relative to controls and 0.2-Gy-irradiated mice, suggesting enhanced anxiety. Compared to previously reported studies, these data suggest that 28Si-radiation exposure damages neurogenesis, but to a lesser extent than 56Fe radiation and that low-dose 28Si exposure induces abnormalities in hippocampal function, disrupting fear memory but also inducing anxiety-like behavior. Furthermore, exposure to 28Si radiation decreased new neuron survival in long-term male groups but not females suggests that sex may be an important factor when performing brain health risk assessment for astronauts traveling in space.


Conditioning, Psychological/radiation effects , Dentate Gyrus/cytology , Fear/psychology , Neurogenesis/radiation effects , Neurons/cytology , Silicon , Whole-Body Irradiation/adverse effects , Animals , Behavior, Animal/physiology , Behavior, Animal/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Cosmic Radiation , Dentate Gyrus/physiology , Dentate Gyrus/radiation effects , Dose-Response Relationship, Radiation , Doublecortin Protein , Fear/radiation effects , Female , Memory/physiology , Memory/radiation effects , Mice , Neurons/radiation effects , Time Factors
6.
Physiol Behav ; 175: 37-46, 2017 06 01.
Article En | MEDLINE | ID: mdl-28341234

The effect of acute irradiation with 5Gy or fractionated exposure with 0.5Gy continuously for 10days (a total dose of 5Gy) was evaluated in an immature BALB/c mouse model. Radioprotective effect of ursolic acid (at 25mg/kg/daily administered 1h after acute or each of fractionated irradiations, and continuously for 30days) was also investigated. We found that both acute and fractionated irradiation at a total dose of 5Gy did not induce any mortality within 30days after exposure to postnatal day 26 (P26) BALB/c mice, but reduced animal weigh gain in the first few weeks. At 90days after irradiation, the weight of animals with acute irradiation was still significantly lower than the control group; no significant difference though was observed for those fractionatedly exposed mice compared to the control group. Behavioral tests indicated that acute irradiation at 5Gy induced deficits in learning and memory in the contextual fear conditioning test. The memory for novel object recognition was also impaired. Similar changes were not observed in mice with fractionated irradiation. Immunohistochemical study demonstrated clearly that acute and fractionated irradiations induced impairment of neurogenesis in the subgranular zone (SGZ) of the dentate gyrus although fractionated exposure induced much lesser loss of newly generated neurons. Ursolic acid administered at 25mg/kg/daily for 30days after irradiation greatly improved acute irradiation-induced deficits in contextual learning and memory and in novel object recognition memory although it exacerbated radiation-induced reduction of neurogenesis in SGZ.


Cyclooxygenase Inhibitors/therapeutic use , Learning Disabilities/drug therapy , Learning Disabilities/etiology , Neurogenesis/drug effects , Radiation Injuries, Experimental/complications , Triterpenes/therapeutic use , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Conditioning, Psychological/drug effects , Conditioning, Psychological/radiation effects , Disease Models, Animal , Doublecortin Domain Proteins , Fear/drug effects , Fear/radiation effects , Hindlimb Suspension , Hippocampus/drug effects , Hippocampus/radiation effects , Ki-67 Antigen/metabolism , Locomotion/drug effects , Locomotion/radiation effects , Mice , Mice, Inbred BALB C , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Radiation Injuries, Experimental/drug therapy , Recognition, Psychology/radiation effects , Swimming/psychology , Ursolic Acid
7.
Behav Brain Res ; 319: 181-187, 2017 02 15.
Article En | MEDLINE | ID: mdl-27865918

