Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4923, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862484

ABSTRACT

Missions into Deep Space are planned this decade. Yet the health consequences of exposure to microgravity and galactic cosmic radiation (GCR) over years-long missions on indispensable visceral organs such as the kidney are largely unexplored. We performed biomolecular (epigenomic, transcriptomic, proteomic, epiproteomic, metabolomic, metagenomic), clinical chemistry (electrolytes, endocrinology, biochemistry) and morphometry (histology, 3D imaging, miRNA-ISH, tissue weights) analyses using samples and datasets available from 11 spaceflight-exposed mouse and 5 human, 1 simulated microgravity rat and 4 simulated GCR-exposed mouse missions. We found that spaceflight induces: 1) renal transporter dephosphorylation which may indicate astronauts' increased risk of nephrolithiasis is in part a primary renal phenomenon rather than solely a secondary consequence of bone loss; 2) remodelling of the nephron that results in expansion of distal convoluted tubule size but loss of overall tubule density; 3) renal damage and dysfunction when exposed to a Mars roundtrip dose-equivalent of simulated GCR.


Subject(s)
Cosmic Radiation , Space Flight , Animals , Humans , Mice , Cosmic Radiation/adverse effects , Rats , Male , Kidney/pathology , Kidney/radiation effects , Kidney/metabolism , Kidney Diseases/pathology , Kidney Diseases/etiology , Weightlessness/adverse effects , Astronauts , Mice, Inbred C57BL , Proteomics , Female , Mars , Weightlessness Simulation/adverse effects
2.
Int J Radiat Biol ; 99(8): 1291-1300, 2023.
Article in English | MEDLINE | ID: mdl-36735963

ABSTRACT

The era of high-throughput techniques created big data in the medical field and research disciplines. Machine intelligence (MI) approaches can overcome critical limitations on how those large-scale data sets are processed, analyzed, and interpreted. The 67th Annual Meeting of the Radiation Research Society featured a symposium on MI approaches to highlight recent advancements in the radiation sciences and their clinical applications. This article summarizes three of those presentations regarding recent developments for metadata processing and ontological formalization, data mining for radiation outcomes in pediatric oncology, and imaging in lung cancer.


Subject(s)
Artificial Intelligence , Lung Neoplasms , Child , Humans , Big Data , Data Mining
3.
Life (Basel) ; 12(11)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36362993

ABSTRACT

Microgravity (modeled by head-tilt bedrest and hind-limb unloading), experienced during prolonged spaceflight, results in neurological consequences, central nervous system (CNS) dysfunction, and potentially impairment during the performance of critical tasks. Similar pathologies are observed in bedrest, sedentary lifestyle, and muscle disuse on Earth. In our previous study, we saw that head-tilt bedrest together with social isolation upregulated the milieu of pro-inflammatory cytokines in the hippocampus and plasma. These changes were mitigated in a MCAT mouse model overexpressing human catalase in the mitochondria, pointing out the importance of ROS signaling in this stress response. Here, we used a head-tilt model in socially housed mice to tease out the effects of head-tilt bedrest without isolation. In order to find the underlying molecular mechanisms that provoked the cytokine response, we measured CD68, an indicator of microglial activation in the hippocampus, as well as changes in normal in-cage behavior. We hypothesized that hindlimb unloading (HU) will elicit microglial hippocampal activations, which will be mitigated in the MCAT ROS-quenching mice model. Indeed, we saw an elevation of the activated microglia CD68 marker following HU in the hippocampus, and this pathology was mitigated in MCAT mice. Additionally, we identified cytokines in the hippocampus, which had significant positive correlations with CD68 and negative correlations with exploratory behaviors, indicating a link between neuroinflammation and behavioral consequences. Unveiling a correlation between molecular and behavioral changes could reveal a biomarker indicative of these responses and could also result in a potential target for the treatment and prevention of cognitive changes following long space missions and/or muscle disuse on Earth.