Typically, in studies designed to assess effects of irradiation on cognitive performance the animals are trained and tested for cognitive function following irradiation. Little is known about post-training effects of irradiation on cognitive performance. In the current study, 3-month-old male mice were irradiated with X-rays 24h following training in a fear conditioning paradigm and cognitively tested starting two weeks later. Average motion during the extinction trials, measures of anxiety in the elevated zero maze, and body weight changes over the course of the study were assessed as well. Exposure to whole body irradiation 24h following training in a fear conditioning resulted in greater freezing levels 2 weeks after training. In addition, motion during both contextual and cued extinction trials was lower in irradiated than sham-irradiated mice. In mice trained for cued fear conditioning, activity levels in the elevated zero maze 12days after sham-irradiation or irradiation were also lower in irradiated than sham-irradiated mice. Finally, the trajectory of body weight changes was affected by irradiation, with lower body weights in irradiated than sham-irradiated mice, with the most profound effect 7days after training. These effects were associated with reduced c-Myc protein levels in the amygdala of the irradiated mice. These data indicate that whole body X ray irradiation of mice at 3 months of age causes persistent alterations in the fear response and activity levels in a novel environment, while the effects on body weight seem more transient.


Conditioning, Psychological/radiation effects , Cues , Fear/radiation effects , Memory/radiation effects , Whole-Body Irradiation , Analysis of Variance , Animals , Body Weight/radiation effects , Brain/radiation effects , Electroshock/adverse effects , Freezing Reaction, Cataleptic/radiation effects , Male , Maze Learning/radiation effects , Mice , Mice, Inbred C57BL , Motor Activity/radiation effects
8.
Life Sci Space Res (Amst) ; 9: 56-61, 2016 Jun.
Article En | MEDLINE | ID: mdl-27345201

The space radiation environment includes energetic charged particles that may impact behavioral and cognitive performance. The relationship between the dose and the ionization density of the various types of charged particles (expressed as linear energy transfer or LET), and cognitive performance is complex. In our earlier work, whole body exposure to (28)Si ions (263 MeV/n, LET=78keV/µm; 1.6 Gy) affected contextual fear memory in C57BL/6J × DBA2/J F1 (B6D2F1) mice three months following irradiation but this was not the case following exposure to (48)Ti ions (1 GeV/n, LET=107keV/µm; 0.2 or 0.4 Gy). As an increased understanding of the impact of charged particle exposures is critical for assessment of risk to the CNS of astronauts during and following missions, in this study we used (40)Ca ion beams (942 MeV/n, LET=90keV/µm) to determine the behavioral and cognitive effects for the LET region between that of Si ions and Ti ions. (40)Ca ion exposure reduced baseline activity in a novel environment in a dose-dependent manner, which suggests reduced motivation to explore and/or a diminished level of curiosity in a novel environment. In addition, exposure to (40)Ca ions had sex-dependent effects on response to shock. (40)Ca ion irradiation reduced the response to shock in female, but not male, mice. In contrast, (40)Ca ion irradiation did not affect fear learning, memory, or extinction of fear memory for either gender at the doses employed in this study. Thus (40)Ca ion irradiation affected behavioral, but not cognitive, performance. The effects of (40)Ca ion irradiation on behavioral performance are relevant, as a combination of novelty and aversive environmental stimuli is pertinent to conditions experienced by astronauts during and following space missions.


Behavior, Animal/physiology , Calcium Radioisotopes/adverse effects , Conditioning, Psychological/radiation effects , Fear/psychology , Memory/physiology , Animals , Behavior, Animal/radiation effects , Dose-Response Relationship, Radiation , Fear/radiation effects , Female , Male , Memory/radiation effects , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Radiation, Ionizing , Sex Factors
9.
Radiat Res ; 185(4): 423-30, 2016 04.
Article En | MEDLINE | ID: mdl-27023259

Cranial X irradiation can severely impair higher brain function, resulting in neurocognitive deficits. Radiation-induced brain injury is characterized by acute, early and late delayed changes, and morbidity is evident more than 6 months after irradiation. While the acute effects of radiation exposure on the brain are known, the underlying mechanisms remain unclear. In this study, we examined the acute effect of X radiation on synaptic function using behavioral analysis and immunohistochemistry. We found that 10 Gy whole-brain irradiation immediately after conditioning (within 30 min) impaired the formation of fear memory, whereas irradiation 24 h prior to conditioning did not. To investigate the mechanisms underlying these behavioral changes, we irradiated one hemisphere of the brain and analyzed synaptic function and adult neurogenesis immunohistochemically. We focused on drebrin, whose loss from dendritic spines is a surrogate marker of synaptopathy. The intensity of drebrin immunoreactivity started to decrease in the irradiated hemisphere 2 h after exposure. The immunostaining intensity recovered to preirradiation levels by 24 h, indicating that X radiation induced transient synaptic dysfunction. Interestingly, the number of newly generated neurons was not changed at 2 h postirradiation, whereas it was significantly decreased at 8 and 24 h postirradiation. Because irradiation 24 h prior to conditioning had no effect on fear memory, our findings suggest that radiation-induced death of newly-generated neurons does not substantially impact fear memory formation. The radiation-induced synaptic dysfunction likely caused a transient memory deficit during the critical period for fear memory formation (approximately 1-3 h after conditioning), which coincides with a change in drebrin immunostaining in the hippocampus, a structure critical for fear memory formation.


Cognitive Dysfunction/physiopathology , Synapses/physiology , Synapses/radiation effects , Animals , Behavior, Animal/radiation effects , Brain/pathology , Brain/physiopathology , Brain/radiation effects , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Conditioning, Psychological/radiation effects , Doublecortin Domain Proteins , Fear/physiology , Male , Memory/radiation effects , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Neurons/radiation effects , Neuropeptides/metabolism , Time Factors , X-Rays/adverse effects
10.
Neurotoxicology ; 53: 290-300, 2016 Mar.
Article En | MEDLINE | ID: mdl-26945731

Although numerous studies have reported the influence of extremely low frequency magnetic field (ELF-MF) exposure on human health, its effects on cognitive deficits in Alzheimer's disease (AD) have remained under debate. Moreover, the influence of ELF-MF on hyperphosphorylated tau, which is one of the most common pathological hallmarks of AD, has not been reported to date. Therefore, transgenic mice (3xTg) were used in the present study. 3xTg mice, which express an APP/PS1 mutation combined with a tau (P301L) mutation and that develop cognitive deficits at 6 months of age, were subjected to ELF-MF (50Hz, 500µT) exposure or sham exposure daily for 3 months. We discovered that ELF-MF exposure ameliorated cognitive deficits and increased synaptic proteins in 3xTg mice. The protective effects of ELF-MF exposure may have also been caused by the inhibition of apoptosis and/or decreased oxidative stress levels that were observed in the hippocampus tissues of treated mice. Furthermore, tau hyperphosphorylation was decreased in vivo because of ELF-MF exposure, and this decrease was induced by the inhibition of GSK3ß and CDK5 activities and activation of PP2Ac. We are the first to report that exposure to ELF-MF can attenuate tau phosphorylation. These findings suggest that ELF-MF exposure could act as a valid therapeutic strategy for ameliorating cognitive deficits and attenuating tau hyperphosphorylation in AD.


Alzheimer Disease/complications , Cognition Disorders/etiology , Cognition Disorders/therapy , Magnetic Field Therapy/methods , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/radiotherapy , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Conditioning, Psychological/physiology , Conditioning, Psychological/radiation effects , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Dose-Response Relationship, Radiation , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Maze Learning/physiology , Maze Learning/radiation effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Phosphorylation/radiation effects , Presenilin-1/genetics , Reactive Oxygen Species/metabolism , tau Proteins/genetics
11.
Behav Brain Res ; 298(Pt B): 1-11, 2016 Feb 01.
Article En | MEDLINE | ID: mdl-26522840