5.
Behav Brain Res ; 419: 113677, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34818568

ABSTRACT

In long-term spaceflight, astronauts will face unique cognitive loads and social challenges which will be complicated by communication delays with Earth. It is important to understand the central nervous system (CNS) effects of deep spaceflight and the associated unavoidable exposure to galactic cosmic radiation (GCR). Rodent studies show single- or simple-particle combination exposure alters CNS endpoints, including hippocampal-dependent behavior. An even better Earth-based simulation of GCR is now available, consisting of a 33-beam (33-GCR) exposure. However, the effect of whole-body 33-GCR exposure on rodent behavior is unknown, and no 33-GCR CNS countermeasures have been tested. Here astronaut-age-equivalent (6mo-old) C57BL/6J male mice were exposed to 33-GCR (75cGy, a Mars mission dose). Pre-/during/post-Sham or 33-GCR exposure, mice received a diet containing a 'vehicle' formulation alone or with the antioxidant/anti-inflammatory compound CDDO-EA as a potential countermeasure. Behavioral testing beginning 4mo post-irradiation suggested radiation and diet did not affect measures of exploration/anxiety-like behaviors (open field, elevated plus maze) or recognition of a novel object. However, in 3-Chamber Social Interaction (3-CSI), CDDO-EA/33-GCR mice failed to spend more time exploring a holder containing a novel mouse vs. a novel object (empty holder), suggesting sociability deficits. Also, Vehicle/33-GCR and CDDO-EA/Sham mice failed to discriminate between a novel stranger vs. familiarized stranger mouse, suggesting blunted preference for social novelty. CDDO-EA given pre-/during/post-irradiation did not attenuate the 33-GCR-induced blunting of preference for social novelty. Future elucidation of the mechanisms underlying 33-GCR-induced blunting of preference for social novelty will improve risk analysis for astronauts which may in-turn improve countermeasures.


Subject(s)
Behavior, Animal , Cognitive Dysfunction , Cosmic Radiation/adverse effects , Oleanolic Acid/analogs & derivatives , Radiation Exposure/adverse effects , Recognition, Psychology , Social Behavior , Animals , Behavior, Animal/drug effects , Behavior, Animal/radiation effects , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Oleanolic Acid/pharmacology , Recognition, Psychology/drug effects , Recognition, Psychology/radiation effects
6.
Front Behav Neurosci ; 15: 722780, 2021.
Article in English | MEDLINE | ID: mdl-34707486

ABSTRACT

Astronauts during interplanetary missions will be exposed to galactic cosmic radiation, including charged particles like 56Fe. Most preclinical studies with mature, "astronaut-aged" rodents suggest space radiation diminishes performance in classical hippocampal- and prefrontal cortex-dependent tasks. However, a rodent cognitive touchscreen battery unexpectedly revealed 56Fe radiation improves the performance of C57BL/6J male mice in a hippocampal-dependent task (discrimination learning) without changing performance in a striatal-dependent task (rule-based learning). As there are conflicting results on whether the female rodent brain is preferentially injured by or resistant to charged particle exposure, and as the proportion of female vs. male astronauts is increasing, further study on how charged particles influence the touchscreen cognitive performance of female mice is warranted. We hypothesized that, similar to mature male mice, mature female C57BL/6J mice exposed to fractionated whole-body 56Fe irradiation (3 × 6.7cGy 56Fe over 5 days, 600 MeV/n) would improve performance vs. Sham conditions in touchscreen tasks relevant to hippocampal and prefrontal cortical function [e.g., location discrimination reversal (LDR) and extinction, respectively]. In LDR, 56Fe female mice more accurately discriminated two discrete conditioned stimuli relative to Sham mice, suggesting improved hippocampal function. However, 56Fe and Sham female mice acquired a new simple stimulus-response behavior and extinguished this acquired behavior at similar rates, suggesting similar prefrontal cortical function. Based on prior work on multiple memory systems, we next tested whether improved hippocampal-dependent function (discrimination learning) came at the expense of striatal stimulus-response rule-based habit learning (visuomotor conditional learning). Interestingly, 56Fe female mice took more days to reach criteria in this striatal-dependent rule-based test relative to Sham mice. Together, our data support the idea of competition between memory systems, as an 56Fe-induced decrease in striatal-based learning is associated with enhanced hippocampal-based learning. These data emphasize the power of using a touchscreen-based battery to advance our understanding of the effects of space radiation on mission critical cognitive function in females, and underscore the importance of preclinical space radiation risk studies measuring multiple cognitive processes, thereby preventing NASA's risk assessments from being based on a single cognitive domain.