The brain might be exposed to irradiation under a variety of situations, including clinical treatments, nuclear accidents, dirty bomb scenarios, and military and space missions. Correctly recalling tasks learned prior to irradiation is important but little is known about post-learning effects of irradiation. It is not clear whether exposure to X-ray irradiation during memory consolidation, a few hours following training, is associated with altered contextual fear conditioning 24h after irradiation and which brain region(s) might be involved in these effects. Brain immunoreactivity patterns of the immediately early gene c-Fos, a marker of cellular activity was used to determine which brain areas might be altered in post-training irradiation memory retention tasks. In this study, we show that post-training gamma irradiation exposure (1 Gy) enhanced contextual fear memory 24h later and is associated with reduced cellular activation in the infralimbic cortex. Reduced GABA-ergic neurotransmission in parvalbumin-positive cells in the infralimbic cortex might play a role in this post-training radiation-enhanced contextual fear memory.


Cerebral Cortex/radiation effects , Fear/radiation effects , Gamma Rays/adverse effects , Memory/radiation effects , Neurons/radiation effects , Animals , Cerebral Cortex/physiopathology , Conditioning, Psychological/physiology , Conditioning, Psychological/radiation effects , Fear/physiology , Immunohistochemistry , Male , Memory/physiology , Mice, Inbred C57BL , Neurons/physiology , Parvalbumins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Random Allocation , gamma-Aminobutyric Acid/metabolism
12.
Hippocampus ; 25(3): 385-92, 2015 Mar.
Article En | MEDLINE | ID: mdl-25330953

Adult animals continue to modify their behavior throughout life, a process that is highly influenced by past experiences. To shape behavior, specific mechanisms of neural plasticity to learn, remember, and recall information are required. One of the most robust examples of adult plasticity in the brain occurs in the dentate gyrus (DG) of the hippocampus, through the process of adult neurogenesis. Adult neurogenesis is strongly upregulated by external factors such as voluntary wheel running (RUN) and environmental enrichment (EE); however, the functional differences between these two factors remain unclear. Although both manipulations result in increased neurogenesis, RUN dramatically increases the proliferation of newborn cells and EE promotes their survival. We hypothesize that the method by which these newborn neurons are induced influences their functional role. Furthermore, we examine how EE-induced neurons may be primed to encode and recognize features of novel environments due to their previous enrichment experience. Here, we gave mice a challenging contextual fear-conditioning (FC) procedure to tease out the behavioral differences between RUN-induced neurogenesis and EE-induced neurogenesis. Despite the robust increases in neurogenesis seen in the RUN mice, we found that only EE mice were able to discriminate between similar contexts in this task, indicating that EE mice might use a different cognitive strategy when processing contextual information. Furthermore, we showed that this improvement was dependent on EE-induced neurogenesis, suggesting a fundamental functional difference between RUN-induced neurogenesis and EE-induced neurogenesis.


Discrimination, Psychological/physiology , Environment , Learning Disabilities/etiology , Learning Disabilities/rehabilitation , Shock/complications , Animals , Conditioning, Psychological/physiology , Conditioning, Psychological/radiation effects , Cranial Irradiation , Discrimination, Psychological/drug effects , Fear/physiology , Female , Hippocampus/pathology , Hippocampus/radiation effects , Learning Disabilities/pathology , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Motor Activity/radiation effects , Neurogenesis , Neurons/metabolism
13.
Behav Brain Res ; 246: 162-7, 2013 Jun 01.
Article En | MEDLINE | ID: mdl-23454674