7.
NPJ Microgravity ; 7(1): 24, 2021 Jul 06.
Article in English | MEDLINE | ID: mdl-34230490

ABSTRACT

Isolation on Earth can alter physiology and signaling of organs systems, including the central nervous system. Although not in complete solitude, astronauts operate in an isolated environment during spaceflight. In this study, we determined the effects of isolation and simulated microgravity solely or combined, on the inflammatory cytokine milieu of the hippocampus. Adult female wild-type mice underwent simulated microgravity by hindlimb unloading for 30 days in single or social (paired) housing. In hippocampus, simulated microgravity and isolation each regulate a discrete repertoire of cytokines associated with inflammation. Their combined effects are not additive. A model for mitochondrial reactive oxygen species (ROS) quenching via targeted overexpression of the human catalase gene to the mitochondria (MCAT mice), are protected from isolation- and/or simulated microgravity-induced changes in cytokine expression. These findings suggest a key role for mitochondrial ROS signaling in neuroinflammatory responses to spaceflight and prolonged bedrest, isolation, and confinement on Earth.

8.
Behav Brain Res ; 407: 113257, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33794227

ABSTRACT

The effects of radiation in space on human cognition are a growing concern for NASA scientists and astronauts as the possibility for long-duration missions to Mars becomes more tangible. Oxygen (16O) radiation is of utmost interest considering that astronauts will interact with this radiation frequently. 16O radiation is a class of galactic cosmic ray (GCR) radiation and also present within spacecrafts. Whole-body exposure to high linear energy transfer (LET) radiation has been shown to affect hippocampal-dependent cognition. To assess the effects of high-LET radiation, we gave 6-month-old female C57BL/6 mice whole-body exposure to 16O at 0.25 or 0.1 Gy at NASA's Space Radiation Laboratory. Three months following irradiation, animals were tested for cognitive performance using the Y-maze and Novel Object Recognition paradigms. Our behavioral data shows that 16O radiation significantly impairs object memory but not spatial memory. Also, dendritic morphology characterized by the Sholl analysis showed that 16O radiation significantly decreased dendritic branch points, ends, length, and complexity in 0.1 Gy and 0.25 Gy dosages. Finally, we found no significant effect of radiation on single nucleotide polymorphisms in hippocampal genes related to oxidative stress, inflammation, and immediate early genes. Our data suggest exposure to heavy ion 16O radiation modulates hippocampal neurons and induces behavioral deficits at a time point of three months after exposure in female mice.


Subject(s)
Cognitive Dysfunction/etiology , Cosmic Radiation/adverse effects , Hippocampus/radiation effects , Maze Learning/radiation effects , Oxygen/adverse effects , Recognition, Psychology/radiation effects , Spatial Memory/radiation effects , Animals , Behavior, Animal/radiation effects , Cognitive Dysfunction/physiopathology , Female , Mice , Mice, Inbred C57BL
9.
Front Behav Neurosci ; 14: 96, 2020.
Article in English | MEDLINE | ID: mdl-32670032

ABSTRACT

The space extending beyond Earth's magnetosphere is subject to a complex field of high-energy charged nuclei, which are capable of traversing spacecraft shielding and human tissues, inducing dense ionization events. The central nervous system is a major area of concern for astronauts who will be exposed to the deep-space radiation environment on a mission to Mars, as charged-particle radiation has been shown to elicit changes to the dendritic arbor within the hippocampus of rodents, and related cognitive-behavioral deficits. We exposed 6-month-old male mice to whole-body 1H (0.5 Gy; 150 MeV/n; 18-19 cGy/minute) and an hour later to 16O (0.1Gy; 600 MeV/n; 18-33 Gy/min) at NASA's Space Radiation Laboratory as a galactic cosmic ray-relevant model. Animals were housed with bedding which provides cognitive enrichment. Mice were tested for cognitive behavior 9 months after exposure to elucidate late radiation effects. Radiation induced significant deficits in novel object recognition and short-term spatial memory (Y-maze). Additionally, we observed opposing morphological differences between the mature granular and pyramidal neurons throughout the hippocampus, with increased dendritic length in the dorsal dentate gyrus and reduced length and complexity in the CA1 subregion of the hippocampus. Dendritic spine analyses revealed a severe reduction in mushroom spine density throughout the hippocampus of irradiated animals. Finally, we detected no general effect of radiation on single-nucleotide polymorphisms in immediate early genes, and genes involved in inflammation but found a higher variant allele frequency in the antioxidants thioredoxin reductase 2 and 3 loci.

10.
Toxicol Sci ; 173(1): 156-170, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31651976

ABSTRACT

Breast cancer (BC) is the most common cancer among women. Fortunately, BC survival rates have increased because the implementation of adjuvant chemotherapy leading to a growing population of survivors. However, chemotherapy-induced cognitive impairments (CICIs) affect up to 75% of BC survivors and may be driven by inflammation and oxidative stress. Chemotherapy-induced cognitive impairments can persist 20 years and hinder survivors' quality of life. To identify early effects of CMF administration in mice, we chose to evaluate adult female mice at 2-week postchemotherapy. Mice received weekly IP administration of CMF (or saline) for 4 weeks, completed behavioral testing, and were sacrificed 2 weeks following their final CMF injection. Behavioral results indicated long-term memory (LTM) impairments postchemotherapy, but did not reveal short-term memory deficits. Dendritic morphology and spine data found increases in overall spine density within CA1 basal and CA3 basal dendrites, but no changes in DG, CA1 apical, or CA3 apical dendrites. Further analysis revealed decreases in arborization across the hippocampus (DG, CA1 apical and basal, CA3 apical and basal). These physiological changes within the hippocampus correlate with our behavioral data indicating LTM impairments following CMF administration in female mice 2-week postchemotherapy. Hippocampal cytokine analysis identified decreases in IL-1α, IL-1ß, IL-3, IL-10, and TNF-α levels.


Subject(s)
Cyclophosphamide/toxicity , Fluorouracil/toxicity , Hippocampus/drug effects , Methotrexate/toxicity , Neurons/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms , CA1 Region, Hippocampal , Chemotherapy, Adjuvant , Dendrites , Dendritic Spines , Disease Models, Animal , Female , Mice , Quality of Life
11.
J Neurochem ; 153(1): 10-32, 2020 04.
Article in English | MEDLINE | ID: mdl-31630412

ABSTRACT

Perception of our environment entirely depends on the close interaction between the central and peripheral nervous system. In order to communicate each other, both systems must develop in parallel and in coordination. During development, axonal projections from the CNS as well as the PNS must extend over large distances to reach their appropriate target cells. To do so, they read and follow a series of axon guidance molecules. Interestingly, while these molecules play critical roles in guiding developing axons, they have also been shown to be critical in other major neurodevelopmental processes, such as the migration of cortical progenitors. Currently, a major hurdle for brain repair after injury or neurodegeneration is the absence of axonal regeneration in the mammalian CNS. By contrasts, PNS axons can regenerate. Many hypotheses have been put forward to explain this paradox but recent studies suggest that hacking neurodevelopmental mechanisms may be the key to promote CNS regeneration. Here we provide a seminar report written by trainees attending the second Flagship school held in Alpbach, Austria in September 2018 organized by the International Society for Neurochemistry (ISN) together with the Journal of Neurochemistry (JCN). This advanced school has brought together leaders in the fields of neurodevelopment and regeneration in order to discuss major keystones and future challenges in these respective fields.