The space radiation environment contains high-energy charged particles such as (56)Fe, which could pose a significant hazard to hippocampal function in astronauts during and after the mission(s). The mechanisms underlying impairments in cognition are not clear but might involve alterations in the percentage of neurons in the dentate gyrus expressing the plasticity-related immediate early gene Arc. Previously, we showed effects of cranial (56)Fe irradiation on hippocampus-dependent contextual freezing and on the percentage of Arc-positive cells in the enclosed, but not free, blade. Because it is unclear whether whole body (56)Fe irradiation causes similar effects on these markers of hippocampal function, in the present study we quantified the effects of whole body (56)Fe irradiation (600MeV, 0.5 or 1Gy) on hippocampus-dependent and hippocampus-independent cognitive performance and determined whether these effects were associated with changes in Arc expression in the enclosed and free blades of the dentate gyrus. Whole body (56)Fe irradiation impacted contextual but not cued fear freezing and the percentage of Arc-positive cells in the enclosed and free blades. In mice tested for contextual freezing, there was a correlation between Arc-positive cells in the enclosed and free blades. In addition, in mice irradiated with 0.5Gy, contextual freezing in the absence of aversive stimuli correlated with the percentage of Arc-positive cells in the enclosed blade. In mice tested for cued freezing, there was no correlation between Arc-positive cells in the enclosed and free blades. In contrast, cued freezing in the presence or absence of aversive stimuli correlated with Arc-positive cells in the free blade. In addition, in mice irradiated with 1Gy cued freezing in the absence of aversive stimuli correlated with the percentage of Arc-positive neurons in the free blade. These data indicate that while whole body (56)Fe radiation affects contextual freezing and Arc-positive cells in the dentate gyrus, the enclosed blade might be more important for contextual freezing while the free blade might be more important for cued freezing.


Conditioning, Psychological/radiation effects , Cytoskeletal Proteins/metabolism , Dentate Gyrus/cytology , Fear/radiation effects , Nerve Tissue Proteins/metabolism , Neurons/radiation effects , Whole-Body Irradiation , Analysis of Variance , Animals , Dentate Gyrus/radiation effects , Dose-Response Relationship, Radiation , Electromagnetic Radiation , Electroshock/adverse effects , Freezing Reaction, Cataleptic/radiation effects , Iron , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neuropsychological Tests
14.
Radiat Res ; 179(4): 493-500, 2013 Apr.
Article En | MEDLINE | ID: mdl-23496055

Apolipoprotein E (ApoE) plays an important role in lipid metabolism and neuronal repair. In humans, there are three major apoE isoforms: apoE2, apoE3 and apoE4. Compared to apoE3, apoE4 increases the risk to develop Alzheimer's disease, particularly in women, and of developing cognitive impairments after specific environmental challenges. ApoE isoform might also be a determinant of cognitive injury after cranial 56Fe irradiation. To assess this possibility, in this study female apoE2, apoE3 and apoE4 mice were cranially irradiated with 56Fe particles (600 MeV, 0, 1 or 2 Gy) and behaviorally tested 3 months later. Exploratory activity and measures of anxiety were also assessed as they can affect performance on cognitive tests. There were no effects of irradiation on exploratory activity or measures of anxiety in the open field or elevated zero maze. However, there were dose- and apoE isoform-dependent effects of irradiation on novel object recognition and spatial memory retention in the water maze. Compared to apoE2 and apoE3 mice, apoE4 mice were more sensitive to 2 Gy induced impairments in hippocampus-dependent spatial memory retention in the water maze after training to locate the first hidden platform location, but less sensitive to 2 Gy induced cortical impairments in novel object recognition. Conversely, of the irradiated mice, apoE4 mice irradiated with 1 Gy were the only group of mice that showed spatial memory retention for the second platform location after reversal learning in the water maze. Together, these data show that cranial 56Fe irradiation causes dose- and apoE isoform-dependent cognitive impairments in female mice and that anatomical specificity might contribute to the relative sensitivity of apoE4 mice to develop space radiation-induced cognitive impairments.