Subject(s)
Axon Guidance/physiology , Axons/physiology , Brain/ultrastructure , Animals , Axons/ultrastructure , Brain/growth & development , Brain/physiology , Humans , Nerve Regeneration , Optic Chiasm/growth & development , Peripheral Nervous System/growth & development , Peripheral Nervous System/physiology , Spinal Cord/growth & development , Spinal Cord/physiology , Spinal Cord/ultrastructure
12.
Life Sci Space Res (Amst) ; 21: 1-21, 2019 May.
Article in English | MEDLINE | ID: mdl-31101151

ABSTRACT

As NASA prepares for the first manned mission to Mars in the next 20 years, close attention has been placed on the cognitive welfare of astronauts, who will likely endure extended durations in confinement and microgravity and be subjected to the radioactive charged particles travelling at relativistic speeds in interplanetary space. The future of long-duration manned spaceflight, thus, depends on understanding the individual hazards associated with the environment beyond Earth's protective magnetosphere. Ground-based single-particle studies of exposed mice and rats have, in the last 30 years, overwhelmingly reported deficits in their cognitive behaviors. However, as particle-accelerator technologies at NASA's Space Radiation Laboratory continue to progress, more realistic representations of space radiation are materializing, including multiple-particle exposures and, eventually, at multiple energy distributions. These advancements help determine how to best mitigate possible hazards due to space radiation. However, risk models will depend on delineating which particles are most responsible for specific behavioral outcomes and whether multiple-particle exposures produce synergistic effects. Here, we review the literature on animal exposures by particle, energy, and behavioral assay to inform future mixed-field radiation studies of possible behavioral outcomes.


Subject(s)
Behavior, Animal/radiation effects , Cosmic Radiation , Space Flight , Animals , Stress, Psychological
13.
Int J Mol Sci ; 20(4)2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30781689

ABSTRACT

Aging is characterized by increased inflammation and deterioration of the cellular stress responses such as the oxidant/antioxidant equilibrium, DNA damage repair fidelity, and telomeric attrition. All these factors contribute to the increased radiation sensitivity in the elderly as shown by epidemiological studies of the Japanese atomic bomb survivors. There is a global increase in the aging population, who may be at increased risk of exposure to ionizing radiation (IR) as part of cancer therapy or accidental exposure. Therefore, it is critical to delineate the factors that exacerbate age-related radiation sensitivity and neurocognitive decline. The transcription factor CCAAT enhancer binding protein delta (C/EBPδ) is implicated with regulatory roles in neuroinflammation, learning, and memory, however its role in IR-induced neurocognitive decline and aging is not known. The purpose of this study was to delineate the role of C/EBPδ in IR-induced neurocognitive decline in aged mice. We report that aged Cebpd-/- mice exposed to acute IR exposure display impairment in short-term memory and spatial memory that correlated with significant alterations in the morphology of neurons in the dentate gyrus (DG) and CA1 apical and basal regions. There were no significant changes in the expression of inflammatory markers. However, the expression of superoxide dismutase 2 (SOD2) and catalase (CAT) were altered post-IR in the hippocampus of aged Cebpd-/- mice. These results suggest that Cebpd may protect from IR-induced neurocognitive dysfunction by suppressing oxidative stress in aged mice.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/deficiency , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Radiation, Ionizing , Aging , Animals , Antioxidants/metabolism , Biomarkers/metabolism , CCAAT-Enhancer-Binding Protein-delta/metabolism , Dendrites/metabolism , Dentate Gyrus/pathology , Inflammation Mediators/metabolism , Maze Learning , Memory, Short-Term , Mice, Inbred C57BL , Oxidative Stress , Spatial Memory , Tissue Extracts
14.
Int J Mol Sci ; 20(1)2019 Jan 07.
Article in English | MEDLINE | ID: mdl-30621014

ABSTRACT

Astronauts traveling to Mars will be exposed to high levels of ionizing radiation upon leaving low-Earth orbit. During prolonged space travel, astronauts are exposed to galactic cosmic rays (GCRs) composed of protons; oxygen molecules; and high energy, high mass charged particles. Notably, oxygen molecules can travel through the shielding of spacecraft, potentially impacting 25% of the hippocampus. The aim of the current study was to assess whether 16O-particle radiation induced a behavioral deficit and histological changes in mice. Mice were sent to the National Aeronautics and Space Administration (NASA) Space Radiation Laboratory at Brookhaven National Laboratory and exposed to particulate 16O radiation at doses of 0 and 0.05 Gy. Nine months after irradiation, the mice were tested for novel object recognition and in the Y-maze, after which the animals were sacrificed. The brains were then dissected along the midsagittal plane for Golgi staining. Exposure to 0.05 Gy significantly impaired novel object recognition. However, short term memory and exploratory activity in the Y-maze were not affected. Micromorphometric analysis revealed significant decreases in mushroom spine density in the dentate gyrus and cornu Ammonis-1 and -3 of the hippocampus. Sholl analysis revealed a significant decrease in dendritic complexity in the dentate gyrus. The present data provide evidence that space radiation has deleterious effects on mature neurons associated with hippocampal learning and memory.


Subject(s)
Cognition/physiology , Oxygen/pharmacology , Animals , Behavior, Animal/drug effects , Biomarkers/metabolism , Cognition/drug effects , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/drug effects , Synapses/metabolism , Time Factors
15.
Radiat Res ; 191(3): 278-294, 2019 03.
Article in English | MEDLINE | ID: mdl-30664396

ABSTRACT

The radiation environment in space remains a major concern for manned space exploration, as there is currently no shielding material capable of fully protecting flight crews. Additionally, there is growing concern for the social and cognitive welfare of astronauts, due to prolonged radiation exposure and confinement they will experience on a mission to Mars. In this artice, we report on the late effects of 16O-particle radiation on social and cognitive behavior and neuronal morphology in the hippocampus of adult female mice. Six-month-old mice received 16O-particle whole-body irradiation at doses of either 0.25 or 0.1 Gy (600 MeV/n; 18-33 cGy/min) at the NASA's Space Radiation Laboratory in Upton, NY. At nine months postirradiation, the animals underwent behavioral testing in the three-chamber sociability, novel object recognition and Y-maze paradigms. Exposure to 0.1 or 0.25 Gy 16O significantly impaired object memory, a 0.25 Gy dose impaired social novelty learning, but neither dosage impaired short-term spatial memory. We observed significant decreases in mushroom spine density and dendrite morphology in the dentate gyrus, cornu ammonis 3, 2 and 1 of the hippocampus, which are critical areas for object novelty and sociability processing. Our data suggest exposure to 16O modulates hippocampal pyramidal and granular neurons and induces behavioral deficits at a time point of nine months after exposure in females.


Subject(s)
Behavior, Animal/radiation effects , Cognition/radiation effects , Hippocampus/physiology , Hippocampus/radiation effects , Oxygen/adverse effects , Social Behavior , Animals , Dendritic Spines/radiation effects , Dose-Response Relationship, Radiation , Extraterrestrial Environment , Female , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Time Factors
16.
Synapse ; 73(6): e22085, 2019 06.
Article in English | MEDLINE | ID: mdl-30586195

ABSTRACT

Cancer survivorship has increased greatly as therapies have become more advanced and effective. Thus, we must now focus on improving the quality of life of patients after treatment. After chemotherapy, many patients experience chemotherapy-induced cognitive decline, indicating a need to investigate pathologies associated with this condition. In this study, we addressed cognitive impairment after thioTEPA treatment by assessing behavior and assaying cytokine production and the structure of dendrites in the hippocampus. Male mice were given three intraperitoneal injections of thioTEPA. Five weeks later, the mice underwent behavior testing, and brains were collected for Golgi staining and cytokine analysis. Behavior tests included y-maze and Morris water maze and licking behavioral task. Cytokines measured include: IL-1α, IL-1ß, IL-2, IL-3, IL-4, IL-5, IL-10, IL-12p70, MCP-1, TNF-α, GMCSF, and RANTES. We observed decreased memory retention in behavioral tasks. Also, dendritic arborization and length were decreased after chemotherapy treatment. Finally, thioTEPA decreased cytokine production in animals treated with chemotherapy, compared to saline-treated controls. Here, we used a mouse model to correlate the decreases in dendritic complexity and inflammatory cytokine production with cognitive impairment after chemotherapy.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Cognitive Dysfunction/chemically induced , Thiotepa/adverse effects , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/pharmacology , Brain/drug effects , Brain/metabolism , Cognition , Cytokines/metabolism , Injections, Intraperitoneal , Male , Maze Learning , Mice , Mice, Inbred C57BL , Movement , Thiotepa/administration & dosage , Thiotepa/pharmacology
17.
Life Sci Space Res (Amst) ; 17: 51-62, 2018 May.
Article in English | MEDLINE | ID: mdl-29753414