Apolipoproteins E/physiology , Cognition/radiation effects , Cranial Irradiation , Animals , Conditioning, Psychological/radiation effects , Dose-Response Relationship, Radiation , Female , Iron Isotopes , Maze Learning/radiation effects , Mice , Mice, Inbred C57BL , Protein Isoforms
15.
Radiat Res ; 179(5): 590-6, 2013 May.
Article En | MEDLINE | ID: mdl-23510274

Relatively little is known about early irradiation effects on hippocampal function in wild-type mice. In this study, the effects of (56)Fe irradiation on hippocampal function were assessed starting 2 weeks after whole-body irradiation. Compared to sham irradiation, radiation impaired novel object recognition in female and male C57BL/6J wild-type mice. There were no effects of irradiation on contextual fear conditioning or spatial memory retention in the water maze. It is possible that oxidative damage might contribute to radiation-induced cognitive changes. Therefore, hippocampal and cortical levels of 3-nitrotyrosine (3NT) and lipid peroxidation, measures of oxidative damage were assessed. There were no effects of irradiation on these measures of oxidative damage. As (56)Fe irradiation can increase reactive oxygen species (ROS) levels, which may contribute to the impairments in novel object recognition, the effects of the antioxidant alpha-lipoic acid (ALA) on cognition following sham irradiation and irradiation were also assessed. ALA did not prevent radiation-induced impairments in novel object recognition and impaired spatial memory retention of sham-irradiated and irradiated mice in the probe trial after the first day of hidden platform training in the water maze. Thus, the novel object recognition test is particularly sensitive to detect early cognitive effects of (56)Fe irradiation through a mechanism unlikely involving ROS or oxidative damage.


Hippocampus/physiology , Hippocampus/radiation effects , Iron/adverse effects , Whole-Body Irradiation/adverse effects , Animals , Cognition/radiation effects , Conditioning, Psychological/radiation effects , Fear/psychology , Fear/radiation effects , Female , Hippocampus/metabolism , Lipid Peroxidation/radiation effects , Male , Maze Learning/radiation effects , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Recognition, Psychology/radiation effects , Time Factors
16.
Behav Brain Res ; 246: 69-75, 2013 Jun 01.
Article En | MEDLINE | ID: mdl-23500678

(56)Fe irradiation affects hippocampus-dependent cognition. The underlying mechanisms may involve alterations in neurogenesis, expression of the plasticity-related immediate early gene Arc, and inflammation. Chemokine receptor-2 (CCR2), which mediates the recruitment of infiltrating and resident microglia to sites of CNS inflammation, is upregulated by (56)Fe irradiation. CCR2 KO and wild-type mice were used to compare effects of (56)Fe radiation (600MeV, 0.25Gy) on hippocampal function using contextual fear conditioning involving tone shock pairing during training (+/+) and exposure to the same environment without tone shock pairings (-/-). In the -/- condition, irradiation enhanced habituation in WT mice, but not CCR2 KO mice, suggesting that a lack of CCR2 was associated with reduced cognitive performance. In the +/+ condition, irradiation reduced freezing but there was no genotype differences. There were no significant correlations between the number of Arc-positive cells in the dentate gyrus and freezing in either genotype. While measures of neurogenesis and gliogenesis appeared to be modulated by CCR2, there were no effects of genotype on the total numbers of newly born activated microglia before or after irradiation, indicating that other mechanisms are involved in the genotype-dependent radiation response.


Electromagnetic Radiation , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Hippocampus/radiation effects , Receptors, CCR2/deficiency , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Conditioning, Psychological/radiation effects , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Radiation , Follow-Up Studies , Hippocampus/cytology , Hippocampus/metabolism , Iron Radioisotopes/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/physiology , Microglia/radiation effects , Nerve Tissue Proteins/metabolism , Neurogenesis/radiation effects , Neuropsychological Tests , Phosphopyruvate Hydratase/metabolism , Receptors, CCR2/genetics
17.
Neurotoxicol Teratol ; 35: 34-45, 2013.
Article En | MEDLINE | ID: mdl-23314114