ABSTRACT

NASA's Missions to Mars and beyond will expose flight crews to potentially dangerous levels of charged-particle radiation. Of all charged nuclei, 1H is the most abundant charged particle in both the galactic cosmic ray (GCR) and solar particle event (SPE) spectra. There are currently no functional spacecraft shielding materials that are able to mitigate the charged-particle radiation encountered in space. Recent studies have demonstrated cognitive injuries due to high-dose 1H exposures in rodents. Our study investigated the effects of 1H irradiation on neuronal morphology in the hippocampus of adult male mice. 6-month-old mice received whole-body exposure to 1H at 0.5 and 1 Gy (150 MeV/n; 0.35-0.55 Gy/min) at NASA's Space Radiation Laboratory in Upton, NY. At 9-months post-irradiation, we tested each animal's open-field exploratory performance. After sacrifice, we dissected the brains along the midsagittal plane, and then either fixed or dissected further and snap-froze them. Our data showed that exposure to 0.5 Gy or 1 Gy 1H significantly increased animals' anxiety behavior in open-field testing. Our micromorphometric analyses revealed significant decreases in mushroom spine density and dendrite morphology in the Dentate Gyrus, Cornu Ammonis 3 and 1 of the hippocampus, and lowered expression of synaptic markers. Our data suggest 1H radiation significantly increased exploration anxiety and modulated the dendritic spine and dendrite morphology of hippocampal neurons at a dose of 0.5 or 1 Gy.


Subject(s)
Cosmic Radiation/adverse effects , Hippocampus/physiology , Hydrogen/adverse effects , Neurons/physiology , Solar Activity , Animals , Biomarkers/metabolism , Dose-Response Relationship, Radiation , Gene Expression Profiling/methods , Hippocampus/radiation effects , Male , Mice , Neurons/radiation effects
18.
Life Sci Space Res (Amst) ; 17: 63-73, 2018 May.
Article in English | MEDLINE | ID: mdl-29753415

ABSTRACT

Astronauts exposed to high linear energy transfer radiation may experience cognitive injury. The pathogenesis of this injury is unknown but may involve glutamate receptors or modifications to dendritic structure and/or dendritic spine density and morphology. Glutamate is the major excitatory neurotransmitter in the central nervous system, where it acts on ionotropic and metabotropic glutamate receptors located at the presynaptic terminal and in the postsynaptic membrane at synapses in the hippocampus. Dendritic spines are sites of excitatory synaptic transmission, and changes in spine structure and dendrite morphology are thought to be morphological correlates of altered brain function associated with hippocampal-dependent learning and memory. The aim of the current study is to assess whether behavior, glutamate receptor gene expression, and dendritic structure in the hippocampus are altered in mice after early exposure to 16O radiation in mice. Two weeks post-irradiation, animals were tested for hippocampus-dependent cognitive performance in the Y-maze. During Y-maze testing, mice exposed to 0.1 Gy and 0.25 Gy radiation failed to distinguish the novel arm, spending approximately the same amount of time in all 3 arms during the retention trial. Exposure to 16O significantly reduced the expression of Nr1 and GluR1 in the hippocampus and modulated spine morphology in the dentate gyrus and cornu Ammon 1 within the hippocampus. The present data provide evidence that 16O radiation has early deleterious effects on mature neurons that are associated with hippocampal learning and memory.