Although maternal cigarette smoking during pregnancy is a well-documented risk factor for a variety of adverse pregnancy outcomes, how prenatal cigarette smoke exposure affects postnatal neurobehavioral/cognitive development remains poorly defined. In order to investigate the cause of an altered behavioral phenotype, mice developmentally exposed to a paradigm of 'active' maternal cigarette smoke is needed. Accordingly, cigarette smoke exposed (CSE) and air-exposed C57BL/6J mice were treated for 6h per day in paired inhalation chambers throughout gestation and lactation and were tested for neurobehavioral effects while controlling for litter effects. CSE mice exhibited less than normal anxiety in the elevated zero maze, transient hypoactivity during a 1h locomotor activity test, had longer latencies on the last day of cued Morris water maze testing, impaired hidden platform learning in the Morris water maze during acquisition, reversal, and shift trials, and impaired retention for platform location on probe trials after reversal but not after acquisition or shift. CSE mice also showed a sexually dimorphic response in central zone locomotion to a methamphetamine challenge (males under-responded and females over-responded), and showed reduced anxiety in the light-dark test by spending more time on the light side. No differences on tests of marble burying, acoustic startle response with prepulse inhibition, Cincinnati water maze, matching-to-sample Morris water maze, conditioned fear, forced swim, or MK-801-induced locomotor activation were found. Collectively, the data indicate that developmental cigarette smoke exposure induces subnormal anxiety in a novel environment, impairs spatial learning and reference memory while sparing other behaviors (route-based learning, fear conditioning, and forced swim immobility). The findings add support to mounting evidence that developmental cigarette smoke exposure has long-term adverse effects on brain function.


Behavior, Animal/physiology , Developmental Disabilities/chemically induced , Developmental Disabilities/physiopathology , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Tobacco Products/toxicity , Age Factors , Animals , Animals, Newborn , Behavior, Animal/drug effects , Body Weight/drug effects , Conditioning, Psychological/radiation effects , Fear/drug effects , Fear/psychology , Female , Inhibition, Psychological , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Pregnancy , Reflex, Acoustic/drug effects , Reflex, Acoustic/physiology , Time Factors
18.
Pharmacol Biochem Behav ; 103(3): 487-93, 2013 Jan.
Article En | MEDLINE | ID: mdl-23051895

Cranial irradiation with (56)Fe, a form of space radiation, causes hippocampus-dependent cognitive changes. (56)Fe irradiation also increases reactive oxygen species (ROS) levels, which may contribute to these changes. Therefore, we investigated the effects of the antioxidant alpha lipoic acid (ALA) on cognition following sham-irradiation and irradiation. Male mice were irradiated (brain only) with (56)Fe (3 Gy) or sham-irradiated at 6-9 months of age. Half of the mice remained fed a regular chow and the other half of the mice were fed a caloric-matched diet containing ALA starting two-weeks prior to irradiation and throughout cognitive testing. Following cognitive testing, levels of 3-nitrotyrosine (3NT), a marker of oxidative protein stress, and levels of microtubule-associated protein (MAP-2), a dendritic protein important for cognition, were assessed using immunohistochemistry and confocal microscopy. ALA prevented radiation-induced impairments in spatial memory retention in the hippocampal and cortical dependent water maze probe trials following reversal learning. However, in sham-irradiated mice, ALA treatment impaired cortical-dependent novel object recognition and amygdala-dependent cued fear conditioning. There was a trend towards lower 3NT levels in irradiated mice receiving a diet containing ALA than irradiated mice receiving a regular diet. In the hippocampal dentate gyrus of mice on regular diet, irradiated mice had higher levels of MAP-2 immunoreactivity than sham-irradiated mice. Thus, ALA might have differential effects on the brain under normal physiological conditions and those involving environmental challenges such as cranial irradiation.