Subject(s)
Cognition/radiation effects , Dendritic Spines/pathology , Hippocampus/pathology , Neurons/pathology , Oxygen Radioisotopes/adverse effects , Animals , Dendritic Spines/radiation effects , Gene Expression Regulation/radiation effects , Hippocampus/radiation effects , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neurons/radiation effects
19.
Radiat Res ; 189(6): 605-617, 2018 06.
Article in English | MEDLINE | ID: mdl-29584587

ABSTRACT

Chemotherapy has been successfully used to reduce radiation dose and volume for most pediatric patients. However, because of the failure of chemotherapeutic agents to cross the blood-brain barrier and the lack of response of some brain tumors to these agents, radiation therapy is still used to treat many childhood cancers with CNS involvement. In this study, we investigated the radiation effects on cognition and dendritic structure in the hippocampus in juvenile male mice. Twenty-one-day-old male C57BL/6 mice were irradiated using the small animal radiation research platform (SARRP). Animals were exposed to either a 10 Gy single dose or 10 Gy × 2 fractionated doses of X-ray cranial radiation. Five weeks after irradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water maze. Significant impairment in spatial memory retention was observed in the probe trial after the first day of hidden-platform training (first probe trial) in animals that received either 10 Gy single-dose or 10 Gy × 2 fractionated doses. However, by day 5, mice that received a 10 Gy single dose showed spatial memory retention in the probe trials, whereas mice that received the 20 Gy fractionated doses remained impaired. During Y-maze testing, animals exposed to radiation were impaired; the irradiated mice were not able to distinguish among the three Y-maze arms and spent approximately the same amount of time in all three arms during the retention trial. Radiation significantly compromised the dendritic architecture and reduced spine density throughout the hippocampal trisynaptic network.


Subject(s)
Behavior, Animal/radiation effects , Cognition/radiation effects , Dendrites/radiation effects , Animals , Dendrites/metabolism , Exploratory Behavior/radiation effects , Hippocampus/cytology , Hippocampus/physiology , Hippocampus/radiation effects , Leukocyte Count , Male , Mice , Mice, Inbred C57BL , Spatial Memory/radiation effects
20.
Behav Brain Res ; 346: 21-28, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29229546

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most prevalent childhood cancer and accounts for 26.8% of cancer diagnoses among children, worldwide-approximately 3000 children each year. While advancements in treating ALL have led to a remission rate of more than 90%, many survivors experience adverse neurocognitive and/or neurobehavioral effects as a result of intrathecal chemotherapy. Methotrexate (MTX) is commonly administered with cytosine arabinoside (AraC, cytarabine) during intrathecal chemotherapy for ALL. To date, few studies exist that test the cognitive effects of intrathecal injections of MTX/AraC on juvenile populations. The purpose of our study was to investigate the combined effects of MTX/AraC on cognition and dendritic structure in the hippocampus in juvenile male mice. Twenty, 21-day-old male C57BL/6 mice were used in this study; 10 mice received intrathecal MTX/AraC treatment, and 10 were given intrathecal saline injections. Five weeks after injections, we tested the animals' hippocampus-dependent cognitive performance in the Morris water maze. After the first day of hidden-platform training, we observed that the mice that received MTX/AraC treatment showed signs of significant impairment in spatial memory retention. MTX/AraC treatment significantly compromised the dendritic architecture and reduced mushroom spine density in the dorsal ganglion (DG), CA1, and CA3 areas of the hippocampus. The present data provided evidence that MTX/AraC compromised the dendritic architecture and impaired hippocampal dependent cognition. This could provide insight into chemotherapy-induced cognitive decline in juvenile patients treated for ALL.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Cognition/drug effects , Cytarabine/toxicity , Dendrites/drug effects , Hippocampus/drug effects , Methotrexate/toxicity , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dendrites/pathology , Hippocampus/pathology , Injections, Spinal , Leukocytes/drug effects , Male , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Spatial Memory/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...