Cranial Irradiation/psychology , Iron/toxicity , Memory Disorders/diet therapy , Memory/drug effects , Memory/radiation effects , Radiation Injuries, Experimental/diet therapy , Thioctic Acid/pharmacology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Brain/drug effects , Brain/metabolism , Brain/radiation effects , Conditioning, Psychological/drug effects , Conditioning, Psychological/radiation effects , Cranial Irradiation/methods , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/radiation effects , Male , Maze Learning/drug effects , Maze Learning/radiation effects , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Radiation Injuries, Experimental/chemically induced , Thioctic Acid/therapeutic use , Tyrosine/analogs & derivatives , Tyrosine/metabolism
19.
Nature ; 484(7394): 381-5, 2012 Mar 22.
Article En | MEDLINE | ID: mdl-22441246

A specific memory is thought to be encoded by a sparse population of neurons. These neurons can be tagged during learning for subsequent identification and manipulation. Moreover, their ablation or inactivation results in reduced memory expression, suggesting their necessity in mnemonic processes. However, the question of sufficiency remains: it is unclear whether it is possible to elicit the behavioural output of a specific memory by directly activating a population of neurons that was active during learning. Here we show in mice that optogenetic reactivation of hippocampal neurons activated during fear conditioning is sufficient to induce freezing behaviour. We labelled a population of hippocampal dentate gyrus neurons activated during fear learning with channelrhodopsin-2 (ChR2) and later optically reactivated these neurons in a different context. The mice showed increased freezing only upon light stimulation, indicating light-induced fear memory recall. This freezing was not detected in non-fear-conditioned mice expressing ChR2 in a similar proportion of cells, nor in fear-conditioned mice with cells labelled by enhanced yellow fluorescent protein instead of ChR2. Finally, activation of cells labelled in a context not associated with fear did not evoke freezing in mice that were previously fear conditioned in a different context, suggesting that light-induced fear memory recall is context specific. Together, our findings indicate that activating a sparse but specific ensemble of hippocampal neurons that contribute to a memory engram is sufficient for the recall of that memory. Moreover, our experimental approach offers a general method of mapping cellular populations bearing memory engrams.


Fear/physiology , Fear/radiation effects , Hippocampus/physiology , Hippocampus/radiation effects , Mental Recall/radiation effects , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Channelrhodopsins , Conditioning, Psychological/physiology , Conditioning, Psychological/radiation effects , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Dentate Gyrus/radiation effects , Female , Freezing Reaction, Cataleptic/physiology , Freezing Reaction, Cataleptic/radiation effects , Gene Expression Regulation , Hippocampus/cytology , Light , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic
20.
Electromagn Biol Med ; 31(2): 151-65, 2012 Jun.
Article En | MEDLINE | ID: mdl-22268919

The kinetics of the acquisition and loss of the use of olfactory and visual cues were previously obtained in six experimental colonies of the ant Myrmica sabuleti meinert 1861, under normal conditions. In the present work, the same experiments were conducted on six other naive identical colonies of M. sabuleti, under electromagnetic radiation similar to those surrounding GSM and communication masts. In this situation, no association between food and either olfactory or visual cues occurred. After a recovery period, the ants were able to make such an association but never reached the expected score. Such ants having acquired a weaker olfactory or visual score and still undergoing olfactory or visual training were again submitted to electromagnetic waves. Not only did they lose all that they had memorized, but also they lost it in a few hours instead of in a few days (as under normal conditions when no longer trained). They kept no visual memory at all (instead of keeping 10% of it as they normally do). The impact of GSM 900 MHz radiation was greater on the visual memory than on the olfactory one. These communication waves may have such a disastrous impact on a wide range of insects using olfactory and/or visual memory, i.e., on bees.


Animal Feed , Ants/radiation effects , Behavior, Animal/physiology , Behavior, Animal/radiation effects , Cell Phone , Cues , Radio Waves/adverse effects , Animals , Ants/physiology , Conditioning, Psychological/physiology , Conditioning, Psychological/radiation effects , Kinetics , Smell/physiology , Smell/radiation effects , Visual Perception/physiology , Visual Perception/radiation effects
...